US20180228750A1 - Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis - Google Patents

Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis Download PDF

Info

Publication number
US20180228750A1
US20180228750A1 US15/516,643 US201515516643A US2018228750A1 US 20180228750 A1 US20180228750 A1 US 20180228750A1 US 201515516643 A US201515516643 A US 201515516643A US 2018228750 A1 US2018228750 A1 US 2018228750A1
Authority
US
United States
Prior art keywords
bone
compound
osteoporosis
alkyl
acceptable salt
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.)
Abandoned
Application number
US15/516,643
Other languages
English (en)
Inventor
Lyne Gagnon
Brigitte Grouix
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.)
Prmoetic Pharma Smt Ltd
Liminal Biosciences Ltd
Original Assignee
Prmoetic Pharma Smt Ltd
Prometic Pharma SMT Ltd
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 Prmoetic Pharma Smt Ltd, Prometic Pharma SMT Ltd filed Critical Prmoetic Pharma Smt Ltd
Priority to US15/516,643 priority Critical patent/US20180228750A1/en
Assigned to PROMETIC BIOSCIENCES INC. reassignment PROMETIC BIOSCIENCES INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GAGNON, LYNE, GROUIX, BRIGITTE
Assigned to PRMOETIC PHARMA SMT LIMITED reassignment PRMOETIC PHARMA SMT LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PROMETIC BIOSCIENCES INC.
Publication of US20180228750A1 publication Critical patent/US20180228750A1/en
Assigned to Liminal Biosciences Limited reassignment Liminal Biosciences Limited CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: PROMETIC PHARMA SMT LIMITED
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C57/00Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms
    • C07C57/30Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms containing six-membered aromatic rings
    • C07C57/32Phenylacetic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C57/00Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms
    • C07C57/52Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms containing halogen
    • C07C57/58Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms containing halogen containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C59/00Compounds having carboxyl groups bound to acyclic carbon atoms and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C59/40Unsaturated compounds
    • C07C59/42Unsaturated compounds containing hydroxy or O-metal groups
    • C07C59/52Unsaturated compounds containing hydroxy or O-metal groups a hydroxy or O-metal group being bound to a carbon atom of a six-membered aromatic ring

