WO2001037876A2 - Methods of ameliorating abnormal bone states - Google Patents

Methods of ameliorating abnormal bone states Download PDF

Info

Publication number
WO2001037876A2
WO2001037876A2 PCT/EP2000/011466 EP0011466W WO0137876A2 WO 2001037876 A2 WO2001037876 A2 WO 2001037876A2 EP 0011466 W EP0011466 W EP 0011466W WO 0137876 A2 WO0137876 A2 WO 0137876A2
Authority
WO
WIPO (PCT)
Prior art keywords
bone
hmg
coa reductase
reductase inhibitor
effective amount
Prior art date
Application number
PCT/EP2000/011466
Other languages
French (fr)
Other versions
WO2001037876A3 (en
Inventor
Cedo M. Bagi
Original Assignee
Bayer Aktiengesellschaft
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 Bayer Aktiengesellschaft filed Critical Bayer Aktiengesellschaft
Priority to AU20007/01A priority Critical patent/AU2000701A/en
Publication of WO2001037876A2 publication Critical patent/WO2001037876A2/en
Publication of WO2001037876A3 publication Critical patent/WO2001037876A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • 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

Definitions

  • This invention relates to methods of using 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone reso ⁇ tion and stimulation of bone formation, comprising administering a therapeutically effective amount of a HMG-Co A reductase inhibitor to a vertebrate in need thereof.
  • HMG-CoA reductase inhibitors for the prevention and for the treatment of conditions ameliorated by a decrease in plasma calcium levels.
  • HMG-CoA reductase catalyzes the conversion of HMG-CoA to mevalonate, which is an early and rate limiting step in the biosynthesis of cholesterol.
  • Compounds from the class of compounds known as HMG-CoA reductase inhibitors (which includes cerivastatin, fluvastatin, lovastatin, mevastatin, pravastatin, simvastatin, atovastatin and others), have been known for some time to be effective anti-cholesteolemic agents in the prevention and treatment of cardiovascular disease. They also slow the progression of atherosclerotic lesions in the coronary and carotid arteries, and reduce the risk of coronary heart disease events including death.
  • Statins are also said to be effective in decreasing beta amyloid protein, a predictor of Alzheimer's disease (WO 99/48488, published September 30, 1999), as well as in inhibiting the l-desoxy-D-xylulose-5 -phosphate biosynthetic pathway and, accordingly, in treating certain parasitic infections (WO 99/52938, published October 21, 1999). These compounds are also reported to have an effect on bone. Generally, after reaching the so-called "peak bone mass" in early adulthood, the bone remains an active tissue. Indeed, throughout adult life, within focal sites scattered throughout the skeleton, microscopic quantities of bone are being replaced. In the normal adult skeleton there are approximately two million of these remodeling units.
  • Bone remodeling is an orderly process, essential for bone strength and occurring in response to mechanical stress (Parfitt, A.M. (1988) Bone remodeling: relationship to the amount and structure of bone, and the pathogenesis and prevention of fractures. In: Riggs, B. & Melton, L.J. Ill, eds. Osteoporosis: Ethiology, diagnosis and management. New York: Raven Press, pp. 133-154.).
  • a balanced sequence of bone resorption and formation is necessary, which in healthy individuals is carefully regulated by a mechanism known as coupling, mediated by a complex interaction of hormones, paracrine growth factors and cytokines (Martin, J.J., Wah, N.K., and Suda, T. (1989) Bone cell physiology. Endocrinol. Metabol. Clin. North. Am. 18:833-858).
  • Bone resorption is mediated by the osteoclast, a multinucleated cell derived from granulocyte-macrophage precursors in the bone marrow, while bone formation requires the presence and function of osteoblasts, derived from mesenchymal fibroblast-like cells (Raisz, L.G. and Rodan, G.A. (1990) Cellular basis for bone turnover. In: Avioli, L.V. and Krane, S.M. eds.: Metabolic bone diseases and clinically related disorders. Philadelphia: W.B. Saunders Company, pp. 1-41.). Many factors are known to stimulate bone reso ⁇ tion, but su ⁇ risingly, none of these factors appears to act directly on osteoclasts.
  • osteoblasts control bone reso ⁇ tion through their effects on osteoclasts (Chambers, T.J. and Fuller, K. (1985) Bone cells predispose endosteal surfaces to reso ⁇ tion by exposure of bone mineral to osteoclastic contact. J. Cell Sci. 76:155- 163) by either direct cell-cell contact or local release of a diffusable osteoblast- derived osteoclast-stimulating factor (McSheehy, P.M.J. and Chambers, T.J. (1986) Osteoblast-like cells in the presence of parathyroid hormone release soluble factors that stimulate osteoclastic bone reso ⁇ tion. Endocrinology 119:1614-1659).
  • the bone remodeling cycle takes place according to a highly organized sequence of events, which is dependent on precise inte ⁇ lay between osteoclasts and osteoblasts.
  • Activation is followed by a reso ⁇ tion phase during which osteoclasts resorb a packet of bone, thus creating a cavity (Howship's lacunae) in the bone surface.
  • a reversal phase follows, separating the completed phase of bone reso ⁇ tion and the subsequent initiation of bone formation.
  • Mononuclear cells in the reso ⁇ tion cavity smooth out the irregular bone surface left behind by the osteoclasts, and cells (possibly osteocytes) are involved in the deposition of a cement layer on the bone surface.
  • a team of new osteoblasts begins to deposit undemineralized bone matrix, the osteoid seam.
  • This newly deposited layer of matrix begins to mineralize after a maturation period of 5-10 days.
  • the lacunae created by the osteoclasts have been filled with newly deposited bone covered by lining cells.
  • the total duration of a remodeling cycle in adults is estimated to be around 250 days. If the osteoblasts have deposited exactly the same amount of bone as has been removed previously by the osteoclasts, the remodeling cycle is in balance. Number of hormonal and/or mechanical factors can change this balance on the tissue or local level resulting in positive or more frequently negative bone balance and loss of bone (Boyce, B.F.
  • Bone formation is coupled to bone reso ⁇ tion and vice versa, so that each time when bone reso ⁇ tion increases or decreases, bone formation follows the same pattern in order to maintain original bone mass. Because bone reso ⁇ tion is much faster process relative to bone formation, the end result of bone remodeling imbalance is often bone loss.
  • Drugs used to treat bone loss of various etiologies can be grouped into those that decrease bone reso ⁇ tion and those that increase bone formation. Both groups of drugs will affect bone reso ⁇ tion and bone formation in the similar fashion due to the coupling between bone reso ⁇ tion and bone formation.
  • Antireso ⁇ tive drugs such as estrogen, calcitonin, bisphosphonates, and selective estrogen receptor modulators (SERMs) will reduce or completely shut down bone reso ⁇ tion which will consequently block bone formation (Riggs, B.L., and Melton III, L.J. Drug Therapy,
  • WO 98/25460 (published June 18, 1998, Zymogenetics) disclosed that certain statins exhibited an in vitro stimulatory effect on osteoblastogenesis, as well as evidence of bone formation in in vivo calavrial studies, an increase in trabecular density and cortical thickness in ovariexctomized (OVX) mice, and in vivo callus formation at osteotomy sites in the forelimbs of rabbits.
  • IL-1 induced bone reso ⁇ tion, however, was not affected by concentrations of statins that corresponded to doses that stimulated in vitro osteoblast activity.
  • II- 1 induced calcium release was decreased by simvastatin at a higher dose, although this decrease in calcium release was attributed to toxicity.
  • HMG-CoA reductase inhibitors are potent agonists of bone formation in women 55 years and older, as evidenced by higher hip bone mineral density and reduced fracture risk.
  • HMG-CoA reductase inhibitor both decreases bone reso ⁇ tion while concurrently stimulating bone growth. It is now known that HMG-Co-A reductase inhibitors simultaneously affect bone formation by promoting osteoblast differentiation in the bone marrow, while reducing formation of multinucleated osteoclasts from their mononuclear precursors in the bone marrow.
  • HMG Co-A reductase inhibitors to effectively decrease plasma calcium levels and, therefore ameliorate conditions caused by high plasma calcium levels, is also heretofore unknown.
  • the delicate balance between normal bone reso ⁇ tion and normal bone formation can be disrupted by, for example but not by way of limitation, chronic glucocorticoid use, immunosuppresive therapy, hormone imbalance, malignancy and the like. Disrupted balance between bone reso ⁇ tion and bone formation can result in such conditions or disease states as osteoporosis of various ethiologies, osteoarthritis, Pagent's disease, hypercalcemia, periprosthetic bone loss or osteolysis, fractures, osteogenesis imperfecta, and aveolar bone loss associated with peridontal disease.
  • the present invention provides the use of HMG-CoA reductase inhibitors for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone reso ⁇ tion and stimulation of bone growth comprising administering a therapeutically effective amount of an HMG-CoA reductase inhibitor to a mammal in need thereof.
  • the present invention relates to a method of preventing or treating osteoporosis, osteoarthritis, Pagent's disease, hypercalcemia, periprosthetic bone loss or osteolysis, osteogenesis imperfecta, and aveolar bone loss associated with peridontal disease comprising administering a therapeutically effective amount of an HMG-CoA reductase inhibitor to a mammal in need thereof.
  • the present invention relates to the use of pharmaceutical compositions for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone reso ⁇ tion and stimulation of bone growth comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor.
  • the present invention also includes a pharmaceutical composition useful for preventing or for treating osteoporosis, osteoarthritis, Pagent's disease, hypercalcemia, periprosthetic bone loss or osteolysis, osteogenesis imperfecta, and aveolar bone loss associated with peridontal disease, comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor.
  • the present invention provides the use of HMG-CoA reductase inhibitors for the prevention and for the treatment of conditions ameliorated by decreasing plasma calcium levels, comprising administering a therapeutically effective amount of an HMG-CoA reductase inhibitor to a mammal in need thereof.
  • the present invention relates to preventing or treating primary hype ⁇ arathyroidism, familial hype ⁇ arathyroid syndrome, familial hypocalcuric hypercalcemia, secondary and tertiary hype ⁇ arathyroidism, humoral hypercalcemia of malignancy, hypoparathyroidism, malignant disease, PTH related protein, ectopic production of 1,25-dihydroxyvitamin D, lytic bone metastases, other factors produced locally or ectopically, endocrine disorders, granulomatous disease, drug induced elevated plasma calcium levels, renal failure, and total parenterai nutrition. Each of these conditions may be ameliorated by decreasing plasma calcium levels.
  • the present invention relates to the use of pharmaceutical compositions for the prevention and for the treatment of abnormal conditions ameliorated by decreasing plasma calcium levels, comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor.
  • the therapeutic agent useful in the present invention is a compound which inhibits HMG-CoA reductase.
  • Compounds which have inhibitory activity for HMG-CoA reductase can be readily identified by using assays well known in the art. See, US. Patent No. 4,231,938, to Monoghan et al, issued November 4, 1980, and US Patent No. 5,354,772, to Kathawal, issued October 11, 1994, both of which are inco ⁇ orated by reference herein in their entirety.
  • HMG-CoA reductase inhibitor generally refers to all classes of compounds included under this term in the prior art.
  • Statins which may be mentioned include atorvastatin (commercially available as Lipitor from Parke-Davis), cerivastatin (commercialy available as Lipobay ® or Baycol ® from Bayer), simvastatin (commercially available as Zocor ® from Merck), pravastatin (commercially available as Lipostat ® from Bristol-Myers Squibb), lovastatin (com- shoutally available as Mevacor from Merck), fluvastatin (commercially available as Lescol ® from Novartis), mevastatin, dihydrocompactin, compactin, itavastatin ("NK-
