WO2014008379A2 - Diamine and meglumine salt forms of fatty acids - Google Patents

Diamine and meglumine salt forms of fatty acids Download PDF

Info

Publication number
WO2014008379A2
WO2014008379A2 PCT/US2013/049290 US2013049290W WO2014008379A2 WO 2014008379 A2 WO2014008379 A2 WO 2014008379A2 US 2013049290 W US2013049290 W US 2013049290W WO 2014008379 A2 WO2014008379 A2 WO 2014008379A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
acid
formula
pharmaceutically acceptable
equivalent
Prior art date
Application number
PCT/US2013/049290
Other languages
French (fr)
Other versions
WO2014008379A3 (en
Inventor
Banavara L. Mylari
Frank C. SCIAVOLINO
Original Assignee
Thetis Pharmaceuticals Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US13/841,667 external-priority patent/US20140011814A1/en
Application filed by Thetis Pharmaceuticals Llc filed Critical Thetis Pharmaceuticals Llc
Publication of WO2014008379A2 publication Critical patent/WO2014008379A2/en
Publication of WO2014008379A3 publication Critical patent/WO2014008379A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C211/00Compounds containing amino groups bound to a carbon skeleton
    • C07C211/01Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms
    • C07C211/02Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • C07C211/09Diamines
    • C07C211/10Diaminoethanes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C215/00Compounds containing amino and hydroxy groups bound to the same carbon skeleton
    • C07C215/02Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C215/04Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated
    • C07C215/06Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated and acyclic
    • C07C215/10Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated and acyclic with one amino group and at least two hydroxy groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C57/00Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms
    • C07C57/02Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms with only carbon-to-carbon double bonds as unsaturation
    • C07C57/03Monocarboxylic acids

