EP1267641A1 - Supplements nutritionnels pour stimuler la croissance osseuse - Google Patents

Supplements nutritionnels pour stimuler la croissance osseuse

Info

Publication number
EP1267641A1
EP1267641A1 EP01927431A EP01927431A EP1267641A1 EP 1267641 A1 EP1267641 A1 EP 1267641A1 EP 01927431 A EP01927431 A EP 01927431A EP 01927431 A EP01927431 A EP 01927431A EP 1267641 A1 EP1267641 A1 EP 1267641A1
Authority
EP
European Patent Office
Prior art keywords
bone
compounds
compound
bone growth
substituted
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP01927431A
Other languages
German (de)
English (en)
Inventor
Gregory R. Mundy
I. Ross Garrett
Gloria E. Gutierrez
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
OsteoScreen Inc
Original Assignee
OsteoScreen Inc
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
Priority claimed from US09/541,943 external-priority patent/US6410521B1/en
Application filed by OsteoScreen Inc filed Critical OsteoScreen Inc
Priority claimed from PCT/US2001/040421 external-priority patent/WO2001074180A1/fr
Publication of EP1267641A1 publication Critical patent/EP1267641A1/fr
Withdrawn legal-status Critical Current

Links

Definitions

  • the invention relates to foodstuffs and nutritional supplements which contain active ingredients that stimulate bone growth in humans and other vertebrates. More specifically, the invention concerns methods to enhance bone growth by supplementing the diet of humans and domesticated animals with edible materials that contain statins or other bone growth enhancers and by formulating such enhancers in edible form.
  • Bone is subject to constant breakdown and resynthesis in a complex process mediated by osteoblasts, which produce new bone, and osteoclasts, which destroy bone.
  • osteoporosis there is a plethora of conditions which are characterized by the need to enhance bone formation. Perhaps the most obvious is the case of bone fractures, where it would be desirable to stimulate bone growth and to hasten and complete bone repair. Agents that enhance bone formation would also be useful in facial reconstruction procedures. Other bone deficit conditions include bone segmental defects, periodontal disease, metastatic bone disease, osteolytic bone disease and conditions where connective tissue repair would be beneficial, such as healing or regeneration of cartilage defects or injury. Also of great significance is the chronic condition of osteoporosis, including age-related osteoporosis and osteoporosis associated with post-menopausal hormone status.
  • BMPs bone morphogenic proteins
  • the BMPs are novel factors in the extended transforming growth factor ⁇ superfamily. Recombinant BMP-2 and BMP-4 can induce new bone formation when they are injected locally into the subcutaneous tissues of rats (Wozney J., Molec ReprodDev (1992) 32:160-67).
  • osteoblasts The cells which are responsible for forming bone are osteoblasts. As osteoblasts differentiate from precursors to mature bone-forming cells, they express and secrete a number of enzymes and structural proteins of the bone matrix, including Type-1 collagen, osteocalcin, osteopontin and alkaline phosphatase (Stein G., et al, Curr Opin Cell Biol (1990) 2:1018-27; Harris S., et al. (1994), supra). They also synthesize a number of growth regulatory peptides which are stored in the bone matrix, and are presumably responsible for normal bone formation. These growth regulatory peptides include the
  • BMPs (Harris S., et al. (1994), supra).
  • BMPs 1, 2, 3, 4, and 6 are expressed by cultured cells prior to the formation of mineralized bone nodules (Harris S., et al. (1994), supra).
  • the BMPs are expressed by cultured osteoblasts as they proliferate and differentiate.
  • BMPs are potent stimulators of bone formation in vitro and in vivo, there are disadvantages to their use as therapeutic agents to enhance bone healing.
  • Receptors for the bone morphogenetic proteins have been identified in many tissues, and the BMPs themselves are expressed in a large variety of tissues in specific temporal and spatial patterns. This suggests that BMPs may have effects on many tissues in addition to bone, potentially limiting their usefulness as therapeutic agents when administered systemically.
  • BMPs since they are peptides, they would have to be administered by injection.
  • Small molecules that are useful in treating bone disorders in vertebrates are of the general formula Ar'-L-Ar 2 wherein Ar 1 and Ar 2 are aromatic moieties and L is a linker that separates them by a specified distance. These are disclosed in PCT application WO98/ 17267 published 30 April 1998. These compounds were assessed for usefulness in treating bone disorders by their ability to enhance the production of a reporter protein when the nucleotide sequence encoding the reporter protein is operably linked to the promoter for
  • HMG-CoA reductase is the principal rate limiting enzyme involved in cellular cholesterol biosynthesis. The pathway is also responsible for the production of dolichol, ubiquinones, isopentenyl adenine and farnesol. HMG-CoA reductase converts 3-hydroxy-3-methyl-glutaryl CoA (HMG-CoA) to mevalonate.
  • Patent 5,280,040 discloses compounds described as useful in the treatment of osteoporosis. These compounds putatively achieve this result by preventing bone resorption.
  • the bisphosphonates ⁇ the methylene bisphosphonic acids. These compounds are comprised of two phosphonic acid residues coupled through a methylene linkage. Typical representatives include the clodronates which are simple compounds wherein the phosphonic acid residues are coupled through dichloromethylene.
  • Other representative bisphosphonates include ibandronates, the risedronates, alandronates and pamidronates. These compounds have been shown to inhibit the resorption of bone, presumably by effecting apoptosis of osteoclasts.
  • the present invention describes means for administering statins and other classes of compounds that enhance bone formation by including them in nutritional supplements, or by administering edible materials that naturally contain these compounds.
  • the invention is directed to an optimum protocol for administering compounds that enhance bone formation.
  • the invention is directed to a method to enhance bone growth in humans or other vertebrate animals by including in their diets foodstuffs or food supplements that contain effective amounts of bone growth enhancing compounds.
  • These compounds can be the small molecule bone enhancing compounds of the formula
  • statins are of the formula: wherein X in each of formulas (1), (2) and (3) represents a substituted or unsubstituted alkylene, alkenylene, or alkynylene linker of 2-6C optionally containing one heteroatom which is O, N or S;
  • Y represents one or more carbocyclic or heterocyclic rings; when two or more rings are present in Y, they may optionally be fused; and
  • R' represents a cation, H or a substituted or unsubstituted alkyl group of 1-6C.
  • the invention is directed to methods to treat bone disorders by directly stimulating bone formation stimulating bone formation by including in the diet of a subject in need of such stimulation, a food or food supplement which contains a bone enhancing compound, especially a statin compound.