  • HMG-CoA reductase inhibitors that are useful in the present invention include but are not limited to those disclosed in US Patent No 5,177,080, US Patent No. 5,006,530, (including, specifically, cerivastatin); US Patent No. 4,231,938 (including, specifically, lovastatin); US Patent No. 4,444,784 (including specifically simvastatin; US Patent No. 4,346,227 (including, specifically, pravastatin); US Patent No. 5,354,772 (including, specifically, fluvastatin); US Patent No. 5,273,995 (including, specifically, atorvastatin); and US Patent No. 3,983,140 (including, specifically, mevastatin).
  • HMG-CoA reductase inhibitor is intended to include all pharmaceutically acceptable salts, esters and lactone forms of compound which have HMG-CoA reductase inhibitor activity. Therefore, the use of such salts, esters and lactone forms is included within the scope of this invention. Furthermore they can be present in the form of their hydrates, alkoholates, or tautomers.
  • preferred HMG Co-A reductase inhibitors are atorvastatin, cerivastatin, simvastatin, pravastatin, lovastatin; fluvastatin, itavastatin and (+)-(3R,5S)-bis(7-(4-(4-fluorophenyl)-6-isopropyl-2-(N-methyl-N-methane- sulfonylamino)pyrimidin-5-yl)-3,5-dihydroxy-6(E)-heptenoic acid.
  • the HMG-CoA reductase inhibitor is selected from the group consisting of cerivastatin, lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, mevastatin, and the pharmaceutically acceptable salts, esters and lactones thereof, and mixtures thereof. More preferably, the HMG-CoA reductase inhibitor is selected from the group consisting of cerivastatin, mevinolin, atorvastatin, simvastatin and the pharmaceutically acceptable salts, esters and lactones thereof, and mixtures thereof.
  • the most preferred compound useful in the present invention is cerivastatin and the pharmaceutically acceptable salts, esters and lactones thereof.
  • agents included within the scope of this invention may have one of the following formulas:
  • R represents an organic radical
  • Y represents hydrogen, a counterion, preferably an alkaline metal ion or an alkaline
  • R preferably represents a substituted hexahydro- or octahydronaphthaline, pyridine, pyrimidine, pyrrole quinoline or indole derivative.
  • HMG Co-A reductase inhibitors can be represented by the chemical formula
  • Z is selected from the group consisting of: a)
  • R is C ⁇ -C ⁇ 0 alkyl
  • R is selected from the group consisting of C1-C 3 alkyl, hydroxy, oxo, and C]-C 3 hydroxy substituted alkyl
  • R 3 is selected from the group consisting of hydrogen, hydroxy, C ⁇ -C 3 alkyl, and C 1 -C 3 hydroxy substituted alkyl
  • a, b, c, and d are all single bonds or a and c are double bonds, or b and d are double bonds, or one of a, b, c, and d is a double bonds
  • n is O, l, or 2;
  • X is selected from the group consisting of N[CH(CH 3 ) 2 ] and CH(CH 2 ) 3 CH 3
  • R 4 and R 5 are each independently selected from the group consisting of hydrogen, fluorine, chorine, bromine, iodine, Cj- alkyl, C ⁇ -C 4 alkoxy, and triflouoromethyl
  • Rs, R 7 , Rs, and R 9 are each independently selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, C ⁇ -C 4 alkyl, and C ⁇ -C 4 alkoxy.
  • the compounds useful in the methods and compositions of the invention can be synthesized by art known methods as they resemble a class of compounds known in the art to behave as HMG Co-A reductase inhibitors.
  • the particular process to be utilized in the preparation of any of the HMG-CoA reductase inhibitors of this invention depends upon the specific compound desired. Those processes are readily recognized by one of ordinary skill in the art with reference to such references including, but not limited to, the US Patents that are inco ⁇ orated herein by reference.
  • Representative salts of the compounds of the present invention include the conventional non-toxic salts and the quaternary ammonium salts which are formed, for example, by reacting the free acid with a suitable organic or incoragnic base.
  • Examples of salt forms of HMG-CoA reductase inhibitors icluded, but are not limited to, acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentane- propionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate
  • Base salts include but are not limited to alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine salts and N-methyl- D-glucamine.
  • basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate; and diamyl sulfates, long chain halides such as decyl, luaryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate
  • diamyl sulfates long chain halides such as decyl, luaryl, my
  • the sodium salt (cerivastatin sodium) is particularly preferably used.
  • esters in the present invention are non-toxic, pharmaceutically acceptable esters used in its standard meaning to denote the condensation product of a carboxylic acid and an alcohol. Ester derivatives of the described compounds can function as products which, when absorbed into the bloodstream of a warm-blooded animal, can cleave in such a manner as to release the drug form and permit the drug to afford improved therapeutic efficacy.
  • Esters of the present invention include but are not limited to such alkyl esters as methyl, ethyl, propyl, isopropyl, butyl, isobutyl or pentyl esters. Additional esters such as phenyl-d-C 5 alkyl may be used, although methyl ester is preferred.
  • the HMG-CoA reductase inhibitor of the present invention may be esterified by a variety of conventional procedures including reacting the appropriate anhydride, carboxylic acid or acid chloride with the alcohol group of the Formula I compound.
  • anhydride is reacted with the alcohol in the presence of an acylation catalyst such as l,8-bis[dimethylamino]napthalene or N,N-dimethylaminopyridine.
  • an acylation catalyst such as l,8-bis[dimethylamino]napthalene or N,N-dimethylaminopyridine.
  • An appropriate carboxylic acid can be reacted with the alcohol in the presence of a dehydrating agent such as dicyclohexylcarbodiimide, l-[3-dimethylaminopropyl]-3- ethylcarbodiimide or other water soluble dehydrating agents which are used to drive the reaction by the removal of water, and, optionally, an acylation catalyst. Esteri- fication can also be reached using the appropriate carboxylic acid in the presence of trifluoroacetic anhydride and, optionally, pyridine, or in the presence of N,N- carbonyldiimidazole with pyridine. Reaction of an acid chloride with the alcohol is carried out with an acylation catalyst such as 4-DMAP or pyridine. Sensitive or reactive groups on the compound of Formula I may need to be protected during any of the above methods for forming esters, and protecting groups may be added and removed by conventional methods well known in the art.
  • a dehydrating agent such as dicyclohex
  • lactones is used herein in referring to the HMG-CoA reductase inhibitors is used in its standard meaning to denote a cyclic condensation product of a carboxylic acid and an alcohol, i.e., a cyclic ester.
  • a cyclic condensation product of a carboxylic acid and an alcohol i.e., a cyclic ester.
  • One skilled in the art would readily know how to successfully carry out these as well as other methods of esterification of alcohols.
  • the HMG-CoA reductase inhibitors of this invention are those HMG Co-A reductase inhibitors that cause the concurrent decrease in bone reso ⁇ tion and stimulation of bone formation.
  • “Decrease in bone formation” means a detectable decrease in the number of osteoclasts from the number of osteoclasts that were present before administration of the HMG Co-A reductase inhibitors of this invention.
  • “Stimulation of bone formation (or of bone growth)” means a detectable increase in the number of osteoblasts from the number of osteoblasts that were present before administration of the HMG Co-A reductase inhibitors of the present invention.
  • an embodiment of the present invention is the administration of the HMG-CoA reductase inhibitors of this invention to a vertebrate, preferably a human or animal, for prevention and for the treatment of conditions ameliorated by concurrent decrease in bone reso ⁇ tion and stimulation of bone growth.
  • “Ameliorated” means its common meaning, that is, for example, to make better or tolerable.
  • HMG-CoA reductase inhibitors of this invention are expected to be valuable as therapeutic agents.
  • An embodiment of this invention includes a method of pre- venting or treating conditions in a vertebrate, preferably a mammal, that are ameliorated by concurrent decrease in bone reso ⁇ tion and stimulation of bone growth which comprises administering to said mammal a composition containing an amount of an HMG-CoA reductase inhibitor that is effective in preventing or treating the target condition.
  • an HMG Co-A reductase inhibitor on bone reso ⁇ tion and bone formation.
  • an OVX rat model can be used to determine the simultaneous effect of a test substance on the decrease in bone reso ⁇ tion and stimulation of bone growth.
  • HMG-CoA reductase inhibitors of the present invention were found to actively, contemporaneously decrease bone reso ⁇ tion while stimulating bone formation, uncoupling these two physiological events.
  • HMG-CoA reductase inhibitors of the present invention were found to actively decrease serum calcium levels.
  • CoA reductase inhibitors of this invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age, weight and or sex of the patient treated, and the nature and extent of the condition treated.
  • a precise pharmaceutically effective amount can be readily determined by the caregiver or clinician. Appropriate amounts can be determined by routine experimentation from animal models and human clinical studies.
  • the amounts of the HMG-CoA reductase inhibitor administered in daily dosing can be the same or similar to those amounts which are employed for anti-hyper- cholesterolemic treatment, and which are described in the Physicians' Desk Reference (PDR), which is inco ⁇ orated by reference herein in its entirety.
  • PDR Physicians' Desk Reference
  • the total amount of the active ingredient to be administered will generally range from about 0.01 micro gram kg to about 100 mg/kg, and preferably from about 1 micro gram/kg to about 20 mg/kg body weight per day.
  • a unit dosage may contain from about 10 micro grams to about 1500 mg of active ingredient, and can be administered one or more times per day.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician.
  • the compounds of this invention can be utilized to achieve the desired pharmacological effect by administration to a patient in need thereof in an appropriately formulated pharmaceutical composition.
  • a patient, for the pu ⁇ ose of this invention is a vertebrate, including a mammal and specifically including a human, in need of treatment for a particular condition or disease that is ameliorated by concurrent decrease in bone reso ⁇ tion and stimulation of bone formation. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of an
  • HMG-CoA reductase inhibitor or a pharmaceutically acceptable salt, ester, or lac- tone thereof.
  • a pharmaceutically acceptable carrier is any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient.
  • a pharmaceutically effective amount of an HMGCoA reductase inhibitor is that amount which produces a result or exerts an influence on the particular condition being treated.
  • HMG-CoA reductase inhibitors of the present invention can be administered by the patient to him/herself or by another, and will be administered with a pharmaceutically- acceptable carrier using any effective conventional dosage unit forms, including immediate and timed release preparations, orally, parenterally, topically, or the like.
  • the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions.
  • the solid unit dosage forms can be a capsule which can be of the ordinary hard- of soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
  • the HMG-CoA reductase inhibitors of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, cornstarch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes coloring agents, and flavoring agents intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient.
  • conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, cornstarch or gelatin
  • disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn
  • Suitable excipients for use in oral liquid dosage forms include diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent.
  • Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavoring and coloring agents described above, may also be present.
  • the pharmaceutical compositions of this invention may also be in the form of oil-in- water emulsions.
  • the oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils.
  • Suitable emulsifying agents may be (1) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, poly- oxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol.
  • the suspensions may also contain one or more preservatives, for example, ethyl or n- propyl p-hydroxybenzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such a sucrose or saccharin.
  • Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative and flavoring and coloring agents.
  • sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, and preservative and flavoring and coloring agents.
  • the HMG-CoA reductase inhibitors of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intramuscularly, or inter- peritoneally, as injectable dosages of the active ingredient in a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2 -dimethyl- 1,1- dioxolane-4-methanol, ethers such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant such as a soap or
  • Suitable fatty acide include oleic acid, stearic acid, and isostearic acid.
  • Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate.
  • Suitable soaps include fatty alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates; nonionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkyl- imidazoline quarternary ammonium salts, as well as mixtures.
  • suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alky
  • compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulation ranges from about 5% to about 15% by weight.
  • the surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
  • surfactants used in parenterai formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed b the condensation of propylene oxide with propylene glycol.
  • compositions may be in the form of sterile injectable aqueous suspensions.
  • suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl- cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acit, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadecaethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • Diluents and solvents that may be employed are, for example, water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile fixed oils are conventionally employed as solvents or suspending media.
  • any bland, fixed oil may be employed including synthetic monoor diglycerides.
  • fatty acids such as oleic acid can be used in the preparation of injectables.
  • composition of the invention may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are, for example, cocoa butter and polyethylene glycol.
  • transdermal delivery devices including transdermal patches. Such devices may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art (See, e.g.,
  • Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • a mechanical delivery device for the delivery of pharmaceutical agents is well known in the art.
  • Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier.
  • One such implantable delivery system used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,011,472, issued April 30, 1991.
  • Such mechanical delivery devices include implantable osmotic pumps.
  • the HMG-CoA reductase inhibitor of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilaellar vesicles and multilamellar vesicles.
  • Liposomes can be formed form a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • This invention can also be delivered by the use of monoclonal antibodies as individ- ual carriers to which the compound molecules are coupled.
  • Active ingredient(s) can also be coupled with soluble polymers as targetable carriers.
  • Such polymers can include polyvinyl-pyrrolidone, pyran copolymer, polyhydroxy-propyl-methacrylamide- phenol, polyhydroxy-ethyl-aspartamide-phenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues.
  • active ingredient(s) may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polyactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, poly- orthoesters, polyacetals, polydihydropyrans, polycycanoacrylates and cross linked or amphipathic block copolymers of hydrogels.
  • biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polyactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, poly- orthoesters, polyacetals, polydihydropyrans, polycycanoacrylates and cross linked or amphipathic block copolymers of hydrogels.
  • compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Any of the compositions of this invention may be preserved by the addition of an antioxidant such as ascorbic acid or by other suitable preservatives. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized.
  • the compound of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects.
  • the compounds of this invention can be combined with estrogen, progesterone, selective estrogen receptor modulators (SERMs), calcitonin, bisphosphonates, prostaglandin, sodium fluoride, PTH, calcium, vitamin D metabolites and the like, or other agents, useful in treating other indications, as well as with admixtures and combinations thereof.
  • SERMs selective estrogen receptor modulators
  • compositions which are comprised of an inert carrier and an effective amount of an HMG-CoA reductase inhibitor, or a salt, ester or lactone thereof are comprised of an inert carrier and an effective amount of an HMG-CoA reductase inhibitor, or a salt, ester or lactone thereof.
  • An inert carrier is any material which does not interact with the active ingredient to be carried and which lends support, means of conveyance, bulk, traceable material, and the like to the compound to be carried.
  • An effective amount of compound is that amount which produces a result or exerts an influence on the particular in vitro, ex vivo, or in vivo test or diagnostic procedure designed to indicate, directly or indirectly, the effect of an HMG-CoA reductase inhibitor on bone reso ⁇ tion or bone growth.
  • the long bones (femurs and tibias) were collected for bone mineral density analyses by pQCT, for static and dynamic bone histomo ⁇ hometry (bone formation) of the cortical and cancellous bone, structural bone analyses under polarized light, and mechanical testing of the cortical bone (3-point bending test). Also, one tibia was demineralized in cold EDTA and embedded in paraffin before stained with TRAP (Tartarate Resistent Acid Phosphatase) for the evaluation of bone reso ⁇ tion and osteoclast analysis.
  • TRAP Temporal Resistent Acid Phosphatase
  • OVX rat model is a small animal model for estrogen deficient bone loss in humans (postmenopausal osteoporosis) (Kalu, D.N. (1991). The ovariectomized rat model of postmenopausal bone loss, Bone Mineral 15:175-192).
  • rats had free access to food (standard laboratory diet, PMI Feeds, Inc., Richmond, IN) and water. Fluorochrome bone markers for histomo ⁇ hometric measurements were administered to all rats in this study as intraperitoneal injections before treatment started, 14 days, and 3 days before euthanasia. Experimental protocol. All rats in this study were dosed daily with an oral dose of vehicle (water) or cerivastatin (0.1; 0.01, and 0.001 mg/kg/day) during the 6 week period. Rats were dosed according to average group weight.
  • Body and Organ Weights were recorded weekly throughout the experiment. At necropsy liver, kidney, adrenal, and uterus was collected, and percent of body weight was determined.
  • pQCT Peripheral Quantitative Computed Tomography
  • Serum chemistry Following euthanasia, serum was collected through cardiac puncture. Serum was submitted to Bayer's comparative medicine department (West).
  • Urine Urine was collected overnight prior to the study, mid study, and prior to euthanasia. The urine was diluted 1:5 and 100 micro liters of the diluent was submitted to Bayer's comparative medicine department (West Haven, CT) to be run on the Technicon Axon system (Bayer Co ⁇ ., Tarrytown, NY) for urine creatinine levels.
  • distal halves were dehydrated with a series of graded ethanol and embedded in methyl- methacrylate.
  • Three consecutive cross sections of distal femoral metaphysis and femoral midshaft were cut with a precision bone saw Isomet 1000 (Buehler, Lake
  • Dynamic histomo ⁇ hometric measurements were performed on unstained, unde- calcified sections (3 sections per animal) at 10 and/or 20 X magnification using a fluorescence microscope (Nikon, Japan) interfaced with a digitizer. The data was collected using a software package ("Stereology", KSS Computer Engineers, Magna, UT) specifically written for bone histomo ⁇ hometry Measurements were made on the entire periosteal surface, on the endocortical surfaces which were free of endocortico-trabecular intersections, and on the entire metaphyseal spongiosa 0.5 mm below the growth plate.
  • Microradiographs of cortical and cancellous bone were analyzed for static and structural indices using "Image Analysis” software (KSS Computer Engineers). For this, images of the bone were captured with a video-camera CCD72 (DAGE-MTI Inc., Michigan City, MI) connected to the microscope, and from these images static mo ⁇ hometric indices were determined. These included cortical and marrow areas, average cortical width, trabecular area, and trabecular thickness, number, and separation.
  • Thyroparathyroid (TPTx) Sprague Dawley rats at 250 gms were ordered from Taconic Labs. At the vendor rats are kept on calcium lactate water 2-3% following surgery. Rats are also be shipped with this water. Upon arrival rats were placed on normal water and no food and were kept this way for the remainder of the study. 0.5 ml of blood was collected and tested for serum calcium levels. Any rat with a time 0 calcium of 10 mg/dl or higher was removed from the study. Alzet osmotic minipumps model 2001D (Alza Co ⁇ ., Palo Alto, Ca.) that were pre-filled with either
  • PTH (0.03 ug/ul, 0.25 ug/hr) or vehicle (saline) were implanted subcutaneously.
  • rats were dosed orally with 0.5 ml of either vehicle (water) or cerivastatin (3, 1, 0.1, 0.01, or 0.001 mg/kg).
  • cerivastatin 3, 1, 0.1, 0.01, or 0.001 mg/kg.
  • rats were euthanized with CO2, and bled via cardiac puncture. Serum was tested for calcium levels and analyzed using the students T-test. Tibias excized and placed in chilled 4° C neutral buffered formalin (Hydrol Chemical Company) for three days to allow for fixation. Washed with cold running tap water for one hour then placed in chilled 5% EDTA for decalcification, and paraffin processed. Sections were cut @ 5um and stained for Tartrate-resistant acid phosphatase (Trap) positive cells using a Leukocyte Acid Phosphatase Kit #387-A (Sigma Diagnostic
  • Example 3 is useful for preventing or treating conditions that can be ameliorated by concurrently decreasing bone reso ⁇ tion and stimulating bone growth, as well as for preventing or treating conditions that can be ameliorated by decreasing plasma calcium levels.
  • Example 3 is useful for preventing or treating conditions that can be ameliorated by concurrently decreasing bone reso ⁇ tion and stimulating bone growth, as well as for preventing or treating conditions that can be ameliorated by decreasing plasma calcium levels.
  • a tablet is prepared from Cervistatin 25 mg
  • the ingredients are mixed and compressed to form tablets.
  • the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
  • a tablet is prepared from
  • the ingredients are combined and blended together and compressed using conventional tableting techniques.
  • the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
  • HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
  • a hard gelatine capsule is prepared from
  • the dry ingredients are combined and blended together and encapsulated in the gelatin coating using standard manufacturing techniques.
  • the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
  • An oral suspension formulation is prepared from Cerivastatin 5 mg
  • An oral suspension is prepared by combining the ingredients using standard formulation techniques.
  • the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
  • HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
  • An intravenous infusion composition is prepared from Cerivastatin 5 mg
  • the ingredients are combined using standard formulation techniques, under sterile conditions.
  • the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
  • a capsule formula is prepared from Cerivastatin 40 mg
  • the components are blended, passed through an appropriate mesh sieve, and filled into hard gelatin capsules.
  • the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.

Landscapes

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

Abstract

This application relates to methods of using 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone resorption and stimulation of bone formation, comprising administering a therapeutically effective amount of a HMG-Co A reductase inhibitor to a vertebrate in need thereof. This invention also relates to methods of using HMG-CoA reductase inhibitors for the prevention and for the treatment of conditions ameliorated by a decrease in plasma calcium levels.

Description

METHODS OF AMELIORATING ABNORMAL BONE STATES
FIELD OF THE INVENTION
This invention relates to methods of using 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone resoφtion and stimulation of bone formation, comprising administering a therapeutically effective amount of a HMG-Co A reductase inhibitor to a vertebrate in need thereof. This invention also relates to methods of using HMG-CoA reductase inhibitors for the prevention and for the treatment of conditions ameliorated by a decrease in plasma calcium levels.
BACKGROUND OF THE INVENTION
HMG-CoA reductase catalyzes the conversion of HMG-CoA to mevalonate, which is an early and rate limiting step in the biosynthesis of cholesterol. Compounds from the class of compounds known as HMG-CoA reductase inhibitors (which includes cerivastatin, fluvastatin, lovastatin, mevastatin, pravastatin, simvastatin, atovastatin and others), have been known for some time to be effective anti-cholesteolemic agents in the prevention and treatment of cardiovascular disease. They also slow the progression of atherosclerotic lesions in the coronary and carotid arteries, and reduce the risk of coronary heart disease events including death.
Statins are also said to be effective in decreasing beta amyloid protein, a predictor of Alzheimer's disease (WO 99/48488, published September 30, 1999), as well as in inhibiting the l-desoxy-D-xylulose-5 -phosphate biosynthetic pathway and, accordingly, in treating certain parasitic infections (WO 99/52938, published October 21, 1999). These compounds are also reported to have an effect on bone. Generally, after reaching the so-called "peak bone mass" in early adulthood, the bone remains an active tissue. Indeed, throughout adult life, within focal sites scattered throughout the skeleton, microscopic quantities of bone are being replaced. In the normal adult skeleton there are approximately two million of these remodeling units. Bone remodeling is an orderly process, essential for bone strength and occurring in response to mechanical stress (Parfitt, A.M. (1988) Bone remodeling: relationship to the amount and structure of bone, and the pathogenesis and prevention of fractures. In: Riggs, B. & Melton, L.J. Ill, eds. Osteoporosis: Ethiology, diagnosis and management. New York: Raven Press, pp. 133-154.). A balanced sequence of bone resorption and formation is necessary, which in healthy individuals is carefully regulated by a mechanism known as coupling, mediated by a complex interaction of hormones, paracrine growth factors and cytokines (Martin, J.J., Wah, N.K., and Suda, T. (1989) Bone cell physiology. Endocrinol. Metabol. Clin. North. Am. 18:833-858).
Bone resorption is mediated by the osteoclast, a multinucleated cell derived from granulocyte-macrophage precursors in the bone marrow, while bone formation requires the presence and function of osteoblasts, derived from mesenchymal fibroblast-like cells (Raisz, L.G. and Rodan, G.A. (1990) Cellular basis for bone turnover. In: Avioli, L.V. and Krane, S.M. eds.: Metabolic bone diseases and clinically related disorders. Philadelphia: W.B. Saunders Company, pp. 1-41.). Many factors are known to stimulate bone resoφtion, but suφrisingly, none of these factors appears to act directly on osteoclasts. The receptors for them are expressed on osteoblasts, and only in the presence of osteoblasts is osteoclastic activity increased following exposure to these stimulating factors. It is now widly believed that osteoblasts control bone resoφtion through their effects on osteoclasts (Chambers, T.J. and Fuller, K. (1985) Bone cells predispose endosteal surfaces to resoφtion by exposure of bone mineral to osteoclastic contact. J. Cell Sci. 76:155- 163) by either direct cell-cell contact or local release of a diffusable osteoblast- derived osteoclast-stimulating factor (McSheehy, P.M.J. and Chambers, T.J. (1986) Osteoblast-like cells in the presence of parathyroid hormone release soluble factors that stimulate osteoclastic bone resoφtion. Endocrinology 119:1614-1659).
The bone remodeling cycle takes place according to a highly organized sequence of events, which is dependent on precise inteφlay between osteoclasts and osteoblasts.
Under normal circumstances and at any one time, 80-95% of the adult bone surfaces is in a quiescent state and covered by residing osteoblasts, while the rest is involved in various stages of the remodeling cycle (Parfitt, A.M. (1988) Bone remodeling: relationship to the amount and structure of bone, and the pathogenesis and prevention of fractures. In: Riggs, B. & Melton, L.J. Ill, eds. Osteoporosis: Ethiology, diagnosis and management. New York: Raven Press, pp. 133-154; Raisz, L.G. and Rodan, G.A. (1990) Cellular basis for bone turnover. In: Avioli, L.V. and Krane, S.M. eds.: Metabolic bone diseases and clinically related disorders. Philadelphia: W.B. Saunders Company, pp. 1-41). Initiation of the remodeling process in microscopic areas of the bone surface is known as the activation phase. Humoral and possibly mechanical stimuli digest the endosteal membrane as it retracts to expose unmineralized bone surface which, in turn, is chemotactic to osteoclast precursors. The digestion of the endosteal membrane probably involves the production by osteoblasts of metalloproteinases and latent collagenase secreted in an active form and subsequently activated by factors released by the lining cells.
Activation is followed by a resoφtion phase during which osteoclasts resorb a packet of bone, thus creating a cavity (Howship's lacunae) in the bone surface. A reversal phase follows, separating the completed phase of bone resoφtion and the subsequent initiation of bone formation. Mononuclear cells in the resoφtion cavity smooth out the irregular bone surface left behind by the osteoclasts, and cells (possibly osteocytes) are involved in the deposition of a cement layer on the bone surface.
After deposition of this cement layer, a team of new osteoblasts begins to deposit undemineralized bone matrix, the osteoid seam. This newly deposited layer of matrix begins to mineralize after a maturation period of 5-10 days. After completion of bone formation, the lacunae created by the osteoclasts have been filled with newly deposited bone covered by lining cells. The total duration of a remodeling cycle in adults is estimated to be around 250 days. If the osteoblasts have deposited exactly the same amount of bone as has been removed previously by the osteoclasts, the remodeling cycle is in balance. Number of hormonal and/or mechanical factors can change this balance on the tissue or local level resulting in positive or more frequently negative bone balance and loss of bone (Boyce, B.F. (1991) Normal bone remodeling and its disruption in metastatic bone disease. In: Rubens, R.D. & Fogelman, I., eds.: Bone metastases - diagnosis and treatment. London: Springer- Verlag, 11-30; Eriksen, E.F., Steinche, T., and Mosekilde, L. et al., (1989)
Histomoφhometric analyses of bone in metabolic bone disease. Endocrinol. Metabol. Clin. North. Am. 18:919-954; Mundy, G.R. (1987) Bone resoφtion and turnover in health and disease. Bone 8 (Suppl. 1):S9-S16). Balanced remodeling cycles, which are necessary to ensure maintanence of the skeleton's structural integrity, suppose to deposit exact amount of bone being previously resorbed by osteoclasts and at the exact same location. It is clear that this process necessitates a highest degree of coordination between the activity of osteoclasts and osteoblasts. Bone formation is coupled to bone resoφtion and vice versa, so that each time when bone resoφtion increases or decreases, bone formation follows the same pattern in order to maintain original bone mass. Because bone resoφtion is much faster process relative to bone formation, the end result of bone remodeling imbalance is often bone loss.
Drugs used to treat bone loss of various etiologies can be grouped into those that decrease bone resoφtion and those that increase bone formation. Both groups of drugs will affect bone resoφtion and bone formation in the similar fashion due to the coupling between bone resoφtion and bone formation. Antiresoφtive drugs such as estrogen, calcitonin, bisphosphonates, and selective estrogen receptor modulators (SERMs) will reduce or completely shut down bone resoφtion which will consequently block bone formation (Riggs, B.L., and Melton III, L.J. Drug Therapy,
The New England Journal of Medicine, 327:620-627; 1992; Patel, S., Lyons, A.R. and Hosking, D.J. Drugs used in the treatment of metabolic bone disease, Drugs, 46:594-617; 1993). Similarly, certain bone forming agents which are in clinical development will demonstrate increase in bone turnover rate.
Wang, G.J., et al, J Formos MedAssoc (1995) 94:589-592 reported that certain lipid clearing agents including lovastatin, were able to inhibit steroid induced bone resoφtion in rabbits. In bone marrow cultures, mevastatin was reported to inhibit in vitro osteoclastogenesis, the formation of specialized, multinucleated osteoclast cells active in bone resoφtion. (S.P. Luckman, et al, Bone, (April, 1997) 20:4(Supp.) Abst No. P378).
Additionally, WO 98/25460 (published June 18, 1998, Zymogenetics) disclosed that certain statins exhibited an in vitro stimulatory effect on osteoblastogenesis, as well as evidence of bone formation in in vivo calavrial studies, an increase in trabecular density and cortical thickness in ovariexctomized (OVX) mice, and in vivo callus formation at osteotomy sites in the forelimbs of rabbits. IL-1 induced bone resoφtion, however, was not affected by concentrations of statins that corresponded to doses that stimulated in vitro osteoblast activity. II- 1 induced calcium release was decreased by simvastatin at a higher dose, although this decrease in calcium release was attributed to toxicity.
Bauer, D.C., et al "Statin Use, Bone Mass and Fracture: An Analysis of Two Prospective Studies," ASBMR, 21st Annual Meeting, (September, 1999), Abstract No. 1188 reported that HMG-CoA reductase inhibitors (statins) are potent agonists of bone formation in women 55 years and older, as evidenced by higher hip bone mineral density and reduced fracture risk. Garrett, I.R., et al (Abstract Number 1189 of the same publication) reports the effect of statins in general in stimulating bone formation in organ cultures of neonatal murine calvaria by stimulating certain bone growth factor (BMP-2) transcription, and that cerivastatin in particular stimulates BMP-2 transcription, BMP-2-mRNA expression in vivo and bone formation in calvarial organ cultures in doses 1000-fold less than other statins. Also WO 99/45923 (Merck & Co., September 16, 1999) disclosed methods of inhibiting abnormal bone resoφtion in mammals, without any mention of concurrent stimulation of bone growth.
No previous publication discloses that the administration of an HMG-CoA reductase inhibitor both decreases bone resoφtion while concurrently stimulating bone growth. It is now known that HMG-Co-A reductase inhibitors simultaneously affect bone formation by promoting osteoblast differentiation in the bone marrow, while reducing formation of multinucleated osteoclasts from their mononuclear precursors in the bone marrow.
Additionally, the use of HMG Co-A reductase inhibitors to effectively decrease plasma calcium levels and, therefore ameliorate conditions caused by high plasma calcium levels, is also heretofore unknown.
SUMMARY OF THE INVENTION
The delicate balance between normal bone resoφtion and normal bone formation can be disrupted by, for example but not by way of limitation, chronic glucocorticoid use, immunosuppresive therapy, hormone imbalance, malignancy and the like. Disrupted balance between bone resoφtion and bone formation can result in such conditions or disease states as osteoporosis of various ethiologies, osteoarthritis, Pagent's disease, hypercalcemia, periprosthetic bone loss or osteolysis, fractures, osteogenesis imperfecta, and aveolar bone loss associated with peridontal disease.