Definitions

  • hyperlipidemia is an elevation of lipids in the blood.
  • lipids include triglycerides, cholesterol, cholesterol esters, and phospholipids.
  • elevated levels of triglycerides in the blood is known as hypertriglyceridemia.
  • hypertriglyceridemia Although a certain amount of triglycerides are necessary for good health, increased triglyceride levels are often associated with increased risk of heart disease.
  • hyperlipidemia is associated with a number of disease states, including coronary artery disease, angina pectoris, carotid artery disease, strokes, cerebral arteriosclerosis, and xanthoma.
  • are compounds useful in the treatment of obesity, cardiovascular disease, and related indications such as such as cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke.
  • R + represents a piperidine or diamine group, wherein the nitrogen of the piperidine and one of the nitrogens of the diamine are protonated;
  • R ++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated
  • R ++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated, and X " is an anion of a pharmaceutically acceptable acid compound.
  • the compounds provided herein are of the Formula I, II, III, IV, V, VI, VII and VIII:
  • a pharmaceutical composition comprising a compound of the invention, and a pharmaceutically acceptable carrier.
  • a kit comprising a) a unit dosage comprising a compound of the invention, and b) instructions on how to use the kit; and at least one container for holding the unit dosage forms.
  • the compounds of the invention can be used to treat obesity, cardiovascular disease, and related indications, in a subject in need thereof.
  • a method of treating hyperlipidemia comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • a method of treating hypertriglyceridemia comprising administering to a subject in need thereof an effective amount of a compound the invention.
  • a method of treating dyslipidemia comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • a method of treating cardiovascular disease comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the cardiovascular disease can be cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, or stroke.
  • the arrhythmia is an atrial fibrillation.
  • a method of treating obesity comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • the subject is human.
  • provided herein is a method of treating prediabetes, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • a method of treating atherosclerosis comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • a method of lowering triglycerides in a subject in need thereof comprising administering to the subject an effective amount of a compound of the invention.
  • provided herein is a method of treating atrial fibrillation, or reducing the probability of an occurrence of an atrial fibrillation, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
  • a combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII and an antihyperlipidemic agent, to treat a metabolic disorder selected from the group consisting of type 2 diabetes (T2D), pre- diabetes, obesity, metabolic syndrome, hypertriglyceridemia and T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including atrial fibrillation, cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients.
  • T2D type 2 diabetes
  • pre- diabetes pre- diabetes
  • obesity metabolic syndrome
  • hypertriglyceridemia and T2D complications
  • T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including atrial fibrillation, cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients.
  • T2D type 2 diabetes
  • provided herein is a method of treating hyperlipidemia, comprising administering to a subject in need thereof an effective amount of the combination therapy described above. In another aspect, provided herein is a method of treating
  • hypertriglyceridemia comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of treating dyslipidemia comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of treating cardiovascular disease comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • the cardiovascular disease can be cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, or stroke.
  • the arrhythmia is an atrial fibrillation.
  • provided herein is a method of treating obesity, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of treating prediabetes comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of treating atherosclerosis comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of lowering triglycerides in a subject in need thereof comprising administering to the subject an effective amount of the combination therapy of the invention.
  • provided herein is a method of treating atrial fibrillation, or reducing the probability of an occurrence of an atrial fibrillation, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • the subject is human.
  • a combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII and an antihyperglycemic agent, to treat a metabolic disorder selected from the group consisting of T2D, pre-diabetes, obesity, metabolic syndrome, hypertriglyceridemia and T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients.
  • a metabolic disorder selected from the group consisting of T2D, pre-diabetes, obesity, metabolic syndrome, hypertriglyceridemia and T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients.
  • PUFAs co-3 polyunsaturated fatty acids
  • PUFAs include eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA).
  • EPA eicosapentaenoic acid
  • DHA docosahexaenoic acid
  • PUFAs have multiple biological functions through lipid-dependent and lipid-independent mechanisms.
  • EPA and mixtures of EPA and DHA have been shown to ameliorate triglycerides (TGs) lipid levels in patients with very high TGs.
  • EPA is shown to increase adiponectin secretion both in obese animals and obese human subjects (Itoh et al. Arteroscler. Thromb. Vase. Biol. 2007, 27, 1918-1925). Increased adiponectin levels are beneficial in regulating both lipid and glucose metabolism in animals as well as in humans.
  • R + represents a piperidine or diamine group, wherein the nitrogen of the piperidine and one of the nitrogens of the diamine are protonated;
  • R ++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated
  • R ++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated, and X " is an anion of a pharmaceutically acceptable acid compound.
  • X " is an anion of a pharmaceutically acceptable acid compound.
  • diamine refers to an organic compound composed of two amino groups.
  • the diamine can be any diamine composed of primary amines, secondary amines, or a combination of a primary amine(s) and a secondary amine(s).
  • the diamine can be a cyclic diamine, such as piperazine, diaminocyclohexane, diphenylethylenediamine, 1,8- diaminonaphthalene, 4,4'-diaminobiphenyl, N,N'-di-2-butyl-l ,4-phenylenediamine, dimethyl- 4-phenylenediamine, p-xylylenediamine (PXD), m-xylylenediamine (MXD), o- xylylenediamine (OXD), p-phenylenediamine (PPD), 2,5-diaminotoluene, m- phenylenediamine (MPD), o-phenylenediamine (OPD).
  • the diamine can also be an acyclic amine, such as ethylenediamine (1,2-diaminoethane), ethambutol,
  • TEDA tetramethylethylenediamine
  • 1,3-diaminopropane propane-1,3- diamine
  • putrescine butane- 1,4-diamine
  • cadaverine pentane- 1 ,5 -diamine
  • hexamethylenediamine hexane-l,6-diamine
  • 1,2-diaminopropane 1,2-diaminopropane
  • piperazine salts of EPA and DHA as well as meglumine salts of EPA and DHA.
  • the compound of Formula I and II are, respectively, the mono-salt of EPA with piperazine and the mono- salt of DHA with piperazine.
  • the compounds of Formula III and IV are, respectively, the di-salt of EPA with piperazine and the di-salt of DHA with piperazine.
  • X " is a pharmaceutically acceptable counter anion
  • the pharmaceutically acceptable counter anion can derived from acid compounds listed in Table 1 , pp 406 - 407, Handbook of Pharmaceutical Salts, P. Heinrich Stahl Camille G. Wermuth (Eds.)-
  • the pharmaceutically acceptable counter anion is selected from mineral acids, such as hydrochloric acid, hydrobromic acid, and phosphoric acid.
  • the pharmaceutically acceptable counter anion is selected from carboxylic acids, poly-carboxylic acids, and poly-hydroxy carboxylic acids, such as acetic acid, propionic acid, succinic acid, maleic acid, malic acid, tartaric acid, lactic acid, citric acid, benzoic acid, and hippuric acid (benzoylaminoethanoic acid).
  • the pharmaceutically acceptable counter anion is selected from sulfonic acids and hydroxyl- sulfonic acids, including, but not limited to, methanesulfonic acid, isethionic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, and benzenesulfonic acid.
  • the pharmaceutically acceptable counter anion is selected from amino acids, including, but not limited to, glycine, alanine, lysine, arginine, aspartic acid, or glutamic acid.
  • X " is an omega-3 polyunsaturated acid, such as eicosapentaenoic acid or docosahexaenoic acid.
  • provided herein is a compound of Formula V wherein X " is mandelic acid. In another embodiment, provided herein is a compound of V wherein X " is hippuric acid. In an embodiment, provided herein are the following compounds: the hydrochloride salt of Formula V, the hydrobromide salt of Formula V, the phosphate salt of Formula V, and the sulfate salt of Formula V.
  • hydrochloride salt of Formula VI the hydrobromide salt of Formula VI, the phosphate salt of Formula VI, and the sulfate salt of Formula VI.
  • the present invention also relates to compounds of the Formula V and Formula VI wherein X " is a pharmaceutically acceptable counter anion derived from naturally occurring amino acids.
  • X " is a pharmaceutically acceptable counter anion derived from naturally occurring amino acids.
  • the amino acids include, but are not limited to, glycine, alanine, lysine, and glutamic acid.
  • X " is a pharmaceutically acceptable counter anion as described above.
  • a compound of Formula XIII wherein X " is methanesulfonic acid.
  • the compounds of the invention e.g., compounds of Formulas I-XIV also include isomers and enantiomers wherever it is applicable.
  • PUFAs and esters of PUFAs are practically insoluble in water. In fact, they form soap-like emulsions when mixed with water. Therefore, the potential to derive optimum therapeutic benefits of PUFAs should be markedly facilitated by delivery of water soluble PUFAs.
  • the compounds of the present invention are more water soluble to achieve high oral absorption and to enable the preparation of intravenous dosage forms.
  • a method of treating obesity, cardiovascular disease, as well as related disorders comprising administering to a subject in need thereof a compound of the invention, e.g., compounds of Formulas I-XIV.
  • a compound of the invention e.g., compounds of Formulas I-XIV.
  • an effective amount of the compound is administered for treatment.
  • Hyperlipidemia is a condition generally characterized by an abnormal increase in serum lipids in the bloodstream and is an important risk factor in developing atherosclerosis and heart disease. Hyperlipidemia is usually classified as primary or secondary
  • hyperlipidemia Primary hyperlipidemia is generally caused by genetic defects, while secondary hyperlipidemia is generally caused by other factors, such as various disease states, drugs, and dietary factors. Alternatively, hyperlipidemia can result from both a combination of primary and secondary causes of hyperlipidemia. Elevated cholesterol levels are associated with a number of disease states, including coronary artery disease, angina pectoris, carotid artery disease, strokes, cerebral arteriosclerosis, and xanthoma.
  • hyperlipidemia is hypertriglyceridemia, which is defined as an elevated level of triglycerides. Elevated triglycerides have been associated with
  • Atherosclerosis even in the absence of high cholesterol levels.
  • High triglycerides can also lead to pancreatitis in excessive concentrations.
  • “obesity” refers to having a body weight more than about 30% greater than ideal body weight, as determined by a medical professional, and/or having a body mass index greater than about 27 as determined by a medical professional.
  • provided herein is a method of treating hyperlipidemia
  • a subject in need thereof comprising administering to the subject an effective amount of a compound of the invention, e.g. , compounds of Formulas I- XIV.
  • a method of treating cardiovascular diseases such as cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the invention, e.g., compounds of Formulas I-XIV.
  • the arrhythmia is an atrial fibrillation.
  • Atrial fibrillation is a type of cardiac arrhythmia where there is disorganized electrical conduction in the atria causing rapid uncoordinated contractions. These contractions can result in ineffective pumping of blood into the ventricle and a lack of synchrony.
  • the atrioventricular node receives electrical impulses from numerous locations throughout the atria instead of only from the sinus node. This overwhelms the atrioventricular node into producing an irregular and rapid heartbeat. As a result, blood pools in the atria that increases a risk for blood clot formation.
  • the major risk factors for atrial fibrillation include age, coronary artery disease, rheumatic heart disease, hypertension, diabetes, and thyrotoxicosis.
  • a method of treating obesity in a subject in need thereof comprising administering to the subject an effective amount of a compound of the invention, e.g., compounds of Formulas I-XIV.
  • Atherosclerosis refers to the buildup of fats and cholesterol in and on artery walls (plaques), which can restrict blood flow. These plaques can also burst, triggering a blood clot.
  • plaques can also burst, triggering a blood clot.
  • atherosclerosis is often considered a heart problem, it can affect arteries anywhere in the body. An animal model of atherosclerosis research is described in
  • treat includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated.
  • the treatment comprises the induction of a disease, followed by the activation of the compound of the invention, which would in turn diminish or alleviate at least one symptom associated or caused by the protein kinase- associated disorder being treated.
  • treatment can be diminishment of one or several symptoms of a disorder or complete eradication of a disorder.
  • subject is intended to include organisms, e.g., prokaryotes and eukaryotes, which are capable of suffering from or afflicted with a disease, disorder or condition.
  • subjects include mammals, e.g. , humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals.
  • the subject is a human, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from obesity; cardiovascular diseases such as cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke; or hyperlipidemia, including hypertriglycreridemia,
  • cardiovascular diseases such as cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke
  • hyperlipidemia including hypertriglycreridemia
  • pharmaceutically acceptable amount of the compound is that amount necessary or sufficient to treat or prevent a disorder, e.g. , prevent the various morphological and somatic symptoms of a disease or condition described herein.
  • an effective amount of a compound of the invention is the amount sufficient to treat obesity, cardiovascular disorders, or a related disorder, in a subject.
  • the effective amount can vary depending on such factors as the size and weight of the subject, the type of illness, or the particular compound of the invention.
  • pharmaceutically acceptable salts includes both pharmaceutically acceptable acid addition salts and pharmaceutically acceptable cationic salts, where appropriate.
  • pharmaceutically-acceptable cationic salts is intended to define but is not limited to such salts as the alkali metal salts, ⁇ e.g.
  • alkaline earth metal salts ⁇ e.g., calcium and magnesium
  • aluminum salts ⁇ e.g., ammonium salts, and salts with organic amines such as benzathine ( ⁇ , ⁇ '-dibenzylethylenediamine), choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), benethamine (N-benzyl phenethylamine) , diethylamine, piperazine, tromethamine (2-amino-2-hydroxymethyl-l ,3- propanediol) and procaine.
  • organic amines such as benzathine ( ⁇ , ⁇ '-dibenzylethylenediamine), choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), benethamine (N-benzyl phenethylamine) , diethylamine, piperazine, tromethamine (2-amino-2-hydroxymethyl-l ,
  • salts are, for example, inorganic acids, such as hydrohalic acids, e. g. hydrochloric, hydrobromic or the like, or sulfuric acid, nitric acid, or phosphoric acid; or suitable organic acids, for example suitable aliphatic acids, like aliphatic mono or dicarboxylic acids, hydro xyalkanoic or hydroxyalkanedioic acids, e.g.
  • acids are e.g. hydrobromic acid, sulphuric acid, phosphoric acid, acetic, benzoic, fumaric, maleic, citric, tartaric, gentisic, dobesilic, methanesulfonic, ethanesulfonic, laurylsulfonic, benzenesulfonic, and para- toluenesulfonic acids.
  • the pharmaceutically acceptable salt is selected from the group consisting of an L-arginine, benenthamine, benzathine, betaine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)ethanol, ethanolamine, ethylenediamine, N-methylglucamine, hydrabamine, 1 H-imidazole, lithium hydroxide, L-lysine, magnesium hydroxide, 4-(2-hydroxyethyl)morpholine, piperazine, potassium hydroxide, l -(2-hydroxyethyl)pyrrolidine, sodium hydroxide, triethanolamine, tromethamine, zinc hydroxide, sodium, calcium, potassium, magnesium, and zinc.
  • the triglyceride lowering efficacy of the compounds of the present invention can be determined in animal models according to the procedure described by Sidika et al in Journal of Lipid Research, 1992, 33, 1-7.
  • Efficacy of the compounds of the present invention in increasing adiponectin secretion in rodent models of obesity and human obese subjects can be determined according the procedures described by Itoh et al. in Arteroscler. Thromb. Vase. Biol. 2007, 27, 1918- 1925.
  • the compounds of the present invention e.g. piperazine and meglumine salts of polyunsaturated fatty acids, (including eicosapentaenoic acid and docosahexaenoic acid, or a mixture thereof) are well-suited to use in combination therapy.
  • combination therapy refers to the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure.
  • 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 containers ⁇ e.g., capsules) for each active ingredient.
  • administration also encompasses use of each type of therapeutic agent in a sequential manner, either at approximately the same time or at different times. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
  • a “mixture of compounds of Formula I, II, III, IV, V, VI, VII or VIII” refers to a mixture of two or more of these compounds present in a and b %, and optionally c, d, e, f, g and/or h % , wherein a and b, and optionally c, d, e, f, g and/or h are not zero, but the sum of a and b, and optionally c, d, e, f, g and/or h, is 100 %.
  • a compound of Formula I is present at 50% and a compound of Formula III is present at 50%.
  • a combination therapy comprising an effective amount of a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a combination thereof, and an antihyperlipidemic or an antihyperglycemic agent.
  • An "effective amount" of a combination of agents is an amount sufficient to provide an observable improvement over the baseline clinically observable signs and symptoms of the disorders treated with the combination.
  • the combination of a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a combination thereof, and an antihyperlipidemic or antihyperglycemic agent described herein displays a synergistic effect.
  • the term "synergistic effect" as used herein refers to action of two agents producing an effect, for example, slowing the symptomatic progression of diabetes or symptoms thereof, which is greater than the simple addition of the effects of each drug administered by themselves.
  • a synergistic effect can be calculated, for example, using suitable methods such as the Sigmoid-Emax equation (Holford, N. H. G. and Scheiner, L. B., Clin. Pharmacokinet.
  • the combination therapy comprises the compounds of the present invention and an antihyperlipidemic agent, to treat a metabolic disorder selected from the group consisting of T2D, pre-diabetes, obesity, metabolic syndrome, hypertriglyceridemia and T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including atrial fibrillation, cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients.
  • a metabolic disorder selected from the group consisting of T2D, pre-diabetes, obesity, metabolic syndrome, hypertriglyceridemia and T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including atrial fibrillation, cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients.
  • An additional aspect provided herein is combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII and an antihyperlipidemic agent, to treat obesity, cardiovascular disease, and related indications in a subject in need thereof.
  • An additional aspect provided herein is combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII or XIV and an antihyperlipidemic agent, to treat obesity, cardiovascular disease, and related indications in a subject in need thereof.
  • provided herein is a method of treating hyperlipidemia, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In another aspect, provided herein is a method of treating
  • hypertriglyceridemia comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of treating dyslipidemia comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of treating cardiovascular disease comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • the cardiovascular disease can be cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, or stroke.
  • the arrhythmia is an atrial fibrillation.
  • provided herein is a method of treating obesity, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of treating prediabetes comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of treating atherosclerosis comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • a method of lowering triglycerides in a subject in need thereof comprising administering to the subject an effective amount of the combination therapy of the invention.
  • a method of treating atrial fibrillation, or reducing the probability of an occurrence of an atrial fibrillation comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
  • provided herein is a method of lowering the cholesterol level and/or the triglyceride level in a mammal comprising administering to the subject an effective amount of the combination therapy of the invention.
  • the subject is human.
  • the invention relates to a combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII or a compound of the structural Formula I, II, III, IV, V or VI and an antihyperlipidemic agent, or a combination of two or more compounds of the structural Formulas I, II, III, IV, V, VI, VII or VIII or compounds of the structural Formulas I, II, III, IV, V or VI and an antihyperlipidemic agent and a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically- acceptable salt of said prodrug.
  • a pharmaceutical composition comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these, an antihyperlipidemic agent and a pharmaceutically-acceptable carrier.
  • the pharmaceutical composition comprises two or more compounds of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these, the two or more compounds are present in x, y, z, ...% etc. with the proviso that x, y, z, ...% are not zero, but the sum x, y, z, ...% is 100%.
  • the antihyperlipidemic agent is about 0.1-1 % by weight of the pharmaceutical composition.
  • the compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof is present in unit dosage strength of 250, 500, 750, 1000 or 1250 mg, and the said antihyperlipidemic agent is present in a unit dosage strength of 1, 2.5, 5, 10, 20, 30, 40, or 50 mg.
  • the said antihyperlipidemic agent is present in a unit dosage strength of 5-100 mg.
  • the components of the combination therapy can be administered in a variety of ways.
  • the components are in separate formulations or unit dosage forms.
  • the components are administered with a pharmaceutically acceptable carrier.
  • the components can be administered separately, at substantially the same time, or administered at different times. When administered separately, they can be administered in any order.
  • the present invention is directed to pharmaceutical compositions comprising compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these and an antihyperlipidemic agent, or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug; and a pharmaceutically- acceptable carrier, vehicle or diluent.
  • Antihyperlipidemic agents that may be used in accordance with the invention may include, for example, statins, which are HMG CoA enzyme inhibitors, cholesterol absorption inhibitors, and cholesterol esterase transfer protein (CETP) inhibitors and pharmaceutically- acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrug, and others.
  • statins which are HMG CoA enzyme inhibitors, cholesterol absorption inhibitors, and cholesterol esterase transfer protein (CETP) inhibitors and pharmaceutically- acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrug, and others.
  • the antihyperlipidemic agent is a statin, cholesterol absorption inhibitor, and CETP inhibitor or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug.
  • the pharmaceutically acceptable salt is selected from the group consisting of a propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne- 1 ,4- dioate, hexyne-l ,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephathal ate, sulfonate, xylenesulfonate, phenyl acetate, phenylpropionate, phenylbutyrate, citrate, lactate, p- hydroxybutyrate, glycolate, tartrate, methanesulfonate, propanesulfonates, naphthalene- 1 -sulf
  • statins are atorvastatin, risuvostatin, simvastatin, or pravastatin, pharmaceutically-acceptable salts or prodrugs thereof, and pharmaceutically- acceptable salts of said prodrugs.
  • a preferred agent among cholesterol absorption inhibitors is ezetimibe also known as Zetia.
  • a preferred agent among CETP inhibitors is anacetrapib.
  • CETP inhibitors include, but are not limited to anacetrapib or a hydrate, and solvate thereof.
  • the pravastatin is present in the amount ranging from 5 mg to
  • the ezetimibe is present 5 mg to 50 mg.
  • kits comprising a first unit dosage form comprising compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these; a second unit dosage form comprising an antihyperlipidemic agent or a hydrate, and solvate thereof; and a container.
  • antihyperlipidemic agents that may be used in accordance with the invention are members of different classes of antihyperlipidemic agents (e.g. , HMG-CoA reductase inhibitors (statins), CETP inhibitors, and cholesterol absorption inhibitors and others), pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
  • HMG-CoA reductase inhibitor refers to a compound that competitively blocks the enzyme 3-hydroxy-3-methyl-glutaryl-co-enzyme A (HMG-CoA) reductase.
  • HMG-CoA reductase inhibitors interfere with cholesterol formation (enzyme catalyzes the conversion ofHMG-CoA to mevalonate). As a result, they decrease total cholesterol, low-density lipoprotein cholesterol (LDL-C), apolipoprotein B (a membrane transport complex for LDL-C), very low-density lipoprotein (VLDL), and plasma triglycerides.
  • LDL-C low-density lipoprotein cholesterol
  • VLDL very low-density lipoprotein
  • HMG-CoA reductase inhibitors which may be used in accordance with the disclosure include, but are not limited to: atorvastatin, which may be prepared as disclosed in U.S. Patent No. 7,030,151 ; pravastatin and related compounds which may be prepared as disclosed in U.S. Patent Nos. 4,346,227 and 4,448,979; rosuvastatin, which may be prepared as disclosed in U.S. Patent No.6, 858,618; simvastatin and related compounds which may be prepared as disclosed U.S. Patent Nos. 4,448,784 and 4,450,171.
  • HMG-CoA reductase inhibitors also include atorvastatin, simvastatin, pravastatin, lovastatin, fluvastatin, rosuvastatin, cerivastatin, mevastatin, rivastatin, pravastatin, nisvastatin, itavastatin, velostatin and fluindo statin.
  • CETP inhibitor refers to a compound which catalyses the transfer of cholesteryl ester from HDL to apolipoprotein B containing lipoproteins in exchange for triglyceride and thereby plays a major role in lipoprotein metabolism.
  • CETP inhibitors see, for example, Curr. Opin. Pharmacal. 6:162 (2006) and references cited therein.
  • CETP inhibitors which may be used in accordance with the disclosure are not limited by any structure or group of CETP inhibitors.
  • CETP inhibitors which may be used in accordance with the disclosure include, but are not limited to: anacetrapib, which may be prepared as disclosed in WO 2007005572. The disclosure thereof is incorporated herein by reference.
  • cholesterol absorption inhibitors refers to a compound that inhibits the absorption ofbiliary and dietary cholesterol from the small intestine without affecting the absorption of fat-soluble vitamins, triglycerides, or bile acids.
  • cholesterol absorption inhibitors see, for example, Nutr. Metab. Cardiovasc. Dis., 13:42 (2004) and references cited therein.
  • Cholesterol absorption inhibitors which may be used in accordance with the disclosure include, but are not limited to ezetimibe (Zetia), which may be prepared as disclosed in U.S. Patent Nos. 5,767,115 and 5, 846,966. The disclosures thereof are incorporated herein by reference. In the practice of the compositions and methods of the disclosure, any HMG Co-A reductase inhibitors (or) or in a pharmaceutically acceptable combination with any flushing inhibiting agent may be employed.
  • compositions comprising a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof; and an antihyperlipidemic agent or a pharmaceutically acceptable salt, hydrate, and solvate thereof.
  • the disclosure provides for unit dose formulations comprising a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof, and an antihyperlipidemic agent or a pharmaceutically acceptable salt, hydrate, and solvate.
  • combination therapies comprising, at least, the following combination of agents:
  • combination therapies of paragraphs 1 , 2, 3, 4, 5, 6, 7 and 8, above can be further combined with compounds of Formula I, II, III, IV, V, VI, VII and/or VIII.
  • a combination therapy comprising the compounds of the present invention and an antihyperglycemic agent, or a pharmaceutically- acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug.
  • the present invention provides a method of treating a metabolic disorder selected from the group consisting of T2D, pre-diabetes, obesity, metabolic syndrome, hypertriglyceridemia and diabetes complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including cardiac arrhythmia, myocardial infarction, and cardiomyopathy in mammals, e.g. , diabetic patients, comprising administering to a subject in need thereof the combination therapy described herein
  • the components of this combination therapy can be administered in a variety of ways.
  • the components are in separate formulations or unit dosage forms.
  • the components are administered with a pharmaceutically acceptable carrier.
  • the components can be administered separately, at substantially the same time, or administered at different times. When administered separately, they can be administered in any order.
  • the present invention is directed to pharmaceutical compositions comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these and an antihyperglycemic agent, or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug; and a
  • the components of this combination therapy can be administered in a variety of ways.
  • the components are in separate formulations or unit dosage forms.
  • the components are administered with a
  • compositions can be administered separately, at substantially the same time, or administered at different times. When administered separately, they can be administered in any order.
  • the present invention is directed to pharmaceutical compositions comprising a compound of the structural Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII or XIV, or a mixture of these and an antihyperglycemic agent, or a
  • the antihyperglycemic agent is about 1-20% by weight of the pharmaceutical composition.
  • said compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof is present in unit dosage strength of 250, 500, 750, 1000 or 1250 mg, and the said antihyperglycemic agent is present in a unit dosage strength of 1, 2.5, 5, 10, 20, 25, 50, 100, 150, or 200 mg.
  • the antihyperglycemic agent is present in a unit dosage strength of 5-100mg.
  • Antihyperglycemic agents that may be used in accordance with the invention may include, for example, sulfonylureas, meglitinides, thiazolidinediones, alpha-glucosidase inhibitors, DPP IV inhibitors, and SGLT-2 inhibitors and pharmaceutically-acceptable salts and prodrug thereof, and pharmaceutically-acceptable salts of said prodrug, and others.
  • the antihyperglycemic agent is a sulfonylurea, meglitinide, thiazolidinedione, alpha-glucosidase inhibitor, DPP IV inhibitor, and SGLT-2 inhibitors or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug.
  • the pharmaceutically acceptable salt is selected from the group consisting of a propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne- 1 ,4-dioate, hexyne-1 ,6- dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephathal ate, sulfonate, xylenesulfonate, phenyl acetate, phenylpropionate, phenylbutyrate, citrate, lactate, p- hydroxybutyrate, glycolate, tartrate, methanesulfonate, propanesulfonates, naphthalene- 1 -s
  • Preferred agents among thiazolidinediones are pioglitazone, pharmaceutically- acceptable salts or a prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
  • Preferred alpha- glucosidase inhibitors include, but are not limited to, acarbose, vaglibose, and miglitol, pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
  • Preferred DPP-IV inhibitors include, but are not limited to, sitagliptin, linagliptin, vildagliptin, saxagliptin, alogliptin, denagliptin, carmegliptin, melogliptin and dutogliptin. and pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
  • Preferred SGLT-2 inhibitors include, but are not limited to, dapagliflozin
  • antihyperglycemic agents that may be used in accordance with the invention are members of different classes of antihyperglycemic agents (e.g., sulfonylureas, meglitinides, thiazolidinediones, alpha-glucosidase inhibitors, DPP-IV inhibitors, SGLT-2 inhibitors and others), pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically- acceptable salts of said prodrugs.
  • antihyperglycemic agents e.g., sulfonylureas, meglitinides, thiazolidinediones, alpha-glucosidase inhibitors, DPP-IV inhibitors, SGLT-2 inhibitors and others
  • pharmaceutically-acceptable salts and prodrugs e.g., sulfonylureas, meglitinides, thiazolidinediones, alpha-glucosidase inhibitors, DPP-IV inhibitors,
  • sulfonylureas refers to a class of compounds that stimulate insulin release by binding to the sulfonylurea receptor, a subunit of the ICATP channel complex. This binding leads to closure of the channel, resulting in voltage change in the beta-cell membrane and, in turn, influx of Ca 2+ ions causing exocytosis of insulin granules.
  • sulfonylureas see, for example, Metabolism, 55, 20 (2006) incorporated by reference, herein, in its entirety and references cited therein, and Lancet, 358, 1709 (2001) incorporated by reference, herein, in its entirety and references cited therein.
  • thiazolidinediones refers to a class of compounds that are selective agonists for the peroxisome proliferator-activated receptor gamma (PPARy), a member of family of nuclear hormone receptors that function as ligand-activated transcription factors.
  • PARy peroxisome proliferator-activated receptor gamma
  • thiazolidinediones see, for example, Trends Endocrin. Met., 10, 9 (1999) and references cited therein.
  • alpha-glucosidase inhibitors refers to a class of compounds having the ability to competitively inhibit brush border enzyme alpha-glucosidase in the GI tract, which has the ability to cleave complex carbohydrates into sugars.
  • alpha-glucosidase inhibitors see, for example, Diabetes Res. Clin. Pr., 40, S51 (1998) and references cited therein.
  • DPP IV inhibitors refers to a class of compounds that have the ability to selectively inactivate the enzyme DPP-IV, and those which have the ability to rapidly inactivate incretin hormones ⁇ e.g. , glucagon-like peptide-1 (GLP-1) and insulinotropic polypeptide (GIP)), that are released by the intestine throughout the day, and whose levels are increased after a meal.
  • GLP-1 glucagon-like peptide-1
  • GIP insulinotropic polypeptide
  • the DPP-IV inhibitor sitagliptin can be prepared according to procedure described by Kim et al. in Journal of Medicinal Chemistry, 48, 141-151 , (2005) and in Journal of Medicinal Chemistry, 51, 589- 602, (2008).
  • SGLT-2 inhibitors refers to a class of compounds that have the ability to selectively inhibit renal sodium-glucose co -transporter 2 and prevent renal glucose reabsorption from the glomerular filtrate and provide an insulin-independent means of controlling hyperglycemia.
  • SGLT-2 inhibitors see, for example, Journal of Medicinal Chemistry, 52, 1785 1794, (2009) and references cited therein.
  • any sulfonylurea, meglitinide, thiazolidinedione, alpha-glucosidase inhibitor, DPP-IV inhibitor, or a SGLT-2 inhibitor or a pharmaceutically- acceptable salt or a prodrug thereof, or a pharmaceutically- acceptable salt of said prodrug, or any combination thereof, may be employed.
  • Sulfonylureas that may be used in accordance with the invention include, but are not limited to, acetohexamide, which may be prepared as described in U.S. Patent No. 3,013 ,072; 1- Butyl-3-methanilyl urea, which may be prepared as described in U.S. Patent No.
  • carbutamide which may be prepared as described in U.S. Patent No. 4,324,796
  • chlorpropamide which may be prepared as described in U.S. Patent No. 4,381 ,304;
  • glibornuride which may be prepared as described in U.S. Patent No. 4, 153,710
  • gliclazide which may be prepared as described in U.S. Patent No. 6,733,782
  • glipizide which may be prepared and its use as oral administration as described in U.S. Patent No. 5,545,413
  • gliquidone has been described and its use as described in U.S. Patent No. 4,708,868;
  • glyburide or glibenclamide which may be prepared and its use as described in U.S. Patent No. 6,830,760; glybuthiazole, which may be prepared as described in U.S. Patent
  • pioglitazone refers to pioglitazone, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salt thereof such as the hydrochloride salt.
  • DPP-IV inhibitors that may be used in accordance with the invention include, but are not limited to, linagliptin, sitagliptin, vildagliptin, alogliptin, saxagliptin, denagliptin, carmegliptin, melogliptin, and dutogliptin, or a pharmaceutically-acceptable salt of one of the beforementioned DPP IV inhibitors, or a prodrug thereof.
  • linagliptin refers to linagliptin and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. Crystalline forms are described in WO 2007/128721. Methods for the manufacture of linagliptin are described in the patent applications WO 2004/018468 and WO 2006/ 048427 for example. Linagliptin is distinguished from structurally comparable DPP IV inhibitors, as it combines exceptional potency and a long- lasting effect with favorable pharmacological properties, receptor selectivity and a favorable side-effect profile or bring about unexpected therapeutic advantages or improvements when used in combination with an SGLT2 inhibitor and a third antidiabetic agent according to this invention.
  • sitagliptin refers to sitagliptin (or MK-0431) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. In one embodiment, sitagliptin is in the form of its
  • sitagliptin phosphate is in the form of a crystalline anhydrate or monohydrate.
  • a class of this embodiment refers to sitagliptin phosphate monohydrate.
  • Sitagliptin free base and pharmaceutically acceptable salts thereof are disclosed in U.S. Patent No. 6,699,871 and in Example 7 of WO 03/004498.
  • Crystalline sitagliptin phosphate monohydrate is disclosed in WO 2005/003135 and in WO 2007/050485. For details, e.g. on a process to manufacture or to formulate this compound or a salt thereof, reference is thus made to these documents.
  • a tablet formulation for sitagliptin is commercially available under the trade name Januvia®.
  • vildagliptin refers to vildagliptin (or LAF-237) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. Specific salts of vildagliptin are disclosed in WO 2007/019255. A crystalline form of vildagliptin as well as a vildagliptin tablet formulation are disclosed in WO 2006/078593. Vildagliptin can be formulated as described in WO 00/34241 or in WO 2005/067976. A modified release vildagliptin formulation is described in WO 2006/135723. For details, e.g.
  • saxagliptin refers to saxagliptin and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof.
  • saxagliptin is in the form of the free base or a HC1 salt (for example as mono- or dihydrochloride, including hydrates thereof), or a mono-benzoate salt as disclosed in WO 2004/052850 and WO 2008/131149.
  • saxagliptin is in the form of the free base.
  • saxagliptin is in the form of the monohydrate of the free base as disclosed in WO 2004/052850.
  • a process for preparing saxagliptin is also disclosed in WO 2005/106011 and WO 2005/ 115982.
  • Saxagliptin can be formulated in a tablet as described in WO 2005/117841.
  • a process to manufacture, to formulate or to use this compound or a salt thereof reference is thus made to these documents and U.S. Patent No. 6,395,767 and WO 01/68603.
  • denagliptin refers to denagliptin (or GSK-823093) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof
  • denagliptin is in the form of its hydrochloride salt as disclosed in Example 2 of WO 03/002531 or its tosylate salt as disclosed in WO 2005/009956.
  • a class of this embodiment refers to denagliptin tosylate.
  • Crystalline anhydrous denagliptin tosylate is disclosed in WO 2005/009956.
  • alogliptin refers to alogliptin (or SYR-322) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof
  • alogliptin is in the form of its benzoate salt, its hydrochloride salt or its tosylate salt each as disclosed in WO 2007/035629.
  • a class of this embodiment refers to alogliptin benzoate.
  • Polymorphs of alogliptin benzoate are disclosed in WO 2007/035372.
  • a process for preparing alogliptin is disclosed in WO 2007/112368 and, specifically, in WO 2007/035629.
  • Alogliptin (namely its benzoate salt) can be formulated in a tablet and administered as described in WO 2007/033266.
  • Alogliptin namely its benzoate salt
  • WO 2007/033266 for details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents and to US 2005/261271 , EP 1586571 and WO 2005/095381.
  • carmegliptin refers to carmegliptin
  • melogliptin refers to melogliptin
  • Specifically claimed salts include the methanesulfonate and p-toluene-sulfonate.
  • methanesulfonate and p-toluene-sulfonate are examples of compounds that are used in the production of salts.
  • p-toluene-sulfonate for details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
  • dutogliptin refers to dutogliptin (or PHX-1149, PHX- 1149T) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. Methods for its preparation are inter alia disclosed in WO 2005/047297. Pharmaceutically acceptable salts include the tartrate. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
  • SGLT-2 inhibitors that may be used in accordance with the invention include, but are not limited to dapagliflozin, canagliflozin, atigliflozin, remogliflozin and sergliflozin.
  • dipagliflozin refers to dapagliflozin, including hydrates and solvates thereof, and crystalline forms thereof.
  • the compound and methods of its synthesis are described in WO 03/099836 for example.
  • Preferred hydrates, solvates and crystalline forms are described in the patent applications WO 2008/116179 and WO 2008/ 002824 for example.
  • canaglifiozin refers to canagliflozin, including hydrates and solvates thereof, and crystalline forms thereof and has the following structure:
  • the compound and methods of its synthesis are described in WO 2005/012326 and WO 2009/035969 for example.
  • Preferred hydrates, solvates and crystalline forms are described in the patent applications WO 2008/069327 for example.
  • atigliflozin refers to atigliflozin, including hydrates and solvates thereof, and crys-talline forms thereof.
  • the compound and methods of its synthesis are described in WO 2004/007517 for example.
  • remogliflozin refers to remogliflozin and prodrugs of remogliflozin, in particular remogliflozin etabonate, including hydrates and solvates thereof, and crystalline forms thereof. Methods of its synthesis are described in the patent applications EP 1213296 and EP 1354888 for example.
  • sergliflozin refers to sergliflozin and prodrugs of sergliflozin, in particular sergliflozin etabonate, including hydrates and solvates thereof, and crystalline forms thereof. Methods for its manufacture are described in the patent applications EP 1344780 and EP 1489089 for example.
  • combination therapies comprising, at least, the following combination of agents:
  • combination therapies of paragraphs 1, 2, 3, 4, 5, 6, 7 and 8, above can be further combined with compounds of Formula I, II, III, IV, V, VI, VII and/or VIII.
  • the compounds of the present invention are suitable as active agents in
  • compositions that are efficacious particularly for treating obesity, cardiovascular disorders, as well as related conditions.
  • the pharmaceutical composition in various embodiments has a pharmaceutically effective amount of the present active agent along with other pharmaceutically acceptable excipients, carriers, fillers, diluents and the like.
  • Pharmaceutical compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in
  • composition includes preparations suitable for administration to mammals, e.g., humans.
  • pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • phrases "pharmaceutically acceptable carrier” is art recognized and includes a pharmaceutically acceptable material, composition or vehicle, suitable for administering compounds of the present invention to mammals.
  • the carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, saffiower oil, sesame oil, olive oil, corn oil and soybean oil;
  • glycols such as propylene glycol
  • polyols such as glycerin, sorbitol, mannitol and polyethylene glycol
  • esters such as ethyl oleate and ethyl laurate
  • agar buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, a- tocopherol, and the like; and metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin
  • Formulations of the present invention include those suitable for oral, nasal, topical, buccal, sublingual, rectal, vaginal and/or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound that produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
  • Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient.
  • a compound of the present invention may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as, for example, cetyl alcohol and glycerol monostea
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydro xypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface- active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydro xypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions that can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents and e
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • compositions of the invention will vary, depending upon the identity, size, and condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100% (w/w) active ingredient.
  • the present invention relates to a pharmaceutical composition for the treatment of dyslipidemia, including hypertriglyceridemia, in mammals comprising an anti-dyslipidemia effective amount of a compound of the present invention and a pharmaceutically acceptable carrier.
  • said mammals are human.
  • the present invention relates to a pharmaceutical composition for the treatment of cardiovascular diseases, including cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke in mammals comprising an effective amount of a compound of the present invention to treat the said cardiovascular diseases and a pharmaceutically acceptable carrier.
  • the arrhythmia is an atrial fibrillation.
  • said mammals are human
  • the present invention relates to a pharmaceutical composition for the treatment of obesity, in mammals comprising an anti-obesity effective amount of a compound of the present invention and a pharmaceutically acceptable carrier. In one embodiment said mammals are human.
  • the total daily dose of the compounds of the invention is typically in the range 0.05 g to 12 g, e.g. , 1 g to 12 g, depending, of course, on the mode of administration.
  • the total daily dose is in the range 1 g to 10 g and in another embodiment the total daily dose is in the range 1 g to 6 g.
  • the daily dose is 0.05 g - 6 g, 0.05 g - 3 g, 0.05 g - 1 g, or 0.05 g - 0.2 g.
  • the total daily dose may be administered in single or divided doses.
  • These dosages are based on an average human subject having a weight of about 65kg to 70kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.
  • a pharmaceutical composition of the invention may be prepared, packaged, or sold in bulk, as a single unit dose, or as a plurality of single unit doses.
  • a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • kits for use by a consumer for treating disease comprise a) a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable carrier, vehicle or diluent; and, optionally, b) instructions describing a method of using the pharmaceutical composition for treating the specific disease.
  • a "kit” as used in the instant application includes a container for containing the separate unit dosage forms such as a divided bottle or a divided foil packet.
  • the container can be in any conventional shape or form as known in the art which is made of a
  • a pharmaceutically acceptable material for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule.
  • the container employed can depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle which is in turn contained within a box.
  • Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material.
  • recesses are formed in the plastic foil.
  • the recesses have the size and shape of individual tablets or capsules to be packed or may have the size and shape to accommodate multiple tablets and/or capsules to be packed.
  • the tablets or capsules are placed in the recesses accordingly and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed.
  • the tablets or capsules are individually sealed or collectively sealed, as desired, in the recesses between the plastic foil and the sheet.
  • the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
  • the written memory aid is of the type containing information and/or instructions for the physician, pharmacist or subject, e.g. , in the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen which the tablets or capsules so specified should be ingested or a card which contains the same type of information.
  • a calendar printed on the card e.g. , as follows "First Week, Monday,
  • a "daily dose” can be a single tablet or capsule or several tablets or capsules to be taken on a given day.
  • Another specific embodiment of a kit is a dispenser designed to dispense the daily doses one at a time.
  • the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen.
  • An example of such a memory-aid is a mechanical counter, which indicates the number of daily doses that, has been dispensed.
  • a memory-aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
  • One embodiment of the present invention relates to a kit comprising a unit dosage comprising a compound of the invention with instructions on how to use the kit and with provision for at least one container for holding the unit dosage form.
  • the salts of the invention can be prepared using any number of synthesis techniques known to the skilled artisan.
  • reaction inert solvent refers to a solvent or a mixture of solvents that does not interact with starting materials, reagents, intermediates or products in a manner that adversely affects the yield of the desired product.
  • Preferred solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile.
  • EPA ethyl methyl ketone
  • Both piperazine and EPA are commercially available.
  • the reaction mixture can be stirred at about ambient temperature to about the reflux temperature of the solvent being used for about two hours to about six hours.
  • the mono salt of piperazine with EPA can be isolated from the mixture by methods well known to those skilled in the art, including according to the method of U.S. Patent No.
  • the di-salt of piperazine with EPA (Formula III) can be prepared according to the above procedure, but by using two equivalents instead of one equivalent of EPA.
  • the mono salt of piperazine with docosahexaenoic acid (DHA) (Formula II) can be prepared as set forth below.
  • One equivalent of piperazine may be dissolved in an appropriate reaction inert solvent.
  • the solvent may be polar such as water.
  • Preferred reaction inert solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile.
  • To this solution may be added a solution of one equivalent of DHA.
  • Both piperazine and DHA are commercially available.
  • the reaction mixture can be stirred at about ambient temperature to about the reflux temperature of the solvent being used for about two hours to about six hours.
  • the mono salt of piperazine with DHA can be isolated from the mixture by methods well known to those skilled in the art, including according to the method of U.S. Patent No. 7,973,073.
  • the di-salt of piperazine with DHA (Formula IV) can be prepared according to the above procedure, but by using two equivalents instead of one equivalent of DHA.
  • the compound of Formula V can be prepared by adding a solution of one equivalent of compound XH to a solution of one equivalent of the compound of the Formula I.
  • Suitable solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile.
  • the compound of Formula VI can be prepared by adding a solution of one equivalent of compound XH to a solution of one equivalent of the compound of the Formula II.
  • Suitable solvents include methanol, ethanol, n- propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile.
  • the meglumine salt of EPA (Formula VII) can be prepared as set forth below.
  • One equivalent of meglumine may be dissolved in an appropriate reaction inert solvent.
  • the solvent may be polar such as water.
  • Preferred reaction inert solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile.
  • To this solution may be added a solution of one equivalent of EPA. Both meglumine and EPA are commercially available.
  • the reaction mixture can be stirred at about ambient temperature to about the reflux temperature of the solvent being used for about two hours to about six hours.
  • the meglumine salt of EPA can be isolated from the mixture by methods well known to those skilled in the art, including according to the method of U.S. Patent No. 7,973,073.
  • the meglumine salt of DHA (Formula VIII) can be prepared as set forth below.
  • One equivalent of meglumine may be dissolved in an appropriate reaction inert solvent.
  • the solvent may be polar such as water.
  • Preferred reaction inert solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile.
  • To this solution may be added a solution of one equivalent of DHA.
  • Both meglumine and DHA are commercially available.
  • the reaction mixture can be stirred at about ambient temperature to about the reflux temperature of the solvent being used for about two hours to about six hours.
  • the meglumine salt of DHA can be isolated from the mixture by methods well known to those skilled in the art, including according to the method of U.S. Patent No. 7,973,073.
  • a method for the manufacture of a compound of Formula I or II comprising: reacting one equivalent of piperazine, and one equivalent of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
  • a method for the manufacture of a compound of Formula III or IV comprising: reacting one equivalent of piperazine, and two equivalents of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
  • a method for the manufacture of a compound of Formula V or VI comprising: reacting one equivalent of piperazine, one equivalent of eicosapentaenoic acid or docosahexaenoic acid, and one equivalent of a pharmaceutically acceptable acid, at a temperature between about 0°C and about 60° C.
  • a method for the manufacture of a compound of Formula IX or X comprising: reacting one equivalent of ethylenediamine, and one equivalent of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
  • a method for the manufacture of a compound of Formula XI or XII comprising: reacting one equivalent of ethylenediamine, and two equivalents of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
  • a method for the manufacture of a compound of Formula XIII or XIV comprising: reacting one equivalent of ethylenediamine, one equivalent of eicosapentaenoic acid or docosahexaenoic acid, and one equivalent of a pharmaceutically acceptable acid, at a temperature between about 0°C and about 60° C.
  • the reaction is performed in the absence of a solvent.
  • Yield 4.5g of ethane- 1 ,2-diaminium (5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l ,14,17-pentaenoate methanesulfonate as a tacky solid.
  • the material is triturated with ice cold CH 3 CN (3 x 20 ml) and dried under hi- vac with P 2 O5 at RT overnight.
  • Yield 3.4g of ethane- 1 ,2- diaminium5Z,8Z,HZ,14Z,17Z)-icosa-5,8,l l,14,17-pentaenoatemethane sulfonate as a tan solid.
  • Oral pharmacokinetic parameters of piperazine di-eicosapentaenoate were determined in Sprague-Dawley rats.
  • Piperazine di- eicosapentaenoate was administered by oral gavage as an aqueous solution in 0.5% carboxymethyl cellulose to 6 Sprague-Dawley rats, 3 males and 3 females. Rats were dosed at 40 mg/kg.
  • Blood samples were obtained from each rat by jugular vein catheter. Samples were collected at 0.25, 0.5, 1, 2, 4, 8, 12, and 24 hours post dose. Blood samples were centrifuged to separate red blood cells and the resulting plasma samples were analyzed for eicosapentaenoic acid. Calculated pharmacokinetic parameters shown below in Table 1 are mean values from 6 rats.
  • Oral pharmacokinetic parameters of ethylene diamine di-eicosapentaenoate were determined in Sprague-Dawley rats. Ethylene diamine di-eicosapentaenoate was administered by oral gavage as an aqueous solution in 0.5% carboxymethyl cellulose to 6 rats, 3 males and 3 females. Rats were dosed at 40 mg/kg. Blood samples were obtained from each rat by jugular vein catheter. Samples were collected at 0.25, 0.5, 1, 2, 4, 8, 12, and 24 hours post dose. Blood samples were centrifuged to separate red blood cells and the resulting plasma samples were analyzed for eicosapentaenoic acid. Calculated pharmacokinetic parameters shown below are mean values from 6 rats.
  • Treating patients with type 2 diabetes with the pharmaceutical composition according to the invention in addition to producing an acute improvement in the glucose metabolic situation, prevents a deterioration in the metabolic situation in the long term. This can be observed when patients are treated for a longer period, e.g. 3 months to 1 year or even 1 to 6 years, with the pharmaceutical composition according to the invention and are compared with patients who have been treated with other antidiabetic medicaments. There is evidence of therapeutic success compared with patients treated with other antidiabetic medicaments if no or only a slight increase in the fasting glucose and/or HbAlc value is observed.
  • the efficacy of a pharmaceutical composition according to the invention can be tested in clinical studies with varying run times (e.g. 12 weeks to 6 years) by determining the fasting glucose or non-fasting glucose (e.g. after a meal or a loading test with oGTT or a defined meal) or the HbAlc value.
  • Examples of this are a reduction in systolic and/or diastolic blood pressure, a lowering of the plasma triglycerides, a reduction in total or LDL cholesterol, an increase in HDL cholesterol or a reduction in weight, either compared with the starting value at the beginning of the study or in comparison with a group of patients treated with placebo or a different therapy.
  • type 2 diabetes or pre-diabetes patients with a pharmaceutical composition according to the invention prevents or reduces or reduces the risk of developing microvascular complications (e.g. diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic foot, diabetic ulcer) or macrovascular complications (e.g. myocardial infarct, acute coronary syndrome, unstable angina pectoris, stable angina pectoris, stroke, peripheral arterial occlusive disease, cardiomyopathy, heart failure, heart rhythm disorders, vascular restenosis).
  • microvascular complications e.g. diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic foot, diabetic ulcer
  • macrovascular complications e.g. myocardial infarct, acute coronary syndrome, unstable angina pectoris, stable angina pectoris, stroke, peripheral arterial occlusive disease, cardiomyopathy, heart failure, heart rhythm disorders, vascular restenosis.
  • Type 2 diabetes or patients with pre-diabetes are treated long-
  • the nerve conduction rate can be mea-isured using a calibrated tuning fork, for example.
  • the following parameters may be investigated before the start, during and at the end of the study: secretion of albumin, creatinine clearance, serum creatinine values, time taken for the serum creatinine values to double, time taken until dialysis becomes necessary.
  • a diabetic rat model that may be used for determination of conditions leading to a method for treatment and prevention of post-ischemic damage of the heart and heart tissue is the spontaneously diabetic bio-bred (BBAV) rats. These rats are a useful model of autoimmune human insulin-dependent diabetes.
  • This example describes an isolated perfused rat heart model used in development of the invention. Studies are performed using an isovolumic isolated rat heart preparation. Acutely diabetic male BBAV rats and non-diabetic age-matched (3-4 months old) control are pretreated with heparin (1000 u; IP), followed by sodium pentobarbital (65 mg/kg; IP). After deep anaesthesia is achieved as determined by the absence of a foot reflex, the hearts are rapidly excised and placed into iced saline. The arrested hearts are retrograde perfused in a non-recirculating model through the aorta within 2 min following their excision.
  • LVDP Left ventricular developed pressure
  • Perfusion pressure is monitored using high pressure tubing off the perfusion line. Hemodynamic measurements are recorded on a 4-channel Gould recorder.
  • the system has two parallel perfusion lines with separate oxygenators, pumps and bubble traps, but common temperature control allowing rapid change perfusion media.
  • the hearts are perfused using an accurate roller pump.
  • the perfusate consists of 118 mM NaCl, .47 mM KC1, 12 mM CaC12, 12 mM MgC12, 25 mM NaHC03, and the substrate 11 mM glucose.
  • the perfusion apparatus is tightly temperature- controlled, with heated baths being used for the perfusate and for the water jacketing around the perfusion tubing to maintain heart temperature at 37 ⁇ 0.5 °C under all conditions.
  • the oxygenated perfusate in the room temperature reservoir is passed through 25 ft. of thin- walled silicone tubing surrounded by distilled water at 37 °C saturated with 95% oxygen.
  • the perfusate then enters the water-jacketed (37 °C) tubing leading to the heart through a water jacketed bubble trap. This preparation provides excellent oxygenation that routinely has been stable for 3-4 hours.
  • Diabetic control (DC) diabetic treated (DZ) normal (C) control and normal treated (CZ) hearts are subjected to 20 min. of normoxic perfusion followed by 20 min. of zero-flow ischemia where the perfusate flow is completely shut off, followed by 60 min. of reperfusion.
  • Hearts are treated with 10 ⁇ of the compounds of the invention.
  • the compounds of the invention treated diabetic group (DZ), hearts are subjected to 10 min. of normoxic perfusion with normal Krebs-Henseleit buffer and 10 min.
  • This example describes a procedure used for study of low-flow ischemia in diabetic controls, diabetic treated, non-diabetic treated and non-diabetic control isolated hearts .
  • Diabetic control hearts DC are subjected to 20 min. of normoxic perfusion at a flow rate of 12.5 mL/min. followed by 30 minutes of low-flow ischemia where the perfusate flow is slowed down to 1.25 mL/min, that is about 10% of normal perfusion, followed by 30 min. of reperfusion at a normal flow rate (12.5 mL/min).
  • DZ or CZ groups treated with the compound of Formula I diabetic or non-diabetic groups
  • hearts are subjected to 10 min. of normoxic perfusion (flow rate 12.5 mL/min) with normal Krebs-Henseleit buffer and 10 min. of normoxic perfusion with Krebs-Henseleit buffer containing 10 ⁇ of a compound of
  • active ingredient denotes two compounds according to the invention, i.e., denotes a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof (first component of the active ingredient) and other antidiabetic agents.
  • the other antidiabetic agents may be statins, cholesterol absorption inhibitors, and CETP inhibitors or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically- acceptable salt of said prodrug (second component of the active ingredient).
  • the antidiabetic agents may be a DPP IV inhibitor, especially sitagliptin, sitagliptin phosphate, alogliptin, vildagliptin, saxagliptin, and linagliptin, sulfonylurea, SGLT-2 inhibitor, or other antidiabetic compounds according to this invention (second component of the active ingredient).
  • DPP IV inhibitor especially sitagliptin, sitagliptin phosphate, alogliptin, vildagliptin, saxagliptin, and linagliptin, sulfonylurea, SGLT-2 inhibitor, or other antidiabetic compounds according to this invention (second component of the active ingredient).
  • Additional suitable formulations may be prepared according to the procedures described in, for example in the application WO 2007/128724, and in the U.S. Patent Application, 2010/032011 the disclosure of which are incorporated herein in its entirety.
  • sulfonylureas, DPP IV inhibitors may be those formulations which are available on the market, or formulations described in the patent applications cited above in paragraph "background of the invention", or those described in the literature, for example as disclosed in current issues of "Rote Liste S” (Germany) or of “Physician's Desk Reference”.
  • (1) is triturated with (3). This trituration is added to the mixture of (2) and (4) with vigorous mixing. This powder mixture is packed into size 3 hard gelatin capsules in a capsule filling machine.
  • (1) is triturated with (3). This trituration is added to the mixture of (2) and (4) with vigorous mixing. This powder mixture is packed into size 3 hard gelatin capsules in a capsule filling machine

Abstract

Provided herein are diamine salts of eicosapentaenoic acid and docosahexaenoic acid, processes for the preparation of such compounds, pharmaceutical compositions comprising such compounds, and the uses of such compounds as agents for treating dyslipidemia, cardiovascular diseases such as cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke and obesity.

Description

DIAMINE AND MEGLUMINE SALT FORMS OF FATTY ACIDS
Related Applications
This application claims priority to U.S. Provisional Application No. 61/668,517, filed July 6, 2012, U.S. Provisional Application No. 61/670,384, filed July 11, 2012, U.S.
Provisional Application No. 61/668,582, filed July 6, 2012, U.S. Provisional Application No. 61/668,512, filed July 6, 2012, and U.S. Patent Application No. 13/841 ,667, filed March 15, 2013. The contents of all of these applications are incorporated herein by reference in their entireties.
Background of the Invention
Of the many health issues plaguing humans, obesity and overall poor cardiovascular health are among the most distressing, due to the fact that these diseases can lead to a number of complications. For example, obesity is often associated with hyperlipidemia, which is an elevation of lipids in the blood. These lipids include triglycerides, cholesterol, cholesterol esters, and phospholipids. Specifically, elevated levels of triglycerides in the blood is known as hypertriglyceridemia. Although a certain amount of triglycerides are necessary for good health, increased triglyceride levels are often associated with increased risk of heart disease. Overall, hyperlipidemia is associated with a number of disease states, including coronary artery disease, angina pectoris, carotid artery disease, strokes, cerebral arteriosclerosis, and xanthoma.
Accordingly, there remains a need for treating obesity, cardiovascular disease, and related indications.
Summary of the Invention
Provided herein are compounds useful in the treatment of obesity, cardiovascular disease, and related indications such as such as cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke.
In an embodiment, provided herein are compounds of the Formula A, B, C, D, E or F:
Figure imgf000003_0001
Figure imgf000003_0002
Figure imgf000003_0003
Figure imgf000003_0004
Figure imgf000003_0005
Formula F
wherein, for Formulas A and B, R+ represents a piperidine or diamine group, wherein the nitrogen of the piperidine and one of the nitrogens of the diamine are protonated;
for Formulas C and D, R++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated; and
for Formulas E and F, R++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated, and X" is an anion of a pharmaceutically acceptable acid compound. In another embodiment, the compounds provided herein are of the Formula I, II, III, IV, V, VI, VII and VIII:
Figure imgf000004_0001
Figure imgf000004_0002
Formula V
Figure imgf000004_0003
Figure imgf000004_0004
Figure imgf000005_0001
Formula VI I I wherein X" is as defined below.
The compounds of Formula A, B, C, D, E, F, I, II, III, IV, V, VI, VII and VIII are referred to herein as the "compounds of the invention."
In an embodiment, provided herein is a pharmaceutical composition comprising a compound of the invention, and a pharmaceutically acceptable carrier. In another embodiment, provided herein is a kit comprising a) a unit dosage comprising a compound of the invention, and b) instructions on how to use the kit; and at least one container for holding the unit dosage forms.
The compounds of the invention can be used to treat obesity, cardiovascular disease, and related indications, in a subject in need thereof. In one aspect, provided herein is a method of treating hyperlipidemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention. In another aspect, provided herein is a method of treating hypertriglyceridemia, comprising administering to a subject in need thereof an effective amount of a compound the invention. In another aspect, provided herein is a method of treating dyslipidemia, comprising administering to a subject in need thereof an effective amount of a compound of the invention. In still another aspect, provided herein is a method of treating cardiovascular disease, comprising administering to a subject in need thereof an effective amount of a compound of the invention. The cardiovascular disease can be cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, or stroke. In an embodiment, the arrhythmia is an atrial fibrillation. In another aspect, provided herein is a method of treating obesity, comprising administering to a subject in need thereof an effective amount of a compound of the invention. In one embodiment of the above methods, the subject is human.
In another aspect, provided herein is a method of treating prediabetes, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
In still another aspect, provided herein is a method of treating atherosclerosis, comprising administering to a subject in need thereof an effective amount of a compound of the invention. Also provided herein is a method of lowering triglycerides in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the invention.
In another embodiment, provided herein is a method of treating atrial fibrillation, or reducing the probability of an occurrence of an atrial fibrillation, comprising administering to a subject in need thereof an effective amount of a compound of the invention.
In another aspect, provided herein is a combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII and an antihyperlipidemic agent, to treat a metabolic disorder selected from the group consisting of type 2 diabetes (T2D), pre- diabetes, obesity, metabolic syndrome, hypertriglyceridemia and T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including atrial fibrillation, cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients. An additional aspect provided herein is combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII and an antihyperlipidemic agent, to treat obesity, cardiovascular disease, and related indications in a subject in need thereof.
In one aspect, provided herein is a method of treating hyperlipidemia, comprising administering to a subject in need thereof an effective amount of the combination therapy described above. In another aspect, provided herein is a method of treating
hypertriglyceridemia, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In another aspect, provided herein is a method of treating dyslipidemia, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In still another aspect, provided herein is a method of treating cardiovascular disease, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. The cardiovascular disease can be cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, or stroke. In an embodiment, the arrhythmia is an atrial fibrillation. In another aspect, provided herein is a method of treating obesity, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In another aspect, provided herein is a method of treating prediabetes, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In still another aspect, provided herein is a method of treating atherosclerosis, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. Also provided herein is a method of lowering triglycerides in a subject in need thereof, comprising administering to the subject an effective amount of the combination therapy of the invention. In another embodiment, provided herein is a method of treating atrial fibrillation, or reducing the probability of an occurrence of an atrial fibrillation, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In one embodiment of the above methods, the subject is human.
In yet another aspect, provided herein is a combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII and an antihyperglycemic agent, to treat a metabolic disorder selected from the group consisting of T2D, pre-diabetes, obesity, metabolic syndrome, hypertriglyceridemia and T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients.
Detailed Description of the Invention
Epidemiological and clinical evidence suggests that an increased intake of co-3 polyunsaturated fatty acids (PUFAs) protects against mortality from coronary artery diseases. PUFAs include eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). It is widely established that PUFAs protect against and can terminate ischemic ventricular arrhythmias (Billman et al. Circulation. 1999, 99, 2452-2457 and Kang et al. Am. J. Clin. Nutr. 2002, 71, 202S-207S). In particular, it is known that EPA is a promising treatment for prevention of major coronary events. PUFAs have multiple biological functions through lipid-dependent and lipid-independent mechanisms. EPA and mixtures of EPA and DHA have been shown to ameliorate triglycerides (TGs) lipid levels in patients with very high TGs. Also, EPA is shown to increase adiponectin secretion both in obese animals and obese human subjects (Itoh et al. Arteroscler. Thromb. Vase. Biol. 2007, 27, 1918-1925). Increased adiponectin levels are beneficial in regulating both lipid and glucose metabolism in animals as well as in humans.
In one aspect, provided herein are compounds of the Formula A, B, C, D, E or F:
Figure imgf000008_0001
Figure imgf000008_0002
Figure imgf000008_0003
Figure imgf000008_0004
Figure imgf000008_0005
Formula F
wherein, for Formulas A and B, R+ represents a piperidine or diamine group, wherein the nitrogen of the piperidine and one of the nitrogens of the diamine are protonated;
for Formulas C and D, R++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated; and
for Formulas E and F, R++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated, and X" is an anion of a pharmaceutically acceptable acid compound. As used herein, the term "diamine" refers to an organic compound composed of two amino groups. The diamine can be any diamine composed of primary amines, secondary amines, or a combination of a primary amine(s) and a secondary amine(s). The diamine can be a cyclic diamine, such as piperazine, diaminocyclohexane, diphenylethylenediamine, 1,8- diaminonaphthalene, 4,4'-diaminobiphenyl, N,N'-di-2-butyl-l ,4-phenylenediamine, dimethyl- 4-phenylenediamine, p-xylylenediamine (PXD), m-xylylenediamine (MXD), o- xylylenediamine (OXD), p-phenylenediamine (PPD), 2,5-diaminotoluene, m- phenylenediamine (MPD), o-phenylenediamine (OPD). The diamine can also be an acyclic amine, such as ethylenediamine (1,2-diaminoethane), ethambutol,
tetramethylethylenediamine (TMEDA or TEMED), 1,3-diaminopropane (propane-1,3- diamine), putrescine (butane- 1,4-diamine), cadaverine (pentane- 1 ,5 -diamine),
hexamethylenediamine (hexane-l,6-diamine), or 1,2-diaminopropane.
In another aspect, provided herein are piperazine salts of EPA and DHA, as well as meglumine salts of EPA and DHA.
In one asp
Figure imgf000009_0001
Formula I
In another aspect, provided herein is a compound of Formula II:
Figure imgf000009_0002
Formula I I
The compound of Formula I and II are, respectively, the mono-salt of EPA with piperazine and the mono- salt of DHA with piperazine.
In another aspect, provided herein are compounds of the Formula III and Formula IV:
Figure imgf000009_0003
Formula I I I
Figure imgf000010_0001
Formula IV
The compounds of Formula III and IV are, respectively, the di-salt of EPA with piperazine and the di-salt of DHA with piperazine.
In still another aspect, provided herein are compounds of the Formula V and VI:
Figure imgf000010_0002
Formula V
Figure imgf000010_0003
Formula VI
wherein X" is a pharmaceutically acceptable counter anion.
The pharmaceutically acceptable counter anion can derived from acid compounds listed in Table 1 , pp 406 - 407, Handbook of Pharmaceutical Salts, P. Heinrich Stahl Camille G. Wermuth (Eds.)- In an embodiment, the pharmaceutically acceptable counter anion is selected from mineral acids, such as hydrochloric acid, hydrobromic acid, and phosphoric acid. In another embodiment, the pharmaceutically acceptable counter anion is selected from carboxylic acids, poly-carboxylic acids, and poly-hydroxy carboxylic acids, such as acetic acid, propionic acid, succinic acid, maleic acid, malic acid, tartaric acid, lactic acid, citric acid, benzoic acid, and hippuric acid (benzoylaminoethanoic acid). In another embodiment, the pharmaceutically acceptable counter anion is selected from sulfonic acids and hydroxyl- sulfonic acids, including, but not limited to, methanesulfonic acid, isethionic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, and benzenesulfonic acid. In another embodiment, the pharmaceutically acceptable counter anion is selected from amino acids, including, but not limited to, glycine, alanine, lysine, arginine, aspartic acid, or glutamic acid. In another embodiment, X" is an omega-3 polyunsaturated acid, such as eicosapentaenoic acid or docosahexaenoic acid.
In an embodiment, provided herein is a compound of Formula V wherein X" is mandelic acid. In another embodiment, provided herein is a compound of V wherein X" is hippuric acid. In an embodiment, provided herein are the following compounds: the hydrochloride salt of Formula V, the hydrobromide salt of Formula V, the phosphate salt of Formula V, and the sulfate salt of Formula V.
In another embodiment, provided herein are the following compounds: the hydrochloride salt of Formula VI, the hydrobromide salt of Formula VI, the phosphate salt of Formula VI, and the sulfate salt of Formula VI.
The present invention also relates to compounds of the Formula V and Formula VI wherein X" is a pharmaceutically acceptable counter anion derived from naturally occurring amino acids. Examples of the amino acids include, but are not limited to, glycine, alanine, lysine, and glutamic acid.
In another aspect, provided herein are compounds of the Formula VII and Formula VIII. These compounds, respectively, are EPA and DHA salts with meglumine:
Figure imgf000011_0001
Formula VII
Figure imgf000011_0002
Formula VIII
In another aspect, provided herein are compounds of Formulas IX - XIV
Figure imgf000011_0003
Formula IX
Figure imgf000011_0004
Formula X
Figure imgf000012_0001
Figure imgf000012_0002
Figure imgf000012_0003
Figure imgf000012_0004
Formula XIV
wherein X" is a pharmaceutically acceptable counter anion as described above.
In an embodiment, provided herein is a compound of Formula XIII, wherein X" is methanesulfonic acid.
The compounds of the invention, e.g., compounds of Formulas I-XIV also include isomers and enantiomers wherever it is applicable.
It is well known in the art that highly water soluble medicinal preparations, when administered orally, result in efficient absorption of such preparations from the
gastrointestinal tract into systemic circulation. Another hallmark of such preparations is the rate at which they are absorbed into systemic circulation resulting in high concentration of the active agent or agents in the blood. Moreover, for delivery of xenobiotics via the intravenous route, they must be presented as a clear solution. PUFAs and esters of PUFAs are practically insoluble in water. In fact, they form soap-like emulsions when mixed with water. Therefore, the potential to derive optimum therapeutic benefits of PUFAs should be markedly facilitated by delivery of water soluble PUFAs. The compounds of the present invention are more water soluble to achieve high oral absorption and to enable the preparation of intravenous dosage forms.
Methods of Treatment
In one aspect, provided herein is a method of treating obesity, cardiovascular disease, as well as related disorders, comprising administering to a subject in need thereof a compound of the invention, e.g., compounds of Formulas I-XIV. In an embodiment, an effective amount of the compound is administered for treatment.
Hyperlipidemia is a condition generally characterized by an abnormal increase in serum lipids in the bloodstream and is an important risk factor in developing atherosclerosis and heart disease. Hyperlipidemia is usually classified as primary or secondary
hyperlipidemia. Primary hyperlipidemia is generally caused by genetic defects, while secondary hyperlipidemia is generally caused by other factors, such as various disease states, drugs, and dietary factors. Alternatively, hyperlipidemia can result from both a combination of primary and secondary causes of hyperlipidemia. Elevated cholesterol levels are associated with a number of disease states, including coronary artery disease, angina pectoris, carotid artery disease, strokes, cerebral arteriosclerosis, and xanthoma.
An example of hyperlipidemia is hypertriglyceridemia, which is defined as an elevated level of triglycerides. Elevated triglycerides have been associated with
atherosclerosis, even in the absence of high cholesterol levels. High triglycerides can also lead to pancreatitis in excessive concentrations.
As used herein, "obesity" refers to having a body weight more than about 30% greater than ideal body weight, as determined by a medical professional, and/or having a body mass index greater than about 27 as determined by a medical professional.
Accordingly, in one aspect, provided herein is a method of treating hyperlipidemia
{e.g. , hypertriglycreridemia) in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the invention, e.g. , compounds of Formulas I- XIV.
In another aspect, provided herein is a method of treating cardiovascular diseases such as cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the invention, e.g., compounds of Formulas I-XIV. In an embodiment, the arrhythmia is an atrial fibrillation. Atrial fibrillation is a type of cardiac arrhythmia where there is disorganized electrical conduction in the atria causing rapid uncoordinated contractions. These contractions can result in ineffective pumping of blood into the ventricle and a lack of synchrony. During atrial fibrillation, the atrioventricular node receives electrical impulses from numerous locations throughout the atria instead of only from the sinus node. This overwhelms the atrioventricular node into producing an irregular and rapid heartbeat. As a result, blood pools in the atria that increases a risk for blood clot formation. The major risk factors for atrial fibrillation include age, coronary artery disease, rheumatic heart disease, hypertension, diabetes, and thyrotoxicosis.
In still another aspect, provided herein is a method of treating obesity in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the invention, e.g., compounds of Formulas I-XIV.
In still another aspect, provided herein is a method of treating atherosclerosis, comprising administering to a subject in need thereof an effective amount of a compound of the invention. Atherosclerosis refers to the buildup of fats and cholesterol in and on artery walls (plaques), which can restrict blood flow. These plaques can also burst, triggering a blood clot. Although atherosclerosis is often considered a heart problem, it can affect arteries anywhere in the body. An animal model of atherosclerosis research is described in
Laboratory Animals (2004) 38, 246-256.
The term "treat," "treated," "treating" or "treatment" includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated. In certain embodiments, the treatment comprises the induction of a disease, followed by the activation of the compound of the invention, which would in turn diminish or alleviate at least one symptom associated or caused by the protein kinase- associated disorder being treated. For example, treatment can be diminishment of one or several symptoms of a disorder or complete eradication of a disorder.
The term "subject" is intended to include organisms, e.g., prokaryotes and eukaryotes, which are capable of suffering from or afflicted with a disease, disorder or condition. Examples of subjects include mammals, e.g. , humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals. In certain embodiments, the subject is a human, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from obesity; cardiovascular diseases such as cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke; or hyperlipidemia, including hypertriglycreridemia, The language "effective amount," "pharmaceutically effective amount" or
"pharmaceutically acceptable amount" of the compound is that amount necessary or sufficient to treat or prevent a disorder, e.g. , prevent the various morphological and somatic symptoms of a disease or condition described herein. In an example, an effective amount of a compound of the invention is the amount sufficient to treat obesity, cardiovascular disorders, or a related disorder, in a subject. The effective amount can vary depending on such factors as the size and weight of the subject, the type of illness, or the particular compound of the invention.
The expression "pharmaceutically acceptable salts" includes both pharmaceutically acceptable acid addition salts and pharmaceutically acceptable cationic salts, where appropriate. The expression "pharmaceutically-acceptable cationic salts" is intended to define but is not limited to such salts as the alkali metal salts, {e.g. , sodium and potassium), alkaline earth metal salts {e.g., calcium and magnesium), aluminum salts, ammonium salts, and salts with organic amines such as benzathine (Ν,Ν'-dibenzylethylenediamine), choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), benethamine (N-benzyl phenethylamine) , diethylamine, piperazine, tromethamine (2-amino-2-hydroxymethyl-l ,3- propanediol) and procaine. The expression "pharmaceutically-acceptable acid addition salts" is intended to define but is not limited to such salts as those with pharmaceutically acceptable mineral or organic acids classically used in pharmacy. Appropriate acids are, for example, inorganic acids, such as hydrohalic acids, e. g. hydrochloric, hydrobromic or the like, or sulfuric acid, nitric acid, or phosphoric acid; or suitable organic acids, for example suitable aliphatic acids, like aliphatic mono or dicarboxylic acids, hydro xyalkanoic or hydroxyalkanedioic acids, e.g. acetic, propanoic, hydroxyacetic, 2-hydroxypropanoic, 2- oxopropanoic, ethanedioic, propanedioic, butanedioic, (Z)-2-butenedioic, (E)-2- butenedioic, 2-hydroxybutanedioic, 2,3-dihydroxybutanedioic, or 2 -hydro y- 1 ,2,3 - propanetricarboxylic acid; phenyl substituted alkanoic acids; or suitable aromatic acids, like 2-hydroxybenzoic, or 4-amino-2-hydroxybenzoic acid; or suitable sulfonic acids, like alkanesulfonic acids, e.g. methanesulfonic, or ethanesulfonic acid, or aromatic sulfonic acids, e.g. benzenesulfonic, or 4-methylbenzenesulfonic acid; or cyclohexanesulfamic acid. In certain embodiments of the disclosure, acids are e.g. hydrobromic acid, sulphuric acid, phosphoric acid, acetic, benzoic, fumaric, maleic, citric, tartaric, gentisic, dobesilic, methanesulfonic, ethanesulfonic, laurylsulfonic, benzenesulfonic, and para- toluenesulfonic acids. In certain embodiments, the pharmaceutically acceptable salt is selected from the group consisting of an L-arginine, benenthamine, benzathine, betaine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)ethanol, ethanolamine, ethylenediamine, N-methylglucamine, hydrabamine, 1 H-imidazole, lithium hydroxide, L-lysine, magnesium hydroxide, 4-(2-hydroxyethyl)morpholine, piperazine, potassium hydroxide, l -(2-hydroxyethyl)pyrrolidine, sodium hydroxide, triethanolamine, tromethamine, zinc hydroxide, sodium, calcium, potassium, magnesium, and zinc.
The triglyceride lowering efficacy of the compounds of the present invention can be determined in animal models according to the procedure described by Sidika et al in Journal of Lipid Research, 1992, 33, 1-7.
Efficacy of the compounds of the present invention in increasing adiponectin secretion in rodent models of obesity and human obese subjects can be determined according the procedures described by Itoh et al. in Arteroscler. Thromb. Vase. Biol. 2007, 27, 1918- 1925.
Combination Therapy
The compounds of the present invention, e.g. piperazine and meglumine salts of polyunsaturated fatty acids, (including eicosapentaenoic acid and docosahexaenoic acid, or a mixture thereof) are well-suited to use in combination therapy.
The term "combination therapy" refers to the administration of two or more therapeutic agents to treat a therapeutic condition or 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 containers {e.g., capsules) for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner, either at approximately the same time or at different times. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
A "mixture of compounds of Formula I, II, III, IV, V, VI, VII or VIII" refers to a mixture of two or more of these compounds present in a and b %, and optionally c, d, e, f, g and/or h % , wherein a and b, and optionally c, d, e, f, g and/or h are not zero, but the sum of a and b, and optionally c, d, e, f, g and/or h, is 100 %. For example, when a compound is made up of a mixture of a compound of Formula I and a compound of Formula III, a compound of Formula I is present at 50% and a compound of Formula III is present at 50%.
In one embodiment, provided herein is a combination therapy comprising an effective amount of a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a combination thereof, and an antihyperlipidemic or an antihyperglycemic agent. An "effective amount" of a combination of agents is an amount sufficient to provide an observable improvement over the baseline clinically observable signs and symptoms of the disorders treated with the combination.
In one embodiment, the combination of a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a combination thereof, and an antihyperlipidemic or antihyperglycemic agent described herein displays a synergistic effect. The term "synergistic effect" as used herein, refers to action of two agents producing an effect, for example, slowing the symptomatic progression of diabetes or symptoms thereof, which is greater than the simple addition of the effects of each drug administered by themselves. A synergistic effect can be calculated, for example, using suitable methods such as the Sigmoid-Emax equation (Holford, N. H. G. and Scheiner, L. B., Clin. Pharmacokinet. 6: 429-453 (1981)) incorporated by reference, herein, in its entirety, the equation of Loewe additivity (Loewe, S. and Muischnek, H., Arch. Exp. Pathol Pharmacol. 114: 313-326 (1926)) incorporated by reference, herein, in its entirety, and the median-effect equation (Chou, T. C. and Talalay, P., Adv. Enzyme Regul. 22: 27-55 (1984)) incorporated by reference, herein, in its entirety. Each equation referred to above can be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination. The corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively.
Combination Therapy with Antihyperlipidemic Agents
In another aspect, the combination therapy comprises the compounds of the present invention and an antihyperlipidemic agent, to treat a metabolic disorder selected from the group consisting of T2D, pre-diabetes, obesity, metabolic syndrome, hypertriglyceridemia and T2D complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including atrial fibrillation, cardiac arrhythmia, myocardial infarction, stroke, and cardiomyopathy in mammals, e.g., diabetic patients.
An additional aspect provided herein is combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII and an antihyperlipidemic agent, to treat obesity, cardiovascular disease, and related indications in a subject in need thereof.
An additional aspect provided herein is combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII or XIV and an antihyperlipidemic agent, to treat obesity, cardiovascular disease, and related indications in a subject in need thereof.
In one aspect, provided herein is a method of treating hyperlipidemia, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In another aspect, provided herein is a method of treating
hypertriglyceridemia, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In another aspect, provided herein is a method of treating dyslipidemia, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In still another aspect, provided herein is a method of treating cardiovascular disease, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. The cardiovascular disease can be cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, or stroke. In an embodiment, the arrhythmia is an atrial fibrillation. In another aspect, provided herein is a method of treating obesity, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In another aspect, provided herein is a method of treating prediabetes, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. In still another aspect, provided herein is a method of treating atherosclerosis, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention. Also provided herein is a method of lowering triglycerides in a subject in need thereof, comprising administering to the subject an effective amount of the combination therapy of the invention. In another embodiment, provided herein is a method of treating atrial fibrillation, or reducing the probability of an occurrence of an atrial fibrillation, comprising administering to a subject in need thereof an effective amount of the combination therapy of the invention.
In an embodiment, provided herein is a method of lowering the cholesterol level and/or the triglyceride level in a mammal comprising administering to the subject an effective amount of the combination therapy of the invention.
In one embodiment of the above methods, the subject is human.
In one aspect, the invention relates to a combination therapy comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII or a compound of the structural Formula I, II, III, IV, V or VI and an antihyperlipidemic agent, or a combination of two or more compounds of the structural Formulas I, II, III, IV, V, VI, VII or VIII or compounds of the structural Formulas I, II, III, IV, V or VI and an antihyperlipidemic agent and a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically- acceptable salt of said prodrug. In another aspect, provided herein is a pharmaceutical composition comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these, an antihyperlipidemic agent and a pharmaceutically-acceptable carrier. In one embodiment, when the pharmaceutical composition comprises two or more compounds of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these, the two or more compounds are present in x, y, z, ...% etc. with the proviso that x, y, z, ...% are not zero, but the sum x, y, z, ...% is 100%.
In one embodiment of the pharmaceutical composition, the antihyperlipidemic agent is about 0.1-1 % by weight of the pharmaceutical composition. In another embodiment of the pharmaceutical composition, the compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof is present in unit dosage strength of 250, 500, 750, 1000 or 1250 mg, and the said antihyperlipidemic agent is present in a unit dosage strength of 1, 2.5, 5, 10, 20, 30, 40, or 50 mg. In another embodiment, the said antihyperlipidemic agent is present in a unit dosage strength of 5-100 mg.
The components of the combination therapy (a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII or a compound of the structural Formula I, II, III, IV, V or VI and an antihyperlipidemic agent, or a combination of two or more compounds of the structural Formulas I, II, III, IV, V, VI, VII or VIII or compounds of the structural Formulas I, II, III, IV, V or VI and an antihyperlipidemic agent) can be administered in a variety of ways. In one embodiment, the components are in separate formulations or unit dosage forms. In another embodiment, the components are administered with a pharmaceutically acceptable carrier. The components can be administered separately, at substantially the same time, or administered at different times. When administered separately, they can be administered in any order.
In one embodiment, the present invention is directed to pharmaceutical compositions comprising compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these and an antihyperlipidemic agent, or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug; and a pharmaceutically- acceptable carrier, vehicle or diluent.
Antihyperlipidemic agents that may be used in accordance with the invention may include, for example, statins, which are HMG CoA enzyme inhibitors, cholesterol absorption inhibitors, and cholesterol esterase transfer protein (CETP) inhibitors and pharmaceutically- acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrug, and others.
In one embodiment of the present invention, the antihyperlipidemic agent is a statin, cholesterol absorption inhibitor, and CETP inhibitor or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug. In certain
embodiments the pharmaceutically acceptable salt is selected from the group consisting of a propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne- 1 ,4- dioate, hexyne-l ,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephathal ate, sulfonate, xylenesulfonate, phenyl acetate, phenylpropionate, phenylbutyrate, citrate, lactate, p- hydroxybutyrate, glycolate, tartrate, methanesulfonate, propanesulfonates, naphthalene- 1 -sulfonate, naphthalene-2-sulfonate, mandelate, hippurate, gluconate, or lactobionate.
Preferred agents among statins are atorvastatin, risuvostatin, simvastatin, or pravastatin, pharmaceutically-acceptable salts or prodrugs thereof, and pharmaceutically- acceptable salts of said prodrugs. A preferred agent among cholesterol absorption inhibitors is ezetimibe also known as Zetia. A preferred agent among CETP inhibitors is anacetrapib.
In certain embodiments, CETP inhibitors include, but are not limited to anacetrapib or a hydrate, and solvate thereof.
In certain embodiments, the pravastatin is present in the amount ranging from 5 mg to
100 mg.
In certain embodiments, the ezetimibe is present 5 mg to 50 mg.
In another aspect, the disclosure provides for kits comprising a first unit dosage form comprising compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these; a second unit dosage form comprising an antihyperlipidemic agent or a hydrate, and solvate thereof; and a container.
The antihyperlipidemic agents that may be used in accordance with the invention are members of different classes of antihyperlipidemic agents (e.g. , HMG-CoA reductase inhibitors (statins), CETP inhibitors, and cholesterol absorption inhibitors and others), pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
The term "HMG-CoA reductase inhibitor" as used herein refers to a compound that competitively blocks the enzyme 3-hydroxy-3-methyl-glutaryl-co-enzyme A (HMG-CoA) reductase. By competitively blocking this enzyme, the HMG-CoA reductase inhibitors interfere with cholesterol formation (enzyme catalyzes the conversion ofHMG-CoA to mevalonate). As a result, they decrease total cholesterol, low-density lipoprotein cholesterol (LDL-C), apolipoprotein B (a membrane transport complex for LDL-C), very low-density lipoprotein (VLDL), and plasma triglycerides. For a review on HMG-CoA inhibitors see, for example, Drug Discovery Today: Therapeutic Strategies, 1 :189 (2004) and references cited therein.
The specific HMG-CoA reductase inhibitors which may be used in accordance with the disclosure include, but are not limited to: atorvastatin, which may be prepared as disclosed in U.S. Patent No. 7,030,151 ; pravastatin and related compounds which may be prepared as disclosed in U.S. Patent Nos. 4,346,227 and 4,448,979; rosuvastatin, which may be prepared as disclosed in U.S. Patent No.6, 858,618; simvastatin and related compounds which may be prepared as disclosed U.S. Patent Nos. 4,448,784 and 4,450,171. HMG-CoA reductase inhibitors also include atorvastatin, simvastatin, pravastatin, lovastatin, fluvastatin, rosuvastatin, cerivastatin, mevastatin, rivastatin, pravastatin, nisvastatin, itavastatin, velostatin and fluindo statin.
The term "CETP inhibitor" as used herein refers to a compound which catalyses the transfer of cholesteryl ester from HDL to apolipoprotein B containing lipoproteins in exchange for triglyceride and thereby plays a major role in lipoprotein metabolism. For a review on CETP inhibitors see, for example, Curr. Opin. Pharmacal. 6:162 (2006) and references cited therein.
CETP inhibitors which may be used in accordance with the disclosure are not limited by any structure or group of CETP inhibitors. CETP inhibitors which may be used in accordance with the disclosure include, but are not limited to: anacetrapib, which may be prepared as disclosed in WO 2007005572. The disclosure thereof is incorporated herein by reference.
The term "cholesterol absorption inhibitors" as used herein refers to a compound that inhibits the absorption ofbiliary and dietary cholesterol from the small intestine without affecting the absorption of fat-soluble vitamins, triglycerides, or bile acids. For a review on cholesterol absorption inhibitors see, for example, Nutr. Metab. Cardiovasc. Dis., 13:42 (2004) and references cited therein.
Cholesterol absorption inhibitors which may be used in accordance with the disclosure include, but are not limited to ezetimibe (Zetia), which may be prepared as disclosed in U.S. Patent Nos. 5,767,115 and 5, 846,966. The disclosures thereof are incorporated herein by reference. In the practice of the compositions and methods of the disclosure, any HMG Co-A reductase inhibitors (or) or in a pharmaceutically acceptable combination with any flushing inhibiting agent may be employed.
In one aspect, the disclosure provides for pharmaceutical compositions comprising a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof; and an antihyperlipidemic agent or a pharmaceutically acceptable salt, hydrate, and solvate thereof.
In one aspect, the disclosure provides for unit dose formulations comprising a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof, and an antihyperlipidemic agent or a pharmaceutically acceptable salt, hydrate, and solvate.
Accordingly, in one embodiment, provided herein are combination therapies comprising, at least, the following combination of agents:
1) a compound of Formula I and atorvastatin; a compound of Formula I and simvastatin; a compound of Formula I and pravastatin; a compound of Formula I and rosuvastatin; and a compound of Formula I and ezitimibe; a compound of Formula I and anacetrapib; a compound of Formula I and atorvastatin calcium;
2) a compound of Formula II and atorvastatin; a compound of Formula II and simvastatin; a compound of Formula II and pravastatin; a compound of Formula II and rosuvastatin; and a compound of Formula II and ezitimibe; a compound of Formula II and anacetrapib; a compound of Formula II and atorvastatin calcium;
3) a compound of Formula III and atorvastatin; a compound of Formula III and simvastatin; a compound of Formula III and pravastatin; a compound of Formula III and rosuvastatin; and a compound of Formula III and ezitimibe; a compound of Formula III and anacetrapib; a compound of Formula III and atorvastatin calcium;
4) a compound of Formula IV and atorvastatin; a compound of Formula IV and simvastatin; a compound of Formula IV and pravastatin; a compound of Formula IV and rosuvastatin; and a compound of Formula IV and ezitimibe; a compound of Formula IV and anacetrapib; a compound of Formula IV and atorvastatin calcium;
5) a compound of Formula V and atorvastatin; a compound of Formula V and simvastatin; a compound of Formula V and pravastatin; a compound of Formula V and rosuvastatin; and a compound of Formula V and ezitimibe; a compound of Formula V and anacetrapib; a compound of Formula V and atorvastatin calcium;
6) a compound of Formula VI and atorvastatin; a compound of Formula VI and simvastatin; a compound of Formula VI and pravastatin; a compound of Formula VI and rosuvastatin; and a compound of Formula VI and ezitimibe; a compound of Formula VI and anacetrapib; a compound of Formula VI and atorvastatin calcium;
7) a compound of Formula VII and atorvastatin; a compound of Formula VII and simvastatin; a compound of Formula VII and pravastatin; a compound of Formula VII and rosuvastatin; and a compound of Formula VII and ezitimibe; a compound of Formula VII and anacetrapib; a compound of Formula VII and atorvastatin calcium;
8) a compound of Formula VIII and atorvastatin; a compound of Formula VIII and simvastatin; a compound of Formula VIII and pravastatin; a compound of Formula VIII and rosuvastatin; and a compound of Formula VIII and ezitimibe; a compound of Formula VIII and anacetrapib; a compound of Formula VIII and atorvastatin calcium;
In an additional embodiment, the combination therapies of paragraphs 1 , 2, 3, 4, 5, 6, 7 and 8, above, can be further combined with compounds of Formula I, II, III, IV, V, VI, VII and/or VIII.
Combination Therapy with Antihyp erg lycemic Agents:
In another aspect, provided herein is a combination therapy comprising the compounds of the present invention and an antihyperglycemic agent, or a pharmaceutically- acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug. In one embodiment, the present invention provides a method of treating a metabolic disorder selected from the group consisting of T2D, pre-diabetes, obesity, metabolic syndrome, hypertriglyceridemia and diabetes complications such as neuropathy, nephropathy, retinopathy, cataracts and cardiovascular complications, including cardiac arrhythmia, myocardial infarction, and cardiomyopathy in mammals, e.g. , diabetic patients, comprising administering to a subject in need thereof the combination therapy described herein
The components of this combination therapy (a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII or a compound of the structural Formula I, II, III, IV, V or VI and an antihyperglycemic agent, or a combination of two or more compounds of the structural Formulas I, II, III, IV, V, VI, VII or VIII or compounds of the structural Formulas I, II, III, IV, V or VI and an antihyperglycemic agent) can be administered in a variety of ways. In one embodiment, the components are in separate formulations or unit dosage forms. In another embodiment, the components are administered with a pharmaceutically acceptable carrier. The components can be administered separately, at substantially the same time, or administered at different times. When administered separately, they can be administered in any order. In one embodiment, the present invention is directed to pharmaceutical compositions comprising a compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture of these and an antihyperglycemic agent, or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug; and a
pharmaceutically-acceptable carrier, vehicle or diluent.
The components of this combination therapy (a compound of the structural Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII or XIV and an antihyperglycemic agent, or a combination of two or more compounds of the structural Formulas I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII or XIV and an antihyperglycemic agent) can be administered in a variety of ways. In one embodiment, the components are in separate formulations or unit dosage forms. In another embodiment, the components are administered with a
pharmaceutically acceptable carrier. The components can be administered separately, at substantially the same time, or administered at different times. When administered separately, they can be administered in any order.
In one embodiment, the present invention is directed to pharmaceutical compositions comprising a compound of the structural Formula I, II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII or XIV, or a mixture of these and an antihyperglycemic agent, or a
pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug; and a pharmaceutically-acceptable carrier, vehicle or diluent.
In one embodiment of the pharmaceutical composition, the antihyperglycemic agent is about 1-20% by weight of the pharmaceutical composition. In another embodiment of the pharmaceutical composition, said compound of the structural Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof is present in unit dosage strength of 250, 500, 750, 1000 or 1250 mg, and the said antihyperglycemic agent is present in a unit dosage strength of 1, 2.5, 5, 10, 20, 25, 50, 100, 150, or 200 mg. In another embodiment, the antihyperglycemic agent is present in a unit dosage strength of 5-100mg.
Antihyperglycemic agents that may be used in accordance with the invention may include, for example, sulfonylureas, meglitinides, thiazolidinediones, alpha-glucosidase inhibitors, DPP IV inhibitors, and SGLT-2 inhibitors and pharmaceutically-acceptable salts and prodrug thereof, and pharmaceutically-acceptable salts of said prodrug, and others.
In one embodiment of the present invention, the antihyperglycemic agent is a sulfonylurea, meglitinide, thiazolidinedione, alpha-glucosidase inhibitor, DPP IV inhibitor, and SGLT-2 inhibitors or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically-acceptable salt of said prodrug. In certain embodiments, the pharmaceutically acceptable salt is selected from the group consisting of a propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne- 1 ,4-dioate, hexyne-1 ,6- dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephathal ate, sulfonate, xylenesulfonate, phenyl acetate, phenylpropionate, phenylbutyrate, citrate, lactate, p- hydroxybutyrate, glycolate, tartrate, methanesulfonate, propanesulfonates, naphthalene- 1 -sulfonate, naphthalene-2-sulfonate, mandelate, hippurate, gluconate, or lactobionate.
Preferred agents among thiazolidinediones are pioglitazone, pharmaceutically- acceptable salts or a prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
Preferred alpha- glucosidase inhibitors include, but are not limited to, acarbose, vaglibose, and miglitol, pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
Preferred DPP-IV inhibitors include, but are not limited to, sitagliptin, linagliptin, vildagliptin, saxagliptin, alogliptin, denagliptin, carmegliptin, melogliptin and dutogliptin. and pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
Preferred SGLT-2 inhibitors include, but are not limited to, dapagliflozin
canagliflozin, atigliflozin, remogliflozin and sergliflozin, and pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically-acceptable salts of said prodrugs.
The antihyperglycemic agents that may be used in accordance with the invention are members of different classes of antihyperglycemic agents (e.g., sulfonylureas, meglitinides, thiazolidinediones, alpha-glucosidase inhibitors, DPP-IV inhibitors, SGLT-2 inhibitors and others), pharmaceutically-acceptable salts and prodrugs thereof, and pharmaceutically- acceptable salts of said prodrugs.
The term "sulfonylureas" refers to a class of compounds that stimulate insulin release by binding to the sulfonylurea receptor, a subunit of the ICATP channel complex. This binding leads to closure of the channel, resulting in voltage change in the beta-cell membrane and, in turn, influx of Ca2+ ions causing exocytosis of insulin granules. For a discussion on sulfonylureas see, for example, Metabolism, 55, 20 (2006) incorporated by reference, herein, in its entirety and references cited therein, and Lancet, 358, 1709 (2001) incorporated by reference, herein, in its entirety and references cited therein.
The term "thiazolidinediones" refers to a class of compounds that are selective agonists for the peroxisome proliferator-activated receptor gamma (PPARy), a member of family of nuclear hormone receptors that function as ligand-activated transcription factors. For a review on thiazolidinediones see, for example, Trends Endocrin. Met., 10, 9 (1999) and references cited therein.
The term "alpha-glucosidase inhibitors" refers to a class of compounds having the ability to competitively inhibit brush border enzyme alpha-glucosidase in the GI tract, which has the ability to cleave complex carbohydrates into sugars. For a review on alpha- glucosidase inhibitors see, for example, Diabetes Res. Clin. Pr., 40, S51 (1998) and references cited therein.
The term "DPP IV inhibitors" refers to a class of compounds that have the ability to selectively inactivate the enzyme DPP-IV, and those which have the ability to rapidly inactivate incretin hormones {e.g. , glucagon-like peptide-1 (GLP-1) and insulinotropic polypeptide (GIP)), that are released by the intestine throughout the day, and whose levels are increased after a meal. For a review on DPP-IV inhibitors see, for example, Expert Opin. Inv. Drug, 12, 87 (2003) and references cited therein. Specifically, the DPP-IV inhibitor sitagliptin can be prepared according to procedure described by Kim et al. in Journal of Medicinal Chemistry, 48, 141-151 , (2005) and in Journal of Medicinal Chemistry, 51, 589- 602, (2008).
The term "SGLT-2 inhibitors" refers to a class of compounds that have the ability to selectively inhibit renal sodium-glucose co -transporter 2 and prevent renal glucose reabsorption from the glomerular filtrate and provide an insulin-independent means of controlling hyperglycemia. For a perspective on SGLT-2 inhibitors see, for example, Journal of Medicinal Chemistry, 52, 1785 1794, (2009) and references cited therein.
In the practice of the compositions and methods of the invention, any sulfonylurea, meglitinide, thiazolidinedione, alpha-glucosidase inhibitor, DPP-IV inhibitor, or a SGLT-2 inhibitor or a pharmaceutically- acceptable salt or a prodrug thereof, or a pharmaceutically- acceptable salt of said prodrug, or any combination thereof, may be employed.
Sulfonylureas that may be used in accordance with the invention include, but are not limited to, acetohexamide, which may be prepared as described in U.S. Patent No. 3,013 ,072; 1- Butyl-3-methanilyl urea, which may be prepared as described in U.S. Patent No.
3,183,260; carbutamide, which may be prepared as described in U.S. Patent No. 4,324,796; chlorpropamide, which may be prepared as described in U.S. Patent No. 4,381 ,304;
glibornuride, which may be prepared as described in U.S. Patent No. 4, 153,710; gliclazide, which may be prepared as described in U.S. Patent No. 6,733,782; glipizide, which may be prepared and its use as oral administration as described in U.S. Patent No. 5,545,413 ; gliquidone, has been described and its use as described in U.S. Patent No. 4,708,868;
glyburide or glibenclamide, which may be prepared and its use as described in U.S. Patent No. 6,830,760; glybuthiazole, which may be prepared as described in U.S. Patent
No.7, 144,900; glybuzole, which may be prepared and its use as described in U.S. Patent No. 7,084,123; glyhexamide, which may be described and its use as described in U.S. Patent No.5859037; glimepiride, which may be prepared and its use as described in U.S. Patent No.4,379,785; glymidine, which may be prepared and its use as described in U.S. Patent No. 4,007,201 ; tolazamide, which may be prepared as described in U.S. Patent No. 3,583,979; tolbutamide, which may be prepared as described in U.S. Patent No. 4,639,436. These patents are incorporated herein by reference.
The term "pioglitazone" as employed herein refers to pioglitazone, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salt thereof such as the hydrochloride salt.
DPP-IV inhibitors that may be used in accordance with the invention include, but are not limited to, linagliptin, sitagliptin, vildagliptin, alogliptin, saxagliptin, denagliptin, carmegliptin, melogliptin, and dutogliptin, or a pharmaceutically-acceptable salt of one of the beforementioned DPP IV inhibitors, or a prodrug thereof.
The term "linagliptin" as employed herein refers to linagliptin and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. Crystalline forms are described in WO 2007/128721. Methods for the manufacture of linagliptin are described in the patent applications WO 2004/018468 and WO 2006/ 048427 for example. Linagliptin is distinguished from structurally comparable DPP IV inhibitors, as it combines exceptional potency and a long- lasting effect with favorable pharmacological properties, receptor selectivity and a favorable side-effect profile or bring about unexpected therapeutic advantages or improvements when used in combination with an SGLT2 inhibitor and a third antidiabetic agent according to this invention.
The term "sitagliptin" as employed herein refers to sitagliptin (or MK-0431) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. In one embodiment, sitagliptin is in the form of its
dihydrogenphosphate salt, i.e. sitagliptin phosphate. In a further embodiment, sitagliptin phosphate is in the form of a crystalline anhydrate or monohydrate. A class of this embodiment refers to sitagliptin phosphate monohydrate. Sitagliptin free base and pharmaceutically acceptable salts thereof are disclosed in U.S. Patent No. 6,699,871 and in Example 7 of WO 03/004498. Crystalline sitagliptin phosphate monohydrate is disclosed in WO 2005/003135 and in WO 2007/050485. For details, e.g. on a process to manufacture or to formulate this compound or a salt thereof, reference is thus made to these documents. A tablet formulation for sitagliptin is commercially available under the trade name Januvia®.
The term "vildagliptin" as employed herein refers to vildagliptin (or LAF-237) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. Specific salts of vildagliptin are disclosed in WO 2007/019255. A crystalline form of vildagliptin as well as a vildagliptin tablet formulation are disclosed in WO 2006/078593. Vildagliptin can be formulated as described in WO 00/34241 or in WO 2005/067976. A modified release vildagliptin formulation is described in WO 2006/135723. For details, e.g. on a process to manufacture or to formulate this compound or a salt thereof, reference is thus made to these documents and U.S. Patent No. 6,166,063. A tablet formulation for vildagliptin is expected to be commercially available under the trade name GALVUS®.
The term "saxagliptin" as employed herein refers to saxagliptin and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. In one embodiment, saxagliptin is in the form of the free base or a HC1 salt (for example as mono- or dihydrochloride, including hydrates thereof), or a mono-benzoate salt as disclosed in WO 2004/052850 and WO 2008/131149. In a further embodiment, saxagliptin is in the form of the free base. In a yet further embodiment, saxagliptin is in the form of the monohydrate of the free base as disclosed in WO 2004/052850. A process for preparing saxagliptin is also disclosed in WO 2005/106011 and WO 2005/ 115982. Saxagliptin can be formulated in a tablet as described in WO 2005/117841. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents and U.S. Patent No. 6,395,767 and WO 01/68603.
The term "denagliptin" as employed herein refers to denagliptin (or GSK-823093) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof In one embodiment, denagliptin is in the form of its hydrochloride salt as disclosed in Example 2 of WO 03/002531 or its tosylate salt as disclosed in WO 2005/009956. A class of this embodiment refers to denagliptin tosylate. Crystalline anhydrous denagliptin tosylate is disclosed in WO 2005/009956. For details on a process to manufacture this compound or a salt thereof, reference is thus made to these documents and to the U.S. Patent No. 7,132,443.
The term "alogliptin" as employed herein refers to alogliptin (or SYR-322) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof In one embodiment, alogliptin is in the form of its benzoate salt, its hydrochloride salt or its tosylate salt each as disclosed in WO 2007/035629. A class of this embodiment refers to alogliptin benzoate. Polymorphs of alogliptin benzoate are disclosed in WO 2007/035372. A process for preparing alogliptin is disclosed in WO 2007/112368 and, specifically, in WO 2007/035629. Alogliptin (namely its benzoate salt) can be formulated in a tablet and administered as described in WO 2007/033266. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents and to US 2005/261271 , EP 1586571 and WO 2005/095381.
The term "carmegliptin" as employed herein refers to carmegliptin and
pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof A process for preparing this compound (specifically its
dihydrochloride salt) is also disclosed in WO 2008/031749, WO 2008/031750 and WO 2008/055814. This compound can be formulated in a pharmaceutical composition as described in WO 2007/017423. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents and to WO 2005/000848.
The term "melogliptin" as employed herein refers to melogliptin and
pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof Methods for its preparation are inter alia disclosed in WO
2006/040625 and WO 2008/001195. Specifically claimed salts include the methanesulfonate and p-toluene-sulfonate. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
The term "dutogliptin" as employed herein refers to dutogliptin (or PHX-1149, PHX- 1149T) and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. Methods for its preparation are inter alia disclosed in WO 2005/047297. Pharmaceutically acceptable salts include the tartrate. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
The disclosure of each of the foregoing documents cited above in connection with the specified DPP IV inhibitors is specifically incorporated herein by reference in its entirety.
SGLT-2 inhibitors that may be used in accordance with the invention include, but are not limited to dapagliflozin, canagliflozin, atigliflozin, remogliflozin and sergliflozin.
The term "dapagliflozin" as employed herein refers to dapagliflozin, including hydrates and solvates thereof, and crystalline forms thereof. The compound and methods of its synthesis are described in WO 03/099836 for example. Preferred hydrates, solvates and crystalline forms are described in the patent applications WO 2008/116179 and WO 2008/ 002824 for example.
The term "canaglifiozin" as employed herein refers to canagliflozin, including hydrates and solvates thereof, and crystalline forms thereof and has the following structure: The compound and methods of its synthesis are described in WO 2005/012326 and WO 2009/035969 for example. Preferred hydrates, solvates and crystalline forms are described in the patent applications WO 2008/069327 for example.
The term "atigliflozin" as employed herein refers to atigliflozin, including hydrates and solvates thereof, and crys-talline forms thereof. The compound and methods of its synthesis are described in WO 2004/007517 for example.
The term "remogliflozin" as employed herein refers to remogliflozin and prodrugs of remogliflozin, in particular remogliflozin etabonate, including hydrates and solvates thereof, and crystalline forms thereof. Methods of its synthesis are described in the patent applications EP 1213296 and EP 1354888 for example.
The term "sergliflozin" as employed herein refers to sergliflozin and prodrugs of sergliflozin, in particular sergliflozin etabonate, including hydrates and solvates thereof, and crystalline forms thereof. Methods for its manufacture are described in the patent applications EP 1344780 and EP 1489089 for example.
Accordingly, in one embodiment, provided herein are combination therapies comprising, at least, the following combination of agents:
1) a compound of Formula I and miglitol; a compound of Formula I and glipizide; a compound of Formula I and glyburide; a compound of Formula I and saxagliptin; a compound of Formula I and sitagliptin; a compound of Formula I and vildagliptin; a compound of Formula I and linagliptin; a compound of Formula I and dutogliptin; a compound of Formula I and metformin; a compound of Formula I, metformin, and sitagliptin;
2) a compound of Formula II and miglitol; a compound of Formula II and glipizide; a compound of Formula II and glyburide; a compound of Formula II and saxagliptin; a compound of Formula II and sitagliptin; a compound of Formula II and vildagliptin; a compound of Formula II and linagliptin; a compound of Formula II and dutogliptin; a compound of Formula II and metformin; a compound of Formula II, metformin, and sitagliptin; 3) a compound of Formula III and miglitol; a compound of Formula III and glipizide; a compound of Formula III and glyburide; a compound of Formula III and saxagliptin; a compound of Formula III and sitagliptin; a compound of Formula III and vildagliptin; a compound of Formula III and linagliptin; a compound of Formula III and dutogliptin; a compound of Formula III and metformin; a compound of Formula III, metformin, and sitagliptin;
4) a compound of Formula IV and miglitol; a compound of Formula IV and glipizide; a compound of Formula IV and glyburide; a compound of Formula IV and saxagliptin; a compound of Formula IV and sitagliptin; a compound of Formula IV and vildagliptin; a compound of Formula IV and linagliptin; a compound of Formula IV and dutogliptin; a compound of Formula IV and metformin; a compound of Formula IV, metformin, and sitagliptin;
5) a compound of Formula V and miglitol; a compound of Formula V and glipizide; a compound of Formula V and glyburide; a compound of Formula V and saxagliptin; a compound of Formula V and sitagliptin; a compound of Formula V and vildagliptin; a compound of Formula V and linagliptin; a compound of Formula V and dutogliptin; a compound of Formula V and metformin; a compound of Formula V, metformin, and sitagliptin;
6) a compound of Formula VI and miglitol; a compound of Formula VI and glipizide; a compound of Formula VI and glyburide; a compound of Formula VI and saxagliptin; a compound of Formula VI and sitagliptin; a compound of Formula VI and vildagliptin; a compound of Formula VI and linagliptin; a compound of Formula VI and dutogliptin; a compound of Formula VI and metformin; a compound of Formula VI, metformin, and sitagliptin;
7) a compound of Formula VII and miglitol; a compound of Formula VII and glipizide; a compound of Formula VII and glyburide; a compound of Formula VII and saxagliptin; a compound of Formula VII and sitagliptin; a compound of Formula VII and vildagliptin; a compound of Formula VII and linagliptin; a compound of Formula VII and dutogliptin; a compound of Formula VII and metformin; a compound of Formula VII, metformin, and sitagliptin;
8) a compound of Formula VIII and miglitol; a compound of Formula VIII and glipizide; a compound of Formula VIII and glyburide; a compound of Formula VIII and saxagliptin; a compound of Formula VIII and sitagliptin; a compound of Formula VIII and vildagliptin; a compound of Formula VIII and linagliptin; a compound of Formula VIII and dutogliptin. a compound of Formula VIII and metformin; a compound of Formula VIII, metformin, and sitagliptin;
In an additional embodiment, the combination therapies of paragraphs 1, 2, 3, 4, 5, 6, 7 and 8, above, can be further combined with compounds of Formula I, II, III, IV, V, VI, VII and/or VIII.
Pharmaceutical Compositions
The compounds of the present invention are suitable as active agents in
pharmaceutical compositions that are efficacious particularly for treating obesity, cardiovascular disorders, as well as related conditions. The pharmaceutical composition in various embodiments has a pharmaceutically effective amount of the present active agent along with other pharmaceutically acceptable excipients, carriers, fillers, diluents and the like. Pharmaceutical compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in
'Remington's Pharmaceutical Sciences', 19th Edition (Mack Publishing Company, 1995).
The language "pharmaceutical composition" includes preparations suitable for administration to mammals, e.g., humans. When the compounds of the present invention are administered as pharmaceuticals to mammals, e.g. , humans, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
The phrase "pharmaceutically acceptable carrier" is art recognized and includes a pharmaceutically acceptable material, composition or vehicle, suitable for administering compounds of the present invention to mammals. The carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, saffiower oil, sesame oil, olive oil, corn oil and soybean oil;
glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
Examples of pharmaceutically acceptable antioxidants include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, a- tocopherol, and the like; and metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Formulations of the present invention include those suitable for oral, nasal, topical, buccal, sublingual, rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound that produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste.
In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; absorbents, such as kaolin and bentonite clay; lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydro xypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface- active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydro xypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
The relative amounts of the active ingredient, the pharmaceutically acceptable carrier, and any additional ingredients in a pharmaceutical composition of the invention will vary, depending upon the identity, size, and condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100% (w/w) active ingredient.
In one embodiment, the present invention relates to a pharmaceutical composition for the treatment of dyslipidemia, including hypertriglyceridemia, in mammals comprising an anti-dyslipidemia effective amount of a compound of the present invention and a pharmaceutically acceptable carrier. In one embodiment said mammals are human.
In one embodiment, the present invention relates to a pharmaceutical composition for the treatment of cardiovascular diseases, including cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, and stroke in mammals comprising an effective amount of a compound of the present invention to treat the said cardiovascular diseases and a pharmaceutically acceptable carrier. In an embodiment, the arrhythmia is an atrial fibrillation. In one embodiment said mammals are human In one embodiment, the present invention relates to a pharmaceutical composition for the treatment of obesity, in mammals comprising an anti-obesity effective amount of a compound of the present invention and a pharmaceutically acceptable carrier. In one embodiment said mammals are human.
Dosage
For administration to human patients, the total daily dose of the compounds of the invention is typically in the range 0.05 g to 12 g, e.g. , 1 g to 12 g, depending, of course, on the mode of administration. In one embodiment the total daily dose is in the range 1 g to 10 g and in another embodiment the total daily dose is in the range 1 g to 6 g. In another embodiment, the daily dose is 0.05 g - 6 g, 0.05 g - 3 g, 0.05 g - 1 g, or 0.05 g - 0.2 g. The total daily dose may be administered in single or divided doses.
These dosages are based on an average human subject having a weight of about 65kg to 70kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.
Methods of preparing various pharmaceutical compositions with a specific amount of active compound are known, or will be apparent, to those skilled in this art. For examples, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easter, Pa., 15th Edition (1975).
A pharmaceutical composition of the invention may be prepared, packaged, or sold in bulk, as a single unit dose, or as a plurality of single unit doses. As used herein, a "unit dose" is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
Kits
Advantageously, the present invention also provides kits for use by a consumer for treating disease. The kits comprise a) a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable carrier, vehicle or diluent; and, optionally, b) instructions describing a method of using the pharmaceutical composition for treating the specific disease.
A "kit" as used in the instant application includes a container for containing the separate unit dosage forms such as a divided bottle or a divided foil packet. The container can be in any conventional shape or form as known in the art which is made of a
pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule. The container employed can depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle which is in turn contained within a box.
An example of such a kit is a so-called blister pack. Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material.
During the packaging process, recesses are formed in the plastic foil. The recesses have the size and shape of individual tablets or capsules to be packed or may have the size and shape to accommodate multiple tablets and/or capsules to be packed. Next, the tablets or capsules are placed in the recesses accordingly and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are individually sealed or collectively sealed, as desired, in the recesses between the plastic foil and the sheet.
Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
It may be desirable to provide a written memory aid, where the written memory aid is of the type containing information and/or instructions for the physician, pharmacist or subject, e.g. , in the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen which the tablets or capsules so specified should be ingested or a card which contains the same type of information. Another example of such a memory aid is a calendar printed on the card e.g. , as follows "First Week, Monday,
Tuesday," . . . etc. . . . "Second Week, Monday, Tuesday, . . . " etc. Other variations of memory aids will be readily apparent. A "daily dose" can be a single tablet or capsule or several tablets or capsules to be taken on a given day. Another specific embodiment of a kit is a dispenser designed to dispense the daily doses one at a time. Preferably, the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen. An example of such a memory-aid is a mechanical counter, which indicates the number of daily doses that, has been dispensed. Another example of such a memory-aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
One embodiment of the present invention relates to a kit comprising a unit dosage comprising a compound of the invention with instructions on how to use the kit and with provision for at least one container for holding the unit dosage form.
Methods of Making
The salts of the invention can be prepared using any number of synthesis techniques known to the skilled artisan.
An example for the synthesis of the mono salt of piperazine with eicosapentaenoic acid (EPA) (Formula I) can be prepared as set forth below.
One equivalent of piperazine may be dissolved in an appropriate reaction inert solvent. The solvent may be polar such as water. As used herein, the expression "reaction inert solvent" refers to a solvent or a mixture of solvents that does not interact with starting materials, reagents, intermediates or products in a manner that adversely affects the yield of the desired product. Preferred solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile. To this solution may be added a solution of one equivalent of EPA. Both piperazine and EPA are commercially available. The reaction mixture can be stirred at about ambient temperature to about the reflux temperature of the solvent being used for about two hours to about six hours. The mono salt of piperazine with EPA can be isolated from the mixture by methods well known to those skilled in the art, including according to the method of U.S. Patent No.
7,973,073.
The di-salt of piperazine with EPA (Formula III) can be prepared according to the above procedure, but by using two equivalents instead of one equivalent of EPA.
The mono salt of piperazine with docosahexaenoic acid (DHA) (Formula II) can be prepared as set forth below.
One equivalent of piperazine may be dissolved in an appropriate reaction inert solvent. The solvent may be polar such as water. Preferred reaction inert solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile. To this solution may be added a solution of one equivalent of DHA. Both piperazine and DHA are commercially available. The reaction mixture can be stirred at about ambient temperature to about the reflux temperature of the solvent being used for about two hours to about six hours. The mono salt of piperazine with DHA can be isolated from the mixture by methods well known to those skilled in the art, including according to the method of U.S. Patent No. 7,973,073.
The di-salt of piperazine with DHA (Formula IV) can be prepared according to the above procedure, but by using two equivalents instead of one equivalent of DHA.
The compound of Formula V can be prepared by adding a solution of one equivalent of compound XH to a solution of one equivalent of the compound of the Formula I. Suitable solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile. The compound of Formula VI can be prepared by adding a solution of one equivalent of compound XH to a solution of one equivalent of the compound of the Formula II. Suitable solvents include methanol, ethanol, n- propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile.
The meglumine salt of EPA (Formula VII) can be prepared as set forth below.
One equivalent of meglumine may be dissolved in an appropriate reaction inert solvent. The solvent may be polar such as water. Preferred reaction inert solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile. To this solution may be added a solution of one equivalent of EPA. Both meglumine and EPA are commercially available. The reaction mixture can be stirred at about ambient temperature to about the reflux temperature of the solvent being used for about two hours to about six hours. The meglumine salt of EPA can be isolated from the mixture by methods well known to those skilled in the art, including according to the method of U.S. Patent No. 7,973,073.
The meglumine salt of DHA (Formula VIII) can be prepared as set forth below. One equivalent of meglumine may be dissolved in an appropriate reaction inert solvent. The solvent may be polar such as water. Preferred reaction inert solvents include methanol, ethanol, n-propanol, isopropanol, acetone, ethyl methyl ketone, diethyl ketone, methyl isobutylketone and acetonitrile. To this solution may be added a solution of one equivalent of DHA. Both meglumine and DHA are commercially available. The reaction mixture can be stirred at about ambient temperature to about the reflux temperature of the solvent being used for about two hours to about six hours. The meglumine salt of DHA can be isolated from the mixture by methods well known to those skilled in the art, including according to the method of U.S. Patent No. 7,973,073.
Accordingly, in one aspect, provided herein is a method for the manufacture of a compound of Formula I or II, comprising: reacting one equivalent of piperazine, and one equivalent of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
In another aspect, provided herein is a method for the manufacture of a compound of Formula III or IV, comprising: reacting one equivalent of piperazine, and two equivalents of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
In still another aspect, provided herein is a method for the manufacture of a compound of Formula V or VI, comprising: reacting one equivalent of piperazine, one equivalent of eicosapentaenoic acid or docosahexaenoic acid, and one equivalent of a pharmaceutically acceptable acid, at a temperature between about 0°C and about 60° C.
In another aspect, provided herein is a method for the manufacture of a compound of Formula IX or X, comprising: reacting one equivalent of ethylenediamine, and one equivalent of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
In yet another aspect, provided herein is a method for the manufacture of a compound of Formula XI or XII, comprising: reacting one equivalent of ethylenediamine, and two equivalents of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
In still another aspect, provided herein is a method for the manufacture of a compound of Formula XIII or XIV, comprising: reacting one equivalent of ethylenediamine, one equivalent of eicosapentaenoic acid or docosahexaenoic acid, and one equivalent of a pharmaceutically acceptable acid, at a temperature between about 0°C and about 60° C.
In one embodiment of any of the above methods, the reaction is performed in the absence of a solvent. Exemplification
Example 1
Preparation of Piperazine Eicosapentaenoate R-(-)-Mandelate
A solution of piperazine (0.450 g, 5.22 mmol) in acetonitrile (30 mL, 600 mmol) was treated with a solution of (5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l,14,17-pentaenoic acid (1.90 g, 6.27 mmol) in acetonitrile (30 mL, 600 mmol). The solution was stirred for 10 minutes, and then cooled at 0 °C. Upon cooling a white precipitate formed. The suspension was treated drop wise over 30 minutes with a solution of R-(-)-mandelic acid (0.795 g, 5.22 mmol) in acetonitrile (24 mL, 460 mmol) and the mixture was stirred an additional 2.5 h at 0-5 °C. The reaction mixture was filtered under nitrogen and the solid was washed with cold acetonitrile. The solid was quickly transferred to a round bottom flask and placed under high vacuum overnight. Yield was 2.40 g. The ιΐΙ NMR spectrum, the 13C NMR spectrum and elemental analysis indicate the material is piperazine eicosapentaenoate R-(-)-mandelate.
Anal Calcd for C32H48N205 plus 0.69% H20: C, 70.59; H, 8.96; N, 5.14. Found: C, 70.27; H, 8.84; N, 5.12.
Ή NMR (300 MHz, MeOD) d ppm 7.47 (m, 2 H), 7.27 (m, 3 H), 5.36 (m, 10 H), 3.05 (s, 8 H), 2.85 (m, 8H), 2.28 (t, J=7.45 Hz, 2 H), 2.10 (m, 4 H), 1.67 (m, 2 H), 0.97 (t, J=7.54 Hz, 3 H)
13C NMR (101 MHz, MeOD) d ppm 179.42, 178.49, 143.34, 132.93, 130.37, 129.88, 129.59, 129.42, 129.31, 129.27, 129.20, 129.08,128.57, 128.33, 128.07, 76.10, 44.22, 35.18, 27.83, 26.70, 26.58, 26.42, 21.65, 14.81
Example 2
Bisr(5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l,14,17-pentaenoate1 piperazine salt (EPA2-Pip)
Figure imgf000041_0001
A stirred solution of piperazine (1.28 g, 14.9 mmol in acetonitrile (30 mL, 600 mmol) is treated dropwise with a solution of (5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l ,14,17-pentaenoic acid (9.00 g, 29.8 mmol) in acetonitrile (73 mL, 1400 mmol) under N2. After 30 minutes, the mixture is stored in the refrigerator overnight. The solid was collected by filtration and dried under hi-vac at RT overnight with P205. Yield = 8.2g of bis[(5Z,8Z,l lZ, 14Z,17Z)-icosa- 5,8, 11 , 14, 17-pentaenoate] piperazine salt as a pink solid.
XH NMR (300 MHz, MeOD) δ 0.99 (t, 6 H) 1.68 (t, 4 H) 2.04 - 2.18 (m, 3 H) 2.11 (d, 5 H) 2.25 (t, 4 H) 2.85 (m, 17 H) 3.07 (s, 9 H) 5.27 - 5.45 (m, 20 H); MS (ESI+) for C20H30O2 m/z 303 (M+H)+; Anal Calcd for C44H70N2O4: C, 76.48; H, 10.21 ; N, 4.05. Found: C, 76.54; H, 10.09; N, 4.04.; MP = 61 - 64°C
Example 3
Ethane- 1.2-diaminium dir(5Z.8Z.l lZ.14Z.17Z)-icosa-5.8.11.14.17-pentaenoatel (EPA2- EDA)
Figure imgf000042_0001
A mixture of (5Z,8Z,l lZ,14Z, 17Z)-icosa-5,8,l l ,14, 17-pentaenoic acid (10.50 g, 34.72 mmol) and ethylenediamine (1.10 mL, 16.5 mmol) in acetonitrile (215.8 mL, 4133 mmol) is stirred with ice bath cooling for 2 hrs and then stored in the refrigerator overnight. The solid is collected by filtration and dried under hi-vac at RT over P2O5 for 12 hrs. Yield = 7.9 g of ethane- 1 ,2-diaminium di[(5Z,8Z,l lZ, 14Z,17Z)-icosa-5 ,8, 11 , 14, 17-pentaenoate] as a brown solid. XH NMR (300 MHz, MeOD) δ 0.92 (t, 6 H) 1.65 (m, 4 H) 2.16 (m, 12 H) 2.89 (m., 16 H) 2.99 (s, 4H) 4.88 (s, 6 H) 5.37 (br. s., 20 H); MS (ESI+) for C20H30O2 m/z 303 (M+H)+; Anal Calcd for C42H68N2O4: C, 75.86; H, 10.31 ; N, 4.21. Found: C, 75.70; H, 10.25; N, 4.07. MP = 30°C
Example 4
(Benzoylamino)acetate (5Z,8Z,l lZ,14Z,17Z)-icosa-5, 8, 11 , 14, 17-pentaenoate piperazine salt
(EPA piperazine hippurate salt)
Figure imgf000042_0002
A solution of (5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l ,14,17-pentaenoic acid (7.00 g, 23.1 mmol) in 100 mL acetonitrile is added to a solution of piperazine (1.661 g, 19.29 mmol) in 90 mL acetonitrile and the mixture is stirred for 30 min. The mixture is cooled in an ice bath and a solution of N-benzoylglycine (3.456 g, 19.29 mmol) in 80 mL of acetonitrile is added drop wise. The mixture is stirred with ice bath cooling for 6 hours, then stored in the refrigerator overnight. The solid is collected by filtration and rinsed with 3 x 20 ml of ice cold CH3CN. The resulting solid is dried at RT under hi- vac with P2O5 overnight. Yield = 9.1g of (benzoylamino) acetate (5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l,14,17-pentaenoate piperazine salt as a pink solid. lU NMR (300 MHz, MeOD) d ppm 0.98 (t, 7=7.54 Hz, 3 H) 1.51 - 1.79 (m, 2 H) 1.94 - 2.22 (m, 4 H) 2.28 (t, 7=7.45 Hz, 2 H) 2.66 - 2.95 (m, 8 H) 5.12 - 5.56 (m, 8 H) 7.28 - 7.63 (m, 3 H) 7.75 - 7.98 (m, 2 H). Anal Calcd for C33H49N3O5 plus 0.28% H20: C, 69.62; H, 8.71 ; N, 7.38. Found: C, 69.26; H, 8.35; N, 7.58.
Example 5
Ethane- 1 ,2-diaminium (5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l,14,17-pentaenoate
methanesulfonat
Figure imgf000043_0001
A mixture of ethylenediamine (0.884 mL, 13.2 mmol) and (5Z,8Z,11Z,14Z,17Z)- icosa-5,8,l l ,14,17-pentaenoic acid (4.800 g, 15.87 mmol; Supplier = Thetis) in acetonitrile (138.1 mL, 2645 mmol) is stirred at RT for 1/2 hr. Methanesulfonic acid (0.858 mL, 13.2 mmol) is added and the mixture is stirred with ice bath cooling for 1 hr, then stored in the refrigerator overnight. The solid is collected by filtration and washed with ice cold CH3CN. Yield = 4.5g of ethane- 1 ,2-diaminium (5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l ,14,17-pentaenoate methanesulfonate as a tacky solid. The material is triturated with ice cold CH3CN (3 x 20 ml) and dried under hi- vac with P2O5 at RT overnight. Yield = 3.4g of ethane- 1 ,2- diaminium5Z,8Z,HZ,14Z,17Z)-icosa-5,8,l l,14,17-pentaenoatemethane sulfonate as a tan solid. XH NMR (300 MHz, DMSO-i 6) d ppm 0.92 (t, 7=7.54 Hz, 3 H) 1.44 - 1.63 (m, 2 H) 1.95 - 2.10 (m, 2 H) 2.16 (t, 7=7.40 Hz, 2 H) 2.34 (s, 3 H) 2.69 - 2.91 (m, 12 H) 5.20 - 5.46 (m, 10 H) 6.60 (br. s., 6 H). Anal Calcd for C23H42N2O5S plus 0.60% H20: C, 59.87; H, 9.24; N, 6.07. Found: C, 59.48; H, 9.13; N, 6.24. Example 6
l-deoxy-l -(methylami -D-glucitol (5Z,8Z,l lZ,14Z,17Z)-icosa-5,8,l l ,14,17-pentaenoate
Figure imgf000044_0001
A suspension of l-deoxy-l-(methylamino)-D-glucitol (0.200 g, 1.02 mmol) in 6.0 mL of methanol was treated drop wise with a solution of (5Z,8Z,1 lZ,14Z,17Z)-icosa- 5,8,11,14, 17-pentaenoic acid (0.325 g, 1.08 mmol) in 7.0 mL tetrahydrofuran and the mixture was stirred at RT for 30 minutes while being protect from light. The slurry became homogeneous upon addition of the EPA solution. The solution was concentrated to pink oil that was placed on high vac overnight to yield a pink foam. Ή NMR (300 MHz, MeOD) δ ppm 5.36 (m, 10 H), 4.04 (dt, J=6.94, 4.96 Hz, 1 H), 3.80 (m, 2 H), 3.68 (m, 3 H), 3.14 (m, 2 H), 2.84 (m, 8 H), 2.68 (s, 3 H), 2.19 (m, 2 H), 2.10 (m, 4 H), 1.66 (m, 2 H), 0.97 (t, J=7.50 Hz, 3 H). Example 7
Pharmacokinetics of Piperazine di-eicosapentaenoate
Oral pharmacokinetic parameters of piperazine di-eicosapentaenoate, prepared by the procedure described in Example 2, were determined in Sprague-Dawley rats. Piperazine di- eicosapentaenoate was administered by oral gavage as an aqueous solution in 0.5% carboxymethyl cellulose to 6 Sprague-Dawley rats, 3 males and 3 females. Rats were dosed at 40 mg/kg. Blood samples were obtained from each rat by jugular vein catheter. Samples were collected at 0.25, 0.5, 1, 2, 4, 8, 12, and 24 hours post dose. Blood samples were centrifuged to separate red blood cells and the resulting plasma samples were analyzed for eicosapentaenoic acid. Calculated pharmacokinetic parameters shown below in Table 1 are mean values from 6 rats.
Table 1
Rat Oral Pharmacokinetic Parameters for Piperazine di-eicosapentaenoate
Figure imgf000044_0002
Example 8
Pharmacokinetics of Ethylene diamine di-eicosapentaenoate
Oral pharmacokinetic parameters of ethylene diamine di-eicosapentaenoate, prepared by the procedure described in Example 3, were determined in Sprague-Dawley rats. Ethylene diamine di-eicosapentaenoate was administered by oral gavage as an aqueous solution in 0.5% carboxymethyl cellulose to 6 rats, 3 males and 3 females. Rats were dosed at 40 mg/kg. Blood samples were obtained from each rat by jugular vein catheter. Samples were collected at 0.25, 0.5, 1, 2, 4, 8, 12, and 24 hours post dose. Blood samples were centrifuged to separate red blood cells and the resulting plasma samples were analyzed for eicosapentaenoic acid. Calculated pharmacokinetic parameters shown below are mean values from 6 rats.
Table 2
Rat Oral Pharmacokinetic Parameters for Ethylene diamine di-eicosapentaenoate
Figure imgf000045_0001
Combination Therapy
Pharmacological Examples
Treatment of Type 2 Diabetes
Treating patients with type 2 diabetes with the pharmaceutical composition according to the invention, in addition to producing an acute improvement in the glucose metabolic situation, prevents a deterioration in the metabolic situation in the long term. This can be observed when patients are treated for a longer period, e.g. 3 months to 1 year or even 1 to 6 years, with the pharmaceutical composition according to the invention and are compared with patients who have been treated with other antidiabetic medicaments. There is evidence of therapeutic success compared with patients treated with other antidiabetic medicaments if no or only a slight increase in the fasting glucose and/or HbAlc value is observed. Further evidence of therapeutic success is obtained if a significantly smaller percentage of the patients treated with a pharmaceutical composition according to the invention, compared with patients who have been treated with other medicaments, undergo a deterioration in the glucose meta-bolic position (e.g. an increase in the HbAlc value to >6.5% or >7%) to the point where treatment with an additional oral antidiabetic medicament or with insulin or with an insulin analogue is indicated. Treatment of Insulin Resistance
In clinical studies running for different lengths of time (e.g. 2 weeks to 12 months) the success of the treatment is checked using a hyperinsulinaemic euglycaemic glucose clamp study. A significant rise in the glucose infusion rate at the end of the study, compared with the initial value or compared with a placebo group, or a group given a different therapy, proves the efficacy of a pharmaceutical composition according to the invention in the treatment of insulin resistance.
Treatment of Hyperglycaemia
In clinical studies running for different lengths of time (e.g. 1 day to 24 months) the success of the treatment in patients with hyperglycaemia is checked by determining the fasting glucose or non-fasting glucose (e.g. after a meal or a loading test with oGTT or a defined meal). A significant fall in these glucose values during or at the end of the study, compared with the initial value or compared with a placebo group, or a group given a different therapy, proves the efficacy of a pharmaceutical composition according to the invention in the treatment of hyperglycaemia. Treatment of Metabolic Syndrome
The efficacy of a pharmaceutical composition according to the invention can be tested in clinical studies with varying run times (e.g. 12 weeks to 6 years) by determining the fasting glucose or non-fasting glucose (e.g. after a meal or a loading test with oGTT or a defined meal) or the HbAlc value. A significant fall in these glucose values or HbAlc values during or at the end of the study, compared with the initial value or compared with a placebo group, or a group given a different therapy, proves the efficacy of an active ingredient or combination of active ingredients in the treatment of Metabolic Syndrome. Examples of this are a reduction in systolic and/or diastolic blood pressure, a lowering of the plasma triglycerides, a reduction in total or LDL cholesterol, an increase in HDL cholesterol or a reduction in weight, either compared with the starting value at the beginning of the study or in comparison with a group of patients treated with placebo or a different therapy. Prevention of Micro- or Macrovascular Complications
The treatment of type 2 diabetes or pre-diabetes patients with a pharmaceutical composition according to the invention prevents or reduces or reduces the risk of developing microvascular complications (e.g. diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic foot, diabetic ulcer) or macrovascular complications (e.g. myocardial infarct, acute coronary syndrome, unstable angina pectoris, stable angina pectoris, stroke, peripheral arterial occlusive disease, cardiomyopathy, heart failure, heart rhythm disorders, vascular restenosis). Type 2 diabetes or patients with pre-diabetes are treated long-term, e.g. for 1-6 years, with a pharmaceutical composition according to the invention or a combination of active ingredients according to the invention and compared with patients who have been treated with other antidiabetic medicaments or with placebo. Evidence of the therapeutic success compared with patients who have been treated with other antidiabetic medicaments or with placebo can be found in the smaller number of single or multiple complications. In the case of macrovascular events, diabetic foot and/or diabetic ulcer, the numbers are counted by anam- nesis and various test methods. In the case of diabetic retinopathy the success of the treatment is determined by com-iputer-controlled illumination and evaluation of the background to the eye or other ophthalmic methods. In the case of diabetic neuropathy, in addition to anamnesis and clinical examination, the nerve conduction rate can be mea-isured using a calibrated tuning fork, for example. With regard to diabetic nephropathy the following parameters may be investigated before the start, during and at the end of the study: secretion of albumin, creatinine clearance, serum creatinine values, time taken for the serum creatinine values to double, time taken until dialysis becomes necessary.
Diabetic Rats Model
A diabetic rat model that may be used for determination of conditions leading to a method for treatment and prevention of post-ischemic damage of the heart and heart tissue is the spontaneously diabetic bio-bred (BBAV) rats. These rats are a useful model of autoimmune human insulin-dependent diabetes.
Isolated Perfused Heart Model
This example describes an isolated perfused rat heart model used in development of the invention. Studies are performed using an isovolumic isolated rat heart preparation. Acutely diabetic male BBAV rats and non-diabetic age-matched (3-4 months old) control are pretreated with heparin (1000 u; IP), followed by sodium pentobarbital (65 mg/kg; IP). After deep anaesthesia is achieved as determined by the absence of a foot reflex, the hearts are rapidly excised and placed into iced saline. The arrested hearts are retrograde perfused in a non-recirculating model through the aorta within 2 min following their excision. Left ventricular developed pressure (LVDP) is determined using a latex balloon in the left ventricle with high pressure tubing connected to a pressure transducer. Perfusion pressure is monitored using high pressure tubing off the perfusion line. Hemodynamic measurements are recorded on a 4-channel Gould recorder. The system has two parallel perfusion lines with separate oxygenators, pumps and bubble traps, but common temperature control allowing rapid change perfusion media. The hearts are perfused using an accurate roller pump. The perfusate consists of 118 mM NaCl, .47 mM KC1, 12 mM CaC12, 12 mM MgC12, 25 mM NaHC03, and the substrate 11 mM glucose. The perfusion apparatus is tightly temperature- controlled, with heated baths being used for the perfusate and for the water jacketing around the perfusion tubing to maintain heart temperature at 37 ± 0.5 °C under all conditions. The oxygenated perfusate in the room temperature reservoir is passed through 25 ft. of thin- walled silicone tubing surrounded by distilled water at 37 °C saturated with 95% oxygen. The perfusate then enters the water-jacketed (37 °C) tubing leading to the heart through a water jacketed bubble trap. This preparation provides excellent oxygenation that routinely has been stable for 3-4 hours. Model for Zero-flow Ischemia
This example describes a procedure used for study of zero-flow ischemia in diabetic control, diabetic treated, non-diabetic treated and control isolated hearts. Diabetic control (DC) diabetic treated (DZ) normal (C) control and normal treated (CZ) hearts are subjected to 20 min. of normoxic perfusion followed by 20 min. of zero-flow ischemia where the perfusate flow is completely shut off, followed by 60 min. of reperfusion. Hearts are treated with 10 μΜ of the compounds of the invention. The compounds of the invention treated diabetic group (DZ), hearts are subjected to 10 min. of normoxic perfusion with normal Krebs-Henseleit buffer and 10 min. of normoxic perfusion with Krebs-Henseleit buffer containing 10 μΜ of the compound of Formula I. The hearts are then subjected to 20 min. of zero-flow ischemia followed by 60 min. of reperfusion. In order to avoid any variability in reperfusion conditions, both DC and DZ hearts are reperfused with normal Krebs-Henseleit buffer. Model for Low-flow Ischemia
This example describes a procedure used for study of low-flow ischemia in diabetic controls, diabetic treated, non-diabetic treated and non-diabetic control isolated hearts .Diabetic control hearts (DC) are subjected to 20 min. of normoxic perfusion at a flow rate of 12.5 mL/min. followed by 30 minutes of low-flow ischemia where the perfusate flow is slowed down to 1.25 mL/min, that is about 10% of normal perfusion, followed by 30 min. of reperfusion at a normal flow rate (12.5 mL/min). In the groups treated with the compound of Formula I diabetic or non-diabetic groups (DZ or CZ), hearts are subjected to 10 min. of normoxic perfusion (flow rate 12.5 mL/min) with normal Krebs-Henseleit buffer and 10 min. of normoxic perfusion with Krebs-Henseleit buffer containing 10 μΜ of a compound of
Formula I. The hearts are subjected to 30 min. of low-flow ischemia (flow rate 1.25 mL/min) and 30 minutes of reperfusion at normal flow rate (12.5 mL/min).
Animal models to determine the effects of compounds of the invention on diabetes and complications of diabetes have been reviewed by Tirabassi et al., ILAR Journal, 2004, 45, 292-302. Antidiabetic activity may also be tested according to protocols described in the following patents: U.S. Patent Nos. 4,340,605; 4,342,771 ; 4,367,234; 4,617;312; 4;687,777 and 4,703,052. Additional references relevant to this application include the following: French Patent 2796551 and U.S. Published Patent Application No. 20030220301.
Formuhtion Examples: Combination Therapy The following examples of formulations, which may be obtained analogously to methods known in the art, serve to illustrate the present invention more fully without restricting it to the contents of these examples.
The term "active ingredient" denotes two compounds according to the invention, i.e., denotes a compound of Formula I, II, III, IV, V, VI, VII or VIII, or a mixture thereof (first component of the active ingredient) and other antidiabetic agents. The other antidiabetic agents may be statins, cholesterol absorption inhibitors, and CETP inhibitors or a pharmaceutically-acceptable salt or prodrug thereof, or a pharmaceutically- acceptable salt of said prodrug (second component of the active ingredient). Alternatively, the antidiabetic agents may be a DPP IV inhibitor, especially sitagliptin, sitagliptin phosphate, alogliptin, vildagliptin, saxagliptin, and linagliptin, sulfonylurea, SGLT-2 inhibitor, or other antidiabetic compounds according to this invention (second component of the active ingredient). Additional suitable formulations may be prepared according to the procedures described in, for example in the application WO 2007/128724, and in the U.S. Patent Application, 2010/032011 the disclosure of which are incorporated herein in its entirety. Additional suitable formulations for the sulfonylureas, DPP IV inhibitors may be those formulations which are available on the market, or formulations described in the patent applications cited above in paragraph "background of the invention", or those described in the literature, for example as disclosed in current issues of "Rote Liste S" (Germany) or of "Physician's Desk Reference".
Example 1
Tablet Containing 1000 mg of Active Ingredient
Composition
(1) Active ingredient 1000 mg
(2) Mannitol 100 mg
(3) Maize starch 50 mg
(4) Polyvinyl pyrrolidone 15 mg
(5) Magnesium stearate 2 mg
1167 mg
Preparation:
(1), (2) and (3) are mixed together and granulated with an aqueous solution of (4). is added to the dried granulated material. From this mixture tablets are pressed, biplanar, faceted on both sides and with a dividing notch on one side.
Diameter of the tablets: 9 mm
Example 2
Tablet Containing 1050 mg of Active Ingredient
Composition
(1) Active ingredient 1050 mg
(2) Mannitol 100 mg
(3) Maize starch 50 mg
(4) Polyvinyl pyrrolidone 15 mg
(5) Magnesium stearate 2 mg
1217 mg
Preparation:
(1), (2) and (3) are mixed together and granulated with an aqueous solution of (4). (5) is added to the dried granulated material. From this mixture tablets are pressed, biplanar, faceted on both sides and with a dividing notch on one side.
Diameter of the tablets: 9 mm
Example 3
Tablet Containing 1100 mg of Active Ingredient
Composition
(1) Active ingredient 1100 mg
(2) Mannitol 100 mg
(3) Maize starch 50 mg
(4) Polyvinyl pyrrolidone 15 mg
(5) Magnesium stearate 2 mg
1267 mg
Preparation:
(1), (2) and (3) are mixed together and granulated with an aqueous solution of (4). is added to the dried granulated material. From this mixture tablets are pressed, biplanar, faceted on both sides and with a dividing notch on one side.
Diameter of the tablets: 9 mm. Example 4
Capsules Containing 1050 mg of Active Ingredient
Composition
(1) Active ingredient 1050 mg
(2) Mannitol 100 mg
(3) Maize starch 50 mg
(4) Polyvinyl pyrrolidone 15 mg
(5) Magnesium stearate 2 mg
1217 mg
Preparation:
(1) is triturated with (3). This trituration is added to the mixture of (2) and (4) with vigorous mixing. This powder mixture is packed into size 3 hard gelatin capsules in a capsule filling machine.
Example 5
Capsules Containing 1100 mg of Active Ingredient
Composition
(1) Active ingredient 1100 mg
(2) Mannitol 100 mg
(3) Maize starch 50 mg
(4) Polyvinyl pyrrolidone 15 mg
(5) Magnesium stearate 2 mg
1267 mg
Preparation:
(1) is triturated with (3). This trituration is added to the mixture of (2) and (4) with vigorous mixing. This powder mixture is packed into size 3 hard gelatin capsules in a capsule filling machine

Claims

Claims
Figure imgf000053_0001
Formula F
wherein, for Formulas A and B, R+ represents a piperidine or diamine group, wherein the nitrogen of the piperidine and one of the nitrogens of the diamine are protonated; for Formulas C and D, R++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated; and
for Formulas E and F, R++ represents a diamine group, wherein two of the nitrogens of the diamine are protonated, and X" is an anion of a pharmaceutically acceptable acid compound.
2. A compound of the structural Formula I, II, III, IV, V, or VI:
Figure imgf000054_0001
Figure imgf000054_0002
Figure imgf000055_0001
Formula VI wherein X" is an anion of a pharmaceutically acceptable acid compound.
A compound of the structural Formula I, II, III, or IV:
Figure imgf000055_0002
Formula I
Figure imgf000055_0003
Formula II
Figure imgf000055_0004
ormu a
Figure imgf000055_0005
Formula IV
wherein X" is an anion of a pharmaceutically acceptable acid compound.
4. A compound of the structural Formula IX, X, XI, XII, XIII or VIV:
Figure imgf000056_0001
Formula XIV
wherein X" is an anion of a pharmaceutically acceptable acid compound.
5. The compound of any of the above claims, wherein X" is selected from mineral acids, carboxylic acids, sulfonic acids, amino acids, or omega-3 polyunsaturated acids.
6. The compound of claim 5, wherein the mineral acid is hydrochloric acid, hydrobromic acid, or phosphoric acid.
7. The compound of claim 5, wherein the carboxylic acid is acetic acid, propionic acid, succinic acid, maleic acid, malic acid, tartaric acid, lactic acid, citric acid, or benzoic acid.
8. The compound of claim 5, wherein the sulfonic acid is methanesulfonic acid, isethionic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, or benzenesulfonic acid.
9. The compound of claim 5, wherein the omega-3 polyunsaturated acid is
eicosapentaenoic acid or docosahexaenoic acid.
10. The compound of claim 5, wherein the amino acid is glycine, alanine, lysine, arginine, aspartic acid, or glutamic acid.
11. A pharmaceutical composition comprising a compound of any of the above claims, and a pharmaceutically acceptable carrier.
12. A kit comprising a) a unit dosage comprising a compound of any of claims 1-10, b) instructions on how to use the kit; and at least one container for holding the unit dosage forms.
13. A method of treating hyperlipidemia, comprising administering to a subject in need thereof an effective amount of a compound of any of claims 1 -10, or a compound of Formula VII or VIII:
Figure imgf000057_0001
Formula VI I
Figure imgf000058_0001
Formula VIII
14. A method of treating hypertriglycerridemia, comprising administering to a subject in need thereof an effective amount of a compound of any of claims 1-10, or a compound of Formula VII or VIII:
Figure imgf000058_0002
15. A method of treating cardiovascular disease, comprising administering to a subject in need thereof an effective amount of a compound of any of claims 1-10, or a compound of Formula VII
Figure imgf000058_0003
Formula VIII
16. The method of claim 15, wherein the cardiovascular disease is cardiac arrhythmia, cardiac ischemia, myocardial infarction, cardiomyopathy, or stroke.
17. A method of treating obesity, comprising administering to a subject in need thereof effective amount of a compound of any of claims 1-10, or a compound of Formula VII or VIII:
Figure imgf000059_0001
F rmula VII
Figure imgf000059_0002
Formula VIII
18. A method of treating atrial fibrillation, or reducing the probability of an occurance of an atrial fibrillation, comprising administering to a subject in need thereof an effective amount of a I:
Figure imgf000059_0003
Formula VIII
The method of any of claims 13-18, wherein the subject is human.
20. A method for the manufacture of a compound of Formula I or II, comprising: reacting one equivalent of piperazine, and one equivalent of eicosapentaenoic acid or
docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
21. A method for the manufacture of a compound of Formula III or IV, comprising: reacting one equivalent of piperazine, and two equivalents of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
22. A method for the manufacture of a compound of Formula V or VI, comprising: reacting one equivalent of piperazine, one equivalent of eicosapentaenoic acid or docosahexaenoic acid, and one equivalent of a pharmaceutically acceptable acid, at a temperature between about 0°C and about 60° C.
23. A method for the manufacture of a compound of Formula IX or X, comprising: reacting one equivalent of ethylenediamine, and one equivalent of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
24. A method for the manufacture of a compound of Formula XI or XII, comprising: reacting one equivalent of ethylenediamine, and two equivalents of eicosapentaenoic acid or docosahexaenoic acid, at a temperature between about 0°C and about 60° C.
25 A method for the manufacture of a compound of Formula XIII or XIV, comprising: reacting one equivalent of ethylenediamine, one equivalent of eicosapentaenoic acid or docosahexaenoic acid, and one equivalent of a pharmaceutically acceptable acid, at a temperature between about 0°C and about 60° C.
26. The method of any of claims 20-25, wherein the reaction is performed in the absence of a solvent. 27. A pharmaceutical composition comprising a compound of any one of claims 1-10 and an antihyperlipidemic agent, and a pharmaceutically acceptable carrier.
28. A pharmaceutical composition comprising a compound of any one of claims 1-10 and an antihyperglycemic agent, and a pharmaceutically acceptable carrier.
PCT/US2013/049290 2012-07-06 2013-07-03 Diamine and meglumine salt forms of fatty acids WO2014008379A2 (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US201261668517P 2012-07-06 2012-07-06
US201261668582P 2012-07-06 2012-07-06
US201261668512P 2012-07-06 2012-07-06
US61/668,512 2012-07-06
US61/668,517 2012-07-06
US61/668,582 2012-07-06
US201261670384P 2012-07-11 2012-07-11
US61/670,384 2012-07-11
US13/841,667 US20140011814A1 (en) 2012-07-06 2013-03-15 Diamine and meglumine salt forms of fatty acids
US13/841,667 2013-03-15

Publications (2)

Publication Number Publication Date
WO2014008379A2 true WO2014008379A2 (en) 2014-01-09
WO2014008379A3 WO2014008379A3 (en) 2014-02-20

Family

ID=49882605

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/049290 WO2014008379A2 (en) 2012-07-06 2013-07-03 Diamine and meglumine salt forms of fatty acids

Country Status (1)

Country Link
WO (1) WO2014008379A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9505709B2 (en) 2014-05-05 2016-11-29 Thetis Pharmaceuticals Llc Compositions and methods relating to ionic salts of peptides
US10563042B2 (en) 2016-12-14 2020-02-18 Ecolab Usa Inc. Quaternary cationic polymers
US11427964B2 (en) 2018-06-12 2022-08-30 Ecolab Usa Inc. Quaternary cationic surfactants and polymers for use as release and coating modifying agents in creping and tissue papers

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996033155A1 (en) * 1995-04-20 1996-10-24 Scotia Holdings Plc Fatty acid derivatives
WO2007091070A1 (en) * 2006-02-07 2007-08-16 Universitetet I Oslo Omega 3
WO2008053331A1 (en) * 2006-11-01 2008-05-08 Pronova Biopharma Norge A/S Alpha-substituted omega-3 lipids that are activators or modulators of the peroxisome proliferators-activated receptor (ppar).

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996033155A1 (en) * 1995-04-20 1996-10-24 Scotia Holdings Plc Fatty acid derivatives
WO2007091070A1 (en) * 2006-02-07 2007-08-16 Universitetet I Oslo Omega 3
WO2008053331A1 (en) * 2006-11-01 2008-05-08 Pronova Biopharma Norge A/S Alpha-substituted omega-3 lipids that are activators or modulators of the peroxisome proliferators-activated receptor (ppar).

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MACKIE S.J. ET AL.: 'Meglumine Eicosapentaenoic acid (MeEPA) a new soluble omega-3 fatty acid formulation: In vitro bladder cancer cytotoxicity tests in combination with epirubicin and mitomycin.' PROSTAGLANDINS, LEUKOTRIENES AND ESSENTIAL FATTY ACIDS vol. 75, 2006, pages 367 - 373 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9505709B2 (en) 2014-05-05 2016-11-29 Thetis Pharmaceuticals Llc Compositions and methods relating to ionic salts of peptides
US10563042B2 (en) 2016-12-14 2020-02-18 Ecolab Usa Inc. Quaternary cationic polymers
US11319427B2 (en) 2016-12-14 2022-05-03 Ecolab Usa Inc. Quaternary cationic polymers
US11427964B2 (en) 2018-06-12 2022-08-30 Ecolab Usa Inc. Quaternary cationic surfactants and polymers for use as release and coating modifying agents in creping and tissue papers

Also Published As

Publication number Publication date
WO2014008379A3 (en) 2014-02-20

Similar Documents

Publication Publication Date Title
EP2872483A1 (en) Tri-salt form of metformin
AU2018304380B2 (en) S-enantiomers of beta-hydroxybutyrate and butanediol and methods for using same
ES2257757T3 (en) TERFENADINE CARBOXYLATE AND THE TREATMENT OF ALLERGIC DISORDERS.
US20140011814A1 (en) Diamine and meglumine salt forms of fatty acids
JP5911514B2 (en) Lipid-lowering antidiabetic drugs
JP2008521827A5 (en)
US20100160408A1 (en) Vildagliptin for treating general peripheral neuropathies
US20080300293A1 (en) Combination of an dpp-iv inhibitor and a ppar-alpha compound
US8765811B2 (en) Tri-salt form of metformin
TW200911732A (en) Compositions and methods of use for treating or preventing lipid related disorders
TWI694069B (en) Pharmaceutical formulation for treating schizophrenia
AU2005304318A1 (en) Lipase inhibitors
WO2014008379A2 (en) Diamine and meglumine salt forms of fatty acids
US20140221467A1 (en) Metformin derivatives for treating diabetes and diabetes complications
WO2014008374A2 (en) Combination therapies comprising metformin salts and antihyperglycemia agents or antihyperlipidemia agents
JP2014528928A (en) Use of organic compounds for the treatment of Noonan syndrome
US9382187B2 (en) Tri-salt form of metformin
WO2008112166A2 (en) Combination of metformin r-(+) lipoate and antihyperlipidemic agents for the treatment of diabetic hyperglycemia and diabetic complications
WO1995022327A1 (en) Use of 1-(arylalkylaminoalkyl) imidazoles for treating neurological damage
EP1247533A2 (en) Combination treatment of multiple sclerosis (MS), other demyelinating conditions and peripheral neuropathy, especially painful neuropathies and diabetic neuropathy
KR20230136627A (en) Use of exogenous ketone esters to induce weight loss in mammals
WO2001034133A2 (en) Oncolytic combinations for the treatment of cancer
WO2009085198A2 (en) Combination of metformin r-(+)-lipoate and antiobesity agents for the treatment of diabetic hyperglycemia and diabetic complications
US20110060156A1 (en) Pyruvate Derivatives with Neuroprotective Effect, Process for Preparing the Same and Pharmaceutical Composition Comprising the Same
WO2009085223A1 (en) Combination of metformin r-(+)-lipoate and antihyperglycemic agents for the treatment of diabetic hyperglycemia and diabetic complications

Legal Events

Date Code Title Description
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13812739

Country of ref document: EP

Kind code of ref document: A2