  • Oral administration of the compounds of the invention, in particular the statins enhances plasma concentration by avoiding hepatic first pass effects. Thus, their effects on bone and non-hepatic tissues are enhanced.
  • the invention relates to administering the compounds that stimulate bone growth on an intermittent schedule in contrast to continuous administration.
  • Figures 1A and IB show the structures and activity of statins in the screening assay of Example 1.
  • the ultimate goal of the methods and compositions of the invention is to treat or ameliorate bone disorders in vertebrate subjects, particularly mammals, and more particularly humans.
  • treat or “treatment” include a postponement of development of bone deficit symptoms and/or a reduction in the severity of such symptoms that will or are expected to develop.
  • the terms further include ameliorating existing bone or cartilage deficit symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, preventing or reversing bone resorption and/or encouraging bone growth.
  • the terms denote that a beneficial result has been conferred on a vertebrate subject with a cartilage, bone or skeletal deficit, or with the potential to develop such deficit.
  • bone deficit is meant an imbalance in the ratio of bone formation to bone reso ⁇ tion, such that, if unmodified, the subject will exhibit less bone than desirable, or the subject's bones will be less intact and coherent than desired. Bone deficit may also result from fracture, from surgical intervention or from dental or periodontal disease.
  • cartilage defect is meant damaged cartilage, less cartilage than desired, or cartilage that is less intact and coherent than desired.
  • Body disorders includes both bone deficits and cartilage defects.
  • Representative uses of the compounds of the present invention include: repair of bone defects and deficiencies, such as those occurring in closed, open and non-union fractures; prophylactic use in closed and open fracture reduction; promotion of bone healing in plastic surgery; stimulation of bone ingrowth into non-cemented prosthetic joints and dental implants; elevation of peak bone mass in pre-menopausal women; treatment of growth deficiencies; treatment of periodontal disease and defects, and other tooth repair processes; increase in bone formation during distraction osteogenesis; and treatment of other skeletal disorders, such as age-related osteoporosis, post-menopausal osteoporosis, glucocorticoid-induced osteoporosis or disuse osteoporosis and arthritis, or any condition that benefits from stimulation of bone formation.
  • bone defects and deficiencies such as those occurring in closed, open and non-union fractures
  • prophylactic use in closed and open fracture reduction promotion of bone healing in plastic surgery
  • stimulation of bone ingrowth into non-cemented prosthetic joints and dental implants elevation of peak bone mass in pre-menopausal women
  • the compounds of the present invention can also be useful in repair of congenital, trauma-induced or surgical resection of bone (for instance, for cancer treatment), and in cosmetic surgery. Further, the compounds of the present invention can be used for limiting or treating cartilage defects or disorders, and may be useful in wound healing or tissue repair.
  • the compositions and the methods of the invention effect the improvement in the foregoing conditions by virtue of their ability to enhance bone formation.
  • the compounds are administered to the appropriate subject as part of the subject's ordinary diet. If the subject is a human, the bone growth enhancing compounds may be artificially added to foodstuffs that are already a part of the individual's diet.
  • a particularly useful approach is to add these compounds to fruit juices or vegetable juices, in particular fruit juices, and most favorably grapefruit juice, which is believed to exert a further beneficial effect.
  • these compounds can suitably be added to liquid components of the diet that are eaten cold, such as the fruit juices mentioned above, or even reconstituted beverages such as iced tea.
  • Those compounds which are active ingredients but which are relatively insoluble in water may also be added to emulsions or lipid-based components such as salad dressings and salad oils.
  • a single compound such as a statin which enhances bone growth, or an active compound in combination with additional agents which stimulate such growth, and, if desired, in combination with other drugs, is formulated into an edible form.
  • the edible compositions can be in the form of tablets or capsules and supplied to the diet as food supplements, as are vitamins and other herbal extracts. Such supplements can also be formulated as syrups or powders to be dissolved in fluids.
  • the cholestin illustrated below may conveniently be formulated in this manner.
  • the tablets or capsules may be swallowed whole or may be designed to be chewable, in which case flavoring elements are desirable.
  • the active compound or compounds are integrated into a foodstuff that becomes a part of the subject's diet.
  • concentrates of fruit juices supplemented with the active compound or compounds can be rehydrated and supplied along with meals or as midmorning or mid-afternoon refreshers.
  • the compounds can be formulated as powders which can be sprinkled onto cold cereal or forms of cold cereal can be produced which have the compounds coated on the granules or flakes that make up the food.
  • the compound or compounds can be integrated into spreads such as cream cheese, butter, margarine and the like and used in contexts where these spreads would be applicable.
  • the compound or compounds are also suitable for blending with emulsions such as salad dressing and mayonnaise or water-based condiments such as catsup, mustard and cocktail sauce.
  • the preparation of the desired foodstuff is performed at room temperature or less to preserve the chemical integrity of the compounds and of the foodstuff.
  • such vehicles as sour cream, cottage cheese and yogurt are convenient.
  • the compounds can also be frozen into ices, sorbets and ice creams. Milkshakes may be a particularly attractive means for administration in view of the solubility characteristics of the compounds. It will be well within the skill of the art to devise attractive recipes for the inclusion of the compounds of the invention and to blend them into foodstuffs in such a manner that patient compliance is facilitated.
  • the bone enhancing compounds can also be provided in the form of dietary supplements where, especially, the active ingredient occurs naturally in the supplement.
  • One particularly preferred supplement is Red Yeast Rice which is known to contain satisfactory concentrations of lovastatin.
  • the lovastatin may be formulated in a cream, for example, by sonicating with whipping cream for a suitable oral dosage.
  • Red Yeast Rice refers to a strain of yeast that grows in rice; it is most familiar as the extract used in the flavoring of Peking duck.
  • the yeast can be grown on a rice substrate and then freeze-dried and pulverized and thus made into a suitable supplement containing lovastatin and related metabolites.
  • the use of grapefruit juice as a vehicle is particularly advantageous, as components of grapefruit juice are known to block the clearance enzyme P450 in serum and thus prevent the clearance of the drug.
  • Another particularly convenient formulation is in the form of a milkshake, taking advantage of the emulsified nature of the liquid to accommodate the solubility characteristics of the compounds of the invention.
  • the active compounds can be formulated into the feed, for example, by coating solid foods with an emulsion of the active ingredient. Enhancement of bone growth may be desirable in such domestic animals as pigs, cows, sheep, domestic animals such as dogs and cats, and even chickens.
  • an aspect of the invention is the administration of the active compounds orally as part of the subject's diet
  • the compounds can, of course, be administered by more traditional routes.
  • Formulations for such routes may include one or more excipients, preservatives, solubilizers, buffering agents, albumin to prevent protein loss on vial surfaces, lubricants, fillers, stabilizers, etc.
  • Methods of formulation are well known in the art and are disclosed, for example, in Remington's Pharmaceutical Sciences, latest edition, Mack Publishing Co., Easton PA, which is incorporated herein by reference.
  • compositions for use within the present invention can be in the form of sterile, non-pyrogenic liquid solutions or suspensions, coated capsules, suppositories, lyophilized powders, transdermal patches or other forms known in the art.
  • Local administration may be by injection at the site of injury or defect, or by insertion or attachment of a solid carrier at the site, or by direct, topical application of a viscous liquid, or the like.
  • the delivery vehicle preferably provides a matrix for the growing bone or cartilage, and more preferably is a vehicle that can be absorbed by the subject without adverse effects.
  • an "effective amount" of a composition is that amount which produces a statistically significant effect.
  • an "effective amount" for therapeutic uses is the amount of the composition comprising an active compound herein required to provide a clinically significant increase in healing rates in fracture repair; reversal of bone loss in osteoporosis; reversal of cartilage defects or disorders; prevention or delay of onset of osteoporosis; stimulation and/or augmentation of bone formation in fracture non-unions and distraction osteogenesis; increase and/or acceleration of bone growth into prosthetic devices; and repair of dental defects.
  • Such effective amounts will be determined using routine optimization techniques and are dependent on the particular condition to be treated, the condition of the patient, the route of administration, the formulation, and the judgment of the practitioner and other factors evident to those skilled in the art.
  • the dosage required for the compounds of the invention is manifested as a statistically significant difference in bone mass between treatment and control groups.
  • This difference in bone mass may be seen, for example, as a 5-20% or more increase in bone mass in the treatment group.
  • Other measurements of clinically significant increases in healing may include, for example, tests for breaking strength and tension, breaking strength and torsion, 4-point bending, increased connectivity in bone biopsies and other biomechamcal tests well known to those skilled in the art.
  • General guidance for treatment regimens is obtained from experiments carried out in animal models of the disease of interest.
  • the dosage of the compounds of the invention will vary according to the extent and severity of the need for treatment, the activity of the administered compound, the general health of the subject, and other considerations well known to the skilled artisan. Generally, they can be administered to a typical human on a daily basis as an oral dose of about 0.1 mg/kg-1000 mg/kg, and more preferably from about 1 mg/kg to about 200 mg/kg. While dietary oral administration may be preferable in most instances (for reasons of ease, patient acceptability, and the like), alternative methods of administration may be appropriate for selected compounds and selected defects or diseases.
  • the administration of the compounds is intermittent rather than continuous.
  • preferred protocols may include administering the compounds once every three days, once every week, optionally in varying amounts at each time of administration. Such forms of administration are preferable to daily administration or consistent several times a day dosage.
  • intermittent administration is meant that the compounds or compositions containing them are provided to the subject only sporadically with substantial periods between dosages or until the results are assessed.
  • Preferred “intermittent" regimens would comprise administering the compounds or compositions every other day, every two days, every five days, or on days unevenly spaced but separated at least by a one day interval between days on which a dosage occurred.
  • intermittent dosage regime is meant either a single bolus dose or multiple dosages that comprise only a small proportion of the timeframe over which the subject is being treated. The timeframe over which treatment is administered is measured by the time between initial administration and assessment of the results. Preferred percentages of the time period occupied by dosage administration are 50% of the time, more preferably 25% of the time, and most preferably 10% of the time.
  • statins Preferred compounds useful in treating bone disorders by enhancing bone growth are generally classified as "statins.”
  • statins The structures of a number of statins, currently used as drugs to inhibit cholesterol formation, are shown in Figure 1. These include cerivastatin, marketed under the name Baycol ® by Bayer (See U.S. patents 5,006,530 and 5,177,080), lovastatin, marketed under the name Mevacor ® by Merck (See U.S. Patent 4,963,538), and simvastatin, marketed under the name of Zocar ® , pravastatin, marketed under the brand name Pravachol ® , atorvastatin, marketed under the name Lipotor ® by Warner-Lambert (See U.S.
  • the compounds that are useful for administration in the methods of the invention can be verified as having bone growth enhancing activity in a number of assays as described for convenience below.
  • the first of these is a high throughput assay that screens for compounds that stimulate the promoter of BMP -2.
  • Other in vitro and in vivo assays more directly assess the ability of the compound to enhance bone growth.
  • This assay is also described as a portion of a study of immortalized murine osteoblasts (derived from a mouse expressing a transgene composed of a BMP-2 promoter driving expression of T-antigen) in Ghosh-Choudhery, N., et al, Endocrinology (1996) 137:331-39.
  • the immortalized cells were stably transfected with a plasmid containing a luciferase reporter gene driven by a mouse BMP-2 promoter (-2736/114 bp), and responded in a dose-dependent manner to recombinant human BMP-2.
  • the assay utilizes cells transformed permanently or transiently with constructs in which the promoter of a bone mo ⁇ hogenetic protein, specifically BMP-2 or BMP -4, is coupled to a reporter gene, typically luciferase. These transformed cells are then evaluated for the production of the reporter gene product; compounds that activate the BMP promoter will drive production of the reporter protein, which can be readily assayed. Many thousands of compounds have been subjected to this rapid screening technique, and only a very small percentage are able to elicit a level of expression of reporter gene 5-fold greater than that produced by vehicle.
  • a bone mo ⁇ hogenetic protein specifically BMP-2 or BMP -4
  • BMP promoter-active compounds can be examined in a variety of other assays that test specificity and toxicity.
  • non-BMP promoters or response elements can be linked to a reporter gene and inserted into an appropriate host cell. Cytotoxicity can be determined by visual or microscopic examination of BMP promoter- and/or non-BMP promoter-reporter gene-containing cells, for instance. Alternatively, nucleic acid and/or protein synthesis by the cells can be monitored.
  • tissues may be removed and examined visually or microscopically, and optionally examined in conjunction with dyes or stains that facilitate histologic examination. In assessing in vivo assay results, it may also be useful to examine biodistribution of the test compound, using conventional medicinal chemistry/animal model techniques.
  • Neonatal Mouse Calvaria Assay An assay for bone reso ⁇ tion or bone formation is similar to that described by
  • the medium used when the assay is conducted to assess bone formation is Fitton and Jackson Modified BGJ Medium (Sigma) supplemented with 6 ⁇ g/ml insulin, 6 ⁇ g/ml transferrin, 6 ng/ml selenous acid, calcium and phosphate concentrations of 1.25 and 3.0 mM, respectively, and ascorbic acid to a concentration of 100 ⁇ g/ml is added every two days.
  • the incubation is conducted at 37°C in a humidified atmosphere of 5% CO 2 and 95% air for 96 hours.
  • the bones are removed from the incubation media and fixed in 10% buffered formalin for 24-48 hours, decalcified in 14% EDTA for 1 week, processed through graded alcohols; and embedded in paraffin wax.
  • Three ⁇ m sections of the calvaria are prepared. Representative sections are selected for histomo ⁇ hometric assessment of bone formation or bone reso ⁇ tion. Bone changes are measured on sections cut 200 ⁇ m apart. Osteoblasts and osteoclasts are identified by their distinctive mo ⁇ hology. Other auxiliary assays can be used as controls to determine non-BMP promoter-mediated effects of test compounds.
  • mitogenic activity can be measured using screening assays featuring a serum-response element (SRE) as a promoter and a luciferase reporter gene. More specifically, these screening assays can detect signaling through SRE-mediated pathways, such as the protein kinase C pathway. For instance, an osteoblast activator SRE-luciferase screen and an insulin mimetic SRE-luciferase screen are useful for this pu ⁇ ose. Similarly, test compound stimulation of cAMP response element (CRE)-mediated pathways can also be assayed.
  • SRE serum-response element
  • cells transfected with receptors for PTH and calcitonin can be used in CRE- luciferase screens to detect elevated cAMP levels.
  • the BMP promoter specificity of a test compound can be examined through use of these types of auxiliary assays.
  • mice Male ICR Swiss white mice, aged 4-6 weeks and weighing 13-26 gm, are employed, using 4-5 mice per group.
  • the calvarial bone growth assay is performed as described in PCT application WO 95/24211, inco ⁇ orated by reference. Briefly, the test compound or appropriate control vehicle is injected into the subcutaneous tissue over the right calvaria of normal mice.
  • the control vehicle is the vehicle in which the compound was solubilized, and is PBS containing 5% DMSO or is PBS containing Tween (2 ⁇ l/10 ml). The animals are sacrificed on day 14 and bone growth measured by histomo ⁇ hometry.
  • Bone samples for quantitation are cleaned from adjacent tissues and fixed in 10% buffered formalin for 24-48 hours, decalcified in 14% EDTA for 1-3 weeks, processed through graded alcohols; and embedded in paraffin wax. Three to five ⁇ m sections of the calvaria are prepared, and representative sections are selected for histomo ⁇ hometric assessment of the effects on bone formation and bone reso ⁇ tion.
  • Sections are measured by using a camera lucida attachment to trace directly the microscopic image onto a digitizing plate. Bone changes are measured on sections cut 200 ⁇ m apart, over 4 adjacent lxl mm fields on both the injected and noninjected sides of the calvaria. New bone is identified by its characteristic woven structure, and osteoclasts and osteoblasts are identified by their distinctive mo ⁇ hology. Histomo ⁇ hometry software
  • Lead compounds can be further tested in intact animals using an in vivo, dosing assay.
  • Prototypical dosing may be accomplished by subcutaneous, intraperitoneal or oral administration, and may be performed by injection, sustained release or other delivery techniques.
  • the time period for administration of test compound may vary (for instance,
  • test compound, positive control compound, PBS, or vehicle alone is administered subcutaneously once per day for 35 days. All animals are injected with calcein nine days and two days before sacrifice (two injections of calcein administered each designated day). Weekly body weights are determined.
  • Lead compounds can also be tested in acute ovariectomized animals (prevention model) using an in vivo dosing assay.
  • Such assays may also include an estrogen-treated group as a control.
  • An exemplary subcutaneous dosing assay is performed as follows: In a typical study, 80 three-month-old female Sprague-Dawley rats are weight-matched and divided into eight groups, with ten animals in each group. This includes a baseline control group of animals sacrificed at the initiation of the study; three control groups (sham ovariectomized (sham ONX) + vehicle only; ovariectomized (ONX) + vehicle only; PBS-treated ONX); and a control ONX group that is administered a compound known to promote bone growth. Three dosage levels of the compound to be tested are administered to the remaining three groups of ONX animals.
  • test compound positive control compound, PBS, or vehicle alone is administered orally or subcutaneously once per day for 35 days.
  • test compound can be formulated in implantable pellets that are implanted for 35 days, or may be administered orally, such as by gastric gavage. All animals, including sham ONX/vehicle and ONX/vehicle groups, are injected intraperitoneally with calcein nine days and two days before sacrifice (two injections of calcein administered each designated day, to ensure proper labeling of newly formed bone). Weekly body weights are determined. At the end of the 35-day cycle, the animals' blood and tissues are processed as described above.
  • Lead compounds may also be tested in chronic ONX animals (treatment model).
  • An exemplary protocol for treatment of established bone loss in ovariectomized animals that can be used to assess efficacy of anabolic agents may be performed as follows. Briefly, 80 to 100 six month old female, Sprague-Dawley rats are subjected to sham surgery (sham ONX) or ovariectomy (ONX) at time 0, and 10 rats are sacrificed to serve as baseline controls. Body weights are recorded weekly during the experiment. After approximately 6 weeks (42 days) or more of bone depletion, 10 sham ONX and 10 ONX rats are randomly selected for sacrifice as depletion period controls. Of the remaining animals, 10 sham ONX and 10 ONX rats are used as placebo-treated controls.
  • ONX animals are treated with 3 to 5 doses of test drug for a period of 5 weeks (35 days).
  • a group of ONX rats can be treated with an agent such as PTH, a known anabolic agent in this model (Kimmel, et al, Endocrinology (1993)
  • the femurs, tibiae and lumbar vertebrae 1 to 4 are excised and collected.
  • the proximal left and right tibiae are used for pQCT measurements, cancellous bone mineral density (BMD) (gravimetric determination), and histology, while the midshaft of each tibiae is subjected to cortical BMD or histology.
  • BMD cancellous bone mineral density
  • the femurs are prepared for pQCT scanning of the midshaft prior to biomechamcal testing.
  • LN LN2 are processed for BMD (pQCT may also be performed); LN3 are prepared for undecalcified bone histology; and LN4 are processed for mechanical testing.
  • statin compounds useful in the methods and compositions of the invention are typically of the formula:
  • X in each of formulas (1), (2) and (3) represents a substituted or unsubstituted alkylene, alkenylene, or alkynylene linker of 2-6C optionally containing one heteroatom which is O, N or S;
  • Y represents one or more carbocyclic or heterocyclic rings; when Y comprises two or more rings, they may optionally be fused; and
  • R' represents a cation, H or a substituted or unsubstituted alkyl group of 1-6C. It is understood that if R' represents a cation with multiple positive charges, the appropriate number of anions is coupled with it.
  • Formulas (1) and (2) are, respectively, the unhydrolyzed and hydro lyzed forms of the statin compounds. Preferred substituents on X (or on R' when R' is alkyl) are hydroxy, alkoxy, phenyl, amino and alkyl- or dialkylamino.
  • the compounds useful in the invention contain at least one and generally several chiral centers. Compounds useful in the invention include mixtures of the various stereoisomers and the stereoisomeric forms of the compounds individually. Preferred stereoisomers with respect to the compound of formula (1) are of the formula:
  • Preferred embodiments of Y comprise ring systems such as naphthyl, polyhydro-naphthyl, monohydro- or dihydrophenyl, quinolyl, pyridyl, quinazolyl, pteridyl, pyrolyl, oxazoyl and the like and the reduced or partially reduced forms thereof.
  • the ring system may contain ⁇ -bonds; wherein R , ⁇ i ⁇ s substituted or unsubstituted alkyl; each R 2 is independently a noninterfering substituent; R 3 is H, hydroxy, or alkoxy (1-6C); each m is independently an integer of 0-6, wherein each R 2 may reside in any of positions 2-7; and p is 0 or 1, depending on the position of any ⁇ -bonds.
  • Particularly preferred embodiments include those of formulas (4a)-(4f) wherein the upper limit of n is adjusted according to the valence requirements appropriate for the particular ring system.
  • R 1 may be substituted alkyl, wherein the substituents may include hydroxy, aikoxy, alkylthiol, phenyl, phenylalkyl, and halo, unsubstituted alkyl is preferred.
  • R 1 are alkyl of 1-6C, including propyl, see-butyl, t-butyl, n-butyl, isobutyl, pentyl, isopentyl, 1-methylbutyl, and 2-methylbutyl. Particularly preferred are propyl and sec-butyl.
  • each n is independently 1 or 2 and preferred positions for substitution are positions 2 and 6 (see formula (4)).
  • Particularly preferred embodiments of R 2 are OH, H, and lower alkyl, in particular CH 3 .
  • Y is 4(a) or 4(b), and especially embodiments having the substitution pattern indicated in formulas 4(g) and 4(h) below.
  • the compounds of the invention may be supplied as individual stereoisomers or as mixtures of stereoisomers.
  • Preferred stereoisomers are those of the formulas (4g) and (4h) as typical and appropriate for those represented by the formulas (4a)-(4f).
  • Preferred embodiments of R 1 in these preferred forms are propyl, sec-butyl, and 2-methyl-but-2-yl.
  • Additional preferred embodiments of Y are:
  • each K comprises a substituted or unsubstituted aromatic or nonaromatic carbocyclic or heterocyclic ring system which may optionally be spaced from the linkage position shown in formula (7) by a linker of 1-2C, including -CHOH-, -CO-, and -CHNH 2 -, for example.
  • Aromatic ring systems are preferred. Particularly preferred are compounds of formula (7), either as shown or wherein Z is contained in a 6-membered, rather than a 5-membered aromatic ring.
  • R is a cyclic alkyl substituent.
  • R 5 is H or linear, branched, cyclic substituted or unsubstituted alkyl, wherein substituents are preferably hydroxy, alkoxy, phenyl, amino and alkyl- or dialkylamino.
  • R 5 is alkyl, it is unsubstituted.
  • Substituents on the aromatic ring systems or nonaromatic ring systems included in Y can be any noninterfering substituents.
  • the non-interfering substituents can be of wide variety.
  • substituents that do not interfere with the beneficial effect of the compounds of the invention on bone formation in treated subjects include alkyl (1-6C, preferably lower alkyl 1-4C), including straight, branched or cyclic forms thereof, alkenyl (2-6C, preferably 2-4C), alkynyl (2-6C, preferably 2-4C), all of which can be straight or branched chains and may contain further substituents; halogens, including F, Cl, Br and I; silyloxy, OR, SR, NR 2 , OOCR, COOR, NCOR, NCOOR, and benzoyl, CF 3 , OCF 3 , SCF 3 , N(CF 3 ) 2 , CN, SO, SO 2 R and SO 3 R wherein R is alkyl (1-6C) or is
  • substituents may form a ring.
  • rings not fused to the aromatic or nonaromatic rings comprising Y may be included as substituents. These rings may be aromatic and may be substituted or unsubstituted.
  • 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 antihypercholesterolemic agents. Typical among these is lovastatin, marketed by Merck as Mevacor ® . The synthesis of lovastatin and various analogs thereof is set forth in U.S. Patent No. 4,963,538, inco ⁇ orated herein by reference. In addition, methods for synthesis of lovastatin and analogous compounds such as compactin (mevastatin), simvastatin, and pravastatin are set forth in U.S. Patent Nos.
  • compositions of the invention may also include the bisphosphonates and their analogs.
  • the bisphosphonates are of the formula
  • Typical salts are those of the inorganic ions, such as sodium ion, potassium ion, calcium ion, magnesium ion and the like; any pharmaceutically acceptable cation may be used.
  • Typical esters are the ethyl, methyl, isobutyl, ethylene glycol, and other typical pharmaceutically acceptable esters; typical amides are the unsubstituted -NH 2 amides as well as the alkyl and dialkyl amides.
  • Embodiments of R 10 include halo, OR, SR, NR 2 , where R is H or alkyl (1-6C) or alkyl or arylalkyl with optional substitutions. Particularly preferred are the amino-substituted alkyl embodiments. Typically, both R 10 are not identical, although in some embodiments, such as clodronate, both R 10 are halo. Particularly preferred compounds among the bisphosphonates are risedronate, alandronate, pamidronate, clodronate and in particular ibandronate. These compounds are particularly useful in combination with the statins.
  • compositions may also contain, as active ingredients, compounds of the formula Ar-L-Ar wherein Ar represents an aryl substituent and L represents a linker.
  • active ingredients may also be supplemented by additional pharmaceuticals complementing the activity of enhancing bone growth, such as the use of estrogens to inhibit bone reso ⁇ tion.
  • the active ingredient compounds are used alone or in mixtures of two or more active compounds in the presence of suitable accompanying excipients which permit oral administration and, preferably, inco ⁇ orate the materials into the diet of the subject.
  • suitable accompanying excipients which permit oral administration and, preferably, inco ⁇ orate the materials into the diet of the subject.
  • vehicle foodstuff or food supplement base will depend on the nature of the active ingredient, and in particular its solubility characteristics. As noted above, it may be necessary to utilize oils or non-aqueous, orally acceptable solvents in order to bring some of the active ingredients into solution, or emulsions or other preparations, such as liposomal preparations can be used.
  • the liquid preparations can be directly ingested or can be coated onto or otherwise inco ⁇ orated into solid food. Suitable approaches are exemplified below. The following examples are intended to illustrate but not to limit the invention.
  • the 2T3-BMP-2-LUC cells a stably transformed osteoblast cell line described in Ghosh-Choudhury, et al, Endocrinology (1996) 137:331-39, referenced above.
  • the cells were cultured using ⁇ -MEM, 10% FCS with 1% penicillin/streptomycin and 1% glutamine ("plating medium"), and were split 1:5 once per week.
  • the cells were resuspended in a plating medium containing 4% FCS, plated in microtiter plates at a concentration of 5 x 10 3 cells (in 50 ⁇ l)/well, and incubated for 24 hours at 37°C in 5% CO 2 .
  • 50 ⁇ l of the test compound or the control in DMSO was added at 2X concentration to each well, so that the final volume was
  • the treated cells were incubated for 24 hours at 37°C and 5% CO 2 .
  • the medium was then removed, and the cells were rinsed three times with PBS.
  • 25 ⁇ l of IX cell culture lysing reagent (Promega #E153A) was added to each well and incubated for at least ten minutes.
  • the plates/samples could be frozen at this point.
  • To each well was added 50 ⁇ l of luciferase substrate (Promega #E152A; 10 ml Promega luciferase assay buffer per 7 mg Promega luciferase assay substrate).
  • Luminescence was measured on an automated 96-well luminometer, and was expressed as either picograms of luciferase activity per well or as picograms of luciferase activity per microgram of protein.
  • compound 59-0008 (3-phenylazo-lH-4,l,2-benzothiadiazine) exhibits a pattern of reactivity which is maximal at a concentration of approximately 3-10 ⁇ M. Accordingly, other tested compounds can be evaluated at various concentrations, and the results compared to the results obtained for 59-0008 at 10 ⁇ M (which value would be normalized to 100). Alternatively, the reactivity of a compound to be tested can be compared directly to a negative control containing no compound.
  • statin compounds were tested in the in vitro BMP-promoter based (designated "ABA") assay (as described at the beginning of this example above), and in some experiments were tested also in a control osteoblast/serum response element (OBSRE) cell-based assay and/or a control glucagon assay.
  • ABA BMP-promoter based
  • a murine osteoblast cell line such as CCC-4 expressing a serum response element (SRE)-luciferase reporter gene is used (see WO96/07733).
  • SRE serum response element
  • a glucagon receptor-positive BHK cell expressing a CRE-luciferase reporter gene is used (see U.S. Patent No. 5,698,672).
  • statins i.e., simvastatin, hydrolyzed simvastatin, mevastatin, lovastatin, and fluvastatin
  • simvastatin demonstrated over a 20-fold induction in the ABA assay, as compared to the non-statin, ABA stimulatory compound 59-0008.
  • the pravastatin that was tested did not exhibit a dose-dependent stimulatory response, but since this compound was extracted from formulated pravastatin, it is possible that insufficient and/or inactive compound was tested.
  • simvastatin and lovastatin stimulated a dose-dependent increase in total bone area.
  • the bone stimulatory effects of these statins were comparable to the bone growth effect observed when 12.5 ⁇ g/kg/day bFGF was tested in the same assay.
  • New bone formation was determined (using a 10X objective) by measuring the new bone area formed in one field in 3 representative sections of each bone (4 bones per group). Each measurement was carried out 14 field distance from the end of the suture. Both total bone and old bone area were measured. Data were expressed as new bone area in mm 2 .
  • Osteoblast numbers were determined by point counting. The number of osteoblast cells lining the bone surface on both sides of the bone were counted in one field using a
  • statins and controls were tested in an antireso ⁇ tive assay. Briefly, 15 day timed pregnant CD-I female mice were injected with 45 Ca (25 ⁇ Ci/mouse). The calvaria from the 4 day old pups were dissected out and cut in half. The excised half calvaria were placed on metal grids (at the surface) in 1 ml of BGJ medium (Sigma) containing 0.1% BSA with glutamine and Pen Strep added. The bones were incubated at 37°C in a 5% humidified incubator for a period of 24 h, and then were transferred to wells containing 1 ml medium with factors added (IL-1, PTH, and/or test compounds).
  • the treated bones were incubated under the above conditions for a further 72 h. After this incubation period, the bones were removed and placed into 20% TCA in a scintillation vial for 1.5 h, and then counted with scintillation fluid. An aliquot of medium (0.4 ml) was also counted. The results were expressed as % 45 Ca release.
  • This assay may be modified by including test compounds/factors or control compounds/factors in the preincubation medium (i.e., during the first 24 h). Since most of the osteoclasts are formed in the calvaria following the preincubation period, compounds or factors that affect osteoclast formation may have a greater effect during the preincubation period.
  • compound toxicity was indicated by obvious death of the cells in the periosteal region and within the marrow cavity of the bone organ cultures. These cells were characterized by pyknotic nuclei and vacuolated cytoplasm, characteristic of cell necrosis and distinct from apoptosis.
  • simvastatin was tested for its ability to inhibit IL-1 induced bone reso ⁇ tion. Briefly, IL-1 (10 " M) was added simultaneously with simvastatin (at 0.1, 1 or 10 ⁇ M) during a 72 h incubation period. Bone reso ⁇ tion was determined by measuring
  • Lovastatin and simvastatin were analyzed in vivo using an acute ONX (prevention model) and/or chronic (treatment model) ONX model system, as described above under "Additional In Vivo Assays". Lovastatin was examined in an acute ONX study. Briefly, 59-0326 was orally administered (35, 7, or 1.4 mg/kg/day; once per day for 35 days) immediately following ovariectomy. At the end of the study, the animals' blood and tissues samples were processed, and the following data were obtained. pQCT analysis of samples removed from animals that received the 7 mg/kg/day dose showed a 20% increase in trabecular density and a 14% increase in cortical thickness.
  • lovastatin was orally administered (0.1 , 1 , 5, 10, 20 or 40 mg/kg/day; once per day for 35 days) immediately following ovariectomy.
  • Two individuals histomo ⁇ hometrically analyzed samples removed from animals that received the 10 mg/kg/day dose.
  • Simvastatin was also examined in a chronic ONX study. Briefly, simvastatin was orally administered (0.1, 1, 10, or 50 mg/kg/day; once per day for 10 weeks) to rats at 6 weeks post-ovariectomy. At the 10 mg/kg/day dose, a 114% (not significant) increase in the bone formation rate in the proximal tibia was measured. At the 50 mg/kg/day dose, an 86% (not significant) increase in the bone formation rate in the proximal tibia was measured.
  • Simvastatin was examined for effects on surgical defects in the rabbit radius. Healing of these defects may be assessed by X-ray, histology and biomechanical strength.
  • the test compound was weighed out in a microcentrifuge tube, and 50 ⁇ l of 1.5% sodium alginate solution was added as a carrier. This test sample was vortexed to wet all of the powder. The sample was sonicated for 20-30 min, and then vortexed again. Disks were created in the top of the microcentrifuge tube (by placing the tube lid or stopper-side down). The indent in the top of the stopper (i.e., lid) was used to form the disk (7.5 mm diameter).
  • CaCl 2 solution (100 ⁇ l of 100 mM) was added to the sodium alginate/drug solution. The samples were allowed to sit for 5-10 min, and then the calcium-alginate disks were carefully removed. The disks were rinsed in a beaker filled with water to rinse off the excess calcium solution, and were saved in tubes using water as vehicle. All solutions and containers were sterile, and all procedures for preparations of disks were performed under a laminar flow hood under sterile conditions.
  • the simvastatin or vehicle was applied to the defect and the defect was closed in layers. No external splinting was needed, as the radius is paired with the ulna, which functions to allow normal ambulation in the rabbit. Disks were cut on strips and inserted in the fracture to cover all the defect. Radiologic evaluation was performed at zero time and at 4 weeks. Because the vehicle (placebo treatment group) prevented full healing in the control group, only X-ray results were obtained and analyzed. Accordingly, X-ray analysis 4 weeks after initiation of treatment showed callus formation at the bone treatment site in the treated (both doses), but not the placebo (vehicle or autologous bone graft) groups.
  • the 2T3 osteoblast progenitor cell line described by Ghosh-Choudhury, et al, Endo (1995) 137:331-339 is BMP-2 responsive and undergoes bone formation spontaneously in culture in vitro.
  • These cells were stably transfected with the human dominant negative H-ras (Dn-ras) cDNA; this negative mutant gene contains an asparagine substitution at position 17 and production of this protein effects inhibition of endogenous ras.
  • the Dn-ras coding sequence was under transcriptional control of the dexamethasone inducible mouse mammary tumor virus promoter. The production of the negative ras protein was confirmed by Western blot.
  • the cells were cultured on 24-well plates at a density of 2x 10 4 /well and cultured with ⁇ MEM medium supplemented with 10% SDS for nodule formation assay indicating bone growth. Bone cell differentiation and module formation were monitored using a mineralized module formation assay described by Bharagava, et al, Bone (1986) 8:155-163 using van Kossa stain. The cultures were washed with phosphate buffered formalin, then with water and dehydrated in 70%, 95% and 100% ethanol, 2x each and then air-dried.
  • the plates were rehydrated before staining; the water was removed, a 2% silver nitrate solution was added and the plates were exposed to sunlight for 20 minutes. The plates were rinsed with water and 5% sodium thiosulfate was added for 3 minutes and then rinsed. Modified van Gleson stain was used as a counterstain for unmineralized collagen matrix. Acid fuschin solution was added for 5 minutes and the plates were then washed and dried for image analysis.
  • the area of van Kossa stained nodules was quantified by an automated image analysis using a video analysis program (Jandel Scientific, San Raphael, California) linked to a video screen camera equipped with metallurgical lenses.
  • the nodule structures were also analyzed by transmission electron microscopy.
  • a dramatic and spontaneous formation of mineralized bone nodules resulted. This result did not occur in wild-type cells in the absence of added BMP.
  • inhibition of ras function causes osteoblasts to differentiate and form bone.
  • Red Yeast Rice is a yeast culture that uses rice as a substrate and is known to contain lovastatin and may contain other HMG-CoA reductase inhibitors.
  • Red Yeast Rice which is marketed as "Cholestin” was tested in the "neonatal mouse calvaria assay (in vitro)" described above and set forth in Example 3 herein.
  • Four bones in each group were treated with either medium alone or with 100 ⁇ g/ml or 50 ⁇ g/ml of Red Yeast Rice.
  • the bone growth was measured in terms of new bone area.
  • the affect on growth was measured both in the absence of mevalonate and in the presence of 15 ⁇ g/ml mevalonate.
  • the formulation was prepared by sonicating lovastatin in whipping cream and diluting the original 1 ⁇ M concentration prepared with additional whipping cream.
  • the preparation was done by preparing a high concentration of lovastatin in DMSO and then diluting in whipping cream to obtain the most concentrated solution followed by sonication. Subsequent dilutions were made with additional whipping cream.
  • Lovastatin was tested for its affects on new bone area in the calvarial assay in vitro as used in Example 9. Four bones were used to obtain each average data point. The results are shown in the table below:
  • lovastatin contained in medium alone enhances bone growth in a manner that becomes significant at 1 ⁇ M.
  • the cream formulation shows bone growth stimulation at 0.04 ⁇ M and is a better bone growth stimulator at both 0.2 ⁇ M and 1 ⁇ M than lovastatin formulated in medium alone.
  • the in vitro neonatal mouse calvaria assay was performed as described except that the media were changed at days 4, 7 and 11 and the calvaria were removed for assessment at day 14.
  • Cerivastatin at a concentration of 0.25 ⁇ M was added to the medium at day 0 in one experimental group, and then removed after 1 day.
  • one-fourth (1/4) of this concentration was added at days 0, 4, 7 and 11.
  • Table 1 where new bone formation area was measured at day 14.
  • cerivastatin administered at a single dose of 0.25 ⁇ M represented an improvement over continuous dosage at one-quarter (1/4) this amount over a longer period.

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

L'invention concerne une aliment ou un supplément nutritif qui comprend un composé qui stimule la croissance osseuse chez les vertébrés. Ledit aliment ou produit alimentaire est préparé de manière à assurer l'effet stimulateur de la croissance osseuse recherché.
EP01927431A 2000-04-03 2001-04-02 Supplements nutritionnels pour stimuler la croissance osseuse Withdrawn EP1267641A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US541943 2000-04-03
US09/541,943 US6410521B1 (en) 1998-06-12 2000-04-03 Nutritional supplements for stimulating bone growth
PCT/US2001/040421 WO2001074180A1 (fr) 2000-01-20 2001-04-02 Supplements nutritionnels pour stimuler la croissance osseuse

Publications (1)

Publication Number Publication Date
EP1267641A1 true EP1267641A1 (fr) 2003-01-02

Family

ID=24161715

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01927431A Withdrawn EP1267641A1 (fr) 2000-04-03 2001-04-02 Supplements nutritionnels pour stimuler la croissance osseuse

Country Status (2)

Country Link
EP (1) EP1267641A1 (fr)
AU (1) AU2001253878A1 (fr)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0174180A1 *

Also Published As

Publication number Publication date
AU2001253878A1 (en) 2001-10-15

Similar Documents

Publication Publication Date Title
WO2001074180A1 (fr) Supplements nutritionnels pour stimuler la croissance osseuse
EP0944312B9 (fr) Compositions visant a stimuler la croissance osseuse et methodes afferentes
US6080779A (en) Compositions and methods for stimulating bone growth
US6342514B1 (en) Compositions and methods for treating bone deficit conditions
WO1998025460A9 (fr) Compositions visant a stimuler la croissance osseuse et methodes afferentes
US5919808A (en) Compositions and methods for treating bone deficit conditions
US5922753A (en) Methods for treating bone deficit conditions with benzothiazole
US5994358A (en) Compositions and methods for treating bone deficit conditions
US6153631A (en) Compositions and methods for treating bone deficit conditions
US6462019B1 (en) Inhibitors of proteasomal activity and production for stimulating bone growth
US5965573A (en) Compositions and methods for treating bone deficit conditions
US5948776A (en) Compositions and methods for treating bone deficit conditions
US6656904B2 (en) Inhibitors of proteasomal activity for stimulating bone and hair growth
US5990169A (en) Compositions and methods for treating bone deficit conditions
US6251901B1 (en) Compositions and methods for treating bone deficit conditions
EP0866710A1 (fr) Compositions et procedes pour le traitement des deficits osseux
WO1997015308A9 (fr) Compositions et procedes pour le traitement des deficits osseux
US6410521B1 (en) Nutritional supplements for stimulating bone growth
US20040254238A1 (en) Bone growth stimulation with NO/statin and other NO modulating combinations
EP1267641A1 (fr) Supplements nutritionnels pour stimuler la croissance osseuse
US20030181374A1 (en) Methods and compositions for stimulating bone growth using inhibitors of microtubule assembly
EP1609469A2 (fr) Compositions et méthodes pour stimuler la croissance osseuse
US6017940A (en) Compositions and methods for treating bone deficit conditions
US5939444A (en) Compositions and methods for treating bone deficit conditions

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20021024

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

17Q First examination report despatched

Effective date: 20040726

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20050208