The present invention provides the use of HMG-CoA reductase inhibitors for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone resoφtion and stimulation of bone growth comprising administering a therapeutically effective amount of an HMG-CoA reductase inhibitor to a mammal in need thereof. In a further embodiment, the present invention relates to a method of preventing or treating osteoporosis, osteoarthritis, Pagent's disease, hypercalcemia, periprosthetic bone loss or osteolysis, osteogenesis imperfecta, and aveolar bone loss associated with peridontal disease comprising administering a therapeutically effective amount of an HMG-CoA reductase inhibitor to a mammal in need thereof.
In further embodiments, the present invention relates to the use of pharmaceutical compositions for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone resoφtion and stimulation of bone growth comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor. The present invention also includes a pharmaceutical composition useful for preventing or for treating osteoporosis, osteoarthritis, Pagent's disease, hypercalcemia, periprosthetic bone loss or osteolysis, osteogenesis imperfecta, and aveolar bone loss associated with peridontal disease, comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor.
In further embodiments, the present invention provides the use of HMG-CoA reductase inhibitors for the prevention and for the treatment of conditions ameliorated by decreasing plasma calcium levels, comprising administering a therapeutically effective amount of an HMG-CoA reductase inhibitor to a mammal in need thereof.
In a further embodiment, the present invention relates to preventing or treating primary hypeφarathyroidism, familial hypeφarathyroid syndrome, familial hypocalcuric hypercalcemia, secondary and tertiary hypeφarathyroidism, humoral hypercalcemia of malignancy, hypoparathyroidism, malignant disease, PTH related protein, ectopic production of 1,25-dihydroxyvitamin D, lytic bone metastases, other factors produced locally or ectopically, endocrine disorders, granulomatous disease, drug induced elevated plasma calcium levels, renal failure, and total parenterai nutrition. Each of these conditions may be ameliorated by decreasing plasma calcium levels.
In further embodiments, the present invention relates to the use of pharmaceutical compositions for the prevention and for the treatment of abnormal conditions ameliorated by decreasing plasma calcium levels, comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor.
DETAILED DESCRIPTION OF THE INVENTION
The therapeutic agent useful in the present invention is a compound which inhibits HMG-CoA reductase. Compounds which have inhibitory activity for HMG-CoA reductase can be readily identified by using assays well known in the art. See, US. Patent No. 4,231,938, to Monoghan et al, issued November 4, 1980, and US Patent No. 5,354,772, to Kathawal, issued October 11, 1994, both of which are incoφorated by reference herein in their entirety.
In the context of the present invention, "HMG-CoA reductase inhibitor" generally refers to all classes of compounds included under this term in the prior art. Statins which may be mentioned include atorvastatin (commercially available as Lipitor from Parke-Davis), cerivastatin (commercialy available as Lipobay® or Baycol® from Bayer), simvastatin (commercially available as Zocor® from Merck), pravastatin (commercially available as Lipostat® from Bristol-Myers Squibb), lovastatin (com- mercially available as Mevacor from Merck), fluvastatin (commercially available as Lescol® from Novartis), mevastatin, dihydrocompactin, compactin, itavastatin ("NK-
104", disclosed in EP-A-304 063), and (+)-(3R,5S)-bis(7-(4-(4-fluoroρhenyl)-6-iso- propyl-2-(N-methyl-N-methanesulfonylamino)pyrimidin-5-yl)-3,5-dihydroxy-6(E)- heptenoic acid, calcium salt (ZD-4522 disclosed in EP-A- 521 471).
Examples of HMG-CoA reductase inhibitors that are useful in the present invention include but are not limited to those disclosed in US Patent No 5,177,080, US Patent No. 5,006,530, (including, specifically, cerivastatin); US Patent No. 4,231,938 (including, specifically, lovastatin); US Patent No. 4,444,784 (including specifically simvastatin; US Patent No. 4,346,227 (including, specifically, pravastatin); US Patent No. 5,354,772 (including, specifically, fluvastatin); US Patent No. 5,273,995 (including, specifically, atorvastatin); and US Patent No. 3,983,140 (including, specifically, mevastatin). The patents cited in the previous sentence that are not already incoφorated by reference previously are incoφorated by reference herein in their entirety. The structural formulae of these and additional HMG-CoA reductase inhibitors that can be used in the present invention are described in the respective US Patents listed above, as well as in M. Yalpani, "Cholesterol Lowering Drugs",
Chemistry & Industry, pp 85 - 89 (February 5, 1996), which is also incoφorated herein by reference in its entirety.
The term HMG-CoA reductase inhibitor is intended to include all pharmaceutically acceptable salts, esters and lactone forms of compound which have HMG-CoA reductase inhibitor activity. Therefore, the use of such salts, esters and lactone forms is included within the scope of this invention. Furthermore they can be present in the form of their hydrates, alkoholates, or tautomers.
For the puφose of this invention, preferred HMG Co-A reductase inhibitors are atorvastatin, cerivastatin, simvastatin, pravastatin, lovastatin; fluvastatin, itavastatin and (+)-(3R,5S)-bis(7-(4-(4-fluorophenyl)-6-isopropyl-2-(N-methyl-N-methane- sulfonylamino)pyrimidin-5-yl)-3,5-dihydroxy-6(E)-heptenoic acid. Most preferably, the HMG-CoA reductase inhibitor is selected from the group consisting of cerivastatin, lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, mevastatin, and the pharmaceutically acceptable salts, esters and lactones thereof, and mixtures thereof. More preferably, the HMG-CoA reductase inhibitor is selected from the group consisting of cerivastatin, mevinolin, atorvastatin, simvastatin and the pharmaceutically acceptable salts, esters and lactones thereof, and mixtures thereof. The most preferred compound useful in the present invention is cerivastatin and the pharmaceutically acceptable salts, esters and lactones thereof. Generally, agents included within the scope of this invention may have one of the following formulas:
Figure imgf000011_0001
wherein
R represents an organic radical
X represents -CH2-CH2- or -CH=CH-, preferably in the trans form
Y represents hydrogen, a counterion, preferably an alkaline metal ion or an alkaline
earth metal ion or a Cι-C6-alkyl group
wherein R preferably represents a substituted hexahydro- or octahydronaphthaline, pyridine, pyrimidine, pyrrole quinoline or indole derivative.
Preferred HMG Co-A reductase inhibitors can be represented by the chemical formula
Figure imgf000011_0002
wherein Z is selected from the group consisting of: a)
Figure imgf000012_0001
wherein R is Cι-Cι0 alkyl, R is selected from the group consisting of C1-C3 alkyl, hydroxy, oxo, and C]-C3 hydroxy substituted alkyl, R3 is selected from the group consisting of hydrogen, hydroxy, Cι-C3 alkyl, and C1-C3 hydroxy substituted alkyl, a, b, c, and d are all single bonds or a and c are double bonds, or b and d are double bonds, or one of a, b, c, and d is a double bonds, and n is O, l, or 2;
b)
Figure imgf000012_0002
wherein X is selected from the group consisting of N[CH(CH3)2] and CH(CH2)3CH3
c)
Figure imgf000012_0003
Figure imgf000013_0001
e)
Figure imgf000013_0002
and
f)
Figure imgf000013_0003
wherein R4 and R5 are each independently selected from the group consisting of hydrogen, fluorine, chorine, bromine, iodine, Cj- alkyl, Cι-C4 alkoxy, and triflouoromethyl, and Rs, R7, Rs, and R9 are each independently selected from the group consisting of hydrogen, fluorine, chlorine, bromine, iodine, Cι-C4 alkyl, and Cι-C4 alkoxy. See U.S. Patent No. 5,650,523, to DeCamp et al., issued July 22, 1997, which is incoφorated by reference herein in its entirety.
The pharmaceutically acceptable salt, ester, and lactone forms or the carboxylic acids or salts of carboxylic acids of the compounds depicted by the preceding chemical formulas are also intended to be within the scope of the present invention.
The compounds useful in the methods and compositions of the invention can be synthesized by art known methods as they resemble a class of compounds known in the art to behave as HMG Co-A reductase inhibitors. The particular process to be utilized in the preparation of any of the HMG-CoA reductase inhibitors of this invention depends upon the specific compound desired. Those processes are readily recognized by one of ordinary skill in the art with reference to such references including, but not limited to, the US Patents that are incoφorated herein by reference.
Representative salts of the compounds of the present invention include the conventional non-toxic salts and the quaternary ammonium salts which are formed, for example, by reacting the free acid with a suitable organic or incoragnic base. Examples of salt forms of HMG-CoA reductase inhibitors icluded, but are not limited to, acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentane- propionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, itaconoate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, sulfonate, tartrate, thiocyanate, tosylate,undecanoate, and mixtures thereof.
Base salts include but are not limited to alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine salts and N-methyl- D-glucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate; and diamyl sulfates, long chain halides such as decyl, luaryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
Preferred are pharmaceutically acceptable and water soluble salts such as the alkaline metal and alkaline-earth metal salts, e.g. the sodium, potassium or calcium salts. In the case of cerivastatin the sodium salt (cerivastatin sodium) is particularly preferably used.
The esters in the present invention are non-toxic, pharmaceutically acceptable esters used in its standard meaning to denote the condensation product of a carboxylic acid and an alcohol. Ester derivatives of the described compounds can function as products which, when absorbed into the bloodstream of a warm-blooded animal, can cleave in such a manner as to release the drug form and permit the drug to afford improved therapeutic efficacy. Esters of the present invention include but are not limited to such alkyl esters as methyl, ethyl, propyl, isopropyl, butyl, isobutyl or pentyl esters. Additional esters such as phenyl-d-C5 alkyl may be used, although methyl ester is preferred.
The HMG-CoA reductase inhibitor of the present invention may be esterified by a variety of conventional procedures including reacting the appropriate anhydride, carboxylic acid or acid chloride with the alcohol group of the Formula I compound.
The appropriate anhydride is reacted with the alcohol in the presence of an acylation catalyst such as l,8-bis[dimethylamino]napthalene or N,N-dimethylaminopyridine.
An appropriate carboxylic acid can be reacted with the alcohol in the presence of a dehydrating agent such as dicyclohexylcarbodiimide, l-[3-dimethylaminopropyl]-3- ethylcarbodiimide or other water soluble dehydrating agents which are used to drive the reaction by the removal of water, and, optionally, an acylation catalyst. Esteri- fication can also be reached using the appropriate carboxylic acid in the presence of trifluoroacetic anhydride and, optionally, pyridine, or in the presence of N,N- carbonyldiimidazole with pyridine. Reaction of an acid chloride with the alcohol is carried out with an acylation catalyst such as 4-DMAP or pyridine. Sensitive or reactive groups on the compound of Formula I may need to be protected during any of the above methods for forming esters, and protecting groups may be added and removed by conventional methods well known in the art.
The term "lactones" is used herein in referring to the HMG-CoA reductase inhibitors is used in its standard meaning to denote a cyclic condensation product of a carboxylic acid and an alcohol, i.e., a cyclic ester. One skilled in the art would readily know how to successfully carry out these as well as other methods of esterification of alcohols.
The HMG-CoA reductase inhibitors of this invention are those HMG Co-A reductase inhibitors that cause the concurrent decrease in bone resoφtion and stimulation of bone formation. "Decrease in bone formation" means a detectable decrease in the number of osteoclasts from the number of osteoclasts that were present before administration of the HMG Co-A reductase inhibitors of this invention. "Stimulation of bone formation (or of bone growth)" means a detectable increase in the number of osteoblasts from the number of osteoblasts that were present before administration of the HMG Co-A reductase inhibitors of the present invention. Accordingly, an embodiment of the present invention is the administration of the HMG-CoA reductase inhibitors of this invention to a vertebrate, preferably a human or animal, for prevention and for the treatment of conditions ameliorated by concurrent decrease in bone resoφtion and stimulation of bone growth. "Ameliorated" means its common meaning, that is, for example, to make better or tolerable.
The HMG-CoA reductase inhibitors of this invention are expected to be valuable as therapeutic agents. An embodiment of this invention includes a method of pre- venting or treating conditions in a vertebrate, preferably a mammal, that are ameliorated by concurrent decrease in bone resoφtion and stimulation of bone growth which comprises administering to said mammal a composition containing an amount of an HMG-CoA reductase inhibitor that is effective in preventing or treating the target condition.
The effectiveness of the HMG-CoA reductase inhibitors of this invention of in concurrently decreasing bone resoφtion and stimulating bone growth can be demonstrated by the following procedures.
Sexually mature ovariectomized rat (Ovx) is a suitable and FDA approved model for studying bone loss due to estrogen deficiency, displaying both increased bone tumover (Wronski, T.J., Cintron, M. and Dann, L.M. (1988) Temporal relationship between bone loss and decreased bone turnover in ovariectomized rats. Calcif. Tissue Int. 43:179-183; Ismail, F., Epstein, S., Fallon, M.D., Thomas, S.B. and
Reinhardt, T.A. (1989) Serum bone gla protein and the vitamin D endocrine system in the ovariectomized rat. Endocrinology 122:2624-2630; Morris, H.A.,Porter, S.J., Durbridge, T.C., Moore, R.J., Need, A.G., Nordin, B.E.C. (1992): Effects of ovariectomy on biochemical and bone variables in rat. Bone Mineral 18:133-142) and rapid bone loss (Wronski, T.J., Dann, L.M., Scott, K.S. and Cintron, M. (1989)
Long-term effects of ovariectomy and aging on the rat skeleton. Calcif. Tissue Int. 45;360-366; Kimmel, D.B. and Wronski, T.J. (1990) Nondestructive measurement of bone mineral in femurs from ovariectomized rats. Calcif. Tissue Int. 46:101-110). Both effects can be blocked by estradiol treatment (Ismail, F., Epstein, S., Fallon, M.D., Thomas, S.B. and Reinhardt, T.A. (1989) Serum bone gla protein and the vitamin D endocrine system in the ovariectomized rat. Endocrinology 122:2624- 2630; Kimmel, D.B. and Wronski, T.J. (1990) nondestructive measurement of bone mineral in femurs from ovariectomized rats. Calcif. Tissue Int. 46:101-110). Wronski et al., (1988) reported increased histomoφhometric markers of bone resoφtion and formation coincident with trabecular bone loss in the proximal tibial metaphysis at 15 days postovariectomy, and biochemical markers of bone turnover have been found to be significantly elevated at 21 days after ovariectomy (Morris, H.A.,Porter, S.J., Durbridge, T.C., Moore, R.J., Need, A.G., Nordin, B.E.C. (1992): Effects of ovariectomy on biochemical and bone variables in rat. Bone Mineral 18:133-142).
One skilled in the art, using one or more of the procedures referenced above, could readily determine the effect of an HMG Co-A reductase inhibitor on bone resoφtion and bone formation. Particularly, an OVX rat model can be used to determine the simultaneous effect of a test substance on the decrease in bone resoφtion and stimulation of bone growth.
In tests utilizing the procedures described above, HMG-CoA reductase inhibitors of the present invention were found to actively, contemporaneously decrease bone resoφtion while stimulating bone formation, uncoupling these two physiological events.
Similarly, one skilled in the art could readily determine the effect of a HMG Co-A reductase inhibitor on plasma calcium levels by procedures well known in the art, particularly by means of standard blood tests.
HMG-CoA reductase inhibitors of the present invention were found to actively decrease serum calcium levels.
Based upon the above and other standard laboratory techniques known to evaluate bone resoφtion and stimulated bone formation, or decrease in plasma calcium levels, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions identified above, and by comparison of these results with the results of known medicaments that are used to treat these conditions, or by the comparison of these results with the appropriate dosage regimen for HMGCoA reductase inhibitors for other uses, the effective dosage of the HMG-
CoA reductase inhibitors of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age, weight and or sex of the patient treated, and the nature and extent of the condition treated. Thus, a precise pharmaceutically effective amount can be readily determined by the caregiver or clinician. Appropriate amounts can be determined by routine experimentation from animal models and human clinical studies.
The amounts of the HMG-CoA reductase inhibitor administered in daily dosing can be the same or similar to those amounts which are employed for anti-hyper- cholesterolemic treatment, and which are described in the Physicians' Desk Reference (PDR), which is incoφorated by reference herein in its entirety.
The total amount of the active ingredient to be administered will generally range from about 0.01 micro gram kg to about 100 mg/kg, and preferably from about 1 micro gram/kg to about 20 mg/kg body weight per day. A unit dosage may contain from about 10 micro grams to about 1500 mg of active ingredient, and can be administered one or more times per day. Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician.
The compounds of this invention can be utilized to achieve the desired pharmacological effect by administration to a patient in need thereof in an appropriately formulated pharmaceutical composition. A patient, for the puφose of this invention, is a vertebrate, including a mammal and specifically including a human, in need of treatment for a particular condition or disease that is ameliorated by concurrent decrease in bone resoφtion and stimulation of bone formation. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of an
HMG-CoA reductase inhibitor, or a pharmaceutically acceptable salt, ester, or lac- tone thereof. A pharmaceutically acceptable carrier is any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient. A pharmaceutically effective amount of an HMGCoA reductase inhibitor is that amount which produces a result or exerts an influence on the particular condition being treated. The HMG-CoA reductase inhibitors of the present invention can be administered by the patient to him/herself or by another, and will be administered with a pharmaceutically- acceptable carrier using any effective conventional dosage unit forms, including immediate and timed release preparations, orally, parenterally, topically, or the like.
Oral administration is preferable. For oral administration, the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions. The solid unit dosage forms can be a capsule which can be of the ordinary hard- of soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
In another embodiment, the HMG-CoA reductase inhibitors of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, cornstarch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid, or magnesium, calcium or zinc stearate, dyes coloring agents, and flavoring agents intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent.
Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavoring and coloring agents described above, may also be present.
The pharmaceutical compositions of this invention may also be in the form of oil-in- water emulsions. The oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils. Suitable emulsifying agents may be (1) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, poly- oxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent such as, for example, beeswax, hard paraffin, or cetyl alcohol. The suspensions may also contain one or more preservatives, for example, ethyl or n- propyl p-hydroxybenzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such a sucrose or saccharin.
Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative and flavoring and coloring agents. The HMG-CoA reductase inhibitors of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intramuscularly, or inter- peritoneally, as injectable dosages of the active ingredient in a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene glycol, glycerol ketals such as 2,2 -dimethyl- 1,1- dioxolane-4-methanol, ethers such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant such as a soap or a detergent, suspending agent such as pectin, carbomers, methycellulose, hydroxypropyl- methylcellulose, or carboxymethylcellulose, or emulsifying agent and other pharmaceutical adjuvants.
Illustrative of oils which can be used in the parenterai formulations of this invention are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum and mineral oil. Suitable fatty acide include oleic acid, stearic acid, and isostearic acid. Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate. Suitable soaps include fatty alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates; nonionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkyl- imidazoline quarternary ammonium salts, as well as mixtures.
The parenterai compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulation ranges from about 5% to about 15% by weight. The surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
Illustrative of surfactants used in parenterai formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed b the condensation of propylene oxide with propylene glycol.
The pharmaceutical compositions may be in the form of sterile injectable aqueous suspensions. Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl- cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acit, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadecaethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride, for example polyoxyethylene sorbitan monooleate.
The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Diluents and solvents that may be employed are, for example, water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as solvents or suspending media. For this puφose, any bland, fixed oil may be employed including synthetic monoor diglycerides. In addition, fatty acids such as oleic acid can be used in the preparation of injectables.
A composition of the invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are, for example, cocoa butter and polyethylene glycol.
Another formulation employed in the methods of the present invention employs transdermal delivery devices including transdermal patches. Such devices may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (See, e.g.,
US Patent No. 5,023,252, issued June 11, 1991, incoφorated herein by reference). Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,011,472, issued April 30, 1991. Such mechanical delivery devices include implantable osmotic pumps.
The HMG-CoA reductase inhibitor of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilaellar vesicles and multilamellar vesicles. Liposomes can be formed form a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
This invention can also be delivered by the use of monoclonal antibodies as individ- ual carriers to which the compound molecules are coupled. Active ingredient(s) can also be coupled with soluble polymers as targetable carriers. Such polymers can include polyvinyl-pyrrolidone, pyran copolymer, polyhydroxy-propyl-methacrylamide- phenol, polyhydroxy-ethyl-aspartamide-phenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues. Furthermore, active ingredient(s) may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polyactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, poly- orthoesters, polyacetals, polydihydropyrans, polycycanoacrylates and cross linked or amphipathic block copolymers of hydrogels.
The compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Any of the compositions of this invention may be preserved by the addition of an antioxidant such as ascorbic acid or by other suitable preservatives. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized.
The compound of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects. For example, the compounds of this invention can be combined with estrogen, progesterone, selective estrogen receptor modulators (SERMs), calcitonin, bisphosphonates, prostaglandin, sodium fluoride, PTH, calcium, vitamin D metabolites and the like, or other agents, useful in treating other indications, as well as with admixtures and combinations thereof.
The use of the HMG-CoA reductase inhibitors of the present invention may also be utilized, in free base form or in compositions, in research and diagnostics, or as analytical references, standards, and the like. Therefore, the present invention includes compositions which are comprised of an inert carrier and an effective amount of an HMG-CoA reductase inhibitor, or a salt, ester or lactone thereof. An inert carrier is any material which does not interact with the active ingredient to be carried and which lends support, means of conveyance, bulk, traceable material, and the like to the compound to be carried. An effective amount of compound is that amount which produces a result or exerts an influence on the particular in vitro, ex vivo, or in vivo test or diagnostic procedure designed to indicate, directly or indirectly, the effect of an HMG-CoA reductase inhibitor on bone resoφtion or bone growth.
The invention is described further by the following specific examples which are included as illustrations only and are not to be construed as limiting the scope of the invention in any way.
Example 1
Determining effect on bone resoφtion and simultaneous stimulation of bone growth
In our studies we were following two protocols utilizing Ovx rat model of postmenopausal osteoporosis: Prevention protocol - dosing with start at the same time with Ovx surgery, and Restoration protocol - in which rats were Ovx-ed 8 weeks before initiation of treatment. Rats were dosed orally with Cerivastatin at 0.1, 0.01, and 0.001 mg/kg/day over 6 weeks. At the end of the study serum and urine were collected for serum and urine chemistry. The long bones (femurs and tibias) were collected for bone mineral density analyses by pQCT, for static and dynamic bone histomoφhometry (bone formation) of the cortical and cancellous bone, structural bone analyses under polarized light, and mechanical testing of the cortical bone (3-point bending test). Also, one tibia was demineralized in cold EDTA and embedded in paraffin before stained with TRAP (Tartarate Resistent Acid Phosphatase) for the evaluation of bone resoφtion and osteoclast analysis.
Laboratory animals. Female Sprague-Dawley rats (Harlan Sprague Dawley, Inc., Indianapolis, IN) were obtained at 90 days of age. Rats were ovariectomized or sham operated using a dorsal approach. At week eight, rats were divided into groups based on body weight with OVX rats in four groups of ten to twelve and Sham rats in one group of ten. OVX rat model is a small animal model for estrogen deficient bone loss in humans (postmenopausal osteoporosis) (Kalu, D.N. (1991). The ovariectomized rat model of postmenopausal bone loss, Bone Mineral 15:175-192). Throughout the 6 weeks of the experiment rats had free access to food (standard laboratory diet, PMI Feeds, Inc., Richmond, IN) and water. Fluorochrome bone markers for histomoφhometric measurements were administered to all rats in this study as intraperitoneal injections before treatment started, 14 days, and 3 days before euthanasia. Experimental protocol. All rats in this study were dosed daily with an oral dose of vehicle (water) or cerivastatin (0.1; 0.01, and 0.001 mg/kg/day) during the 6 week period. Rats were dosed according to average group weight.
Body and Organ Weights. Body weights were recorded weekly throughout the experiment. At necropsy liver, kidney, adrenal, and uterus was collected, and percent of body weight was determined.
Peripheral Quantitative Computed Tomography (pQCT). At the end of the study bone mineral density and bone mineral content were determined at distal femoral metaphysis and femoral midshaft by using pQCT (Norland). The Bone Mineral Density (BMD) was expressed in g/cm .
Serum chemistry. Following euthanasia, serum was collected through cardiac puncture. Serum was submitted to Bayer's comparative medicine department (West
Haven, CT) to be run on the Technicon Axon system (Bayer Coφ. Taπytown, NY) for the
Urine. Urine was collected overnight prior to the study, mid study, and prior to euthanasia. The urine was diluted 1:5 and 100 micro liters of the diluent was submitted to Bayer's comparative medicine department (West Haven, CT) to be run on the Technicon Axon system (Bayer Coφ., Tarrytown, NY) for urine creatinine levels.
Histology. Right tibias were cleaned of soft tissues, fixed in Neutral buffered formalin (NBF), demineralized in 10% Ethylenedinitrilo tetraacetic acid, disodium salt, dihydrate in cold (EDTA) and embedded in paraffin. Longitudinal 5μm thick sections of the proximal tibial metaphyses were cut with a Supercut 2050 microtome (Reinchart Jung, Germany). Sections were routinely stained with H&E and TRAP prior to examination under the light microscope (Nikon, Japan). Histomorphometry. Following DEXA measurements, the left femurs and tibias were cut in two halves. Following fixation with NBF for 48 hours, the distal halves were dehydrated with a series of graded ethanol and embedded in methyl- methacrylate. Three consecutive cross sections of distal femoral metaphysis and femoral midshaft were cut with a precision bone saw Isomet 1000 (Buehler, Lake
Bluff, 111), and ground to 25 μm in thickness (Pethro-thin, Buehler, Lake Bluff, 111) before being used for microradiography (Faxitron, Faxitron X-Ray Coφ., Buffalo Grove IL). Microradiographs were taken using 1A ultra microradiography plates (IMTEC, Sunnyvale, CA). The distal metaphises were used for static bone histomoφhometry of the cancellous bone while diaphyses wereused for the cortical bone.
Dynamic histomoφhometric measurements were performed on unstained, unde- calcified sections (3 sections per animal) at 10 and/or 20 X magnification using a fluorescence microscope (Nikon, Japan) interfaced with a digitizer. The data was collected using a software package ("Stereology", KSS Computer Engineers, Magna, UT) specifically written for bone histomoφhometry Measurements were made on the entire periosteal surface, on the endocortical surfaces which were free of endocortico-trabecular intersections, and on the entire metaphyseal spongiosa 0.5 mm below the growth plate.
Microradiographs of cortical and cancellous bone were analyzed for static and structural indices using "Image Analysis" software (KSS Computer Engineers). For this, images of the bone were captured with a video-camera CCD72 (DAGE-MTI Inc., Michigan City, MI) connected to the microscope, and from these images static moφhometric indices were determined. These included cortical and marrow areas, average cortical width, trabecular area, and trabecular thickness, number, and separation.
Statistical analyses. Differences among groups were tested for significance in a oneway analysis of variance. When the analysis of variance indicated significant differences among means, the differences were evaluated using Fisher's protected least significant difference (PLSD) method for multiple comparisons. Statistical significance was considered at P<0.05, and results are expressed as the mean ± standard error (SE).
Example 2
Determining effect on plasma calcium levels
Thyroparathyroid (TPTx) Sprague Dawley rats at 250 gms were ordered from Taconic Labs. At the vendor rats are kept on calcium lactate water 2-3% following surgery. Rats are also be shipped with this water. Upon arrival rats were placed on normal water and no food and were kept this way for the remainder of the study. 0.5 ml of blood was collected and tested for serum calcium levels. Any rat with a time 0 calcium of 10 mg/dl or higher was removed from the study. Alzet osmotic minipumps model 2001D (Alza Coφ., Palo Alto, Ca.) that were pre-filled with either
PTH (0.03 ug/ul, 0.25 ug/hr) or vehicle (saline) were implanted subcutaneously. Immediately after surgery rats were dosed orally with 0.5 ml of either vehicle (water) or cerivastatin (3, 1, 0.1, 0.01, or 0.001 mg/kg). Approximately 24 hours post surgery, rats were euthanized with CO2, and bled via cardiac puncture. Serum was tested for calcium levels and analyzed using the students T-test. Tibias excized and placed in chilled 4° C neutral buffered formalin (Hydrol Chemical Company) for three days to allow for fixation. Washed with cold running tap water for one hour then placed in chilled 5% EDTA for decalcification, and paraffin processed. Sections were cut @ 5um and stained for Tartrate-resistant acid phosphatase (Trap) positive cells using a Leukocyte Acid Phosphatase Kit #387-A (Sigma Diagnostics, Inc. St.
Louis, Mo.).
The following examples are useful for preventing or treating conditions that can be ameliorated by concurrently decreasing bone resoφtion and stimulating bone growth, as well as for preventing or treating conditions that can be ameliorated by decreasing plasma calcium levels. Example 3
A tablet is prepared from Cervistatin 25 mg
Cellulose, microcrystaline 200 mg
Colloidal silicon dioxide lO mg
Stearic acid 5.0 mg
The ingredients are mixed and compressed to form tablets.
In alternative formulations, the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
Example 4
A tablet is prepared from
Cerivastatin 10 micro grams Microcrystalline cellulose 116.9 mg
Lactose anhydrate 116.9 mg
Crosmellose sodium 7.5 mg
Magnesium stearate 3.7 mg
The ingredients are combined and blended together and compressed using conventional tableting techniques.
In alternative formulations, the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin. Example 5
A hard gelatine capsule is prepared from
Cerivastatin 5 mg Microcrystalline cellulose 47 mg
Lactose anhydrate 47 mg Magnesium stearate 1 mg
Hard gelatin capsule 1 capsule
The dry ingredients are combined and blended together and encapsulated in the gelatin coating using standard manufacturing techniques.
In alternative formulations, the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
Example 6
An oral suspension formulation is prepared from Cerivastatin 5 mg
Polyvinylpyrrolidone 150 mg
Polyoxyethylene sorbitan monolaurate 2.5 mg
Benzoic acid lO mL
Aqueous sorbitol solution (70%) 5 mL
An oral suspension is prepared by combining the ingredients using standard formulation techniques.
In alternative formulations, the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin. Example 7
An intravenous infusion composition is prepared from Cerivastatin 5 mg
Polyethylene oxide 400 0.2 mg
Sodium cholride 1.8 mg
Purified water to 200 mL
The ingredients are combined using standard formulation techniques, under sterile conditions.
In alternative formulations, the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin.
Example 8
A capsule formula is prepared from Cerivastatin 40 mg
Starch 109 mg
Magnesium steatrate 1 mg
The components are blended, passed through an appropriate mesh sieve, and filled into hard gelatin capsules.
In alternative formulations, the cerivastatin is replaced by an HMG-CoA reductase inhibitor selected from atorvastatin, fluvastatin, lovastatin, mevastatin, pravastatin, and simvastatin. It should be apparent to one of ordinary skill in the art that changes and modifications can be made to this invention without departing from the spirit or scope of the invention as it is set forth herein.

Claims

WHAT IS CLAIMED IS:
1. Use of HMG-CoA reductase inhibitors for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone resoφtion and stimulation of bone growth comprising administering a therapeutically effective amount of an HMG-CoA reductase inhibitor to a mammal in need thereof.
2. A method of preventing or treating osteoporosis, osteoarthritis, Pagent's disease, hypercalcemia, periprosthetic bone loss or osteolysis, osteogenesis imperfecta, and aveolar bone loss associated with peridontal disease comprising administering a therapeutically effective amount of an HMG-CoA, reductase inhibitor to a mammal in need thereof.
3. Use of pharmaceutical compositions for the prevention and for the treatment of abnormal conditions ameliorated by concurrent decrease in bone resoφtion and stimulation of bone growth comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor.
4. Pharmaceutical compositions useful for preventing or for treating osteoporosis, osteoarthritis, Pagent's disease, hypercalcemia, periprosthetic bone loss or osteolysis, osteogenesis imperfecta, and aveolar bone loss associated with peridontal disease, comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor.
5. Use of HMG-CoA reductase inhibitors for the prevention and for the treatment of conditions ameliorated by decreasing plasma calcium levels, comprising administering a therapeutically effective amount of an HMG-CoA reductase inhibitor to a mammal in need thereof.
6. A method of preventing or treating primary hypeφarathyroidism, familial hypeφarathyroid syndrome, familial hypocalcuric hypercalcemia, secondary and tertiary hypeφarathyroidism, humoral hypercalcemia of malignancy, hypoparathyroidism, malignant disease, PTH related protein, ectopic production of 1,25-dihydroxyvitamin D, lytic bone metastases, other factors produced locally or ectopically, endocrine disorders, granulomatous disease, drug induced elevated plasma calcium levels, renal failure, and total parenterai nutrition.
7. Pharmaceutical compositions useful for the prevention and for the treatment of abnormal conditions ameliorated by decreasing plasma calcium levels, comprising a therapeutically effective amount of an HMG-CoA reductase inhibitor.
8. Pharmaceutical compositions according to claims 4 and 7 comprising statins.
9. Pharmaceutical compositions according to claim 7 comprising at least one HMG-CoA reductase inhibitor selected from the group consisting of atorvastatin, cerivastatin, simvastatin, pravastatin, lovastatin; fluvastatin, itavastatin and (+)-(3R,5S)-bis(7-(4-(4-fluorophenyl)-6-isopropyl-2-(N-methyl-N- memanesulfonylammo)pyrimidin-5-yl)-3,5-dihyd_roxy-6(E)-heptenoic acid and/or the pharmaceutically acceptable salts, esters and lactones thereof,.
10. Pharmaceutical compositions according to claim 7 comprising cerivastatin and/or the pharmaceutically acceptable salts, esters and lactones thereof.
11. Use of HMG-CoA reductase inhibitors for the preparation of pharmaceutical compositions for stimulating bone formation.
PCT/EP2000/011466 1999-11-24 2000-11-17 Methods of ameliorating abnormal bone states WO2001037876A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU20007/01A AU2000701A (en) 1999-11-24 2000-11-17 Methods of ameliorating abnormal bone states

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16726799P 1999-11-24 1999-11-24
US60/167,267 1999-11-24

Publications (2)

Publication Number Publication Date
WO2001037876A2 true WO2001037876A2 (en) 2001-05-31
WO2001037876A3 WO2001037876A3 (en) 2002-03-21

Family

ID=22606650

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2000/011466 WO2001037876A2 (en) 1999-11-24 2000-11-17 Methods of ameliorating abnormal bone states

Country Status (2)

Country Link
AU (1) AU2000701A (en)
WO (1) WO2001037876A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001052829A2 (en) * 2000-01-20 2001-07-26 Osteoscreen, Inc. Statin-type bone growth stimulators
JP2002370982A (en) * 2001-06-12 2002-12-24 Kowa Co Osteogenesis promoter
EP1291017A2 (en) * 2001-09-10 2003-03-12 Warner-Lambert Company Use of statins to inhibit formation of osteoclasts
WO2004071402A2 (en) * 2003-02-12 2004-08-26 Lek Pharmaceuticals D.D. STABLE PHARMACEUTICAL DOSAGE FORM COMPRISING HMG-CoA REDUCTASE INHIBITOR
WO2004071403A2 (en) 2003-02-12 2004-08-26 Lek Pharmaceuticals D.D. Coated particles and pharmaceutical dosage forms
WO2006008091A2 (en) 2004-07-16 2006-01-26 Lek Pharmaceuticals D.D. Oxidative degradation products of atorvastatin calcium
US7163945B2 (en) 2004-04-29 2007-01-16 Pharmix Corp. Compositions and treatments for inhibiting kinase and/or HMG-CoA reductase
US7183285B2 (en) 2004-04-29 2007-02-27 Pharmix Corp. Compositions and treatments for inhibiting kinase and/or HMG-CoA reductase
US7199126B2 (en) 2004-04-29 2007-04-03 Pharmix Corporation Compositions and treatments for inhibiting kinase and/or HMG-CoA reductase
US8691796B2 (en) * 2010-10-29 2014-04-08 China Medical University Pharmaceutical composition and method for inhibiting inflammation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998025460A1 (en) * 1996-12-13 1998-06-18 Zymogenetics, Inc. Compositions and methods for stimulating bone growth
WO1999045923A1 (en) * 1998-03-13 1999-09-16 Merck & Co., Inc. Methods of inhibiting bone resorption

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998025460A1 (en) * 1996-12-13 1998-06-18 Zymogenetics, Inc. Compositions and methods for stimulating bone growth
WO1999045923A1 (en) * 1998-03-13 1999-09-16 Merck & Co., Inc. Methods of inhibiting bone resorption

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6376476B1 (en) 1996-12-13 2002-04-23 Zymogenetics Corporation Isoprenoid pathway inhibitors for stimulating bone growth
US7101907B2 (en) 1996-12-13 2006-09-05 Zymogenetics Corporation Topical administration of statins for treatment of bone disorders
WO2001052829A2 (en) * 2000-01-20 2001-07-26 Osteoscreen, Inc. Statin-type bone growth stimulators
WO2001052829A3 (en) * 2000-01-20 2002-05-02 Osteoscreen Inc Statin-type bone growth stimulators
JP2002370982A (en) * 2001-06-12 2002-12-24 Kowa Co Osteogenesis promoter
EP1291017A2 (en) * 2001-09-10 2003-03-12 Warner-Lambert Company Use of statins to inhibit formation of osteoclasts
EP1291017A3 (en) * 2001-09-10 2003-07-02 Warner-Lambert Company Use of statins to inhibit formation of osteoclasts
WO2004071402A3 (en) * 2003-02-12 2004-10-21 Lek Pharmaceuticals STABLE PHARMACEUTICAL DOSAGE FORM COMPRISING HMG-CoA REDUCTASE INHIBITOR
WO2004071403A2 (en) 2003-02-12 2004-08-26 Lek Pharmaceuticals D.D. Coated particles and pharmaceutical dosage forms
WO2004071402A2 (en) * 2003-02-12 2004-08-26 Lek Pharmaceuticals D.D. STABLE PHARMACEUTICAL DOSAGE FORM COMPRISING HMG-CoA REDUCTASE INHIBITOR
US7163945B2 (en) 2004-04-29 2007-01-16 Pharmix Corp. Compositions and treatments for inhibiting kinase and/or HMG-CoA reductase
US7183285B2 (en) 2004-04-29 2007-02-27 Pharmix Corp. Compositions and treatments for inhibiting kinase and/or HMG-CoA reductase
US7199126B2 (en) 2004-04-29 2007-04-03 Pharmix Corporation Compositions and treatments for inhibiting kinase and/or HMG-CoA reductase
WO2006008091A2 (en) 2004-07-16 2006-01-26 Lek Pharmaceuticals D.D. Oxidative degradation products of atorvastatin calcium
US8044086B2 (en) 2004-07-16 2011-10-25 Lek Pharmaceuticals D.D. Oxidative degradation products of atorvastatin calcium
US8691796B2 (en) * 2010-10-29 2014-04-08 China Medical University Pharmaceutical composition and method for inhibiting inflammation

Also Published As

Publication number Publication date
WO2001037876A3 (en) 2002-03-21
AU2000701A (en) 2001-06-04

Similar Documents

Publication Publication Date Title
US5691375A (en) Method for preventing or reducing risk of onset of cardiovascular events employing an HMG CoA reductase inhibitor
US5622985A (en) Method for preventing a second heart attack employing an HMG CoA reductase inhibitor
US5593971A (en) Method for preventing onset of hypertension employing a cholesterol lowering drug
US20060035867A1 (en) Methods of treating cancer and the pain associated therewith using endothelin antagonists
AU2002210521A1 (en) Use of statins (HMG-CoA reductase inhibitors) for the preparation of medicament as a novel type of immunomodulator, immunosuppressor and anti-inflammatory agent
CZ301583B6 (en) Medicament containing rosuvastatin intended for treating heterozygous familial hypercholesterolemia
JP2005523255A (en) Use of benzothiazepine with activity as an inhibitor of ileal bile acid transport to reduce cholesterololemia
WO2005102381A1 (en) Bone densifying agent characterized by use of cathepsin k inhibitor with pth
WO2001037876A2 (en) Methods of ameliorating abnormal bone states
JP2002506030A (en) How to inhibit bone resorption
JP4901218B2 (en) Concomitant medication
JP2007063288A (en) Method of treating cancer and pain associated therewith using endothelin antagonist
MXPA01004683A (en) Methods and compositions to lower plasma cholesterol levels.
SK14102002A3 (en) New combination of a betablocker and a cholesterol-lowering agent
JPH04243839A (en) Drug containing hmg coa reductase inhibitor
AU2001281134A1 (en) Methods of treating cancer and the pain associated therewith using endothelin antagonists
EP1621210B1 (en) Adiponectin production enhancer
US20020159974A1 (en) Treatment of multiple sclerosis with statins (HMG-CoA reductase inhibitors)
US20030060502A1 (en) Treatment of diabetes with statins (HMG-COa reductase inhibitors)
KR20070011288A (en) Abca1 stabilizer
US20070004681A1 (en) Methods for treating arthritic conditions
WO2003031597A2 (en) Methods of inhibiting osteoclast activity
JP2003321365A (en) Hyperlipemia treating agent
AU2003206016A1 (en) Statins (HMG-CoA reductase inhibitors) as a novel type of immunomodulator, immunsuppressor and anti-inflammatory agent

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase