WO2015171345A1 - N-aryl pyridinones modulators of fibrosis and/or collagen infiltration - Google Patents

N-aryl pyridinones modulators of fibrosis and/or collagen infiltration Download PDF

Info

Publication number
WO2015171345A1
WO2015171345A1 PCT/US2015/027765 US2015027765W WO2015171345A1 WO 2015171345 A1 WO2015171345 A1 WO 2015171345A1 US 2015027765 W US2015027765 W US 2015027765W WO 2015171345 A1 WO2015171345 A1 WO 2015171345A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
recited
fibrosis
deuterium
acid
Prior art date
Application number
PCT/US2015/027765
Other languages
French (fr)
Inventor
Chengzhi Zhang
Original Assignee
Auspex Pharmaceuticals, 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
Application filed by Auspex Pharmaceuticals, Inc. filed Critical Auspex Pharmaceuticals, Inc.
Publication of WO2015171345A1 publication Critical patent/WO2015171345A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/64One oxygen atom attached in position 2 or 6

Definitions

  • fibrosis and/or collagen infiltration in a subject are also provided for the treatment of disorders such as idiopathic pulmonary fibrosis, uterine fibroids, multiple sclerosis, renal fibrosis, diabetic kidney disease, endotoxin-induced liver injury after partial hepatectomy or hepatic ischemia, allograft injury after organ transplantation, cystic fibrosis, atrial fibrilation, neutropenia, scleroderma, dermatomyositis, cirrhosis, diffuse parenchymal lung disease, mediastinal fibrosis, tuberculosis, spleen fibrosis caused by sickle-cell anemia, rheumatoid arthritis, radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, radiation-
  • F-351 (F-351, CAS # 851518-71-3), l-(4-hydroxyphenyl)-5-methyl- 2(lH)-pyridinone, l-(4-fluorophenyl)-5-methylpyridin-2(lH)-one (CAS # 199386- 13-5), l-phenyl-5-(trifluoromethyl)pyridin-2(lH)-one (CAS # 914918-70-0), l-(4- hydroxyphenyl)-5-(trifluoromethyl)pyridin-2(lH)-one (CAS # 914918-71-1), and l-(4-fluorophenyl)-5-(trifluoromethyl)pyridin-2(lH)-one (CAS # 914918-72-2) (hereafter collectively referred to as "5 -methyl- l-phenylpyridin-2(lH)-one derivatives”) is a fibrosis and/or collagen infiltration modulator.
  • F-351 is under investigation for the treatment of fibrotic disorders such as radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, diabetic kidney disease, radiation-induced lung injury, fibrotic lesions, renal fibrosis, and interstitial cardiac fibrosis.
  • fibrotic disorders such as radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, diabetic kidney disease, radiation-induced lung injury, fibrotic lesions, renal fibrosis, and interstitial cardiac fibrosis.
  • 5-methyl-l- phenylpyridin-2(lH)-one derivatives have shown promise in treating fibrotic disorders such as radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, diabetic kidney disease, radiation- induced lung injury, fibrotic lesions, renal fibrosis, interstitial cardiac fibrosis, idiopathic pulmonary fibrosis, uterine fibroids, multiple sclerosis, renal fibrosis, endotoxin-induced liver injury after partial hepatectomy or hepatic ischemia, allograft injury after organ transplantation, cystic fibrosis, atrial fibrilation, neutropenia, scleroderma, dermatomyositis, cirrhosis, diffuse parenchymal lung disease, mediastinal fibrosis, tuberculosis, spleen fibrosis caused by sickle-cell anemia, rheumatoid arthritis, pulmonary fibrosis of Her
  • 5-methyl-l-phenylpyridin-2(lH)-one derivatives contain a number of features that we posit will produce inactive or toxic metabolites, the formation of which can be reduced by the approach described herein.
  • 5-methyl-l-phenylpyridin-2(lH)-one derivatives are likely metabolized via CYP45o-mediated oxidation of the methyl group and/or ring hydroxylation. These, as well as other metabolic transformations, occur in part through polymorphically- expressed enzymes, exacerbating interpatient variability.
  • 5-methyl-l-phenylpyridin-2(lH)-one derivatives may have undesirable side effects.
  • the drugs In order to overcome its short half-life, the drugs likely must be taken several times per day, which increases the probability of patient incompliance and discontinuance. Further, abruptly stopping treatment with 5- methyl-l-phenylpyridin-2(lH)-one derivatives can lead to withdrawal or discontinuation syndrome. Medicines with longer half-lives will likely attenuate these deleterious effects.
  • the animal body expresses various enzymes, such as the cytochrome P450 enzymes (CYPs), esterases, proteases, reductases, dehydrogenases, and monoamine oxidases, to react with and convert these foreign substances to more polar intermediates or metabolites for renal excretion.
  • CYPs cytochrome P450 enzymes
  • esterases proteases
  • reductases reductases
  • dehydrogenases dehydrogenases
  • monoamine oxidases monoamine oxidases
  • Such metabolic reactions frequently involve the oxidation of a carbon-hydrogen (C-H) bond to either a carbon-oxygen (C-O) or a carbon-carbon (C-C) ⁇ -bond.
  • C-H carbon-hydrogen
  • C-O carbon-oxygen
  • C-C carbon-carbon
  • the resultant metabolites may be stable or unstable under physiological conditions, and can have substantially different
  • the Arrhenius equation states that, at a given temperature, the rate of a chemical reaction depends exponentially on the activation energy (Eact).
  • the transition state in a reaction is a short lived state along the reaction pathway during which the original bonds have stretched to their limit.
  • the activation energy Eact for a reaction is the energy required to reach the transition state of that reaction. Once the transition state is reached, the molecules can either revert to the original reactants, or form new bonds giving rise to reaction products.
  • a catalyst facilitates a reaction process by lowering the activation energy leading to a transition state. Enzymes are examples of biological catalysts.
  • Carbon-hydrogen bond strength is directly proportional to the absolute value of the ground-state vibrational energy of the bond. This vibrational energy depends on the mass of the atoms that form the bond, and increases as the mass of one or both of the atoms making the bond increases. Since deuterium (D) has twice the mass of protium (3 ⁇ 4), a C-D bond is stronger than the corresponding C- l R bond. If a C- 1 !! bond is broken during a rate-determining step in a chemical reaction (i.e. the step with the highest transition state energy), then substituting a deuterium for that protium will cause a decrease in the reaction rate. This phenomenon is known as the Deuterium Kinetic Isotope Effect (DKIE).
  • DKIE Deuterium Kinetic Isotope Effect
  • the magnitude of the DKIE can be expressed as the ratio between the rates of a given reaction in which a C- 1 ! bond is broken, and the same reaction where deuterium is substituted for protium.
  • the DKIE can range from about 1 (no isotope effect) to very large numbers, such as 50 or more. Substitution of tritium for hydrogen results in yet a stronger bond than deuterium and gives numerically larger isotope effects [0009]
  • Deuterium ( 2 H or D) is a stable and non-radioactive isotope of hydrogen which has approximately twice the mass of protium (3 ⁇ 4), the most common isotope of hydrogen.
  • Deuterium oxide (D2O or "heavy water”) looks and tastes like H2O, but has different physical properties.
  • Metabolic switching is enabled by the relatively vast size of binding pockets in many Phase I enzymes and the promiscuous nature of many metabolic reactions. Metabolic switching can lead to different proportions of known metabolites as well as altogether new metabolites. This new metabolic profile may impart more or less toxicity. Such pitfalls are non- obvious and are not predictable a priori for any drug class.
  • 5 -methyl- l-phenylpyridin-2(lH)-one derivatives are fibrosis and/or collagen infiltration modulators.
  • the carbon-hydrogen bonds of 5-methyl-l- phenylpyridin-2(lH)-one derivatives contain a naturally occurring distribution of hydrogen isotopes, namely l H or protium (about 99.9844%), 2 H or deuterium (about 0.0156%), and 3 H or tritium (in the range between about 0.5 and 67 tritium atoms per 10 18 protium atoms).
  • DKIE Deuterium Kinetic Isotope Effect
  • a medicine with a longer half-life may result in greater efficacy and cost savings.
  • Various deuteration patterns can be used to (a) reduce or eliminate unwanted metabolites, (b) increase the half-life of the parent drug, (c) decrease the number of doses needed to achieve a desired effect, (d) decrease the amount of a dose needed to achieve a desired effect, (e) increase the formation of active metabolites, if any are formed, (f) decrease the production of deleterious metabolites in specific tissues, and/or (g) create a more effective drug and/or a safer drug for
  • Novel compounds and pharmaceutical compositions certain of which have been found to modulate fibrosis and/or collagen infiltration have been discovered, together with methods of synthesizing and using the compounds, including methods for the treatment of fibrosis-mediated disorders and/or collagen infiltration-mediated disorders in a patient by administering the compounds.
  • Ri is selected from the group consisting of -CH3, -CH2D, -CD2H, -CD3, and
  • R2-R8 are independently selected from the group consisting of hydrogen and deuterium
  • R9 is selected from the group consisting of hydrogen, deuterium, fluorine, chlorine, -OH, and -OD;
  • R1-R9 is deuterium or contains deuterium
  • Ri is -CF3.
  • Certain compounds disclosed herein may possess useful fibrosis and/or collagen infiltration modulating activity, and may be used in the treatment or prophylaxis of a disorder in which fibrosis and/or collagen infiltration plays an active role.
  • certain embodiments also provide pharmaceutical compositions comprising one or more compounds disclosed herein together with a
  • Certain embodiments provide methods for modulating fibrosis and/or collagen infiltration.
  • Other embodiments provide methods for treating a fibrosis-mediated disorder and/or a collagen infiltration- mediated disorder in a patient in need of such treatment, comprising administering to said patient a therapeutically effective amount of a compound or composition according to the present invention.
  • certain compounds disclosed herein for use in the manufacture of a medicament for the prevention or treatment of a disorder ameliorated by the modulation of fibrosis and/or collagen infiltration.
  • composition comprising compounds of structural Formula I:
  • Ri is selected from the group consisting of -CH3, -CH2D, -CD2H, -CD3, and
  • R2-R8 are independently selected from the group consisting of hydrogen and deuterium
  • R9 is selected from the group consisting of hydrogen, deuterium, fluorine, chlorine, -OH, and -OD;
  • At least one of the positions R1-R9 is enriched with deuterium
  • Ri is -CF3.
  • the compounds as disclosed herein may also contain less prevalent isotopes for other elements, including, but not limited to, 13 C or 14 C for carbon, 33 S, 34 S, or 36 S for sulfur, 15 N for nitrogen, and 17 0 or 18 0 for oxygen.
  • the compound disclosed herein may expose a patient to a maximum of about 0.000005% D2O or about 0.00001% DHO, assuming that all of the C-D bonds in the compound as disclosed herein are metabolized and released as D2O or DHO.
  • the levels of D2O shown to cause toxicity in animals is much greater than even the maximum limit of exposure caused by administration of the deuterium enriched compound as disclosed herein.
  • the deuterium-enriched compound disclosed herein should not cause any additional toxicity due to the formation of D2O or DHO upon drug metabolism.
  • the deuterated compounds disclosed herein maintain the beneficial aspects of the corresponding non-isotopically enriched molecules while substantially increasing the maximum tolerated dose, decreasing toxicity, increasing the half-life (T1/2), lowering the maximum plasma concentration (Cmax) of the minimum efficacious dose (MED), lowering the efficacious dose and thus decreasing the non-mechanism-related toxicity, and/or lowering the probability of drug-drug interactions.
  • All publications and references cited herein are expressly incorporated herein by reference in their entirety. However, with respect to any similar or identical terms found in both the incorporated publications or references and those explicitly put forth or defined in this document, then those terms definitions or meanings explicitly put forth in this document shall control in all respects.
  • deuterium enrichment refers to the percentage of incorporation of deuterium at a given position in a molecule in the place of hydrogen. For example, deuterium enrichment of 1% at a given position means that 1% of molecules in a given sample contain deuterium at the specified position. Because the naturally occurring distribution of deuterium is about 0.0156%, deuterium enrichment at any position in a compound synthesized using non- enriched starting materials is about 0.0156%. The deuterium enrichment can be determined using conventional analytical methods known to one of ordinary skill in the art, including mass spectrometry and nuclear magnetic resonance spectroscopy.
  • deuterium when used to describe a given position in a molecule such as R1-R9 or the symbol "D", when used to represent a given position in a drawing of a molecular structure, means that the specified position is enriched with deuterium above the naturally occurring distribution of deuterium.
  • deuterium enrichment is no less than about 1%, in another no less than about 5%, in another no less than about 10%, in another no less than about 20%, in another no less than about 50%, in another no less than about 70%, in another no less than about 80%, in another no less than about 90%, or in another no less than about 98% of deuterium at the specified position.
  • isotopic enrichment refers to the percentage of incorporation of a less prevalent isotope of an element at a given position in a molecule in the place of the more prevalent isotope of the element.
  • non-isotopically enriched refers to a molecule in which the percentages of the various isotopes are substantially the same as the naturally occurring percentages.
  • bond refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure.
  • a bond may be single, double, or triple unless otherwise specified.
  • a dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
  • disorder as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disease” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms.
  • treat are meant to include alleviating or abrogating a disorder or one or more of the symptoms associated with a disorder; or alleviating or eradicating the cause(s) of the disorder itself.
  • treatment'Of a disorder is intended to include prevention.
  • prevent refer to a method of delaying or precluding the onset of a disorder; and/or its attendant symptoms, barring a subject from acquiring a disorder or reducing a subject's risk of acquiring a disorder.
  • terapéuticaally effective amount refers to the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disorder being treated.
  • therapeutically effective amount also refers to the amount of a compound that is sufficient to elicit the biological or medical response of a cell, tissue, system, animal, or human that is being sought by a researcher, veterinarian, medical doctor, or clinician.
  • subject refers to an animal, including, but not limited to, a primate (e.g., human, monkey, chimpanzee, gorilla, and the like), rodents (e.g., rats, mice, gerbils, hamsters, ferrets, and the like), lagomorphs, swine (e.g., pig, miniature pig), equine, canine, feline, and the like.
  • a primate e.g., human, monkey, chimpanzee, gorilla, and the like
  • rodents e.g., rats, mice, gerbils, hamsters, ferrets, and the like
  • lagomorphs e.g., pig, miniature pig
  • swine e.g., pig, miniature pig
  • equine canine
  • feline feline
  • combination therapy means the administration of two or more therapeutic agents to treat a therapeutic disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the disorders described herein.
  • fibrosis refers to the development of excessive fibrous connective tissue within an organ or tissue.
  • collagen infiltration refers to the entry of the connective tissue collagen into cells or into the extracellular matrix around cells. This occurs in organs and tissues naturally and under normal circumstances but can occur excessively and accompany or cause disease.
  • fibrosis and “collagen infiltration” are not necessarily synonymous but can, in certain contexts, be used interchangeably.
  • collagen infiltration-mediated disorder refers to a disorder that is characterized by abnormal or undesired collagenic infiltration, that when collagen infiltration activity is modified, leads to the desired responses depending on the route of administration and desired end result.
  • a collagen-mediated disorder may be completely or partially mediated through the modulation of collagen infiltration.
  • a collagen-mediated disorder is one in which modulation of collagen infiltration activity results in some effect on the underlying disorder, e.g., administering a collagen-infiltration modulator results in some improvement in at least some of the patients being treated.
  • fibrosis-mediated disorder refers to a disorder that is characterized by abnormal or undesired fibrotic activity, that when fibrosis activity is modified, leads to the desired responses depending on the route of administration and desired end result.
  • a fibrosis- mediated disorder may be completely or partially mediated through the modulation of fibrosis.
  • a fibrosis- mediated disorder is one in which modulation of fibrosis activity results in some effect on the underlying disorder, e.g., administering a fibrosis modulator results in some improvement in at least some of the patients being treated.
  • fibrosis modulator or “modulating fibrosis” are meant to be interchangeable and refer to the ability of a compound disclosed herein to alter the occurrence and/or amount of fibrosis.
  • a fibrosis modulator may increase the occurrence or level of fibrosis, may increase or decrease the occurrence and/or amount of fibrosis depending on the concentration of the compound exposed to the adrenergic receptor, or may decrease the occurrence and/or amount of fibrosis.
  • Such activation or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
  • collagen infiltration modulator or “modulating collagen infiltration” are meant to be interchangeable and refer to the ability of a compound disclosed herein to alter the occurrence and/or amount of collagen infiltration.
  • a fibrosis modulator may increase the occurrence or level of collagen infiltration, may increase or decrease the occurrence and/or amount of collagen infiltration depending on the concentration of the compound exposed to the adrenergic receptor, or may decrease the occurrence and/or amount of collagen infiltration.
  • Such activation or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
  • fibrosis and/or collagen infiltration-mediated disorder refers to a disorder that is characterized by the development of excessive fibrous connective tissue within an organ or tissue or abnormal entry of the connective tissue collagen into cells or into the extracellular matrix around cells.
  • a fibrosis and/or collagen infiltration-mediated disorder may be completely or partially mediated by modulating fibrosis and/or collagen infiltration.
  • a fibrosis and/or collagen infiltration-mediated disorder is one in which modulation of fibrosis and/or collagen infiltration results in some effect on the underlying disorder e.g., administration of a fibrosis and/or collagen infiltration modulator results in some improvement in at least some of the patients being treated.
  • modulation of fibrosis and/or collagen infiltration may be assessed using the method described in Gosselin et al, Muscle & Nerve, 2007, 35(2), 208-216; US 20080161361 ; WO 2007053685: and WO 2007062167.
  • terapéuticaally acceptable refers to those compounds (or salts, prodrugs, tautomers, zwitterionic forms, etc.) which are suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, immunogenecity, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • pharmaceutically acceptable carrier refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material.
  • pharmaceutically-acceptable material such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material.
  • Each component must be “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation. It must also be suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenecity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • active ingredient refers to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients or carriers, to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder.
  • drug refers to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder.
  • release controlling excipient refers to an excipient whose primary function is to modify the duration or place of release of the active substance from a dosage form as compared with a conventional immediate release dosage form.
  • nonrelease controlling excipient refers to an excipient whose primary function do not include modifying the duration or place of release of the active substance from a dosage form as compared with a conventional immediate release dosage form.
  • prodrug refers to a compound functional derivative of the compound as disclosed herein and is readily convertible into the parent compound in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound. A prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221-294; Morozowich et al. in "Design of Biopharmaceutical Properties through Prodrugs and Analogs," Roche Ed., APHA Acad. Pharm. Sci. 1977; "Bioreversible Carriers in Drug in Drug Design, Theory and Application,” Roche Ed., APHA Acad. Pharm. Sci. 1987; "Design of
  • the compounds disclosed herein can exist as therapeutically acceptable salts.
  • the term "therapeutically acceptable salt,” as used herein, represents salts or zwitterionic forms of the compounds disclosed herein which are therapeutically acceptable as defined herein.
  • the salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound with a suitable acid or base.
  • Therapeutically acceptable salts include acid and basic addition salts.
  • Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid,
  • benzenesulfonic acid benzoic acid, 4-acetamidobenzoic acid, boric acid, (+)- camphoric acid, camphorsulfonic acid, (+)-(lS)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, D-gluconic acid, D-glucuronic acid, L-glutamic acid, a-oxo-glutaric acid, glycolic acid, hippuric acid,
  • Suitable bases for use in the preparation of pharmaceutically acceptable salts including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2- (diethylamino)-ethanol, ethanolamine, ethylamine, ethylenediamine,
  • inorganic bases such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide
  • organic bases such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine,
  • compositions which comprise one or more of certain compounds disclosed herein, or one or more pharmaceutically acceptable salts, prodrugs, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients.
  • pharmaceutical compositions which comprise one or more of certain compounds disclosed herein, or one or more pharmaceutically acceptable salts, prodrugs, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients.
  • Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences.
  • compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • the pharmaceutical compositions may also be formulated as a modified release dosage form, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modifled-Release Drug Deliver Technology, Rathbone et al, Eds., Drugs and the Pharmaceutical Science, Marcel Dekker, Inc.: New York, NY, 2002; Vol. 126).
  • compositions include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient.
  • parenteral including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary
  • intraperitoneal including transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient.
  • topical including dermal, buccal, sublingual and intraocular
  • these methods include the step of bringing into association a compound of the subject invention or a pharmaceutically salt, prodrug, or solvate thereof ("active ingredient") with the carrier which constitutes one or more accessory ingredients.
  • active ingredient a compound of the subject invention or a pharmaceutically salt, prodrug, or solvate thereof
  • the carrier which constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • Formulations of the compounds disclosed herein suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a nonaqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • compositions which can be used orally include tablets, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Dragee cores are provided with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried
  • lyophilized condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen- free water, immediately prior to use.
  • sterile liquid carrier for example, saline or sterile pyrogen- free water
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Formulations for parenteral administration include aqueous and nonaqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneous ly or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner. Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
  • Certain compounds disclosed herein may be administered topically, that is by non-systemic administration. This includes the application of a compound disclosed herein externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream.
  • systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
  • Formulations suitable for topical administration include liquid or semi- liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose.
  • compounds may be delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray.
  • Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
  • Preferred unit dosage formulations are those containing an effective dose, as herein below recited, or an appropriate fraction thereof, of the active ingredient.
  • Compounds may be administered orally or via injection at a dose of from 0.1 to 500 mg/kg per day.
  • the dose range for adult humans is generally from 5 mg to 2 g/day.
  • Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of one or more compounds which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the compounds can be administered in various modes, e.g. orally, topically, or by injection.
  • the precise amount of compound administered to a patient will be the responsibility of the attendant physician.
  • the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of administration, route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the disorder being treated. Also, the route of administration may vary depending on the disorder and its severity.
  • the administration of the compounds may be administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disorder.
  • the administration of the compounds may be given continuously or temporarily suspended for a certain length of time (i.e., a "drug holiday").
  • a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, can be reduced, as a function of the symptoms, to a level at which the improved disorder is retained. Patients can, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • Disclosed herein are methods of treating a fibrosis-mediated disorder and/or a collagen infiltration-mediated disorder comprising administering to a subject having or suspected to have such a disorder, a therapeutically effective amount of a compound as disclosed herein or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • Fibrosis and/or collagen infiltration-mediated disorders include, but are not limited to, idiopathic pulmonary fibrosis, uterine fibroids, multiple sclerosis, renal fibrosis, diabetic kidney disease, endotoxin-induced liver injury after partial hepatectomy or hepatic ischemia, allograft injury after organ transplantation, cystic fibrosis, atrial fibrilation, neutropenia, scleroderma, dermatomyositis, cirrhosis, diffuse parenchymal lung disease, mediastinal fibrosis, tuberculosis, spleen fibrosis caused by sickle-cell anemia, rheumatoid arthritis, radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, radiation-induced lung injury, fibrotic lesions, pulmonary fibrosis of Hermansky- Pudlak Syndrome, focal segmental glomerulosclerosis, hypertrophic
  • cardiomyopathy neurofibromatosis, interstitial cardiac fibrosis, and/or any disorder which can lessened, alleviated, or prevented by administering a fibrosis and/or collagen infiltration modulator.
  • a method of treating a fibrosis and/or collagen infiltration-mediated disorder comprises administering to the subject a
  • a compound of as disclosed herein or a pharmaceutically acceptable salt, solvate, or prodrug thereof, so as to affect: (1) decreased inter- individual variation in plasma levels of the compound or a metabolite thereof; (2) increased average plasma levels of the compound or decreased average plasma levels of at least one metabolite of the compound per dosage unit; (3) decreased inhibition of, and/or metabolism by at least one cytochrome P450 or monoamine oxidase isoform in the subject; (4) decreased metabolism via at least one polymorphically-expressed cytochrome P450 isoform in the subject; (5) at least one statistically-significantly improved disorder-control and/or disorder-eradication endpoint; (6) an improved clinical effect during the treatment of the disorder, (7) prevention of recurrence, or delay of decline or appearance, of abnormal alimentary or hepatic parameters as the primary clinical benefit, or (8) reduction or elimination of deleterious changes in any diagnostic hepatobiliary function endpoints, as compared to the corresponding non- isotop
  • inter-individual variation in plasma levels of the compounds as disclosed herein, or metabolites thereof is decreased; average plasma levels of the compound as disclosed herein are increased; average plasma levels of a metabolite of the compound as disclosed herein are decreased; inhibition of a cytochrome P450 or monoamine oxidase isoform by a compound as disclosed herein is decreased; or metabolism of the compound as disclosed herein by at least one polymorphically-expressed cytochrome P450 isoform is decreased; by greater than about 5%, greater than about 10%, greater than about 20%, greater than about 30%, greater than about 40%, or by greater than about 50% as compared to the corresponding non-isotopically enriched compound.
  • Plasma levels of the compound as disclosed herein, or metabolites thereof, may be measured using the methods described by Li et al. Rapid
  • Examples of cytochrome P450 isoforms in a mammalian subject include, but are not limited to, CYP1A1, CYP1A2, CYP1B 1, CYP2A6, CYP2A13, CYP2B6, CYP2C8, CYP2C9, CYP2C18, CYP2C19, CYP2D6, CYP2E1, CYP2G1, CYP2J2, CYP2R1, CYP2S1, CYP3A4, CYP3A5, CYP3A5P 1, CYP3A5P2, CYP3A7, CYP4A11, CYP4B 1, CYP4F2, CYP4F3, CYP4F8, CYP4F1 1, CYP4F12, CYP4X1, CYP4Z1, CYP5A1, CYP7A1, CYP7B 1, CYP8A1, CYP8B 1,
  • Examples of monoamine oxidase isoforms in a mammalian subject include, but are not limited to, MAOA, and MAOB.
  • the inhibition of the cytochrome P450 isoform is measured by the method of Ko et al. (British Journal of Clinical Pharmacology, 2000, 49, 343-351).
  • the inhibition of the MAOA isoform is measured by the method of Weyler et al. (J. Biol Chem. 1985, 260, 13199-13207).
  • the inhibition of the MAOB isoform is measured by the method of Uebelhack et al. (Pharmacopsychiatry, 1998, 31, 187- 192).
  • Examples of polymorphically-expressed cytochrome P450 isoforms in a mammalian subject include, but are not limited to, CYP2C8, CYP2C9, CYP2C19, and CYP2D6.
  • liver microsomes cytochrome P450 isoforms
  • monoamine oxidase isoforms are measured by the methods described herein.
  • improved disorder-control and/or disorder-eradication endpoints include, but are not limited to, change in forced vital capacity, change in pulmonary function parameters, progression to renal failure, reduced proteinuria, progression-free survival, change in shortness-of- breath, change in oxygen saturation during the six minute walk test, change in distance walked during the six minute walk test, tumor volume, GFR as calculated using forty -variable Levey equation, and reduced adverse effects as compared to the non-isotopically enriched compound.
  • diagnostic hepatobiliary function endpoints include, but are not limited to, alanine aminotransferase ("ALT”), serum glutamic-pyruvic transaminase (“SGPT”), aspartate aminotransferase ("AST" or "SGOT”),
  • ALT alanine aminotransferase
  • SGPT serum glutamic-pyruvic transaminase
  • AST aspartate aminotransferase
  • SGOT aspartate aminotransferase
  • ALT/AST ratios serum aldolase, alkaline phosphatase ("ALP"), ammonia levels, bilirubin, gamma-glutamyl transpeptidase ("GGTP,” “ ⁇ -GTP,” or “GGT”), leucine aminopeptidase (“LAP”), liver biopsy, liver ultrasonography, liver nuclear scan, 5'- nucleotidase, and blood protein. Hepatobiliary endpoints are compared to the stated normal levels as given in "Diagnostic and Laboratory Test Reference", 4 th edition, Mosby, 1999. These assays are run by accredited laboratories according to standard protocol.
  • certain compounds and formulations disclosed herein may also be useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
  • the compounds disclosed herein may also be combined or used in combination with other agents useful in the treatment of fibrosis-mediated disorders and/or collagen infiltration-mediated disorders.
  • the therapeutic effectiveness of one of the compounds described herein may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced).
  • Such other agents, adjuvants, or drugs may be administered, by a route and in an amount commonly used therefor, simultaneously or sequentially with a compound as disclosed herein.
  • a pharmaceutical composition containing such other drugs in addition to the compound disclosed herein may be utilized, but is not required.
  • the compounds disclosed herein can be combined with one or more steroidal drugs and non-steroidal anti-inflammatory drugs.
  • the compounds disclosed herein can be combined with a steroidal drug selected from the group consisting of aldosterone, beclometasone, betamethasone, deoxycorticosterone acetate, fludrocortisone acetate, hydrocortisone, prednisolone, prednisone, methylprenisolone,
  • a steroidal drug selected from the group consisting of aldosterone, beclometasone, betamethasone, deoxycorticosterone acetate, fludrocortisone acetate, hydrocortisone, prednisolone, prednisone, methylprenisolone,
  • dexamethasone and triamcinolone.
  • the compounds disclosed herein can be combined with a non-steroidal anti-inflammatory drug selected from the group consisting of aceclofenac, acemetacin, amoxiprin, aspirin, azapropazone, benorilate, bromfenac, carprofen, celecoxib, choline magnesium salicylate, diclofenac, diflunisal, etodolac, etoracoxib, chloramine, fenbuten, fenoprofen, flurbiprofen, ibuprofen, indometacin, ketoprofen, ketorolac, lornoxicam, loxoprofen,
  • a non-steroidal anti-inflammatory drug selected from the group consisting of aceclofenac, acemetacin, amoxiprin, aspirin, azapropazone, benorilate, bromfenac, carprofen, celecoxib, choline magnesium salicylate, diclofenac, diflun
  • lumiracoxib meclofenamic acid, mefenamic acid, meloxicam, metamizole, methyl salicylate, magnesium salicylate, nabumetone, naproxen, nimesulide,
  • oxyphenbutazone parecoxib, phenylbutazone, piroxicam, salicyl salicylate, sulindac, sulfinprazone, suprofen, tenoxicam, tiaprofenic acid, and tolmetin.
  • norepinephrine reuptake inhibitors such as atomoxetine
  • DARIs dopamine reuptake inhibitors
  • SNRIs serotonin-norepinephrine reuptake inhibitors
  • sedatives such as diazepham
  • NDRIs norepinephrine-dopamine reuptake inhibitor
  • SNDRIs serotonin-norepinephrine-dopamine-reuptake-inhibitors
  • SNDRIs serotonin-norepinephrine-dopamine-reuptake-inhibitors
  • monoamine oxidase inhibitors such as selegiline
  • hypothalamic phospholipids such as hypothalamic phospholipids
  • ECE endothelin converting enzyme
  • phosphoramidon opioids, such as tramadol; thromboxane receptor antagonists, such as ifetroban; potassium channel openers; thrombin inhibitors, such as hirudin; hypothalamic phospholipids; growth factor inhibitors, such as modulators of PDGF activity; platelet activating factor (PAF) antagonists; anti-platelet agents, such as GPIIb/IIIa blockers (e.g., abdximab, eptifibatide, and tirofiban), P2Y(AC) antagonists (e.g., clopidogrel, ticlopidine and CS-747), and aspirin; anticoagulants, such as warfarin; low molecular weight heparins, such as enoxaparin; Factor Vila Inhibitors and Factor Xa Inhibitors; renin inhibitors; neutral endopeptidase (NEP) inhibitors; vasopepsidase inhibitors (dual NEP-ACE inhibitors
  • squalene synthetase inhibitors include fibrates; bile acid sequestrants, such as questran; niacin; anti- atherosclerotic agents, such as ACAT inhibitors; MTP Inhibitors; calcium channel blockers, such as amlodipine besylate; potassium channel activators; alpha- muscarinic agents; beta-muscarinic agents, such as carvedilol and metoprolol; antiarrhythmic agents; diuretics, such as chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichioromethiazide, polythiazide, benzothlazide, ethacrynic acid,
  • metformin glucosidase inhibitors
  • glucosidase inhibitors e.g., acarbose
  • insulins meglitinides (e.g., repaglinide)
  • meglitinides e.g., repaglinide
  • sulfonylureas e.g., glimepiride, glyburide, and glipizide
  • thiozolidinediones e.g.
  • troglitazone, rosiglitazone and pioglitazone), and PPAR-gamma agonists mineralocorticoid receptor antagonists, such as spironolactone and eplerenone; growth hormone secretagogues; aP2 inhibitors; phosphodiesterase inhibitors, such as PDE III inhibitors (e.g., cilostazol) and PDE V inhibitors (e.g., sildenafil, tadalafil, vardenafil); protein tyrosine kinase inhibitors; antiinflammatories; antiproliferatives, such as methotrexate, FK506 (tacrolimus, Prograf), mycophenolate mofetil; chemotherapeutic agents;
  • mineralocorticoid receptor antagonists such as spironolactone and eplerenone
  • growth hormone secretagogues aP2 inhibitors
  • phosphodiesterase inhibitors such as PDE III inhibitors
  • immunosuppressants include anticancer agents and cytotoxic agents (e.g., alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes); antimetabolites, such as folate antagonists, purine analogues, and pyrridine analogues; antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes, such as L-asparaginase; farnesyl-protein transferase inhibitors; hormonal agents, such as glucocorticoids (e.g., cortisone), estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone anatagonists, and octreotide acetate; microtubule- disruptor agents, such as ecteinascidins; microtub
  • certain embodiments provide methods for treating fibrosis-mediated disorders and/or collagen infiltration-mediated disorders in a human or animal subject in need of such treatment comprising administering to said subject an amount of a compound disclosed herein effective to reduce or prevent said disorder in the subject, in combination with at least one additional agent for the treatment of said disorder that is known in the art.
  • certain embodiments provide therapeutic compositions comprising at least one compound disclosed herein in combination with one or more additional agents for the treatment of fibrosis-mediated disorders and/or collagen infiltration-mediated disorders.
  • Isotopic hydrogen can be introduced into a compound as disclosed herein by synthetic techniques that employ deuterated reagents, whereby incorporation rates are pre-determined; and/or by exchange techniques, wherein incorporation rates are determined by equilibrium conditions, and may be highly variable depending on the reaction conditions.
  • Synthetic techniques where tritium or deuterium is directly and specifically inserted by tritiated or deuterated reagents of known isotopic content, may yield high tritium or deuterium abundance, but can be limited by the chemistry required.
  • Exchange techniques on the other hand, may yield lower tritium or deuterium incorporation, often with the isotope being distributed over many sites on the molecule.
  • Compound 1 is reacted with compound 2 in the presence of an appropriate metalating agent, such as n-butyl lithium, in an appropriate solvent, such as tetrahydrofuran, to give compound 3.
  • an appropriate metalating agent such as n-butyl lithium
  • an appropriate solvent such as tetrahydrofuran
  • Compound 3 is treated with an appropriate acid, such as hydrochloric acid, in an appropriate solvent, such as a combination of water and 1,4-dioxane, to give compound 4.
  • Compound 4 is reacted with compound 5, in the presence of an appropriate catalyst, such as cupric acetate, in the presence of an appropriate base, such as triethylamine, in an appropriate solvent, such as tetrahydrofuran, to give compound 6 of formula I.
  • Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme I, by using appropriate deuterated intermediates.
  • compound 1 with the corresponding deuterium substitutions can be used.
  • compound 2 with the corresponding deuterium substitutions can be used.
  • compound 5 with the corresponding deuterium substitutions can be used.
  • Deuterium can be incorporated to various positions having an exchangeable proton, such as the hydroxyl O-H, via proton-deuterium equilibrium exchange.
  • an exchangeable proton such as the hydroxyl O-H
  • this proton may be replaced with deuterium selectively or non-selectively through a proton- deuterium exchange method known in the art.
  • Compound 4 is reacted with compound 8, in the presence of an appropriate catalyst, such as cupric acetate, in the presence of an appropriate base, such as triethylamine, in an appropriate solvent, such as tetrahydrofuran, to give compound 9.
  • an appropriate catalyst such as cupric acetate
  • an appropriate base such as triethylamine
  • an appropriate solvent such as tetrahydrofuran
  • Compound 9 is treated with an appropriate acid, such as hydrochloric acid, in an appropriate solvent, such as a combination of water and 1,4-dioxane, to give compound 6 of formula I.
  • Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme II, by using appropriate deuterated intermediates.
  • compound 4 with the corresponding deuterium substitutions can be used.
  • compound 9 with the corresponding deuterium substitutions can be used.
  • Deuterium can be incorporated to various positions having an exchangeable proton, such as the hydroxyl O-H, via proton-deuterium equilibrium exchange.
  • an exchangeable proton such as the hydroxyl O-H
  • this proton may be replaced with deuterium selectively or non-selectively through a proton- deuterium exchange method known in the art.
  • Compound 10 is reacted with compound 11 in the presence of an appropriate base, such as sodium hydride, in an appropriate solvent, such as toluene, to give compound 12.
  • Compound 12 is reacted with an appropriate deprotecting reagent, such as a combination of hydrogen gas and an appropriate catalyst, such as palladium on carbon, in an appropriate solvent, such as methanol, to give compound 4.
  • Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme III, by using appropriate deuterated intermediates. For example, to introduce deuterium at one or more positions of R1-R4, compound 8 with the corresponding deuterium substitutions can be used.
  • Compound 13 is reacted with compound 11 in the presence of an appropriate base, such as potassium tert-butoxide, to give compound 14.
  • Compound 14 is reacted with compound 2 in the presence of an appropriate metalating agent, such as n-butyl lithium, in an appropriate solvent, such as tetrahydrofuran, to give compound 4.
  • Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme IV, by using appropriate deuterated intermediates.
  • compound 13 with the corresponding deuterium substitutions can be used.
  • compound 2 with the corresponding deuterium substitutions can be used.
  • Compound 15 is reacted with compound 16 in the presence of 1.1 equivalents of n-butyl lithium in tetrahydrofuran at -40°C to -20°C, to give compound 17.
  • Compound 17 is refluxed in a 1 : 1 mixture of concentrated aqueous hydrochloric acid and 1,4-dioxane, to give compound 18.
  • Compound 18 is reacted with compound 19, in the presence of cupric acetate and triethylamine in tetrahydrofuran at room temperature to give compound 20 of formula I.
  • Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme IX, by using appropriate deuterated intermediates.
  • compound 4 with the corresponding deuterium substitutions can be used.
  • compound 27 with the corresponding deuterium substitutions can be used.
  • Liver microsomal stability assays are conducted at 1 mg per mL liver microsome protein with an NADPH-generating system in 2% NaHCC (2.2 mM NADPH, 25.6 mM glucose 6-phosphate, 6 units per mL glucose 6-phosphate dehydrogenase and 3.3 mM MgCk).
  • Test compounds are prepared as solutions in 20% acetonitrile-water and added to the assay mixture (final assay concentration 5 microgram per mL) and incubated at 37 °C. Final concentration of acetonitrile in the assay should be ⁇ 1%. Aliquots (50 ⁇ ) are taken out at times 0, 15, 30, 45, and 60 min, and diluted with ice cold acetonitrile (200 ⁇ ) to stop the reactions.
  • the cytochrome P450 enzymes are expressed from the corresponding human cDNA using a baculovirus expression system (BD Biosciences, San Jose, CA).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to new N-Aryi pyridinones modulators of fibrosis and/or collagen infiltration, pharmaceutical compositions thereof, and methods of use thereof. Disclosed herein are new N-aryl pyridinones compounds and compositions and their application as pharmaceuticals for the treatment of disorders. Methods of modulation of fibrosis and/or collagen infiltration in a subject are also provided for the treatment of disorders.

Description

N-ARYL PYRIDINONES MODULATORS OF FIBROSIS AND/OR COLLAGEN INFILTRATION
[0001] This application claims the benefit of priority of United States provisional application No. 61/986,454, filed April 30, 2014, the disclosure of which is hereby incorporated by reference as if written herein in its entirety.
[0002] Disclosed herein are new N-aryl pyridinones compounds and compositions and their application as pharmaceuticals for the treatment of disorders. Methods of modulation of fibrosis and/or collagen infiltration in a subject are also provided for the treatment of disorders such as idiopathic pulmonary fibrosis, uterine fibroids, multiple sclerosis, renal fibrosis, diabetic kidney disease, endotoxin-induced liver injury after partial hepatectomy or hepatic ischemia, allograft injury after organ transplantation, cystic fibrosis, atrial fibrilation, neutropenia, scleroderma, dermatomyositis, cirrhosis, diffuse parenchymal lung disease, mediastinal fibrosis, tuberculosis, spleen fibrosis caused by sickle-cell anemia, rheumatoid arthritis, radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, radiation- induced lung injury, fibrotic lesions, pulmonary fibrosis of Hermansky-Pudlak Syndrome, focal segmental glomerulosclerosis, hypertrophic cardiomyopathy, neurofibromatosis, and interstitial cardiac fibrosis.
[0003] F-351 (F-351, CAS # 851518-71-3), l-(4-hydroxyphenyl)-5-methyl- 2(lH)-pyridinone, l-(4-fluorophenyl)-5-methylpyridin-2(lH)-one (CAS # 199386- 13-5), l-phenyl-5-(trifluoromethyl)pyridin-2(lH)-one (CAS # 914918-70-0), l-(4- hydroxyphenyl)-5-(trifluoromethyl)pyridin-2(lH)-one (CAS # 914918-71-1), and l-(4-fluorophenyl)-5-(trifluoromethyl)pyridin-2(lH)-one (CAS # 914918-72-2) (hereafter collectively referred to as "5 -methyl- l-phenylpyridin-2(lH)-one derivatives") is a fibrosis and/or collagen infiltration modulator. F-351 is under investigation for the treatment of fibrotic disorders such as radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, diabetic kidney disease, radiation-induced lung injury, fibrotic lesions, renal fibrosis, and interstitial cardiac fibrosis. US 20080161361. 5-methyl-l- phenylpyridin-2(lH)-one derivatives have shown promise in treating fibrotic disorders such as radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, diabetic kidney disease, radiation- induced lung injury, fibrotic lesions, renal fibrosis, interstitial cardiac fibrosis, idiopathic pulmonary fibrosis, uterine fibroids, multiple sclerosis, renal fibrosis, endotoxin-induced liver injury after partial hepatectomy or hepatic ischemia, allograft injury after organ transplantation, cystic fibrosis, atrial fibrilation, neutropenia, scleroderma, dermatomyositis, cirrhosis, diffuse parenchymal lung disease, mediastinal fibrosis, tuberculosis, spleen fibrosis caused by sickle-cell anemia, rheumatoid arthritis, pulmonary fibrosis of Hermansky-Pudlak Syndrome, focal segmental glomerulosclerosis, hypertrophic cardiomyopathy,
neurofibromatosis, and interstitial cardiac fibrosis. US 20080161361, WO
2007053685, and WO 2007062167.
1 -(4-hydroxyp
yl)pyridin-2(1 /- )-one
1 -(4-fluoroph
)pyridin-2(1 /- )-one
Figure imgf000003_0001
1 -phenyl-5-(trifluoromethyl)pyridin-2(1 H)-one [0004] The chemical structures of 5-methyl-l-phenylpyridin-2(lH)-one derivatives contain a number of features that we posit will produce inactive or toxic metabolites, the formation of which can be reduced by the approach described herein. 5-methyl-l-phenylpyridin-2(lH)-one derivatives are likely metabolized via CYP45o-mediated oxidation of the methyl group and/or ring hydroxylation. These, as well as other metabolic transformations, occur in part through polymorphically- expressed enzymes, exacerbating interpatient variability. Additionally, some metabolites of 5-methyl-l-phenylpyridin-2(lH)-one derivatives may have undesirable side effects. In order to overcome its short half-life, the drugs likely must be taken several times per day, which increases the probability of patient incompliance and discontinuance. Further, abruptly stopping treatment with 5- methyl-l-phenylpyridin-2(lH)-one derivatives can lead to withdrawal or discontinuation syndrome. Medicines with longer half-lives will likely attenuate these deleterious effects.
Deuterium Kinetic Isotope Effect
[0005] In order to eliminate foreign substances such as therapeutic agents, the animal body expresses various enzymes, such as the cytochrome P450 enzymes (CYPs), esterases, proteases, reductases, dehydrogenases, and monoamine oxidases, to react with and convert these foreign substances to more polar intermediates or metabolites for renal excretion. Such metabolic reactions frequently involve the oxidation of a carbon-hydrogen (C-H) bond to either a carbon-oxygen (C-O) or a carbon-carbon (C-C) π-bond. The resultant metabolites may be stable or unstable under physiological conditions, and can have substantially different
pharmacokinetic, pharmacodynamic, and acute and long-term toxicity profiles relative to the parent compounds. For most drugs, such oxidations are generally rapid and ultimately lead to administration of multiple or high daily doses.
[0006] The relationship between the activation energy and the rate of reaction may be quantified by the Arrhenius equation, k = Ae~Eact/RT. The Arrhenius equation states that, at a given temperature, the rate of a chemical reaction depends exponentially on the activation energy (Eact).
[0007] The transition state in a reaction is a short lived state along the reaction pathway during which the original bonds have stretched to their limit. By definition, the activation energy Eact for a reaction is the energy required to reach the transition state of that reaction. Once the transition state is reached, the molecules can either revert to the original reactants, or form new bonds giving rise to reaction products. A catalyst facilitates a reaction process by lowering the activation energy leading to a transition state. Enzymes are examples of biological catalysts.
[0008] Carbon-hydrogen bond strength is directly proportional to the absolute value of the ground-state vibrational energy of the bond. This vibrational energy depends on the mass of the atoms that form the bond, and increases as the mass of one or both of the atoms making the bond increases. Since deuterium (D) has twice the mass of protium (¾), a C-D bond is stronger than the corresponding C-lR bond. If a C-1!! bond is broken during a rate-determining step in a chemical reaction (i.e. the step with the highest transition state energy), then substituting a deuterium for that protium will cause a decrease in the reaction rate. This phenomenon is known as the Deuterium Kinetic Isotope Effect (DKIE). The magnitude of the DKIE can be expressed as the ratio between the rates of a given reaction in which a C-1!! bond is broken, and the same reaction where deuterium is substituted for protium. The DKIE can range from about 1 (no isotope effect) to very large numbers, such as 50 or more. Substitution of tritium for hydrogen results in yet a stronger bond than deuterium and gives numerically larger isotope effects [0009] Deuterium (2H or D) is a stable and non-radioactive isotope of hydrogen which has approximately twice the mass of protium (¾), the most common isotope of hydrogen. Deuterium oxide (D2O or "heavy water") looks and tastes like H2O, but has different physical properties.
[0010] When pure D2O is given to rodents, it is readily absorbed. The quantity of deuterium required to induce toxicity is extremely high. When about 0-15% of the body water has been replaced by D2O, animals are healthy but are unable to gain weight as fast as the control (untreated) group. When about 15-20% of the body water has been replaced with D2O, the animals become excitable. When about 20-25% of the body water has been replaced with D2O, the animals become so excitable that they go into frequent convulsions when stimulated. Skin lesions, ulcers on the paws and muzzles, and necrosis of the tails appear. The animals also become very aggressive. When about 30% of the body water has been replaced with D2O, the animals refuse to eat and become comatose. Their body weight drops sharply and their metabolic rates drop far below normal, with death occurring at about 30 to about 35% replacement with D2O. The effects are reversible unless more than thirty percent of the previous body weight has been lost due to D2O. Studies have also shown that the use of D2O can delay the growth of cancer cells and enhance the cytotoxicity of certain antineoplastic agents.
[001 1] Deuteration of pharmaceuticals to improve pharmacokinetics (PK), pharmacodynamics (PD), and toxicity profiles has been demonstrated previously with some classes of drugs. For example, the DKIE was used to decrease the hepatotoxicity of halothane, presumably by limiting the production of reactive species such as trifluoroacetyl chloride. However, this method may not be applicable to all drug classes. For example, deuterium incorporation can lead to metabolic switching. Metabolic switching occurs when xenogens, sequestered by Phase I enzymes, bind transiently and re-bind in a variety of conformations prior to the chemical reaction (e.g., oxidation). Metabolic switching is enabled by the relatively vast size of binding pockets in many Phase I enzymes and the promiscuous nature of many metabolic reactions. Metabolic switching can lead to different proportions of known metabolites as well as altogether new metabolites. This new metabolic profile may impart more or less toxicity. Such pitfalls are non- obvious and are not predictable a priori for any drug class.
[0012] 5 -methyl- l-phenylpyridin-2(lH)-one derivatives are fibrosis and/or collagen infiltration modulators. The carbon-hydrogen bonds of 5-methyl-l- phenylpyridin-2(lH)-one derivatives contain a naturally occurring distribution of hydrogen isotopes, namely lH or protium (about 99.9844%), 2H or deuterium (about 0.0156%), and 3H or tritium (in the range between about 0.5 and 67 tritium atoms per 1018 protium atoms). Increased levels of deuterium incorporation may produce a detectable Deuterium Kinetic Isotope Effect (DKIE) that could effect the pharmacokinetic, pharmacologic and/or toxicologic profiles of such 5-methyl-l- phenylpyridin-2(lH)-one derivatives in comparison with the compound having naturally occurring levels of deuterium.
[0013] Based on discoveries made in our laboratory, as well as considering the literature, 5-methyl-l-phenylpyridin-2(lH)-one derivatives are likely metabolized in humans at the methyl group and/or various ring C-H bonds. The current approach has the potential to prevent metabolism at these sites. Other sites on the molecule may also undergo transformations leading to metabolites with as-yet- unknown pharmacology/toxicology. Limiting the production of these metabolites has the potential to decrease the danger of the administration of such drugs and may even allow increased dosage and/or increased efficacy. All of these transformations can occur through polymorphically-expressed enzymes, exacerbating interpatient variability. Further, some disorders are best treated when the subject is medicated around the clock or for an extended period of time. For all of the foregoing reasons, a medicine with a longer half-life may result in greater efficacy and cost savings. Various deuteration patterns can be used to (a) reduce or eliminate unwanted metabolites, (b) increase the half-life of the parent drug, (c) decrease the number of doses needed to achieve a desired effect, (d) decrease the amount of a dose needed to achieve a desired effect, (e) increase the formation of active metabolites, if any are formed, (f) decrease the production of deleterious metabolites in specific tissues, and/or (g) create a more effective drug and/or a safer drug for
polypharmacy, whether the polypharmacy be intentional or not. The deuteration approach has the strong potential to slow the metabolism of 5 -methyl- 1- phenylpyridin-2(lH)-one derivatives and attenuate interpatient variability.
[0014] Novel compounds and pharmaceutical compositions, certain of which have been found to modulate fibrosis and/or collagen infiltration have been discovered, together with methods of synthesizing and using the compounds, including methods for the treatment of fibrosis-mediated disorders and/or collagen infiltration-mediated disorders in a patient by administering the compounds.
[0015] In certain embodiments of the present invention, compounds have structural Formula I:
Figure imgf000007_0001
(I) or a salt thereof, wherein: Ri is selected from the group consisting of -CH3, -CH2D, -CD2H, -CD3, and
R2-R8 are independently selected from the group consisting of hydrogen and deuterium;
R9 is selected from the group consisting of hydrogen, deuterium, fluorine, chlorine, -OH, and -OD;
at least one of R1-R9 is deuterium or contains deuterium; and
if R9 is hydrogen or deuterium, then Ri is -CF3.
[0016] Certain compounds disclosed herein may possess useful fibrosis and/or collagen infiltration modulating activity, and may be used in the treatment or prophylaxis of a disorder in which fibrosis and/or collagen infiltration plays an active role. Thus, certain embodiments also provide pharmaceutical compositions comprising one or more compounds disclosed herein together with a
pharmaceutically acceptable carrier, as well as methods of making and using the compounds and compositions. Certain embodiments provide methods for modulating fibrosis and/or collagen infiltration. Other embodiments provide methods for treating a fibrosis-mediated disorder and/or a collagen infiltration- mediated disorder in a patient in need of such treatment, comprising administering to said patient a therapeutically effective amount of a compound or composition according to the present invention. Also provided is the use of certain compounds disclosed herein for use in the manufacture of a medicament for the prevention or treatment of a disorder ameliorated by the modulation of fibrosis and/or collagen infiltration.
[0017] In certain embodiments of the present invention, disclosed herein is a composition comprising compounds of structural Formula I:
Figure imgf000008_0001
(I) or a salt thereof, wherein in each compound of Formula I:
Ri is selected from the group consisting of -CH3, -CH2D, -CD2H, -CD3, and
R2-R8 are independently selected from the group consisting of hydrogen and deuterium;
R9 is selected from the group consisting of hydrogen, deuterium, fluorine, chlorine, -OH, and -OD;
and in the composition as a whole, at least one of the positions R1-R9 is enriched with deuterium; and
if R9 is hydrogen or deuterium, then Ri is -CF3.
[0018] The compounds as disclosed herein may also contain less prevalent isotopes for other elements, including, but not limited to, 13C or 14C for carbon, 33S, 34S, or 36S for sulfur, 15N for nitrogen, and 170 or 180 for oxygen.
[0019] In certain embodiments, the compound disclosed herein may expose a patient to a maximum of about 0.000005% D2O or about 0.00001% DHO, assuming that all of the C-D bonds in the compound as disclosed herein are metabolized and released as D2O or DHO. In certain embodiments, the levels of D2O shown to cause toxicity in animals is much greater than even the maximum limit of exposure caused by administration of the deuterium enriched compound as disclosed herein. Thus, in certain embodiments, the deuterium-enriched compound disclosed herein should not cause any additional toxicity due to the formation of D2O or DHO upon drug metabolism.
[0020] In certain embodiments, the deuterated compounds disclosed herein maintain the beneficial aspects of the corresponding non-isotopically enriched molecules while substantially increasing the maximum tolerated dose, decreasing toxicity, increasing the half-life (T1/2), lowering the maximum plasma concentration (Cmax) of the minimum efficacious dose (MED), lowering the efficacious dose and thus decreasing the non-mechanism-related toxicity, and/or lowering the probability of drug-drug interactions. [0021] All publications and references cited herein are expressly incorporated herein by reference in their entirety. However, with respect to any similar or identical terms found in both the incorporated publications or references and those explicitly put forth or defined in this document, then those terms definitions or meanings explicitly put forth in this document shall control in all respects.
[0022] As used herein, the terms below have the meanings indicated.
[0023] The singular forms "a," "an," and "the" may refer to plural articles unless specifically stated otherwise.
[0024] The term "about," as used herein, is intended to qualify the numerical values which it modifies, denoting such a value as variable within a margin of error. When no particular margin of error, such as a standard deviation to a mean value given in a chart or table of data, is recited, the term "about" should be understood to mean that range which would encompass the recited value and the range which would be included by rounding up or down to that figure as well, taking into account significant figures.
[0025] When ranges of values are disclosed, and the notation "from ni ... to m" or "m-m" is used, where ni and m are the numbers, then unless otherwise specified, this notation is intended to include the numbers themselves and the range between them. This range may be integral or continuous between and including the end values.
[0026] The term "deuterium enrichment" refers to the percentage of incorporation of deuterium at a given position in a molecule in the place of hydrogen. For example, deuterium enrichment of 1% at a given position means that 1% of molecules in a given sample contain deuterium at the specified position. Because the naturally occurring distribution of deuterium is about 0.0156%, deuterium enrichment at any position in a compound synthesized using non- enriched starting materials is about 0.0156%. The deuterium enrichment can be determined using conventional analytical methods known to one of ordinary skill in the art, including mass spectrometry and nuclear magnetic resonance spectroscopy.
[0027] The term "is/are deuterium," when used to describe a given position in a molecule such as R1-R9 or the symbol "D", when used to represent a given position in a drawing of a molecular structure, means that the specified position is enriched with deuterium above the naturally occurring distribution of deuterium. In one embodiment deuterium enrichment is no less than about 1%, in another no less than about 5%, in another no less than about 10%, in another no less than about 20%, in another no less than about 50%, in another no less than about 70%, in another no less than about 80%, in another no less than about 90%, or in another no less than about 98% of deuterium at the specified position.
[0028] The term "isotopic enrichment" refers to the percentage of incorporation of a less prevalent isotope of an element at a given position in a molecule in the place of the more prevalent isotope of the element.
[0029] The term "non-isotopically enriched" refers to a molecule in which the percentages of the various isotopes are substantially the same as the naturally occurring percentages.
[0030] Asymmetric centers exist in the compounds disclosed herein. These centers are designated by the symbols "R" or "S," depending on the configuration of substituents around the chiral carbon atom. It should be understood that the invention encompasses all stereochemical isomeric forms, including diastereomeric, enantiomeric, and epimeric forms, as well as d-isomers and 1 -isomers, and mixtures thereof. Individual stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or
recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art. Starting compounds of particular stereochemistry are either commercially available or can be made and resolved by techniques known in the art. Additionally, the compounds disclosed herein may exist as geometric isomers. The present invention includes all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the appropriate mixtures thereof. Additionally, compounds may exist as tautomers; all tautomeric isomers are provided by this invention. Additionally, the compounds disclosed herein can exist in unsolvated as well as solvated forms with
pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms.
[0031] The term "bond" refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure. A bond may be single, double, or triple unless otherwise specified. A dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
[0032] The term "disorder" as used herein is intended to be generally synonymous, and is used interchangeably with, the terms "disease" and "condition" (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms.
[0033] The terms "treat," "treating," and "treatment" are meant to include alleviating or abrogating a disorder or one or more of the symptoms associated with a disorder; or alleviating or eradicating the cause(s) of the disorder itself. As used herein, reference to "treatment'Of a disorder is intended to include prevention. The terms "prevent," "preventing," and "prevention" refer to a method of delaying or precluding the onset of a disorder; and/or its attendant symptoms, barring a subject from acquiring a disorder or reducing a subject's risk of acquiring a disorder.
[0034] The term "therapeutically effective amount" refers to the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disorder being treated. The term "therapeutically effective amount" also refers to the amount of a compound that is sufficient to elicit the biological or medical response of a cell, tissue, system, animal, or human that is being sought by a researcher, veterinarian, medical doctor, or clinician.
[0035] The term "subject" refers to an animal, including, but not limited to, a primate (e.g., human, monkey, chimpanzee, gorilla, and the like), rodents (e.g., rats, mice, gerbils, hamsters, ferrets, and the like), lagomorphs, swine (e.g., pig, miniature pig), equine, canine, feline, and the like. The terms "subject" and "patient" are used interchangeably herein in reference, for example, to a mammalian subject, such as a human patient.
[0036] The term "combination therapy" means the administration of two or more therapeutic agents to treat a therapeutic disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the disorders described herein.
[0037] The term "fibrosis" refers to the development of excessive fibrous connective tissue within an organ or tissue.
[0038] The term "collagen infiltration" refers to the entry of the connective tissue collagen into cells or into the extracellular matrix around cells. This occurs in organs and tissues naturally and under normal circumstances but can occur excessively and accompany or cause disease.
[0039] The terms "fibrosis" and "collagen infiltration" are not necessarily synonymous but can, in certain contexts, be used interchangeably.
[0040] The terms "collagen infiltration-mediated disorder" refers to a disorder that is characterized by abnormal or undesired collagenic infiltration, that when collagen infiltration activity is modified, leads to the desired responses depending on the route of administration and desired end result. A collagen-mediated disorder may be completely or partially mediated through the modulation of collagen infiltration. In particular, a collagen-mediated disorder is one in which modulation of collagen infiltration activity results in some effect on the underlying disorder, e.g., administering a collagen-infiltration modulator results in some improvement in at least some of the patients being treated.
[0041] The terms "fibrosis-mediated disorder" refers to a disorder that is characterized by abnormal or undesired fibrotic activity, that when fibrosis activity is modified, leads to the desired responses depending on the route of administration and desired end result. A fibrosis- mediated disorder may be completely or partially mediated through the modulation of fibrosis. In particular, a fibrosis- mediated disorder is one in which modulation of fibrosis activity results in some effect on the underlying disorder, e.g., administering a fibrosis modulator results in some improvement in at least some of the patients being treated.
[0042] The terms "fibrosis modulator" or "modulating fibrosis" are meant to be interchangeable and refer to the ability of a compound disclosed herein to alter the occurrence and/or amount of fibrosis. A fibrosis modulator may increase the occurrence or level of fibrosis, may increase or decrease the occurrence and/or amount of fibrosis depending on the concentration of the compound exposed to the adrenergic receptor, or may decrease the occurrence and/or amount of fibrosis. Such activation or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
[0043] The terms "collagen infiltration modulator" or "modulating collagen infiltration" are meant to be interchangeable and refer to the ability of a compound disclosed herein to alter the occurrence and/or amount of collagen infiltration. A fibrosis modulator may increase the occurrence or level of collagen infiltration, may increase or decrease the occurrence and/or amount of collagen infiltration depending on the concentration of the compound exposed to the adrenergic receptor, or may decrease the occurrence and/or amount of collagen infiltration. Such activation or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
[0044] The term "fibrosis and/or collagen infiltration-mediated disorder," refers to a disorder that is characterized by the development of excessive fibrous connective tissue within an organ or tissue or abnormal entry of the connective tissue collagen into cells or into the extracellular matrix around cells. A fibrosis and/or collagen infiltration-mediated disorder may be completely or partially mediated by modulating fibrosis and/or collagen infiltration. In particular, a fibrosis and/or collagen infiltration-mediated disorder is one in which modulation of fibrosis and/or collagen infiltration results in some effect on the underlying disorder e.g., administration of a fibrosis and/or collagen infiltration modulator results in some improvement in at least some of the patients being treated.
[0045] In some embodiments, modulation of fibrosis and/or collagen infiltration may be assessed using the method described in Gosselin et al, Muscle & Nerve, 2007, 35(2), 208-216; US 20080161361 ; WO 2007053685: and WO 2007062167.
[0046] The term "therapeutically acceptable" refers to those compounds (or salts, prodrugs, tautomers, zwitterionic forms, etc.) which are suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, immunogenecity, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
[0047] The term "pharmaceutically acceptable carrier," "pharmaceutically acceptable excipient," "physiologically acceptable carrier," or "physiologically acceptable excipient" refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material. Each component must be "pharmaceutically acceptable" in the sense of being compatible with the other ingredients of a pharmaceutical formulation. It must also be suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenecity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice of Pharmacy, 21st Edition; Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 5th Edition; Rowe et al, Eds., The Pharmaceutical Press and the American Pharmaceutical Association: 2005; and Handbook of Pharmaceutical Additives, 3rd Edition; Ash and Ash Eds., Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, Gibson Ed., CRC Press LLC: Boca Raton, FL, 2004).
[0048] The terms "active ingredient," "active compound," and "active substance" refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients or carriers, to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder.
[0049] The terms "drug," "therapeutic agent," and "chemotherapeutic agent" refer to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder.
[0050] The term "release controlling excipient" refers to an excipient whose primary function is to modify the duration or place of release of the active substance from a dosage form as compared with a conventional immediate release dosage form.
[0051] The term "nonrelease controlling excipient" refers to an excipient whose primary function do not include modifying the duration or place of release of the active substance from a dosage form as compared with a conventional immediate release dosage form.
[0052] The term "prodrug" refers to a compound functional derivative of the compound as disclosed herein and is readily convertible into the parent compound in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound. A prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221-294; Morozowich et al. in "Design of Biopharmaceutical Properties through Prodrugs and Analogs," Roche Ed., APHA Acad. Pharm. Sci. 1977; "Bioreversible Carriers in Drug in Drug Design, Theory and Application," Roche Ed., APHA Acad. Pharm. Sci. 1987; "Design of
Prodrugs," Bundgaard, Elsevier, 1985; Wang et al, Curr. Pharm. Design 1999, 5, 265-287; Pauletti et al, Adv. Drug. Delivery Rev. 1997, 27, 235-256; Mizen et al, Pharm. Biotech. 1998, 11, 345-365; Gaignault et al, Pract. Med. Chem. 1996, 671- 696; Asgharnejad in "Transport Processes in Pharmaceutical Systems," Amidon et al, Ed., Marcell Dekker, 185-218, 2000; Balant et al., Eur. J. Drug Metab.
Pharmacokinet. 1990, 15, 143-53; Balimane and Sinko, Adv. Drug Delivery Rev. 1999, 39, 183-209; Browne, Clin. Neuropharmacol. 1997, 20, 1-12; Bundgaard, Arch. Pharm. Chem. 1979, 86, 1-39; Bundgaard, Controlled Drug Delivery 1987, 17, 179-96; Bundgaard, Adv. Drug Delivery Rev.1992, 8, 1-38; Fleisher et al, Adv. Drug Delivery Rev. 1996, 19, 115-130; Fleisher et al, Methods Enzymol. 1985, 112, 360-381 ; Farquhar et al, J. Pharm. Sci. 1983, 72, 324-325; Freeman et al, J. Chem. Soc, Chem. Commun. 1991, 875-877; Friis and Bundgaard, Eur. J. Pharm. Sci. 1996, 4, 49-59; Gangwar et al, Des. Biopharm. Prop. Prodrugs Analogs, 1977, 409-421; Nathwani and Wood, Drugs 1993, 45, 866-94; Sinhababu and Thakker, Adv. Drug Delivery Rev. 1996, 19, 241-273; Stella et al, Drugs 1985, 29, 455-73; Tan et al, Adv. Drug Delivery Rev. 1999, 39, 117-151 ; Taylor, Adv. Drug Delivery Rev. 1996, 19, 131-148; Valentino and Borchardt, Drug Discovery Today 1997, 2, 148-155; Wiebe and Knaus, Adv. Drug Delivery Rev. 1999, 39, 63-80; Waller et al, Br. J. Clin. Pharmac. 1989, 28, 497-507.
[0053] The compounds disclosed herein can exist as therapeutically acceptable salts. The term "therapeutically acceptable salt," as used herein, represents salts or zwitterionic forms of the compounds disclosed herein which are therapeutically acceptable as defined herein. The salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound with a suitable acid or base.Therapeutically acceptable salts include acid and basic addition salts. For a more complete discussion of the preparation and selection of salts, refer to "Handbook of Pharmaceutical Salts, Properties, and Use," Stah and Wermuth, Ed.;( Wiley-VCH and VHCA, Zurich, 2002) and Berge et al, J. Pharm. Set 1977, 66, 1-19.
[0054] Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid,
benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+)- camphoric acid, camphorsulfonic acid, (+)-(lS)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, D-gluconic acid, D-glucuronic acid, L-glutamic acid, a-oxo-glutaric acid, glycolic acid, hippuric acid,
hydrobromic acid, hydrochloric acid, hydroiodic acid, (+)-L-lactic acid, (±)-DL- lactic acid, lactobionic acid, lauric acid, maleic acid, (-)-L-malic acid, malonic acid, (±)-DL-mandelic acid, methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene- 1,5-disulfonic acid, l-hydroxy-2 -naphthoic acid, nicotinic acid, nitric acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, perchloric acid, phosphoric acid, L-pyroglutamic acid, saccharic acid, salicylic acid, 4-amino- salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid, undecylenic acid, and valeric acid.
[0055] Suitable bases for use in the preparation of pharmaceutically acceptable salts, including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2- (diethylamino)-ethanol, ethanolamine, ethylamine, ethylenediamine,
isopropylamine, N-methyl-glucamine, hydrabamine, lH-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, l-(2-hydroxyethyl)-pyrrolidine, pyridine, quinuclidine, quinoline, isoquinoline, secondary amines, triethanolamine, trimethylamine, triethylamine, N-methyl-D-glucamine, 2-amino-2-(hydroxymethyl)-l,3- propanediol, and tromethamine. [0056] While it may be possible for the compounds of the subject invention to be administered as the raw chemical, it is also possible to present them as a pharmaceutical composition. Accordingly, provided herein are pharmaceutical compositions which comprise one or more of certain compounds disclosed herein, or one or more pharmaceutically acceptable salts, prodrugs, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences. The pharmaceutical compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes. The pharmaceutical compositions may also be formulated as a modified release dosage form, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modifled-Release Drug Deliver Technology, Rathbone et al, Eds., Drugs and the Pharmaceutical Science, Marcel Dekker, Inc.: New York, NY, 2002; Vol. 126).
[0057] The compositions include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient. The compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
Typically, these methods include the step of bringing into association a compound of the subject invention or a pharmaceutically salt, prodrug, or solvate thereof ("active ingredient") with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
[0058] Formulations of the compounds disclosed herein suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a nonaqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
[0059] Pharmaceutical preparations which can be used orally include tablets, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[0060] The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried
(lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen- free water, immediately prior to use.
Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
[0061] Formulations for parenteral administration include aqueous and nonaqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
[0062] In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneous ly or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[0063] For buccal or sublingual administration, the compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner. Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth. [0064] The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
[0065] Certain compounds disclosed herein may be administered topically, that is by non-systemic administration. This includes the application of a compound disclosed herein externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream. In contrast, systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
[0066] Formulations suitable for topical administration include liquid or semi- liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose.
[0067] For administration by inhalation, compounds may be delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray. Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount.
Alternatively, for administration by inhalation or insufflation, the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch. The powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
[0068] Preferred unit dosage formulations are those containing an effective dose, as herein below recited, or an appropriate fraction thereof, of the active ingredient.
[0069] Compounds may be administered orally or via injection at a dose of from 0.1 to 500 mg/kg per day. The dose range for adult humans is generally from 5 mg to 2 g/day. Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of one or more compounds which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg. [0070] The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
[0071] The compounds can be administered in various modes, e.g. orally, topically, or by injection. The precise amount of compound administered to a patient will be the responsibility of the attendant physician. The specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of administration, route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the disorder being treated. Also, the route of administration may vary depending on the disorder and its severity.
[0072] In the case wherein the patient's condition does not improve, upon the doctor's discretion the administration of the compounds may be administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disorder.
[0073] In the case wherein the patient's status does improve, upon the doctor's discretion the administration of the compounds may be given continuously or temporarily suspended for a certain length of time (i.e., a "drug holiday").
[0074] Once improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, can be reduced, as a function of the symptoms, to a level at which the improved disorder is retained. Patients can, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.
[0075] Disclosed herein are methods of treating a fibrosis-mediated disorder and/or a collagen infiltration-mediated disorder comprising administering to a subject having or suspected to have such a disorder, a therapeutically effective amount of a compound as disclosed herein or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[0076] Fibrosis and/or collagen infiltration-mediated disorders, include, but are not limited to, idiopathic pulmonary fibrosis, uterine fibroids, multiple sclerosis, renal fibrosis, diabetic kidney disease, endotoxin-induced liver injury after partial hepatectomy or hepatic ischemia, allograft injury after organ transplantation, cystic fibrosis, atrial fibrilation, neutropenia, scleroderma, dermatomyositis, cirrhosis, diffuse parenchymal lung disease, mediastinal fibrosis, tuberculosis, spleen fibrosis caused by sickle-cell anemia, rheumatoid arthritis, radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, radiation-induced lung injury, fibrotic lesions, pulmonary fibrosis of Hermansky- Pudlak Syndrome, focal segmental glomerulosclerosis, hypertrophic
cardiomyopathy, neurofibromatosis, interstitial cardiac fibrosis, and/or any disorder which can lessened, alleviated, or prevented by administering a fibrosis and/or collagen infiltration modulator.
[0077] In certain embodiments, a method of treating a fibrosis and/or collagen infiltration-mediated disorder comprises administering to the subject a
therapeutically effective amount of a compound of as disclosed herein, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, so as to affect: (1) decreased inter- individual variation in plasma levels of the compound or a metabolite thereof; (2) increased average plasma levels of the compound or decreased average plasma levels of at least one metabolite of the compound per dosage unit; (3) decreased inhibition of, and/or metabolism by at least one cytochrome P450 or monoamine oxidase isoform in the subject; (4) decreased metabolism via at least one polymorphically-expressed cytochrome P450 isoform in the subject; (5) at least one statistically-significantly improved disorder-control and/or disorder-eradication endpoint; (6) an improved clinical effect during the treatment of the disorder, (7) prevention of recurrence, or delay of decline or appearance, of abnormal alimentary or hepatic parameters as the primary clinical benefit, or (8) reduction or elimination of deleterious changes in any diagnostic hepatobiliary function endpoints, as compared to the corresponding non- isotopically enriched compound.
[0078] In certain embodiments, inter-individual variation in plasma levels of the compounds as disclosed herein, or metabolites thereof, is decreased; average plasma levels of the compound as disclosed herein are increased; average plasma levels of a metabolite of the compound as disclosed herein are decreased; inhibition of a cytochrome P450 or monoamine oxidase isoform by a compound as disclosed herein is decreased; or metabolism of the compound as disclosed herein by at least one polymorphically-expressed cytochrome P450 isoform is decreased; by greater than about 5%, greater than about 10%, greater than about 20%, greater than about 30%, greater than about 40%, or by greater than about 50% as compared to the corresponding non-isotopically enriched compound.
[0079] Plasma levels of the compound as disclosed herein, or metabolites thereof, may be measured using the methods described by Li et al. Rapid
Communications in Mass Spectrometry 2005, 19, 1943-1950, Hughes et al, Xenobiotica 1992, 22(7), 859-69, Varma et al, Journal of Pharmaceutical and Biomedical Analysis 2004, 36(3), 669-674, Massoud et al, Journal of
Chromatography, B: Biomedical Sciences and Applications 1999, 734(1), 163- 167, Kim et al, Journal of Pharmaceutical and Biomedical Analysis 2003, 31(2), 341-349, and Lindeke et al, Acta Pharmaceutica Suecica 1981, 18(1), 25-34.
[0080] Examples of cytochrome P450 isoforms in a mammalian subject include, but are not limited to, CYP1A1, CYP1A2, CYP1B 1, CYP2A6, CYP2A13, CYP2B6, CYP2C8, CYP2C9, CYP2C18, CYP2C19, CYP2D6, CYP2E1, CYP2G1, CYP2J2, CYP2R1, CYP2S1, CYP3A4, CYP3A5, CYP3A5P 1, CYP3A5P2, CYP3A7, CYP4A11, CYP4B 1, CYP4F2, CYP4F3, CYP4F8, CYP4F1 1, CYP4F12, CYP4X1, CYP4Z1, CYP5A1, CYP7A1, CYP7B 1, CYP8A1, CYP8B 1, CYPl lAl, CYP11B1, CYP 11B2, CYP17, CYP19, CYP21, CYP24, CYP26A1, CYP26B 1, CYP27A1, CYP27B 1, CYP39, CYP46, and CYP51.
[0081] Examples of monoamine oxidase isoforms in a mammalian subject include, but are not limited to, MAOA, and MAOB.
[0082] The inhibition of the cytochrome P450 isoform is measured by the method of Ko et al. (British Journal of Clinical Pharmacology, 2000, 49, 343-351). The inhibition of the MAOA isoform is measured by the method of Weyler et al. (J. Biol Chem. 1985, 260, 13199-13207). The inhibition of the MAOB isoform is measured by the method of Uebelhack et al. (Pharmacopsychiatry, 1998, 31, 187- 192).
[0083] Examples of polymorphically-expressed cytochrome P450 isoforms in a mammalian subject include, but are not limited to, CYP2C8, CYP2C9, CYP2C19, and CYP2D6.
[0084] The metabolic activities of liver microsomes, cytochrome P450 isoforms, and monoamine oxidase isoforms are measured by the methods described herein.
[0085] Examples of improved disorder-control and/or disorder-eradication endpoints, or improved clinical effects include, but are not limited to, change in forced vital capacity, change in pulmonary function parameters, progression to renal failure, reduced proteinuria, progression-free survival, change in shortness-of- breath, change in oxygen saturation during the six minute walk test, change in distance walked during the six minute walk test, tumor volume, GFR as calculated using forty -variable Levey equation, and reduced adverse effects as compared to the non-isotopically enriched compound.
[0086] Examples of diagnostic hepatobiliary function endpoints include, but are not limited to, alanine aminotransferase ("ALT"), serum glutamic-pyruvic transaminase ("SGPT"), aspartate aminotransferase ("AST" or "SGOT"),
ALT/AST ratios, serum aldolase, alkaline phosphatase ("ALP"), ammonia levels, bilirubin, gamma-glutamyl transpeptidase ("GGTP," "γ-GTP," or "GGT"), leucine aminopeptidase ("LAP"), liver biopsy, liver ultrasonography, liver nuclear scan, 5'- nucleotidase, and blood protein. Hepatobiliary endpoints are compared to the stated normal levels as given in "Diagnostic and Laboratory Test Reference", 4th edition, Mosby, 1999. These assays are run by accredited laboratories according to standard protocol.
[0087] Besides being useful for human treatment, certain compounds and formulations disclosed herein may also be useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
Combination Therapy
[0088] The compounds disclosed herein may also be combined or used in combination with other agents useful in the treatment of fibrosis-mediated disorders and/or collagen infiltration-mediated disorders. Or, by way of example only, the therapeutic effectiveness of one of the compounds described herein may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced).
[0089] Such other agents, adjuvants, or drugs, may be administered, by a route and in an amount commonly used therefor, simultaneously or sequentially with a compound as disclosed herein. When a compound as disclosed herein is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound disclosed herein may be utilized, but is not required.
[0090] In certain embodiments, the compounds disclosed herein can be combined with one or more steroidal drugs and non-steroidal anti-inflammatory drugs.
[0091] In certain embodiments, the compounds disclosed herein can be combined with a steroidal drug selected from the group consisting of aldosterone, beclometasone, betamethasone, deoxycorticosterone acetate, fludrocortisone acetate, hydrocortisone, prednisolone, prednisone, methylprenisolone,
dexamethasone, and triamcinolone.
[0092] In certain embodiments, the compounds disclosed herein can be combined with a non-steroidal anti-inflammatory drug selected from the group consisting of aceclofenac, acemetacin, amoxiprin, aspirin, azapropazone, benorilate, bromfenac, carprofen, celecoxib, choline magnesium salicylate, diclofenac, diflunisal, etodolac, etoracoxib, faislamine, fenbuten, fenoprofen, flurbiprofen, ibuprofen, indometacin, ketoprofen, ketorolac, lornoxicam, loxoprofen,
lumiracoxib, meclofenamic acid, mefenamic acid, meloxicam, metamizole, methyl salicylate, magnesium salicylate, nabumetone, naproxen, nimesulide,
oxyphenbutazone, parecoxib, phenylbutazone, piroxicam, salicyl salicylate, sulindac, sulfinprazone, suprofen, tenoxicam, tiaprofenic acid, and tolmetin.
[0093] The compounds disclosed herein can also be administered in
combination with other classes of compounds, including, but not limited to, norepinephrine reuptake inhibitors (NRIs) such as atomoxetine; dopamine reuptake inhibitors (DARIs), such as methylphenidate; serotonin-norepinephrine reuptake inhibitors (SNRIs), such as milnacipran; sedatives, such as diazepham;
norepinephrine-dopamine reuptake inhibitor (NDRIs), such as bupropion;
serotonin-norepinephrine-dopamine-reuptake-inhibitors (SNDRIs), such as venlafaxine; monoamine oxidase inhibitors, such as selegiline; hypothalamic phospholipids; endothelin converting enzyme (ECE) inhibitors, such as
phosphoramidon; opioids, such as tramadol; thromboxane receptor antagonists, such as ifetroban; potassium channel openers; thrombin inhibitors, such as hirudin; hypothalamic phospholipids; growth factor inhibitors, such as modulators of PDGF activity; platelet activating factor (PAF) antagonists; anti-platelet agents, such as GPIIb/IIIa blockers (e.g., abdximab, eptifibatide, and tirofiban), P2Y(AC) antagonists (e.g., clopidogrel, ticlopidine and CS-747), and aspirin; anticoagulants, such as warfarin; low molecular weight heparins, such as enoxaparin; Factor Vila Inhibitors and Factor Xa Inhibitors; renin inhibitors; neutral endopeptidase (NEP) inhibitors; vasopepsidase inhibitors (dual NEP-ACE inhibitors), such as omapatrilat and gemopatrilat; HMG CoA reductase inhibitors, such as pravastatin, lovastatin, atorvastatin, simvastatin, NK-104 (a.k.a. itavastatin, nisvastatin, or nisbastatin), and ZD-4522 (also known as rosuvastatin, or atavastatin or visastatin); squalene synthetase inhibitors; fibrates; bile acid sequestrants, such as questran; niacin; anti- atherosclerotic agents, such as ACAT inhibitors; MTP Inhibitors; calcium channel blockers, such as amlodipine besylate; potassium channel activators; alpha- muscarinic agents; beta-muscarinic agents, such as carvedilol and metoprolol; antiarrhythmic agents; diuretics, such as chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichioromethiazide, polythiazide, benzothlazide, ethacrynic acid, tricrynafen, chlorthalidone, furosenilde, musolimine, bumetanide, triamterene, amiloride, and spironolactone; thrombolytic agents, such as tissue plasminogen activator (tPA), recombinant tPA, streptokinase, urokinase, prourokinase, and anisoylated plasminogen streptokinase activator complex (APSAC); anti-diabetic agents, such as biguanides (e.g. metformin), glucosidase inhibitors (e.g., acarbose), insulins, meglitinides (e.g., repaglinide), sulfonylureas (e.g., glimepiride, glyburide, and glipizide), thiozolidinediones (e.g. troglitazone, rosiglitazone and pioglitazone), and PPAR-gamma agonists; mineralocorticoid receptor antagonists, such as spironolactone and eplerenone; growth hormone secretagogues; aP2 inhibitors; phosphodiesterase inhibitors, such as PDE III inhibitors (e.g., cilostazol) and PDE V inhibitors (e.g., sildenafil, tadalafil, vardenafil); protein tyrosine kinase inhibitors; antiinflammatories; antiproliferatives, such as methotrexate, FK506 (tacrolimus, Prograf), mycophenolate mofetil; chemotherapeutic agents;
immunosuppressants; anticancer agents and cytotoxic agents (e.g., alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes); antimetabolites, such as folate antagonists, purine analogues, and pyrridine analogues; antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes, such as L-asparaginase; farnesyl-protein transferase inhibitors; hormonal agents, such as glucocorticoids (e.g., cortisone), estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone anatagonists, and octreotide acetate; microtubule- disruptor agents, such as ecteinascidins; microtubule-stablizing agents, such as pacitaxel, docetaxel, and epothilones A-F; plant-derived products, such as vinca alkaloids, epipodophyllotoxins, and taxanes; and topoisomerase inhibitors; prenyl- protein transferase inhibitors; and cyclosporins; steroids, such as prednisone and dexamethasone; cytotoxic drugs, such as azathiprine and cyclophosphamide; TNF- alpha inhibitors, such as tenidap; anti-TNF antibodies or soluble TNF receptor, such as etanercept, rapamycin, and leflunimide; and cyclooxygenase-2 (COX-2) inhibitors, such as celecoxib and rofecoxib; and miscellaneous agents such as, hydroxyurea, procarbazine, mitotane, hexamethylmelamine, gold compounds, platinum coordination complexes, such as cisplatin, satraplatin, and carboplatin.
[0094] Thus, in another aspect, certain embodiments provide methods for treating fibrosis-mediated disorders and/or collagen infiltration-mediated disorders in a human or animal subject in need of such treatment comprising administering to said subject an amount of a compound disclosed herein effective to reduce or prevent said disorder in the subject, in combination with at least one additional agent for the treatment of said disorder that is known in the art. In a related aspect, certain embodiments provide therapeutic compositions comprising at least one compound disclosed herein in combination with one or more additional agents for the treatment of fibrosis-mediated disorders and/or collagen infiltration-mediated disorders.
General Synthetic Methods for Preparing Compounds
[0095] Isotopic hydrogen can be introduced into a compound as disclosed herein by synthetic techniques that employ deuterated reagents, whereby incorporation rates are pre-determined; and/or by exchange techniques, wherein incorporation rates are determined by equilibrium conditions, and may be highly variable depending on the reaction conditions. Synthetic techniques, where tritium or deuterium is directly and specifically inserted by tritiated or deuterated reagents of known isotopic content, may yield high tritium or deuterium abundance, but can be limited by the chemistry required. Exchange techniques, on the other hand, may yield lower tritium or deuterium incorporation, often with the isotope being distributed over many sites on the molecule. [0096] The compounds as disclosed herein can be prepared by methods known to one of skill in the art and routine modifications thereof, and/or following procedures similar to those described in the Example section herein and routine modifications thereof, and/or procedures found in US 20080161361, which are hereby incorporated in their entirety, and references cited therein and routine modifications thereof. Compounds as disclosed herein can also be prepared as shown in any of the following schemes and routine modifications thereof.
[0097] The following schemes can be used to practice the present invention. Any position shown as hydrogen may optionally be replaced with deuterium.
Scheme I
Figure imgf000029_0001
6
[0098] Compound 1 is reacted with compound 2 in the presence of an appropriate metalating agent, such as n-butyl lithium, in an appropriate solvent, such as tetrahydrofuran, to give compound 3. Compound 3 is treated with an appropriate acid, such as hydrochloric acid, in an appropriate solvent, such as a combination of water and 1,4-dioxane, to give compound 4. Compound 4 is reacted with compound 5, in the presence of an appropriate catalyst, such as cupric acetate, in the presence of an appropriate base, such as triethylamine, in an appropriate solvent, such as tetrahydrofuran, to give compound 6 of formula I. [0099] Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme I, by using appropriate deuterated intermediates. For example, to introduce deuterium at one or more positions of R2-R4, compound 1 with the corresponding deuterium substitutions can be used. To introduce deuterium at one or more positions of Ri, compound 2 with the corresponding deuterium substitutions can be used. To introduce deuterium at one or more positions of Rs-Rs, compound 5 with the corresponding deuterium substitutions can be used.
[00100] Deuterium can be incorporated to various positions having an exchangeable proton, such as the hydroxyl O-H, via proton-deuterium equilibrium exchange. For example, to introduce deuterium at the hydroxyl O-H, this proton may be replaced with deuterium selectively or non-selectively through a proton- deuterium exchange method known in the art.
Scheme II
Figure imgf000030_0001
[00101] Compound 4 is reacted with compound 8, in the presence of an appropriate catalyst, such as cupric acetate, in the presence of an appropriate base, such as triethylamine, in an appropriate solvent, such as tetrahydrofuran, to give compound 9. Compound 9 is treated with an appropriate acid, such as hydrochloric acid, in an appropriate solvent, such as a combination of water and 1,4-dioxane, to give compound 6 of formula I.
[00102] Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme II, by using appropriate deuterated intermediates. For example, to introduce deuterium at one or more positions of R1-R4, compound 4 with the corresponding deuterium substitutions can be used. To introduce deuterium at one or more positions of Rs-Rs, compound 9 with the corresponding deuterium substitutions can be used.
[00103] Deuterium can be incorporated to various positions having an exchangeable proton, such as the hydroxyl O-H, via proton-deuterium equilibrium exchange. For example, to introduce deuterium at the hydroxyl O-H, this proton may be replaced with deuterium selectively or non-selectively through a proton- deuterium exchange method known in the art.
Scheme III
Figure imgf000031_0001
[00104] Compound 10 is reacted with compound 11 in the presence of an appropriate base, such as sodium hydride, in an appropriate solvent, such as toluene, to give compound 12. Compound 12 is reacted with an appropriate deprotecting reagent, such as a combination of hydrogen gas and an appropriate catalyst, such as palladium on carbon, in an appropriate solvent, such as methanol, to give compound 4.
[00105] Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme III, by using appropriate deuterated intermediates. For example, to introduce deuterium at one or more positions of R1-R4, compound 8 with the corresponding deuterium substitutions can be used.
Scheme IV
Figure imgf000032_0001
[00106] Compound 13 is reacted with compound 11 in the presence of an appropriate base, such as potassium tert-butoxide, to give compound 14. Compound 14 is reacted with compound 2 in the presence of an appropriate metalating agent, such as n-butyl lithium, in an appropriate solvent, such as tetrahydrofuran, to give compound 4.
[00107] Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme IV, by using appropriate deuterated intermediates. For example, to introduce deuterium at one or more positions of R2-R4, compound 13 with the corresponding deuterium substitutions can be used. To introduce deuterium at one or more positions of Ri, compound 2 with the corresponding deuterium substitutions can be used.
Scheme V
Figure imgf000032_0002
15 17 18
Figure imgf000032_0003
20
[00108] Compound 15 is reacted with compound 16 in the presence of 1.1 equivalents of n-butyl lithium in tetrahydrofuran at -40°C to -20°C, to give compound 17. Compound 17 is refluxed in a 1 : 1 mixture of concentrated aqueous hydrochloric acid and 1,4-dioxane, to give compound 18. Compound 18 is reacted with compound 19, in the presence of cupric acetate and triethylamine in tetrahydrofuran at room temperature to give compound 20 of formula I.
Scheme VI
Figure imgf000033_0001
[00109] Compound 18 is reacted with compound 21, in the presence of cupric acetate and triethylamine in tetrahydrofuran at room temperature to give compound 22. Compound 22 is refluxed in a 1 : 1 mixture of concentrated aqueous hydrochloric acid and 1,4-dioxane, to give compound 20 of formula I.
Scheme VII
Figure imgf000033_0002
[001 10] Compound 22 is reacted with di-potassium hydroxide in d6-dimethyl sulfoxide to give compound 23. Compound 23 is reacted with two equivalents of compound 11 and 2.1 equivalents of sodium hydride in refluxing toluene to give compound 24. Compound 23 is reacted with hydrogen gas and palladium on carbon in methanol to give compound 18.
Scheme VIII
Figure imgf000034_0001
[001 11] Compound 25 is reacted with compound 11 and potassium tert-butoxide to give compound 26. Compound 26 is reacted with compound 16 in the presence of 1.1 equivalents of n-butyl lithium in tetrahydrofuran at -40°C to -20°C, to give compound 24.
Scheme IX
Figure imgf000034_0002
[001 12] Compound 4 is reacted with compound 27, in the presence of an appropriate catalyst, such as cupric acetate, in the presence of an appropriate base, such as triethylamine, in an appropriate solvent, such as tetrahydrofuran, to give compound 28 of formula I.
[001 13] Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme IX, by using appropriate deuterated intermediates. For example, to introduce deuterium at one or more positions of R1-R4, compound 4 with the corresponding deuterium substitutions can be used. To introduce deuterium at one or more positions of R5-R9, compound 27 with the corresponding deuterium substitutions can be used.
[001 14] The invention is further illustrated by the following examples. All IUPAC names were generated using CambridgeSoft's ChemDraw 10.0.
[001 15] The following compounds can generally be made using the methods described above. It is expected that these compounds when made will have activity similar to those described in the examples above.
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001

Figure imgf000039_0001

Figure imgf000040_0001

Figure imgf000041_0001
40
Figure imgf000042_0001
41
Figure imgf000043_0001
42 [001 16] Changes in the metabolic properties of the compounds disclosed herein as compared to their non-isotopically enriched analogs can be shown using the following assays. Compounds listed above which have not yet been made and/or tested are predicted to have changed metabolic properties as shown by one or more of these assays as well.
Biological Activity Assays
In vitro Liver Microsomal Stability Assay
[001 17] Liver microsomal stability assays are conducted at 1 mg per mL liver microsome protein with an NADPH-generating system in 2% NaHCC (2.2 mM NADPH, 25.6 mM glucose 6-phosphate, 6 units per mL glucose 6-phosphate dehydrogenase and 3.3 mM MgCk). Test compounds are prepared as solutions in 20% acetonitrile-water and added to the assay mixture (final assay concentration 5 microgram per mL) and incubated at 37 °C. Final concentration of acetonitrile in the assay should be <1%. Aliquots (50μί) are taken out at times 0, 15, 30, 45, and 60 min, and diluted with ice cold acetonitrile (200 μί) to stop the reactions.
Samples are centrifuged at 12,000 RPM for 10 min to precipitate proteins.
Supernatants are transferred to microcentrifuge tubes and stored for LC/MS/MS analysis of the degradation half-life of the test compounds.
In vitro metabolism using human cytochrome P450 enzymes
[001 18] The cytochrome P450 enzymes are expressed from the corresponding human cDNA using a baculovirus expression system (BD Biosciences, San Jose, CA). A 0.25 milliliter reaction mixture containing 0.8 milligrams per milliliter protein, 1.3 millimolar NADP+, 3.3 millimolar glucose-6-phosphate, 0.4 U/mL glucose-6-phosphate dehydrogenase, 3.3 millimolar magnesium chloride and 0.2 millimolar of a compound of Formula I, the corresponding non-isotopically enriched compound or standard or control in 100 millimolar potassium phosphate (pH 7.4) is incubated at 37 °C for 20 min. After incubation, the reaction is stopped by the addition of an appropriate solvent (e.g., acetonitrile, 20% trichloroacetic acid, 94% acetonitrile/6% glacial acetic acid, 70% perchloric acid, 94% acetonitrile/6% glacial acetic acid) and centrifuged (10,000 g) for 3 min. The supernatant is analyzed by HPLC/MS/MS. Cytochrome P450 Standard
CYP 1A2 Phenacetin
CYP2A6 Coumarin
CYP2B6 [13C]-(S)-mephenytoin
CYP2C8 Paclitaxel
CYP2C9 Diclofenac
CYP2C19 [13C]-(S)-mephenytoin
CYP2D6 (+/-)-Bufuralol
CYP2E1 Chlorzoxazone
CYP3A4 Testosterone
CYP4A [13C]-Lauric acid
Monoamine Oxidase A Inhibition and Oxidative Turnover
[001 19] The procedure is carried out using the methods described by Weyler, Journal of Biological Chemistry 1985, 260, 13199-13207, which is hereby incorporated by reference in its entirety. Monoamine oxidase A activity is measured spectrophotometrically by monitoring the increase in absorbance at 314 nm on oxidation of kynuramine with formation of 4-hydroxyquinoline. The measurements are carried out, at 30 °C, in 50mM NaPi buffer, pH 7.2, containing 0.2% Triton X-100 (monoamine oxidase assay buffer), plus 1 mM kynuramine, and the desired amount of enzyme in 1 mL total volume.
Monooamine Oxidase B Inhibition and Oxidative Turnover
[00120] The procedure is carried out as described in Uebelhack,
Pharmacopsychiatry 1998, 37(5), 187-192, which is hereby incorporated by reference in its entirety.
Dystrophic (mdx) Mouse Muscle Fibrosis Assay
[00121] The procedure is carried out as described in Gosselin et al, Muscle & Nerve, 2007, 35(2), 208-216, which is hereby incorporated by reference in its entirety.
Effect on Radiation-Induced Lung Injury [00122] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety.
Pirfenidone-Induced Increased Tolerance to Therapeutic Irradiation Dosage
[00123] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety.
Treatment of Adriamycin-Induced Renal Toxicity
[00124] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety.
Treatment of Bleomycin-Induced Lung Injury
[00125] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety.
Prevention and/or Treatment of Radiation-Induced Lung Fibrosis
[00126] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety.
Prophylactic and/or Therapeutic Use in Drug-Induced Nephrotoxicity
[00127] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety.
Treatment of Cyclosporine A-Induced Nephrotoxicity
[00128] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety.
Treatment of Adriamycin-Induced Nephrotoxicity
[00129] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety.
Side Effects of Dexamethasone Compared with Pirfenidone
[00130] The procedure is carried out as described in US 20080161361, which is hereby incorporated by reference in its entirety. In Vitro p38 ATF2 Phosphorylation Assay
[00131] The procedure is carried out as described in WO 2007062167, which is hereby incorporated by reference in its entirety.
TNFot Assay in THP-1 Cells
[00132] The procedure is carried out as described in WO 2007062167, which is hereby incorporated by reference in its entirety.
TNFot Assay in Primary Human Peripheral Blood Mononuclear Cells
[00133] The procedure is carried out as described in WO 2007062167, which is hereby incorporated by reference in its entirety.
In Vivo TNFot Assay
[00134] The procedure is carried out as described in WO 2007062167, which is hereby incorporated by reference in its entirety.
Kinase Assay
[00135] The procedure is carried out as described in WO 2007062167, which is hereby incorporated by reference in its entirety.
Canine Atrial Fibrilation Model
[00136] The procedure is carried out as described in WO 2007053685, which is hereby incorporated by reference in its entirety.
Human Atrial Fibrilation Trial
[00137] The procedure is carried out as described in WO 2007053685, which is hereby incorporated by reference in its entirety.
Transgenic Mouse Atrial Fibrilation Model
[00138] The procedure is carried out as described in WO 2007053685, which is hereby incorporated by reference in its entirety. [00139] From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.

Claims

What is claimed is:
1. A compound of structural Formula I
Figure imgf000049_0001
(I)
or a salt thereof, wherein:
Ri is selected from the group consisting of -CH3, -CH2D, -CD2H, -CD3, and
Figure imgf000049_0002
R2-R8 are independently selected from the group consisting of hydrogen and deuterium;
R9 is selected from the group consisting of hydrogen, deuterium, fluorine, chlorine, -OH, and -OD;
at least one of R1-R9 is deuterium or contains deuterium; and
if R9 is hydrogen or deuterium, then Ri is -CF3.
2. The compound as recited in Claim 1 wherein at least one of R1-R9 independently has deuterium enrichment of no less than about 10%.
3. The compound as recited in Claim 1 wherein at least one of R1-R9 independently has deuterium enrichment of no less than about 50%.
4. The compound as recited in Claim 1 wherein at least one of R1-R9 independently has deuterium enrichment of no less than about 90%.
5. The compound as recited in Claim 1 wherein at least one of R1-R9 independently has deuterium enrichment of no less than about 98%.
6. The compound as recited in Claim 1 wherein said compound has a structural formula selected from the group consisting of
Figure imgf000050_0001

Figure imgf000051_0001
Figure imgf000052_0001
51
Figure imgf000053_0001
52
Figure imgf000054_0001
Figure imgf000055_0001
54
Figure imgf000056_0001
Figure imgf000057_0001
56
Figure imgf000058_0001
Figure imgf000059_0001
58
Figure imgf000060_0001
8. The compound as recited in Claim 7 wherein each position represented as D has deuterium enrichment of no less than about 10%.
9. The compound as recited in Claim 7 wherein each position represented as D has deuterium enrichment of no less than about 50%.
10. The compound as recited in Claim 7 wherein each position represented as D has deuterium enrichment of no less than about 90%.
11. The compound as recited in Claim 7 wherein each position represented as D has deuterium enrichment of no less than about 98%.
12. The compound as recited in Claim 7 wherein said compound has the structural formula:
Figure imgf000060_0002
13. The compound as recited in Claim 7 wherein said compound has the structural formula:
Figure imgf000061_0001
14. The compound as recited in Claim 7 wherein said compound has the structural formula:
Figure imgf000061_0002
15. The compound as recited in Claim 7 wherein said compound has the stmctural formula:
Figure imgf000061_0003
16. The compound as recited in Claim 7 wherein said compound has the structural formula:
Figure imgf000061_0004
17. The compound as recited in Claim 7 wherein said compound has the stmctural formula:
Figure imgf000062_0001
18. The compound as recited in Claim 7 wherein said compound has the structural formula:
Figure imgf000062_0002
19. A pharmaceutical composition comprising a compound as recited in Claim 1 together with a pharmaceutically acceptable carrier.
20. A method of treatment of a fibrosis-mediated disorder and/or a collagen
infiltration-mediated disorder comprising the administration of a therapeutically effective amount of a compound as recited in Claim 1 to a patient in need thereof.
21. The method as recited in Claim 20 wherein said disorder is selected from the group consisting of idiopathic pulmonary fibrosis, uterine fibroids, multiple sclerosis, renal fibrosis, diabetic kidney disease, endotoxin-induced liver injury after partial hepatectomy or hepatic ischemia, allograft injury after organ transplantation, cystic fibrosis, atrial fibrilation, neutropenia, scleroderma, dermatomyositis, cirrhosis, diffuse parenchymal lung disease, mediastinal fibrosis, tuberculosis, spleen fibrosis caused by sickle-cell anemia, rheumatoid arthritis, radiation pneumonitis, fibrosis caused by radiation, fibrosis caused by administration of cytotoxic agents, radiation-induced lung injury, fibrotic lesions, pulmonary fibrosis of Hermansky-Pudlak Syndrome, focal segmental glomerulosclerosis, hypertrophic cardiomyopathy, neurofibromatosis, and interstitial cardiac fibrosis.
22. The method as recited in Claim 20 further comprising the administration of an additional therapeutic agent.
23. The method as recited in Claim 22 wherein said additional therapeutic agent is selected from the group consisting of steroidal drugs and non-steroidal antiinflammatory drugs.
24. The method as recited in Claim 23 wherein said additional therapeutic agent is a steroidal drug selected from the group consisting of aldosterone, beclometasone, betamethasone, deoxycorticosterone acetate, fludrocortisone acetate, hydrocortisone, prednisolone, prednisone, methylprenisolone, dexamethasone, and triamcinolone.
25. The method as recited in Claim 23 wherein said additional therapeutic agent is a non-steroidal anti-inflammatory drug selected from the group consisting of aceclofenac, acemetacin, amoxiprin, aspirin, azapropazone, benorilate, bromfenac, carprofen, celecoxib, choline magnesium salicylate, diclofenac, diflunisal, etodolac, etoracoxib, faislamine, fenbuten, fenoprofen, flurbiprofen, ibuprofen, indometacin, ketoprofen, ketorolac, lornoxicam, loxoprofen, lumiracoxib, meclofenamic acid, mefenamic acid, meloxicam, metamizole, methyl salicylate, magnesium salicylate, nabumetone, naproxen, nimesulide, oxyphenbutazone, parecoxib, phenylbutazone, piroxicam, salicyl salicylate, sulindac, sulfinprazone, suprofen, tenoxicam, tiaprofenic acid, and tolmetin.
26. The method as recited in Claim 20, further resulting in at least one effect
selected from the group consisting of:
a. decreased inter-individual variation in plasma levels of said compound or a metabolite thereof as compared to the non- isotopically enriched compound;
b. increased average plasma levels of said compound per dosage unit thereof as compared to the non-isotopically enriched compound; c. decreased average plasma levels of at least one metabolite of said compound per dosage unit thereof as compared to the non- isotopically enriched compound;
d. increased average plasma levels of at least one metabolite of said compound per dosage unit thereof as compared to the non- isotopically enriched compound; and
e. an improved clinical effect during the treatment in said subject per dosage unit thereof as compared to the non-isotopically enriched compound.
27. The method as recited in Claim 20, further resulting in at least two effects selected from the group consisting of:
a. decreased inter-individual variation in plasma levels of said compound or a metabolite thereof as compared to the non- isotopically enriched compound;
b. increased average plasma levels of said compound per dosage unit thereof as compared to the non-isotopically enriched compound; c. decreased average plasma levels of at least one metabolite of said compound per dosage unit thereof as compared to the non- isotopically enriched compound;
d. increased average plasma levels of at least one metabolite of said compound per dosage unit thereof as compared to the non- isotopically enriched compound; and
e. an improved clinical effect during the treatment in said subject per dosage unit thereof as compared to the non-isotopically enriched compound.
28. The method as recited in Claim 20, wherein the method effects a decreased metabolism of the compound per dosage unit thereof by at least one
polymorphically-expressed cytochrome P450 isoform in the subject, as compared to the corresponding non-isotopically enriched compound.
29. The method as recited in Claim 28, wherein the cytochrome P450 isoform is selected from the group consisting of CYP2C8, CYP2C9, CYP2C19, and CYP2D6.
30. The method as recited Claim 20, wherein said compound is characterized by decreased inhibition of at least one cytochrome P450 or monoamine oxidase isoform in said subject per dosage unit thereof as compared to the non- isotopically enriched compound.
31. The method as recited in Claim 30, wherein said cytochrome P450 or
monoamine oxidase isoform is selected from the group consisting of CYP1A1, CYP1A2, CYP1B 1, CYP2A6, CYP2A13, CYP2B6, CYP2C8, CYP2C9, CYP2C18, CYP2C19, CYP2D6, CYP2E1, CYP2G1, CYP2J2, CYP2R1, CYP2S1, CYP3A4, CYP3A5, CYP3A5P 1, CYP3A5P2, CYP3A7, CYP4A1 1, CYP4B1, CYP4F2, CYP4F3, CYP4F8, CYP4F11, CYP4F12, CYP4X1, CYP4Z1, CYP5A1, CYP7A1, CYP7B1, CYP8A1, CYP8B 1, CYP1 1A1, CYPl lBl, CYP11B2, CYP 17, CYP19, CYP21, CYP24, CYP26A1, CYP26B 1, CYP27A1, CYP27B 1, CYP39, CYP46, CYP51, MAOA, and MAOB.
32. The method as recited in Claim 20, wherein the method reduces a deleterious change in a diagnostic hepatobiliary function endpoint, as compared to the corresponding non-isotopically enriched compound.
33. The method as recited in Claim 32, wherein the diagnostic hepatobiliary
function endpoint is selected from the group consisting of alanine
aminotransferase ("ALT"), serum glutamic -pyruvic transaminase ("SGPT"), aspartate aminotransferase ("AST," "SGOT"), ALT/AST ratios, serum aldolase, alkaline phosphatase ("ALP"), ammonia levels, bilirubin, gamma-glutamyl transpeptidase ("GGTP," "γ-GTP," "GGT"), leucine aminopeptidase ("LAP"), liver biopsy, liver ultrasonography, liver nuclear scan, 5 '-nucleotidase, and blood protein.
34. A compound as recited in Claim 1 for use as a medicament.
35. A compound as recited in Claim 1 for use in the manufacture of a medicament for the prevention or treatment of a disorder ameliorated by the modulation of fibrosis and/or collagen infiltration.
PCT/US2015/027765 2014-04-30 2015-04-27 N-aryl pyridinones modulators of fibrosis and/or collagen infiltration WO2015171345A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461986454P 2014-04-30 2014-04-30
US61/986,454 2014-04-30

Publications (1)

Publication Number Publication Date
WO2015171345A1 true WO2015171345A1 (en) 2015-11-12

Family

ID=54392851

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/027765 WO2015171345A1 (en) 2014-04-30 2015-04-27 N-aryl pyridinones modulators of fibrosis and/or collagen infiltration

Country Status (1)

Country Link
WO (1) WO2015171345A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9504677B2 (en) 2007-06-20 2016-11-29 Auspex Pharmaceuticals, Inc. Substituted N-aryl pyridinones
US10047077B2 (en) 2016-04-13 2018-08-14 Skyline Antiinfectives, Inc. Deuterated O-sulfated beta-lactam hydroxamic acids and deuterated N-sulfated beta-lactams

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110313004A1 (en) * 2008-12-04 2011-12-22 Concert Pharmaceuticals, Inc. Deuterated pyridinones
US20130018193A1 (en) * 2007-09-10 2013-01-17 Concert Pharmaceuticals Inc. Deuterated pirfenidone
US8383823B2 (en) * 2007-06-20 2013-02-26 Auspex Pharmaceuticals Substituted N-aryl pyridinones
US8426407B2 (en) * 2009-05-25 2013-04-23 Central South University Preparation of 1-(substituted aryl)-5-trifluoromethyl-2-(1H)pyridone compounds and salts thereof and their applications

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8383823B2 (en) * 2007-06-20 2013-02-26 Auspex Pharmaceuticals Substituted N-aryl pyridinones
US20130018193A1 (en) * 2007-09-10 2013-01-17 Concert Pharmaceuticals Inc. Deuterated pirfenidone
US20110313004A1 (en) * 2008-12-04 2011-12-22 Concert Pharmaceuticals, Inc. Deuterated pyridinones
US8426407B2 (en) * 2009-05-25 2013-04-23 Central South University Preparation of 1-(substituted aryl)-5-trifluoromethyl-2-(1H)pyridone compounds and salts thereof and their applications

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9504677B2 (en) 2007-06-20 2016-11-29 Auspex Pharmaceuticals, Inc. Substituted N-aryl pyridinones
US10047077B2 (en) 2016-04-13 2018-08-14 Skyline Antiinfectives, Inc. Deuterated O-sulfated beta-lactam hydroxamic acids and deuterated N-sulfated beta-lactams
US10093666B2 (en) 2016-04-13 2018-10-09 Arixa Pharmaceuticals, Inc. Deuterated O-sulfated beta lactam hydroxamic acids and deuterated N-sulfated beta lactams

Similar Documents

Publication Publication Date Title
US9260424B2 (en) 4,6-diaminopyrimidine stimulators of soluble guanylate cyclase
US10568965B2 (en) Aminopyrimidine inhibitors of tyrosine kinase
US20110206661A1 (en) Trimethoxyphenyl inhibitors of tyrosine kinase
WO2010144477A2 (en) Sulfonylurea modulators of endothelin receptor
US20100143507A1 (en) Carboxylic acid inhibitors of histone deacetylase, gaba transaminase and sodium channel
US20110257260A1 (en) 3,4-methylenedioxyphenyl inhibitors of gaba aminotransferase and/or gaba reuptake transporter inhibitor
US20100113496A1 (en) Piperidine modulators of vmat2
WO2011123524A2 (en) Macrolide inhibitors of mtor
US20100076074A1 (en) Carbamate reducers of skeletal muscle tension
US20100075950A1 (en) Phenylpropanone modulators of dopamine receptor
US20100129366A1 (en) Thiazole inhibitors of cyclooxygenase
US20100120861A1 (en) Benzoic acid inhibitors of atp-sensitive potassium channels
US20100124541A1 (en) Hydroxyadamantyl inhibitors of dipeptidylpeptidase iv
US20100150899A1 (en) Pyrazolinone scavengers of free radical
US8227451B2 (en) Phenylacetic acid inhibitors of cyclooxygenase
WO2015171345A1 (en) N-aryl pyridinones modulators of fibrosis and/or collagen infiltration
US20100113405A1 (en) Methylindazole modulators of 5-ht3 receptors
US20100129311A1 (en) Phenylalanine amide inhibitors of atp-sensitive potassium channels
US20100120744A1 (en) Acetamidopropane modulators of nmda receptors
US20100113478A1 (en) Indolone modulators of 5-ht3 receptor
US20100130617A1 (en) Ethanolamine modulators of nmda receptor and muscarinic acetylcholine receptor
US20100093758A1 (en) Pyridine sulfonamide modulators of endothelin-a receptor
US20100144880A1 (en) Amino acid inhibitors of plasmin
US20100099701A1 (en) Isoquinolinone modulators of 5-ht3 receptors
US20100137332A1 (en) Piperazine modulators of nk-1 receptors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15788929

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15788929

Country of ref document: EP

Kind code of ref document: A1