Definitions

  • the present invention relates to the field of medicine. Particular aspects of the invention relates to compounds, pharmaceutical compositions and uses thereof for the prevention or treatment of osteoporosis.
  • Bone is a highly dynamic tissue which is constantly turned over and replaced by a process which is known as bone remodeling.
  • the ability to remodel bone ensures that old bone or damaged tissue is renewed and that the architecture of the skeleton can most efficiently adapt to mechanical demands.
  • Bone remodeling commences with the removal of old bone by osteoclast cells in a resorption phase lasting several weeks. Osteoblasts then migrate to the erosion cavity and deposit new bone over three or four months.
  • bone remodeling couples the activities of the osteoclast and osteoblast cells such that the amount of new bone laid down is equal to the bone that was removed, thereby maintaining a healthy bone mass.
  • bone resorption exceeds bone formation, there is a net loss of bone.
  • the resultant condition, osteoporosis is characterized by excessive bone resorption and subsequent low bone mass with increased bone fragility.
  • Osteoporosis is the general term used for diseases of the bone of diverse etiology that are characterized by a reduction in the mass of bone per unit volume to a level below that required for adequate mechanical support (Krane, S. M. et al., “Metabolic Bone Disease” in Harrison's Principles of Internal Medicine, page 1889, Edition 11 (1987)).
  • One form of osteoporosis is senile osteoporosis which is responsible for a large portion of the health dollars spent on the geriatric population (Resnick, N. M. et al. “Senile Osteoporosis Reconsidered”, JAMA 261, 1025-1029 (1989)).
  • osteoporosis The two other most common forms of osteoporosis are pre- or postmenopausal osteoporosis and corticosteroid induced osteoporosis.
  • Patients with chronic kidney disease (CKD) may develop bone diseases which can include osteoporosis as a result of changes in mineral metabolism and subsequent alterations in bone structure. Most often these changes worsen with progressive loss of kidney function. Indeed, and as summarized in the paragraph below, a number of pathological conditions can occur which increase the probability of the development of osteoporosis.
  • Osteomalacia-like osteoporosis shares many of the symptoms of osteoporosis such as loss of calcium. Osteopenia refers to a lower than normal bone density but not as low as that observed in osteoporosis.
  • Osteogenesis imperfecta is a congenital bone disorder characterized by brittle bones prone to bone fracture.
  • Osteopetrosis is a rare inherited disease whereby the bones harden but are more brittle than normal.
  • Osteonecrosis is a disease that causes bone death and collapse due to loss of blood supply to the bone. Paget's disease of bone is caused by excessive degradation and formation of bone followed by disorganized bone remodeling.
  • autoimmune diseases which include rheumatoid arthritis, lupus and multiple sclerosis
  • gastrointestinal disorders which include celiac disease, inflammatory bowel disease, gastrectomy and gastrointestinal by-pass procedures
  • endocrine/hormonal disorders which include diabetes, hyperparathyroidism, thyrotoxicosis and Cushing's syndrome
  • hematological disorders which include leukemia, lymphoma, multiple myeloma, sickle cell disease anemia (bone marrow disorders) and thalassemia
  • cancer which includes breast and prostate cancer
  • neurological disorders which include depression, Parkinson's disease and spinal cord injury
  • organ diseases which include lung (COPD, emphysema), liver and chronic kidney diseases; ankylosing spondylitis; AIDS/HIV; bone fracture; poor diet which includes eating disorders and malnutrition; and menopause (Pre-Menopause and Post-Menopause).
  • the osteoblast has been considered the master cell in the control of osteoclast development and, therefore, bone resorption.
  • the interactions between cells of the immune system and bone cells have redefined the thinking on the regulation of bone resorption.
  • the identification of the osteoclast and its role in bone destruction permits targeted therapy to reduce its resorptive capacity.
  • Such therapies include the use of agents that can interfere with receptor activator of NF ⁇ B ligand (RANKL), one of the key cytokines promoting osteoclast differentiation. This may be achieved through the use of recombinant Fcosteoprotegerin (Fc-OPG) or a humanised anti-RANKL antibody (Denosumab) that is being developed by Amgen.
  • Fc-OPG recombinant Fcosteoprotegerin
  • Denosumab humanised anti-RANKL antibody
  • osteoclast activity include the bisposhonates, c-src inhibitors, cathepsin K inhibitors and inhibitors of the chloride channel CLC7 (Gillespie, M. T. (2007) Arthritis Research & Therapy, Volume 9, No. 2, pp. 103-105).
  • bisphosphonates have been successful in limiting bone loss in rodent models of arthritis, although it should be noted that the nitrogen-containing bisphosphonates (which include aldronate, ibandronate, pamidronate and zoledronate) enhance proliferation of y/6 T lymphocytes, while non nitrogen-containing bisphosphonates (for example, clondronate) do not (Gillespie, M. T. (2007) Arthritis Research & Therapy, Volume 9, No. 2, pp. 103-105).
  • nitrogen-containing bisphosphonates which include aldronate, ibandronate, pamidronate and zoledronate
  • non nitrogen-containing bisphosphonates for example, clondronate
  • Parathyroid hormone (1-84 PTH) plays a central role in calcium homeostasis and, upon intermittent administration, an anabolic effect on bone remodeling.
  • Teriparatide approved by the US FDA (Forteo), is a recombinant form of a portion (amino acids 1-34) of PTH used for the treatment of osteoporosis in men and postmenopausal women who are at a high risk of a bone fracture. It may find some use off-label to speed healing of a bone fracture. Teriparatide enhances osteoblast formation and prevents osteoblast apoptosis.
  • Teriparatide is not recommended for use in patients with increased risk of bone tumors.
  • Denosumab is a fully human monoclonal antibody for the treatment of osteoporosis, treatment-induced bone loss, bone metastases, multiple myeloma and giant cell tumor of bone. Denosumab was approved by the US FDA (Prolia) for prevention of osteoporosis in postmenopausal women and (Xgeva) for the prevention of skeleton-related events in patients with bone metastases from solid tumors. This antibody binds to and inhibits RANKL (RANK ligand), a protein that acts as the primary signal for bone removal in many bone loss conditions. Osteoclast cell precursors (pre-osteoclasts) express RANK receptors.
  • RANKL RANK ligand
  • U.S. Pat. No. 6,372,728 (2002) assigned to AstraZeneca AB describes an improved oral formulation of bisphosphonates, e.g. Alendronate. According to this patent, the oral bioavailability of many bisphosphonates is 1%-10% between meals. The improved formulation employs a medium-chain glyceride absorption enhancer.
  • the present invention aims to address the need for new treatment methods, compounds and pharmaceutical compositions for patients afflicted by, or susceptible to, osteoporosis.
  • Particular aspects of the invention relates to the use of compounds and compositions for the prevention and/or treatment of osteoporosis.
  • Certain aspects concern compounds according to Formula I as defined herein and pharmaceutically acceptable salts thereof, as prophylactically effective and/or therapeutically effective agents against various forms of osteoporosis in subjects.
  • the subject is afflicted by, or susceptible to be afflicted by, bone loss, bone fracture and the like.
  • the compounds and compositions of the invention are useful for stimulating bone formation and/or for stimulating bone remodeling and/or for stimulating the differentiation and mineralization of osteoblasts and/or for inhibiting bone resorption.
  • a particular aspect of the invention relates to a method for the prevention and/or treatment of osteoporosis, comprising the step of administering to a subject in need thereof a compound represented by Formula I, or a pharmaceutical acceptable salt thereof, as defined herein.
  • the osteoporosis is selected from the group consisting of post-menopausal osteoporosis (primary type 1), primary type 2 osteoporosis, secondary osteoporosis abnormally high osteoclastogenesis, osteomalacia-like osteoporosis, osteopenia, osteogenesis imperfecta, osteopetrosis, osteonecrosis, Paget's disease of bone, hypophosphatemia and combinations thereof.
  • the osteoporosis is post-menopausal osteoporosis (primary type 1), primary type 2 osteoporosis or secondary osteoporosis.
  • the osteoporosis is post-menopausal osteoporosis (primary type 1).
  • the invention also relates to treatment methods wherein the compounds of the invention exhibit one or more of the following biological activities in a subject: inhibition of osteoclastogenesis; stimulation of interleukin-12 (IL-12) production by a stimulated osteoclast precursor cell; reduction of acid phosphatase activity in bone cells (demonstrates a reduction in osteoclastogenesis); reduction of the Receptor activator of NF- ⁇ B ligand (RANKL) over Osteoprotegerin (OPG) ratio (RANKL/PG ratio) in bone, which indicates a reduction in osteoclastogenesis; increase of collagen content in bone; and modulation (e.g. increase) of the serum level of adiponectin.
  • IL-12 interleukin-12
  • OPG Osteoprotegerin
  • aspect of the invention relates to a method for preventing and/or reducing bone loss, comprising the step of administering to a subject in need thereof a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein.
  • the administration of the compound reduces loss of calcium.
  • the subject is afflicted by or susceptible of osteoporosis.
  • the subject is a postmenopausal women.
  • the invention relates to a method for inhibiting osteoclastogenesis, comprising contacting an osteoclast precursor cell with a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein, wherein the compound inhibits differentiation of the precursor cell into an osteoclast cell.
  • the invention relates to a method for stimulating interleukin-12 (IL-12) production by a stimulated osteoclast precursor cell, comprising contacting said stimulated osteoclast precursor cell with a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein, wherein an increased IL-12 production is measurable in presence of the compound.
  • IL-12 interleukin-12
  • the invention relates to a method for reducing acid phosphatase activity in bone cells, comprising contacting the bone cells with a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein, wherein a reduced phosphatase activity is measurable in presence of the compound.
  • the invention relates to a method for reducing expression and/or activity of receptor activator of NF-43 ligand/Osteoprotegerin ratio (RANKL/OPG ratio) in bone cells, comprising contacting the bone cells with a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein.
  • a method for reducing expression and/or activity of receptor activator of NF-43 ligand/Osteoprotegerin ratio (RANKL/OPG ratio) in bone cells comprising contacting the bone cells with a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein.
  • the invention relates to a method for increasing collagen content in bone, comprising contacting the bone with a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein.
  • the invention relates to a method for stimulating bone formation and/or for stimulating bone remodeling and/or for stimulating the differentiation and mineralization of osteoblasts and/or for inhibiting bone resorption, comprising contacting osteoblasts in said bone with a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein.
  • the invention relates to a method for modulating serum level of adiponectin in a subject, comprising the step of administering to a subject in need thereof a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein.
  • the subject is obese and/or diabetic.
  • Additional aspects of the invention relates to the methods mentioned hereinabove, further comprising the step of administering concomitantly a drug selected from the group consisting of: bisphosphonates, Odanacatib, Alendronate, Risedronate, Etidronate, Zoledronate, Pamidronate, Teriparatide, Tamoxifen, Raloxifene, and Denosumab.
  • a drug selected from the group consisting of: bisphosphonates, Odanacatib, Alendronate, Risedronate, Etidronate, Zoledronate, Pamidronate, Teriparatide, Tamoxifen, Raloxifene, and Denosumab.
  • Another related aspect of the invention relates to pharmaceutical compositions comprising compounds of Formula I for the manufacture of a medicament, e.g. a medicament for the prevention and/or treatment of osteoporosis.
  • a pharmaceutical composition for preventing or treating osteoporosis comprising a compound represented by Formula I as defined herein, and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for use in preventing or treating osteoporosis comprising a compound as defined in Table 1, and more particularly, a pharmaceutical composition comprising Compound I and/or Compound XXXI.
  • Related aspect concerns methods for the prevention and/or treatment of osteoporosis, comprising administering to a patient a therapeutically effective amount of a pharmaceutical composition as defined herein.
  • the invention relates to a compound represented by Formula I or a pharmaceutical acceptable salt thereof as defined herein or to a composition comprising the same, for use in the prevention and/or treatment of osteoporosis.
  • FIG. 1 is a panel with pictures illustrating the effect of Compound I on LPS-induced osteoclastogenesis in RAW264.7 cells, according to Example 2.
  • FIG. 2 is a bar graph demonstrating that Compound I induces the production of IL-12 in LPS-stimulated RAW264.7 cells, according to Example 2.
  • FIG. 3 is a line graph demonstrating the effect of Compound I on body weight of ovariectomized (OVX) rats, according to Example 3.
  • FIG. 4 a panel with bar graphs demonstrating the effect of Compound I on calcium in urine of ovariectomized (OVX) rats, according to Example 4.
  • FIG. 5 a panel with bar graphs demonstrating the effect of Compound I on acid phosphatase activity in serum of ovariectomized (OVX) rats, according to Example 4.
  • FIG. 6 is a bar graph demonstrating the effect of Compound I on RANKL/OPG mRNA expression of osteoclast marker in ovariectomized (OVX) rat tibia, according to Example 4.
  • FIG. 7 is a bar graph demonstrating the effect of Compound I on collagen content in metaphyse of rat's femur, according to Example 4.
  • FIG. 8 is a panel with pictures illustrating the effect of Compound I on collagen content in metaphyse of rat's femur, according to Example 4.
  • FIG. 9 is a dot graph demonstrating that Compound XIV increases serum adiponectin level, a marker of modulation of bone formation and remodeling, according to Example 5.
  • the present discloses compounds of Formula I, pharmaceutically acceptable salts thereof, compositions comprising same and uses thereof.
  • Various embodiments of the present invention include:
  • the invention concerns the pharmaceutical uses of a compound represented by Formula I, or a pharmaceutically acceptable salt thereof:
  • A is C 5 alkyl, C 6 alkyl, C 5 alkenyl, C 6 alkenyl, C(O)—(CH 2 ) n —CH 3 or CH(OH)—(CH 2 ) n —CH 3 wherein n is 3 or 4; or is preferably C 5 alkyl, C 5 alkenyl, C(O)—(CH 2 ) n —CH 3 or CH(OH)—(CH 2 ) n —CH 3 wherein n is 3; or is preferably C 6 alkyl, C 6 alkenyl, C(O)—(CH 2 ) n —CH 3 or CH(OH)—(CH 2 ) n —CH 3 wherein n is 4; or is preferably straight chain C 5 alkyl, C 6 alkyl, C 5 alkenyl, C 6 alkenyl, C(O)—(CH 2 ) n —CH 3 or CH(OH)—(CH 2 ) n —CH 3 wherein n is
  • A is C 5 alkyl or C 6 alkyl.
  • R 1 is H or OH.
  • R 2 is H, F, OH, C 5 alkyl or C 6 alkyl.
  • R 3 is H or OH.
  • R 4 is H or OH.
  • Q is:
  • Q is:
  • Q is:
  • Q is (CH 2 ) m C(O)OH where m is 1 or 2.
  • the compound is of Formula I, wherein A is C 5 alkyl or C 6 alkyl; R 1 is H, F or OH; R 2 is H, F, OH, C 5 alkyl or C 6 alkyl; R 3 is H, OH or CH 2 Ph; R 4 is H, F or OH; and Q is (CH 2 ) m C(O)OH where m is 1 or 2.
  • the compound is of Formula I; wherein A is C 5 alkyl; R 1 is H; R 2 is H or C 5 alkyl; R 3 is H; R 4 is H; and Q is (CH 2 ) m C(O)OH where m is 1.
  • alkyl is intended to include a straight chain saturated aliphatic hydrocarbon group having the specified number of carbon atoms in a linear arrangement.
  • alkenyl is intended to mean unsaturated straight chain hydrocarbon groups having the specified number of carbon atoms therein, and in which at least two of the carbon atoms are bonded to each other by a double bond, and having either E or Z regiochemistry and combinations thereof.
  • Examples of compounds of Formula I include, but are not limited to, Compounds I to XXIV listed in Table 1 hereinbelow.
  • the compound is represented by the acid form or a pharmaceutically acceptable salt of any one of Compounds I to XXIV.
  • salts are intended to mean base addition salts.
  • Example of pharmaceutically acceptable salts are also described, for example, in Berge et al., “Pharmaceutical Salts”, J. Pharm. Sci. 66, 1-19 (1977).
  • Pharmaceutically acceptable salts may be synthesized from the parent agent that contains an acidic moiety, by conventional chemical methods. Generally, such salts are prepared by reacting the free acid forms of these agents with a stoichiometric amount of the appropriate base in water or in an organic solvent, or in a mixture of the two. Salts may be prepared in situ, during the final isolation or purification of the agent or by separately reacting a purified compound of the invention in its free acid form with the desired corresponding base, and isolating the salt thus formed.
  • the pharmaceutically acceptable salt of the compounds of Formula I may be selected from the group consisting of base addition salts of sodium, potassium, calcium, magnesium, lithium, ammonium, manganese, zinc, iron, or copper.
  • the pharmaceutically acceptable salt of the compounds according to the invention may be the sodium, potassium, calcium, magnesium or lithium salt. More preferably the pharmaceutically acceptable salt is sodium.
  • All acid, salt and other ionic and non-ionic forms of the compounds described are included as compounds of the invention.
  • the salt forms of the compound are also included.
  • a compound is shown as a salt and the acid forms are also included.
  • the compounds of the present invention as represented by generalized Formula I, wherein said compounds are present in the free carboxylic acid form may also include all pharmaceutically acceptable salts, isosteric equivalents such as tetrazole and prodrug forms thereof.
  • examples of the latter include the pharmaceutically acceptable esters or amides obtained upon reaction of alcohols or amines, including amino acids, with the free acids defined by Formula I.
  • the compounds of the present invention may contain one or more asymmetric centers, chiral axes and chiral planes and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms and may be defined in terms of absolute stereochemistry, such as (R)- or (S)-.
  • the present invention is intended to include all such possible isomers, as well as, their racemic and optically pure forms.
  • Optically active (+) and ( ⁇ ), (R)- and (S)-, isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, such as reverse phase HPLC.
  • the racemic mixtures may be prepared and thereafter separated into individual optical isomers or these optical isomers may be prepared by chiral synthesis.
  • the enantiomers may be resolved by methods known to those skilled in the art, for example by formation of diastereoisomeric salts which may then be separated by crystallization, gas-liquid or liquid chromatography, selective reaction of one enantiomer with an enantiomer specific reagent. It will also be appreciated by those skilled in the art that where the desired enantiomer is converted into another chemical entity by a separation technique, an additional step is then required to form the desired enantiomeric form. Alternatively specific enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts, or solvents or by converting one enantiomer to another by asymmetric transformation.
  • Certain compounds of the present invention may exist in Zwitterionic form and the present invention includes Zwitterionic forms of these compounds and mixtures thereof.
  • the compounds of the invention also may exist in hydrated and anhydrous forms. Hydrates of any of the formulas described herein are included as compounds of the invention which may exist as a monohydrate or in the form of a polyhydrate.
  • all compounds of the present invention may be prepared by any conventional methods, using readily available and/or conventionally preparable starting materials, reagents and conventional synthesis procedures.
  • any conventional methods using readily available and/or conventionally preparable starting materials, reagents and conventional synthesis procedures.
  • Of particular interest is the work of Hundertmark, T.; Littke, A. F.; Buchwald, S. L.; Fu, G. C. Org. Lett. 12, 1729-1731 (2000).
  • the compounds of the present invention have beneficial pharmaceutical properties and these compounds may have useful pharmaceutical applications in subjects.
  • Medical and pharmaceutical applications contemplated by the inventors include, but are not limited to, prevention and/or treatment of various forms of osteoporosis.
  • osteoporosis refers to a progressive bone disease that is characterized by a decrease in bone mass and density which can lead to an increased risk of bone fracture.
  • osteoporosis encompasses primary type 1 osteoporosis or postmenopausal osteoporosis (most common in women after menopause), primary type 2 osteoporosis (occurs in both females and males generally after age 75), and secondary osteoporosis (which may arise at any age, a form that results from chronic predisposing medical problems or disease, or prolonged use of medications such as glucocorticoids (the disease may then be called steroid- or glucocorticoid-induced osteoporosis)).
  • osteoporosis also includes bone disorders involving a loss in bone mass and/or density such as abnormally high osteoclastogenesis, osteomalacia-like osteoporosis, osteopenia, osteogenesis imperfecta, osteopetrosis, osteonecrosis, Paget's disease of bone, hypophosphatemia and combinations thereof.
  • osteoporosis As is known, various diseases and conditions may cause osteoporosis, and the present invention may be useful in preventing and/or treating osteoporosis related, directly or indirectly to one or more of these causes:
  • the osteoporosis is primary type 1 osteoporosis or postmenopausal osteoporosis. In another embodiment the osteoporosis is primary type 2 osteoporosis.
  • subject includes living organisms in which osteoporosis can occur, or which are susceptible to such disease.
  • subject includes animals such as mammals or birds.
  • the subject is a mammal, including but not limited to human, horse, dog and cat. In some embodiments mouse is excluded from the scope of a mammal. More preferably, the subject is a human. Most preferably, the subject is a human patient in need of treatment.
  • the subject is a person having or suffering from any of the following conditions: primary type 1 osteoporosis, postmenopausal osteoporosis, menopause (pre-menopause and post-menopause), primary type 2 osteoporosis, more than 75 years old, bone fracture, osteoporosis, chronic predisposing medical problems or disease, prolonged use of medications such as glucocorticoids, abnormally high osteoclastogenesis, osteomalacia-like osteoporosis, osteopenia, osteogenesis imperfecta, osteopetrosis, osteonecrosis, Paget's disease of bone, hypophosphatemia and combinations thereof.
  • the subject is a postmenopausal woman.
  • preventing or “prevention” is intended to refer to at least the reduction of likelihood of the risk of (or susceptibility to) acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a patient that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease).
  • Biological and physiological parameters for identifying such patients are provided herein and are also well known by physicians.
  • “preventing” or “prevention” refers to preventing decrease in bone mass and/or bone density, and/or to reducing risk of bone fracture.
  • treatment includes the application or administration of a compound of the invention to a subject (or application or administration of a compound of the invention to a cell or tissue from a subject) with the purpose of delaying, stabilizing, curing, healing, alleviating, relieving, altering, remedying, less worsening, ameliorating, improving, or affecting the disease or condition, the symptom of the disease or condition, or the risk of (or susceptibility to) the disease or condition.
  • treating refers to any indication of success in the treatment or amelioration of an injury, pathology or condition, including any objective or subjective parameter such as abatement; remission; lessening of the rate of worsening; lessening severity of the disease; stabilization, diminishing of symptoms or making the injury, pathology or condition more tolerable to the subject; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; or improving a subject's physical or mental well-being.
  • the term “treating” can include increasing a subject's life expectancy and/or delay before additional treatments are required.
  • treatment or “treating” refers to increasing bone mass and/or bone density, and/or increasing healing of bone fracture.
  • the compounds of the invention are for used monotherapy for the treatment prevention and/or treatment of osteoporosis.
  • the compounds of the invention are used in combination with already approved drugs, including but not limited to drugs used for the treatment of osteoporosis.
  • known osteoporosis-related agents which may be used in combination with the compounds of the present invention include, but are not limited to bisphosphonates, Odanacatib, Alendronate, Risedronate, Etidronate, Zoledronate, Pamidronate, Teriparatide, Tamoxifen, Raloxifene, and Denosumab.
  • methods of treatment according to the present invention may also include co-administration of the at least one compound according to the invention, or a pharmaceutically acceptable salt thereof, together with the administration of another therapeutically effective agent. Therefore, an additional aspect of the invention relates to methods of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a first agent and a second agent, wherein the first agent is as defined in Formula I, and the second agent is for the prevention or treatment of any one of disorder or disease as defined hereinbefore.
  • the term “concomitant” or “concomitantly” as in the phrases “concomitant therapeutic treatment” or “concomitantly with” includes administering a first agent in the presence of a second agent.
  • a concomitant therapeutic treatment method includes methods in which the first, second, third or additional agents are co-administered.
  • a concomitant therapeutic treatment method also includes methods in which the first or additional agents are administered in the presence of a second or additional agents, wherein the second or additional agents, for example, may have been previously administered.
  • a concomitant therapeutic treatment method may be executed step-wise by different actors. For example, one actor may administer to a subject a first agent and as a second actor may administer to the subject a second agent and the administering steps may be executed at the same time, or nearly the same time, or at distant times, so long as the first agent (and/or additional agents) are after administration in the presence of the second agent (and/or additional agents).
  • the actor and the subject may be the same entity (e.g., a human).
  • the invention also relates to a method for preventing, reducing or eliminating a symptom or complication of any one of the above mentioned diseases or conditions.
  • the method comprises administering, to a subject in need thereof, a first pharmaceutical composition comprising at least one compound of the invention and a second pharmaceutical composition comprising one or more additional active ingredients, wherein all active ingredients are administered in an amount sufficient to inhibit, reduce, or eliminate one or more symptoms or complications of the disease or condition to be treated.
  • the administration of the first and second pharmaceutical composition is temporally spaced apart by at least about two minutes.
  • the first agent is a compound of Formula I as defined herein, or a pharmaceutically acceptable salt thereof, e.g., sodium salt.
  • the second agent may be selected from the list of compounds given hereinbefore (e.g. agents or drugs used for the prevention and/or treatment of osteoporosis).
  • Osteoclasts are a type of bone cell that resorbs bone tissue.
  • the osteoclast disassembles the bone at a molecular level by secreting acid and a collagenase. This process is known as bone resorption.
  • Osteoclastogenesis refers to the differentiation of precursor of osteoclasts into osteoclasts. In the prevention and/or treatment of osteoporosis, it is desirable to reduce osteoclastogenesis.
  • Osteoblast is a type of cells that synthesizes bone. Osteoblasts arise from mesenchymal stem cells. In the prevention and/or treatment of osteoporosis, it is desirable is to stimulate osteoblastic differentiation.
  • the compounds of the invention are capable of inhibiting and/or reducing osteoclastogenesis. This is demonstrated for example by: the absence of TRAP cells (Example 2; FIG. 1 ); a strong induction of IL-12 production in LPS-stimulated RAW264.7 cells (Examples 2 and 3); a reduction of calcium loss in vivo (Example 4, FIG. 4 ); a reduction of acid phosphatase activity in vivo (Example 4, FIG. 5 ); a decrease of mRNA expression of RANKL/OPG in vivo (Example 4, FIG. 6 ); an increase in collagen content in vivo (Example 4, FIGS. 7 and 8 ); and an increase of the serum level of adiponectin (Example 5, FIG. 9 ).
  • results demonstrate an ability of the compounds of the present invention to prevent and/or reduce bone loss, including but not limited to, calcium loss. Accordingly, these results further suggest an ability of the compounds of the present invention to prevent/treat osteoporosis, via the stimulation of osteoblastic differentiation.
  • the compounds of the invention induce the production of IL-1 2 in the presence of LPS.
  • the compounds and compositions of the invention are useful for the stimulation of interleukin-12 (IL-12) production including, but not limited to, production by a stimulated osteoclast precursor cell.
  • IL-12 interleukin-12
  • the compounds of the invention reduce enzymatic acid phosphatase activity, as measured in the serum of ovariectomized rats. These results suggest an ability of these compounds to prevent and/or treat osteoporosis as a result of reduction of enzymatic acid phosphatase activity.
  • the compounds and compositions of the invention are useful for reducing acid phosphatase activity in bone cells.
  • the compounds of the invention reduce mRNA expression of RANKL, as measured in the tibia of ovariectomized rats. These results suggest an ability of these compounds to prevent and/or treat osteoporosis as a result of reduction of RANKL's expression and/or biological activity.
  • the compounds and compositions of the invention are useful for reducing expression and/or activity of RANKL in bone cells.
  • the compounds of the invention increase the content of collagen in bone, as measured in the metaphyse of the femur of ovariectomized rats. These results suggest an ability of these compounds to prevent and/or treat osteoporosis as demonstrated by the increase of the collagen content in the bone.
  • the compounds and compositions of the invention are useful for increasing collagen content in living bone.
  • the compounds of the invention increase the level of adiponectin, as measured in the serum of obese diabetic mice. These results suggest an ability of these compounds to prevent and/or treat osteoporosis by stimulating bone formation, remodeling and mineralization and/or by inhibiting bone resorption.
  • the compounds and compositions of the invention are useful for stimulating bone formation and/or for stimulating bone remodeling and/or for stimulating the differentiation and mineralization of osteoblasts and/or for inhibiting bone resorption.
  • a related aspect of the invention concerns pharmaceutical compositions comprising a therapeutically effective amount one or more of the compounds of the invention described herein (e.g., a compound of Formula I).
  • the pharmaceutical compositions of the invention may be useful: in the prevention and/or treatment of osteoporosis; in the inhibition of osteoclastogenesis; in stimulating interleukin-12 (IL-12) production by a stimulated osteoclast precursor cell; in the reduction of acid phosphatase activity in bone cells; in the reduction of expression of Receptor activator of NF- ⁇ B ligand (RANKL) in bone cells; in increasing collagen content in bone, in stimulating bone formation; in stimulating bone remodeling; in stimulating bone mineralization; and/or in inhibiting bone resorption.
  • IL-12 interleukin-12
  • RNKL Receptor activator of NF- ⁇ B ligand
  • the term “therapeutically effective amount” means the amount of compound that, when administered to a subject for treating or preventing a particular disorder, disease or condition, is sufficient to effect such treatment or prevention of that disorder, disease or condition. Dosages and therapeutically effective amounts may vary for example, depending upon a variety of factors including the activity of the specific agent employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, and any drug combination, if applicable, the effect which the practitioner desires the compound to have upon the subject (e.g., total or partial response as evidenced by factors which include increasing bone mass and/or bone density (or reduction of a decrease thereof), reducing a risk of bone fracture, etc.), the properties of the compounds (e.g., bioavailability, stability, potency, toxicity, etc.), and the particular disorder(s) the subject is suffering from.
  • the effect which the practitioner desires the compound to have upon the subject e.g., total or partial response as evidenced by factors which include
  • the therapeutically effective amount may depend on the subject's blood parameters (e.g., calcium levels, lipid profile, insulin levels, glycemia), the severity of the disease state, organ function, or underlying disease or complications.
  • blood parameters e.g., calcium levels, lipid profile, insulin levels, glycemia
  • Such appropriate doses may be determined using any available assays including the assays described herein.
  • a physician may for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained.
  • the dose to be administered will ultimately be at the discretion of the oncologist. In general, however, it is envisioned that the dose for the present compounds may be in the range of about 1 to about 50 mg/kg per day in human.
  • the range may be between 1 to 30 mg/kg per day in human. In selected embodiments, the range may be between 1 to 20 mg/kg per day in human. In selected embodiments, the range may be between 5 to 18 mg/kg per day in human. In selected embodiments, the range may be between 1 to 18 mg/kg per day in human.
  • the term “pharmaceutical composition” refers to the presence of at least one compound of the invention according to Formula I as defined herein and at least one pharmaceutically acceptable carrier, diluent, vehicle or excipient.
  • pharmaceutically acceptable carrier pharmaceutically acceptable diluent or pharmaceutically acceptable excipient
  • the pharmaceutically acceptable vehicle can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • compositions include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glyco
  • antibacterial and antifungal agents for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents are included, for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
  • composition of the present invention may include one or more compounds of Formula I as defined herein or pharmaceutically acceptable derivatives, salts prodrugs, analogues and isomers or enantiomers thereof.
  • Formulations of the active compound may be prepared so as to provide a pharmaceutical composition in a form suitable for enteral, mucosal (including sublingual, pulmonary and rectal), parenteral (including intramuscular, intradermal, subcutaneous and intravenous) or topical (including ointments, creams or lotions) administration.
  • the formulation may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods well-known in the art of pharmaceutical formulation. All methods include the step of bringing together the active pharmaceutical ingredient with liquid carriers or finely divided solid carriers or both as the need dictates.
  • the above-described formulations may be adapted so as to provide sustained release of the active pharmaceutical ingredient. Sustained release formulations well-known to the art include the use of a bolus injection, continuous infusion, biocompatible polymers or lipo
  • the compound(s) of the invention may be packaged as part of a kit, optionally including a container (e.g., packaging, a box, a vial, etc.).
  • the kit may be commercially used according to the methods described herein and may include instructions for use in a method of the invention.
  • Additional kit components may include acids, bases, buffering agents, inorganic salts, solvents, antioxidants, preservatives, or metal chelators.
  • the additional kit components are present as pure compositions, or as aqueous or organic solutions that incorporate one or more additional kit components. Any or all of the kit components optionally further comprise buffers.
  • the compound(s) of the invention may or may not be administered to a patient at the same time or by the same route of administration. Therefore, the methods of the invention encompass kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of two or more active ingredients to a patient.
  • a typical kit of the invention comprises a unit dosage form of at least one compound according to the invention as defined by Formula I as defined herein, or a pharmaceutically acceptable salt thereof, and a unit dosage form of at least one additional active ingredient.
  • additional active ingredients include, but are not limited to, any of the drugs indicated hereinbefore (e.g. drugs used for the treatment of osteoporosis) that could be used in combination with the compound(s) of the invention.
  • Kits of the invention can further comprise pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients.
  • the kit can comprise a sealed container or a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration.
  • suitable vehicles are provided hereinbefore.
  • the Propyl triphenylphosphonium Bromide (3.2 g, 8.2 mmol) was placed in a round bottom flask, under nitrogen, and dry THF (5 mL) was added. The flask is cooled in an ice/acetone ( ⁇ 10° C.) bath, and nButyllithium (2.5 M in Hexanes, 3.28 mL, 8.2 mmol) was added slowly. The mixture turn dark colored with stirring for 30 minutes. In an ice/acetone ( ⁇ 10° C.) bath was placed the crude reaction mixture from the previous step in dry THF (5 mL) under nitrogen.
  • the above compound was prepared as for Compound I starting with E-(3-pent-1-enyl-phenyl)acetic acid methyl ester.
  • the latter was prepared by reacting 3-bromophenyl acetic acid methyl ester with trans-1-pentenylboronic acid pinacol ester under Suzuki conditions.
  • aqueous phase was extracted with a further portion of ethyl acetate (50 mL); and the combined extracts were washed with water (2 ⁇ 50 mL) and with saturated aqueous sodium chloride (50 mL); dried over sodium sulfate; filtered, and evaporated in vacuo to give the crude product.
  • the above compound was prepared from methyl 2-(3-bromo-4-fluorophenyl)acetate by Suzuki coupling as for Compound VII; followed by hydrogenation, ester hydrolysis and salt formation as for Compound I.
  • the starting ester was prepared by reaction of 2-(3-bromo-4-fluorophenyl)acetic acid with methanol in the presence of sulfuric acid.
  • the above compound was prepared from ethyl 2-fluoro-2-(3-pentylphenyl)acetate as for Compound I.
  • the ester was prepared by reaction of ethyl 2-(3-pentylphenyl)acetate with lithium diisopropylamide and N-fluorobenzenesulfonimide at ⁇ 78° C. in Tetrahydrofuran.
  • the reaction was cooled to room temperature and was partitioned between ethyl acetate (200 mL) and 1M aqueous hydrochloric acid (150 mL). The organic phase was washed with 5% aqueous sodium bicarbonate (150 mL), and with saturated aqueous sodium chloride (150 mL); then dried over sodium sulphate; filtered, and evaporated in vacuo to give the crude product.
  • Methyl 2-(2-hydroxy-3,5-dipentylphenyl)acetate (0.2 g, 0.53 mmol) was hydrolysed as for Compound I, step 4, to give the crude product mixed with lactonised material. A small portion was purified on a BiotageTM SP1 system (120 g silica cartridge), eluting with 0-100% ethyl acetate in hexanes, to give 2-(2-hydroxy-3,5-dipentylphenyl)acetic acid (13.5 mg).
  • LPS a bacteria-derived cell wall product
  • LPS plays an important role in bone resorption, which involves recruitment of inflammatory cells, synthesis of cytokines (such as interleukin-6 (IL-6), IL-12 and tumor necrosis factor-a (TNF-a)), and activation of osteoclast formation and differentiation.
  • IL-6 interleukin-6
  • TNF-a tumor necrosis factor-a
  • RAW264.7 cells are precursors of osteoclasts and can be differentiated by several factors including Receptor activator of NF- ⁇ B ligand (RANKL) or Lipopolysaccharide (LPS). Osteoclasts are characterized by high expression or tartrate resistant acid phosphatase (TRACP) and Matrix Metalloproteinase-9 (MMP-9) which can be used as markers for osteoclasts. It has been shown that RAW264.7 cells incubated in presence of capric acid resulted in an increase in IL-12 production and in a reduction of phosphatase (TRAP)-positive cells (TRAP expression, an osteoclast differentiation marker) (Wang et al., J. Biol. Chem. (2006), Vol.
  • TRAP phosphatase
  • TRAP expression an osteoclast differentiation marker
  • LPS strongly upregulated inducible nitric oxide synthase (iNOS) mRNA levels and nitric oxide (NO) production, whereas capric acid inhibited them. Additionally, capric acid also inhibited monocyte chemoattractant protein-1 (MCP-1) mRNA expression.
  • iNOS inducible nitric oxide synthase
  • NO nitric oxide
  • MCP-1 monocyte chemoattractant protein-1
  • RAW264.7 cells a murine osteoclast precursor cell line, in comparison with capric acid, as a positive control for reduction of osteoclastogenesis (Park et al. (2011) PLOS One Volume 6, Issue 11, 8 pages).
  • RAW264.7 cells were differentiated by the incubation with LPS (1 ug/ml) in presence or not of capric acid or Compound I.
  • LPS 1 ug/ml
  • osteoclast formation was evaluated using tartrate-resistant acid phosphatase (TRAP) staining.
  • TRAP tartrate-resistant acid phosphatase
  • FIG. 1 represents the osteoclastogenesis effect of LPS in RAW264.7 cells as demonstrated by the high expression of TRAP (dark staining). Incubation with capric acid and Compound I indicates a different phenotype that the control and LPS-induced cells and no TRAP cells are observable indicating that the cells are not differentiated in osteoclasts.
  • capric acid increases the production of IL-12 in LPS-stimulated RAW264.7 (Wang et al., J. Biol. Chem. (2006), Vol. 281, No. 45, pp. 34457-64). Since capric acid is also a known osteoclastogenesis inhibitor, an experiment was undertaken to determine if Compound I was capable of promoting an increase in IL-12 production.
  • RAW264.7 cells were cultured with 100 ng/mL of LPS in presence or absence of compounds for 21 hours in a humidified atmosphere of 95% air-5% carbon dioxide at 37° C.
  • IL-12 concentration in the culture medium was measured using the IL-12 ELISA according to the manufacturer (BD Biosciences) recommendations.
  • FIG. 2 illustrates the strong induction of IL-12 production in LPS-stimulated RAW264.7 cells in presence of various concentrations of Compound I.
  • IL-12 is also reported to inhibit osteoclast formation (Horwood and al. (2001) J. of Immunology, Volume 166, No. 8, pp. 4915-4921).
  • Example 1 LPS-stimulated RAW264.7 incubated in presence of Compound I increases IL-12 and reduces osteoclastogenesis (TRAP).
  • An in vitro IL-12 production assay was used for screening potential inhibitors of osteoclastogenesis.
  • Table 2 represents the effect of representative compounds of Formula I on IL-12 production. All these compounds induced a significant increase in IL-12 production.
  • rats can be ovariectomized to make them sex-hormone deficient, and to stimulate the accelerated loss of bone that occurs in women following menopause.
  • Ovariectomy induces bone loss in the rat and postmenopausal bone loss share many similar characteristics. These include: increased rate of bone turnover with resorption exceeding formation; an initial rapid phase of bone loss followed by a much slower phase; greater loss of cancellous than cortical bone; decreased intestinal absorption of calcium; some protection against bone loss by obesity; and similar skeletal response to therapy with estrogen, tamoxifen, bisphosphonates, parathyroid hormone, calcitonin and exercise. These wide-ranging similarities are strong evidence that the ovariectomized rat bone loss model is suitable for studying problems that are relevant to postmenopausal bone loss.
  • Rats were ovariectomized (OVX) at day 0. Rats were treated by oral gavage with Compound I (200 mg/kg) from day 14 to day 68. Evaluation of different parameters (body weight, calcium loss, osteoclast markers (RANKL and TRAP mRNA expression), collagen content and histology) was performed at day 68.
  • FIG. 3 illustrates the increase in body weight of the ovariectomized rats (similar to the “postmenopausal obesity”).
  • Compound I reduced ovariectomized-induced obesity.
  • FIG. 4 represents the effect of Compound I on calcium loss in ovariectomized rats. Calcium loss is detected in urine from ovariectomized rats from day 28 to day 56. Compound I reduced significantly the calcium loss in ovariectomized rats.
  • FIG. 6 represents the effect of Compound I on the ratio of mRNA expression of RANKL/OPG at day 68 in rat tibia.
  • RANKL/OPG mRNA expression increased in rats developing osteoporosis while it decreased with the treatment with Compound I; a decrease is indicative of a successful reduction of osteoclastogenesis.
  • FIG. 8 shows representative pictures of histological bone section of the metaphysis of the femur.
  • Compound I reduced bone loss in the metaphysis portion of the femur.
  • adiponectin stimulates bone formation and remodeling as well as inhibits bone resorption, suggesting that adiponectin may be a negative regulator of bone mass (Lubkowska et al. (2014) Disease Markers, Volume 2014, Article ID975178, 14 pages). Also, most in vitro studies have demonstrated that adiponectin stimulates the differentiation and mineralization of osteoblasts as well as the expression of osteocalcin, which acts as a hormone regulating glucose metabolism and fat mass (Lubkowska et al. (2014)). The potential influence of adiponectin on osteoblasts and osteoclasts and consequently on bone remodeling is possibly connected with the interrelationship between the endocrine system and fat metabolism (Lubkowska et al. (2014)).
  • mice (6 weeks-old were uninephrectomized and treated with Compound XIV for 113 days (structure is shown in Table 1 hereinbefore). Serum adiponectin levels were determined with a mouse adiponectin enzyme-linked immunosorbant assay kit (R&D Systems).
  • FIG. 9 illustrates the serum adiponectin level as measured in obese db/db diabetic mice.
  • Serum adiponectin level was decreased in non-treated db/db mice while treatment with Compound XIV increased the level of adiponectin to the level of the sham animals, suggesting that this compound may prevent or reduce osteoporosis.
  • Variants of Compound XIV particularly compounds with a similar chemical structure, may also increase the level of adiponectin and prevent and/or reduce osteoporosis.
  • a non-exhaustive list of chemical variants of Compound XIV include, but is not limited to, Compounds XV to XXIV (and any pharmaceutical acceptable salts thereof) shown in Table 1 above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Organic Chemistry (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Child & Adolescent Psychology (AREA)
  • Endocrinology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US15/516,643 2014-10-10 2015-10-08 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis Abandoned US20180228750A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/516,643 US20180228750A1 (en) 2014-10-10 2015-10-08 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462062597P 2014-10-10 2014-10-10
PCT/CA2015/000540 WO2016054728A1 (en) 2014-10-10 2015-10-08 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis
US15/516,643 US20180228750A1 (en) 2014-10-10 2015-10-08 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2015/000540 A-371-Of-International WO2016054728A1 (en) 2014-10-10 2015-10-08 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/844,813 Continuation US11185521B2 (en) 2014-10-10 2020-04-09 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis

Publications (1)

Publication Number Publication Date
US20180228750A1 true US20180228750A1 (en) 2018-08-16

Family

ID=55652429

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/516,643 Abandoned US20180228750A1 (en) 2014-10-10 2015-10-08 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis
US16/844,813 Active US11185521B2 (en) 2014-10-10 2020-04-09 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/844,813 Active US11185521B2 (en) 2014-10-10 2020-04-09 Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis

Country Status (23)

Country Link
US (2) US20180228750A1 (ru)
EP (1) EP3203999B1 (ru)
JP (1) JP6700639B2 (ru)
KR (1) KR20170066476A (ru)
CN (1) CN106999456B (ru)
AR (1) AR102246A1 (ru)
AU (1) AU2015330645B2 (ru)
BR (1) BR112017007426A2 (ru)
CA (1) CA2963354A1 (ru)
DK (1) DK3203999T3 (ru)
ES (1) ES2785317T3 (ru)
IL (1) IL251404B (ru)
MX (1) MX2017004733A (ru)
MY (1) MY194308A (ru)
PH (1) PH12017500638A1 (ru)
PL (1) PL3203999T3 (ru)
PT (1) PT3203999T (ru)
RU (1) RU2709205C2 (ru)
SG (1) SG11201702868VA (ru)
TW (1) TWI699200B (ru)
UY (1) UY36356A (ru)
WO (1) WO2016054728A1 (ru)
ZA (1) ZA201702639B (ru)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20170072352A (ko) 2014-11-12 2017-06-26 프로메틱 파마 에스엠티 리미티드 조직 자가-회복 및 재생을 위한 치환된 방향족 화합물 및 약제학적 조성물
JP2018507914A (ja) 2015-03-09 2018-03-22 インテクリン・セラピューティクス・インコーポレイテッド 非アルコール性脂肪肝疾患および/またはリポジストロフィーの処置のための方法
CA3033971A1 (en) * 2016-08-18 2018-02-22 Intekrin Therapeutics, Inc. Ppar.gamma. agonist for treatment of bone disorders
JP2020515639A (ja) 2017-04-03 2020-05-28 コヒラス・バイオサイエンシズ・インコーポレイテッド 進行性核上性麻痺の処置のためのPPARγアゴニスト
EP4225724A1 (en) * 2020-10-06 2023-08-16 Pharmaceutique Ingenew Inc. Substituted aromatic compounds and pharmaceutical compositions thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010127440A1 (en) * 2009-05-04 2010-11-11 Prometic Biosciences Inc. Substituted aromatic compounds and pharmaceutical uses thereof
US8637574B2 (en) * 2009-05-04 2014-01-28 Prometic Biosciences, Inc. Salts of 3-pentylphenylacetic acid and pharmaceutical uses thereof
US9439882B2 (en) * 2010-10-27 2016-09-13 Prometic Biosciences Inc. Compounds and compositions for the treatment of cancer
US9884802B2 (en) * 2013-03-15 2018-02-06 Prometic Pharma Smt Limited Substituted aromatic compounds for the treatment pulmonary fibrosis, liver fibrosis, skin fibrosis and cardiac fibrosis

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200720256A (en) * 2005-05-13 2007-06-01 Synta Pharmaceuticals Corp IL-12 modulatory compounds
WO2012055014A1 (en) * 2010-10-27 2012-05-03 Prometic Biosciences Inc. Phenylketone carboxylate compounds and pharmaceutical uses thereof
TWI689490B (zh) * 2013-03-15 2020-04-01 英商邊緣生物科技有限公司 用於治療纖維化之經取代之芳族化合物及相關方法

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010127440A1 (en) * 2009-05-04 2010-11-11 Prometic Biosciences Inc. Substituted aromatic compounds and pharmaceutical uses thereof
US20120088784A1 (en) * 2009-05-04 2012-04-12 Boulos Zacharie Substituted Aromatic Compounds and Pharmaceutical Uses thereof
US8637574B2 (en) * 2009-05-04 2014-01-28 Prometic Biosciences, Inc. Salts of 3-pentylphenylacetic acid and pharmaceutical uses thereof
US8927765B2 (en) * 2009-05-04 2015-01-06 Prometic Biosciences Inc. Substituted aromatic compounds and pharmaceutical uses thereof
US9938221B2 (en) * 2009-05-04 2018-04-10 Prometic Pharma Smt Limited Substituted aromatic compounds and pharmaceutical uses thereof
US9439882B2 (en) * 2010-10-27 2016-09-13 Prometic Biosciences Inc. Compounds and compositions for the treatment of cancer
US9884802B2 (en) * 2013-03-15 2018-02-06 Prometic Pharma Smt Limited Substituted aromatic compounds for the treatment pulmonary fibrosis, liver fibrosis, skin fibrosis and cardiac fibrosis

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Bone et al. "Ten Years' Experience with Alendronate for Osteoporosis in Postmenopausal Women". N Engl J Med. 2004; 350:1189-1199. (Year: 2004) *
Dobbs et al. "Osteoporosis: The Increasing Role of the Orthopaedist". Iowa Orthop J. 1999; 19:43-52. (Year: 1999) *
Hayashi et al. "Distinct Osteoclast Precursors in the Bone Marrow and Extramedullary Organs Characterized by Responsiveness to Toll-Like Receptor Ligands and TNF-Alpha". The Journal of Immunology. 2003; 171:5130-5139. (Year: 2003) *
Rao et al. [Online] "Oxidative Stress and Antioxidants in the Risk of Osteoporosis - Role of the Antioxidants Lycopene and Polyphenols". <https://www.intechopen.com/books/topics-in-osteoporosis/oxidative-stress-and-antioxidants-in-the-risk-of-osteoporosis-role-of-the-antioxidants->.Published 05/15/13. (Year: 2013) *
Rao Oxidative Stress and Antioxidants in the Risk of Osteoporosis Role of the Antioxidants Lycopene and Polyphenols, Published May 15, 2013 *

Also Published As

Publication number Publication date
EP3203999A1 (en) 2017-08-16
TW201618762A (zh) 2016-06-01
AU2015330645B2 (en) 2020-03-12
BR112017007426A2 (pt) 2018-01-16
AU2015330645A1 (en) 2017-05-04
SG11201702868VA (en) 2017-05-30
AR102246A1 (es) 2017-02-15
PH12017500638A1 (en) 2017-09-25
WO2016054728A1 (en) 2016-04-14
IL251404A0 (en) 2017-05-29
TWI699200B (zh) 2020-07-21
US20200237693A1 (en) 2020-07-30
UY36356A (es) 2016-06-01
IL251404B (en) 2020-11-30
CN106999456B (zh) 2020-07-10
PL3203999T3 (pl) 2020-07-27
EP3203999B1 (en) 2020-01-08
MX2017004733A (es) 2017-07-17
RU2017116178A (ru) 2018-11-19
CA2963354A1 (en) 2016-04-14
RU2017116178A3 (ru) 2019-05-16
EP3203999A4 (en) 2018-06-27
PT3203999T (pt) 2020-04-23
ES2785317T3 (es) 2020-10-06
DK3203999T3 (da) 2020-03-30
US11185521B2 (en) 2021-11-30
RU2709205C2 (ru) 2019-12-17
MY194308A (en) 2022-11-27
KR20170066476A (ko) 2017-06-14
ZA201702639B (en) 2018-06-27
CN106999456A (zh) 2017-08-01
JP6700639B2 (ja) 2020-05-27
JP2017530180A (ja) 2017-10-12

Similar Documents

Publication Publication Date Title
US11185521B2 (en) Substituted aromatic compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis
EP1773315B1 (en) Arylacetic acids and related compounds for treatment of alzheimer&#39;s disease
DE602005002647T2 (de) Hexensäurederivate, verfahren zu deren herstellung, pharmazeutische zusammensetzungen, die diese enthalten, und therapeutische anwendungen davon
US8227456B2 (en) Pharmaceutical composition for preventing and treating metabolic bone diseases containing alpha-arylmethoxyacrylate derivatives
US20150313856A1 (en) Compounds and compositions for the treatment of cancer
JP4594099B2 (ja) ビタミンdレセプターモジュレーター
CN101434517B (zh) 治疗免疫疾病的1,2-二苯基乙烯衍生物
US11964938B2 (en) 3-phenyl-4-hexynoic acid derivatives as GPR40 agonists
US20080015253A1 (en) Pentenoic Acid Derivatives, Processes For The Preparation Thereof, Pharmaceutical Compositions Comprising Them, And Therapeutic Applications Thereof
US10925846B2 (en) Phenylketone carboxylate compounds and pharmaceutical compositions for the prevention and treatment of osteoporosis
KR20170066614A (ko) 당뇨병의 예방 및 치료를 위한 치환된 방향족 화합물 및 약제학적 조성물
PL233938B1 (pl) Pochodne kwasu 3-fenylo-4-heksynowego jako agoniści GPR40

Legal Events

Date Code Title Description
AS Assignment

Owner name: PROMETIC BIOSCIENCES INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GAGNON, LYNE;GROUIX, BRIGITTE;REEL/FRAME:042212/0565

Effective date: 20151009

Owner name: PRMOETIC PHARMA SMT LIMITED, GREAT BRITAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PROMETIC BIOSCIENCES INC.;REEL/FRAME:042212/0637

Effective date: 20160722

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

AS Assignment

Owner name: LIMINAL BIOSCIENCES LIMITED, UNITED KINGDOM

Free format text: CHANGE OF NAME;ASSIGNOR:PROMETIC PHARMA SMT LIMITED;REEL/FRAME:052501/0416

Effective date: 20191011

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION