Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF HYPERGLYCEMIA
Document Type and Number:
WIPO Patent Application WO/2013/167995
Kind Code:
A2
Abstract:
The invention relates to the compounds of formula (I) or its pharmaceutical acceptable salts, as well as polymorphs, solvates, enantiomers, stereoisomers and hydrates thereof The pharmaceutical compositions comprising an effective amount of compounds of formula (I), and methods for the treatment of hyperglycemia may be formulated for oral, buccal, rectal, topical, transdermal, transmucosal, intravenous, parenteral administration, syrup, or injection. Such compositions may be used to treatment of insulin resistance, diabetes mellitus, diabetes insipidus, type 1 diabetes, type 2 diabetes, microvascular complications, macrovascular complications, lipid disorders, prediabetes, obesity, arrhythmia, myocardial infarction, stroke, neuropathy, renal complications, hypertriglyceridemia, cardiovascular complications, and post prandial hyperglycemia.

Inventors:
KANDULA MAHESH (IN)
Application Number:
PCT/IB2013/050903
Publication Date:
November 14, 2013
Filing Date:
February 03, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
KANDULA MAHESH (IN)
Download PDF:
Claims:
CLAIMS

1. A compound of formula I:

Formula. 1

or a pharmaceutically acceptable salt, hydrate, polymorph, solvate, prodrug, enantiomer, or stereoisomer thereof;

Wherein,

R1 independently represents .D, H, methyl, ethyl, CH3CO- or each n is independently 0, 1 , 2, or 3;

R2 independently represents H, D,

44

46

a is independently 2,3 or 7; each b is independently 3, 5 or 6; e is independently 1, 2 or 6;

c and d are each independently H, D, -OH, -OD, CrCf,-alkyI, -N¾ or -COC¾;

R4, R5, Rb, Rf each independently are selected from the groups consisting of H, D, -OH, -OD, methyl, ethyl, -OCH?, ~OCD3 or acetyl .

2. A Pharmaceutical composition comprising a compound of claim 1 and a pharmaceutically acceptable carrier

3. The pharmaceutical composition of claim 2, which is formulated to treat the underlying etiology with an effective amount administering the patient in need by oral administration, delayed release or sustained release, transmucosal, syrup, topical, parenteral administration, injection, subdermai, oral solution, rectal administration, buccai administration or transdermal administration.

4. Compounds and compositions of claim 3 are formulated for the treatment of hyperglycemia, insulin resistance, diabetes roeilirus, diabetes insipidus, type I diabetes, type 2 diabetes, microvascular complications, macrovascular complications, lipid disorders, prediabetes, obesity, arrhythmia, myocardial infarction, stroke, neuropathy, renal com plications, hyperinsulinemia, hypertriglyceridemia, cardiovascular complications. and post prandial hyperglycemia,

5. A molecular conjugate of miglitol and R-Lipoic acid.

6. A molecular conjugate o . miglitol and eicosapentaenoic acid.

7. A molecular conjugate of miglitol and docosahexaenoic acid.

8. A molecular conjugate of miglitol and acetyl cysteine.

9. A molecular conjugate of miglitol and saJsalate.

Description:
COMPOSITIONS AND METHODS FOR THE

TREATMENT OF HYPERGLYCEMIA

PRIORITY

[0001 j The present application claims the benefit of Indian Provisional Patent Application No. 181 i/CHE/2012 filed on 08~May~2012, the entire disclosure of which is relied on for all purposes and is incorporated into this application by reference.

FIELD OF THE INVENTION j ' 0002| This disclosure generally relates to compounds and compositions for the treatment of hyperglycemia, diabetes and insulin resistance More particularly, this invention relates to treating subjects with a pharmaceutically acceptable dose of compounds, crystals, esters, stereoisomers, enantiomers, salts, hydrates, prodrugs, or mixtures thereof.

BACKGROUND OF THE INVENTION jOOOS] The muitifaceted metabolic syndrome is defined as a number of major metabolic disorders that enhances the risk of cardiovascular disease (CVD) - still the most important cause of death in the Western world -- and type 2 diabetes melHtus. It is also known as the insulin resistance syndrome, syndrome X, dysmetabolic syndrome, or the deadly quartet, and is characterized by aberrations in a wide variety of metabolic risk markers such as hyperinsulinemia, impaired glucose metabolism., elevated plasma leveis of triglycerides, decreased levels of high-density lipoprotein cholesteroi (HDL-C), raised blood pressure, centrally distributed obesity, impaired endothelial and haemostatic function, and a low-grade inflammatory state.

[0004] Type 2 Diabetes Mellitus (T2DM) is characterized by fasting and postprandial hyperglycemia and relative insulin insufficiency. If left untreated, then hyperglycemia may cause long term microvascular and macrovascular complications, such as nephropathy, neuropathy, retinopathy, and atherosclerosis. This disease causes significant morbidity and mortality at considerable expense to patients, their families and society. The incidence of T2DM worldwide is now increasing at more rapid rates in Africa, Asia and South America than in Europe or the U.S. Thus, T2D is now considered worldwide epidemic.

10005] Oxidative stress has long been associated with the late complications of diabetes, and has been implicated in their etiology. The reactive oxygen intermediates, produced in mitochondria, peroxisomes, and the cytosol, are scavenged by cellular defending systems, including enzymatic (ex. superoxide dismutase, glutathione peroxidase GPx, glutathione reductase and cataJase) and nonenzymatic antioxidants (ex. glutathione G-SH, thioredoxin, lipoic acid, ubiquinol, albumin, uric acid, flavonoids, vitamins A, C and E, etc.). Some are located in cell membranes, others in the cytosol, and others in the blood plasma, in hyperglycemia, an altered oxidative metabolism is a consequence either of the chronic exposure to hyperglycaeraia or of the absolute or relative insulin deficit; insulin regulates several reactions involved in oxi do-reductive metabolism. Despite strong experimental evidence indicating that oxidative stress may determine the onset and progression of late-diabetic complications controversy exists about whether the increased oxidative stress is merely associative rather than causal in hyperglycemia.

10006] Managing acute pathology of ofte relies on the addressing underlying pathology and symptoms of the disease. There is currently a need in the art for new compositions to treatment or delay of the onset of diabetes related hyperglycemi and its associated complications progression ,

SU j V.MA.RY OF TH E INVENTION

|0007| The present invention provides compounds, compositions containing these compounds and methods for using the same to treat, prevent and/or ameliorate the effects of the conditions such as hyperglycemia. [0008) The invention herein provides compositions comprising of formula I or pharmaceutical acceptable salts thereof. The invention also provides pharmaceutical compositions comprising one or more compounds of formul ί or intermediates thereof and one or more of pharmaceutically acceptable carriers, vehicles or diluents. These compositions may be used in the treatment of hyperglycemia and its associated complications.

Formula 1

[0009) in certain embodiments, the present invention relates to the compounds and compositions of formula I, or pharmaceutically acceptable salts thereof,

Formula I

Wherein,

R independently represents D, H, methyl, ethyl, C¾CO- or each n is independently 0, 1 , 2, or 3; independently represents H, D, 

a is independently 2,3 or 7;

each b is independently 3, 5 or 6;

e is independently 1 , 2 or 6;

e and d are each independently H, D, -OH, -OD, d-CV.-a kyi, - H? or -COCH,,;

Κ R' , R'\ R' each independentl are selected from the groups consisting of H, D, -OH, -01 methyl, ethyl, -0C¾, -OCI¾ or acetyl .

jOOlOj in the illustrative embodiments, examples of compounds of formula 1 are as set forth below:

(1 -1 )

( 1-2)

(1 -3)

[0011] Herein the application also provides a kit comprising any of the pharmaceutical compositions disclosed herein. The kit may comprise instructions for use in the treatment of hyperglycemia or its related complications.

[0012] The application also discloses a pharmaceutical composition comprising a pharmaceutically acceptable carrier and any of the compositions herein. In some aspects, the pharmaceutical composition is formulated for systemic administration, oral administration, sustained release, parenteral administration, injection, subdemsal administration, or transdermal administration.

[0013] Herein, the application additionally provides kits comprising the pharmaceutical compositions described herein. The kits may further comprise instructions for use in the treatment of hyperglycemia or its related complications.

[0014] The compositions described herein have several uses. The present application provides, for example, methods of treating a patient suffering from hyperglycemia or its related complications manifested from metabolic conditions or disorders, metabolic syndrome, chronic diseases or disorders; HypermsuUnemia, Insulin resistance, Glucose intolerance,. Hepatology, Cancer, Respiratory, Hematological, Orthopedic, Cardiovascular, Renal, Skin, Vascular or Ocular complications. DETAI LED DESCRIPTION OF THE ' INVENTION

Definitions

|0015] As used herein, the following terms and phrases shall have the meanings set forth below. Unless defined otherwise, ail technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art.

|0016 ' ] The compounds of the present invention can be present in the form of pharmaceutically acceptable salts. The compounds of the present invention can also be present in the form of pharmaceutically acceptable esters (i.e., the methyl and ethyl esters of the acids of formula I to be used as prodrugs). The compounds of the present invention can also be solvated, i.e. hydrated. The solvation can be affected in the course of the manufacturing process or can take place i .e. as a consequence of hygroscopic properties of an initially anhydrous compound of formula 1 (hydration).

|0017] Compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed "isomers." Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers," Diastereomers are stereoisomers with opposite configuration at one or more chiral centers which are not enantiomers. Stereoisomers bearing one or more asymmetric centers that are non- superimposable mirror images of each other are termed "enantiomers." When a compound has an asymmetric center, for example, if a carbon atom is bonded to four different groups, a pair of enantiomers is possible. An enantiomer can be characterized by the absolute configuration of its asymmetric center or centers and. is described by the - and S-sequencing rules of Calm, Ingold and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as {+) or (~)~Isomers respectively). A chiral compound can exist as either individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a "racemic mixture" . [0018) As used herein, the term "metabolic conditi n" refers to an inborn errors of metabolism (or genetic metabolic conditions) are genetic disorders that result from a defect i one or more metabolic pathways; specifically, the function of an enzyme is affected and is either deficient or completely absent. 001.91 some embodiments, a molecular conjugate comprises of compounds selected from the group consisting of R-iipoie acid (CAS No, 1200-22-2), salsalate (CAS No. 552-94-3). acetylcysteine (CAS No. 61 -91-.1 ), Eieosapentaenoic acid (CAS No. 10417- 94-4), Docosahexaenoic acid (CAS No. 6217-54-5).

{002 1 The term "polymorph" as used herein is art-recognized and refers to one crystal structure of a given compound.

[0021] The phrases "parenteral administration" and "administered parenteral! " as used herein refer to modes of administration other than enteral and topical administration, such as injections, and include without limitation intravenous, intramuscular, intrapleural, intravascular, intrapericardiai, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intrademial, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intra sternal injection and infusion.

[0022] A "patient,' " "subject," or "host" to be treated by the subject method may mean either a human or non-human animal, such as primates, mammals, and vertebrates.

[0023] The phrase "pharmaceutically acceptable" is art-recognized. In certain embodiments, the term includes compositions, polymers and other materials and/or dosage fonns which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of mammals, human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. |0024| The phrase "pharmaceutically acceptable carrier" is art-recognized, and includes, for example, pharmaceutically acceptable materials, compositions or vehicles, such as a. liquid or solid .filler, diluent, so! ent or encapsulating material involved in carrying or transporting any subject composition, from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must he "acceptable" in the sense of being compatible with the other ingredients of a subject composition and not injurious to the patient, in certain embodiments, a pharmaceutically acceptable carrier is non-pyrogenic. Some examples of materials which may serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carhoxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, com oil and soybean oil; ( 10) glycols, such as propylene glycol; (11) polyols. such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyi lattrate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (.18) Ringer ' s solution; (19) ethyl alcohol; (20) phosphate buffer soiutions; and (21 ) other non-toxic compatible substances employed in pharmaceutical formulations.

[00251 The term "prodrug" is intended to encompass compounds that, under physiological conditions, are converted into the therapeutically active agents of the present invention. A common method for making a prodrug is to include selected moieties that are hydro!yzed under physiological conditions to reveal the desired molecule. In other embodiments, the prodrug is converted by an enzymatic activity of the host animal . j0026 ' | he term "prophylactic or therapeutic" treatment is art-recognized and includes administration to the host of one or more of the subject compositions, if it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, i.e., it protects the host against developing the unwanted condition, whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i .e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).

|0027] The term "predicting" as used herein refers to assessing the probability related diseases patient will suffer from abnormalities or complication and/or terminal platelet aggregation or failure and/or death (i.e. mortality) within a defined time window (predictive window) in the future. The mortality may be caused by the central nervous system or complication. The predictive window is an interval in which the subject will develop one or more of the said complications according to the predicted probability. The predictive window may be the entire remaining lifespan of the subject upon analysis by the method of the present i nvention.

|002S] The term "treating " is art -recognized and includes preventing a disease, disorder or condition from occurring in an animal which may be predisposed to the disease, disorder and/or condition but has not yet been diagnosed as having it; inhibiting the disease, disorder or condition, e.g., impeding its progress; and relieving the disease, disorder, or condition, e.g., causing regression of the disease, disorder and/or condition. Treating the disease or condition includes ameliorating at least one symptom of the particular disease or condition, even if the underlying pathophysiology is not affected, such as treating the hyperglycemia, insulin resistance, diabetes mellitus, diabetes insipidus, type 1 diabetes, type 2 diabetes, microvascular complications, macrovascular complications, lipid disorders, hypertriglyceridemia, cardiovascular complications, and post prandsal hyperglycemia of a subject by administration of an agent even though such agent does not treat the cause of the condition. The term "treating", "treat" or "treatment" as used herein includes curative, preventative (e.g., prophylactic), adjunct and palliative treatment. |0029 The phrase "therapeutically effective amount" is an art-recognized term. In certain embodiments, the term refers to an amount of a salt or composition disclosed herei that produces some desired effect at a reasonable benefit/risk ratio applicable to any medical treatment, in certain embodiments, the term refers to that amount necessary or sufficient to eliminate or reduce medical symptoms for a period of time The effective amount may vary depending on such factors as the disease or condition being treated, the particular targeted constructs being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art may empirically determine the effective amount of a particular composition without necessitating undue experimentation.

1 03 1 In certain embodiments, the pharmaceutical composition described herein are formulated in a manner such that said compositions will be delivered to a patient in a therapeutically effective amount, as part of a prophylactic or therapeutic treatment. The desired amount of the composition to be administered to a patient will depend on absorption, inactivation, and excretion rates of the drug as well as the delivery rate of the salts and compositions from the subject compositions. It is to be noted that dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. Typically, dosing will be determined using techniques known to one skilled in the art.

|0 3i) Additionally, the optimal concentration and/or quantities or amounts of any particular salt or composition may be adjusted to accommodate variations in the treatment parameters. Such treatment parameters include the clinical use to which the preparation is put, e.g., the site treated, the type of patient, e.g., human or non-human, adult or child, and the nature of the disease or condition. [0032] in certain embodiments, the dosage of the subject compositi ns provided herein may be determined by reference to the plasma concentrations of the therapeutic composition or other encapsulated materials. For example, the maximum plasma concentration (Crnax) and the area under the plasma conce tration-time curve from time 0 to infinity may he used.

(0033] When used with respect to a pharmaceutical composition or other material, the term "sustained release" is art-recognized. For example, a subject composition which releases a substance over time may exhibit sustained release characteristics, in contrast to a bolus type administration in which the entire amount of the substance is made biologically available at one time. For example, in particular embodiments, upon contact with body fluids including blood, spinal fluid, mucus secretions, lymph or the like, one or more of the pharmaceutically acceptable excspients may undergo gradual or delayed degradation (e.g., through hydrolysis) with concomitant release of any material incorporated therein, e.g., an therapeutic and/or biologicall active salt and/or composition, for a sustained or extended period (as compared to the release from a bolus). This release may result in prolonged delivery of therapeutically effective amounts of any of the therapeutic agents disclosed herein.

[0034] The phrases "systemic administration," "administered systemicalry," "peripheral administration" and "administered peripherally" are art-recognized, and include the administration of a subject composition, therapeutic or other material at a site remote from the disease being treated. Administration of an agent for the disease being treated, even if the agent is subsequently distributed systemicaily, may be termed "local" or "topical" or "regional" administration, other than directly into the central nervous system, e.g., by subcutaneous administration, such that it enters the patient's system and, thus, i subject to metabolism and other like processes.

[0035] The phrase "therapeutically effective amount" is an art-recognized term. In certain embodiments, the term refers to an amount of a salt or composition disclosed herein that produces some desired effect at a reasonable benefit/risk ratio applicable to any medical treatment, in certain embodiments, the term refers to that amount necessary or sufficient to eliminate or reduce medical symptoms for a period of time. The effective amount ma vary depending on such factors as the disease or condition being treated, the particular targeted constructs being admi n stered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art may empirically determine the effective amount of a particular composition without necessitating undue experimentation. fOO j The present disclosure also contemplates prodrugs of the compositions disclosed herein, as wel l as pharmaceuticall acceptable salts of said prodrugs.

|0037| This application also discloses a pharmaceutical composition comprising a pharmaceutically acceptable carrier and the composition of a compound of Formula. I may be formulated for systemic or topical or oral administration. The pharmaceutical composition may be also formulated for oral administration, oral solution, injection, subdermal administration, or transdermal administration. The pharmaceutical composition may further comprise at least one of a pharmaceutically acceptable stabilizer, diluent, surfactant, filler, binder, and lubricant.

[0038J In many embodiments, the pharmaceutical compositions described herein will incorporate the disclosed compounds and compositions (Formula 1) to be delivered in an amount sufficient to deliver to a patient a therapeutically effective amount of a compound of formula I or composition as part of a prophylactic or therapeutic treatment. The desired concentration of formula I or its pharmaceutical acceptable salts will depend on absorption, inactivation, and excretion rates of the drug as well as the delivery rate of the salts and compositions from the subject, compositions. It is to be noted that dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. Typically, dosing will be determined using techniques known to one skilled in the art.

[0039] Additionally, the optimal concentration and/or quantities or amounts of any particular compoimd of formul Ϊ may be adjusted to accommodate variations in the treatment parameters. Such treatment parameters include the clinical use to which the preparation is put, e.g., the site treated, the type of patient, e.g., human or non-human, adult or child, and the nature of the disease or condition.

[0040] The concentration and/or amount of any compound of formula Ϊ may be readily identified by routine screening in animals, e.g., rats, by screening a range of concentration and/or amounts of the materia! in question using appropriate assays. Known methods axe also available to assay local tissue concentrations, diffusion rates of the salts or compositions, and local blood flow before and after administration of therapeutic formulations disclosed herein. One such method is niicrodialysss, as reviewed by T. E, Robinson et al., 1 91 , microdtaiysis in the oeurosctences, Techniques, volume 7, Chapter . The methods reviewed by Robinson may be applied, in brief as follows. A microdialysis loop is placed in situ in a test animal. Dialysis fluid is pumped through the loop. When compounds with formula 1 suc as those disclosed herein are injected adjacent to the loop, released drugs are collected in the dialysate in proportion, to their local tissue concentrations. The progress of diffusion of the salts or compositions may be determined thereby with suitable calibration procedures using known concentrations of salts or compositions.

[0041 J In certain embodiments, the dosage of the subject compounds of formula 1 provided herein may be determined by reference to the plasma concentrations of the therapeutic composition or other encapsulated materials. For example, the maximum plasma concentration (Cmax) and the area under the plasma concentration-time curve from time 0 to infinity ma be used. [0042] Generally, in carrying out the methods detailed in this application, an effective dosage for the compounds of Formulas I is in the range of about 0.01 mg kg day to about 100 mg/kg/day in single or divided doses, for instance 0.01 mg kg day to about 50 mg kg day in single or divided doses. The compounds of Formulas I may be administered at a dose of, for example, less than 0.2 mg kg day, 0.5 mg kg/day, 1.0 mg/kg/day, 5 mg/kg/day, 10 mg/kg/day, 20 mg/kg/day, 30 mg kg/day, or 40 mg kg day. Compounds of Formula Ϊ may also be administered to a human patient at a dose of, for example, between 0.1 mg and 1000 mg, between 5 mg and 80 mg, or less than .1.0, 9.0. 12.0, 20.0, 50.0, 75.0, 100, 300, 400, 500, 800, 1000, 2000, 5000 mg per day. In certain embodiments, the composiiions herein are administered at an amount that is less than 95%, 90%, 80% 70%, 60%, 50%, 40%, 30%, 20%, or 10% of the compound of formula I required for the same therapeutic benefit. 0043] An effective amount of the compounds of formula 1 described herein refers to the amount of one of said salts or compositions which is capable of inhibiting or preventing a disease.

{ ' 004 J An effective amount may be sufficient to prohibit, treat, alleviate, ameliorate, halt, restrain, slow or reverse the progression, or reduce the severity of a complication resulting from, nerve damage or demyeiization and/or elevated reactive oxidative- nitrosative species and/or abnormalities in physiological homeostasis' s, in patients who are at risk for such complicati ns. As such, these methods include both medical therapeutic (acute) and/or prophylactic (prevention) administration as appropriate The amount and timing of compositions administered will, of course, be dependent on the subject being treated, on the severity of the affliction, on the manner of administration and on the judgment of the prescribing physician. Thus, because of patient-to-patient variability, the dosages given above are a guideline and the physician may titrate doses of the drug to achieve the treatment that the physician considers appropriate for the patient. In considering the degree of treatment desired, the physician must balance a variety of factors such as age of the patient, presence of preexisting disease, as well as presence of other diseases.

|0045J The compositions provided by this application may be administered to a subject in need of treatment by a variety of conventional routes of administration, including orally, topically, parenteraiiy, e.g., intravenously, subcutaneously or intramedullary. Further, the compositions may be administered intranasal! y, as a rectal suppository, or using a "flash" formulation, i.e., allowing the medication to dissolve i the mouth without the need to use water Furthermore, the compositions may be administered to a subject in need of treatment by controlled release dosage forms, site specific drug delivery, transdermal drug delivery, patch (active/passive) mediated drug delivery, by stereotactic injection, or in nanoparticles.

{0046 ' | The compositions may be administered alone or in combination with pharmaceutically acceptable carriers, vehicles or diluents, in either single or multiple doses. Suitable pharmaceutical carriers, vehicles and diluents include inert solid diluents or fillers, sterile aqueous solutions and various organic solvents. The pharmaceutical compositions formed by combining the compositions and the pharmaceutically acceptable carriers, vehicles or diluents are then readily administered in a variety of dosage forms such as tablets, powders, lozenges, syrups, injectable solutions and the like. These pharmaceutical compositions can, if desired, contain additional ingredients such as flavorings, binders, excipients and the like. Thus, for purposes of oral administration, tablets containing various excipients such as L-arginine, sodium citrate, calcium carbonate and calcium phosphate may be employed along with various disintegrates such as starch, alginic acid and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, sodium iauryl sulfate and talc are often useful for tahletting purposes. Solid compositions of a similar type may also be employed as fillers in soft and hard filled gelatin capsules. Appropriate materials for this include lactose or milk sugar and high molecular weight polyethylene glycols. When aqueous suspensions or elixirs are desired for oral administration, the essentia! active ingredient therein may be combined with various sweetening or flavoring agents, coloring matter or dyes and. if desired, emulsifying or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin and combinations thereof. The compounds of formula 1 may also comprise enterically coated comprising of various excipients, as is well known in the pharmaceutical art. 0047J For parenteral administration, solutions of the compositions may be prepared in (for example) sesame or peanut oil, aqueous propylene glycol, or in sterile aqueous solutions may be employed. Such aqueous solutions should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, the sterile aqueous media employed are all readily available by standard techniques known to those skilled in the art. 0048] The formulations, for instance tablets, may contain e.g. 10 to 100, 50 to 250, 150 to 500 mg, or 350 to 800 mg e.g. 10, 50, 100, 300, 500, 700, 800 nig of the compounds of formula 1 disclosed herein, for instance, compounds of formula 1 or pharmaceutical acceptable salts of a compounds of Formula 1

[0049) Generally, a composition as described herein may be administered orally, or parenierally (e.g., intravenous, intramuscular, subcutaneous or intramedullary). Topical administration ma also be indicated, for example, where the patient is suffering from gastrointestinal disorder that prevent oral administration., or whenever the medication is best applied to the surface of a tissue or organ as determined by the attending physician. Localized administration may also he indicated, for example, when a high dose is desired at the target tissue or organ. For buccal administration the active composition, may take the form of tabl ets or lozenges formulated in a conventional manner. |0050 The dosage administered will be dependent upon the identity of the metabolic disease; the type of .host involved, including its age, health and weight; the kind of concurrent treatment, if any, the frequency of treatment, and therapeutic ratio.

[0051] illustratively, dosage levels of the administered active ingredients are: intravenous, 0.1 to about 200 mg kg; .intramuscular, 1 to about 500 mg kg; orally, 5 to about 1000 mg/kg; intranasal instillation, 5 to about 1000 mg/kg; and aerosol, 5 to about 1 00 mg/kg of host body weight.

[0052] Expressed in terms of concentration, an active ingredient can be present in the compositions of the present invention for localized use about the cutis, intranasaiiy, pharyngolaryngeaily, bronchi ally, intra vaginally, rectaliy, or ocularly in a concentration of from about 0.01 to about 50% w/w of the composition; preferably about 1 to about 20% w/w of the composition; and for parenteral use in a concentration of from about 0.05 to about 50% w/v of the composition and preferably from about 5 to about 20% w/v.

[0053] The compositions of the present invention are preferably presented for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, suppositories, sterile parenteral solutions or suspensions, sterile non-parenterai solutions of suspensions, and oral solutions or suspensions and the like, containing suitable quantities of an active ingredient. For oral administration either solid or fluid unit dosage forms can be prepared.

[0054] As discussed above, the tablet core contains one or more I dropliilic polymers. Suitable hydrophilic polymers include, but are not limited to, water swellable cellulose derivatives, poly alky lene glycols, thermoplastic polyalkylene oxides, acrylic polymers, hydrocolloids., clays, gelling starches, swelling cross-linked polymers, and mixtures thereof. Examples of suitable water swellable cellulose derivatives include, but are not limited to, sodium carboxymethylcellulose, cross-linked hydroxypropylcellulose, hydroxy propyl eel lul ose (HPC), h droxypropy Imethylcel 1 ul ose (HP C), h droxy isopropylcellul ose, hy droxy buty ! ce!iul ose, hydroxyphenylcellulose, hydroxy ethyl cell utose (HEC), hydroxypentylcellulose, hydroxypropylethy lcell u!ose, hy droxy propylbutylcel lulose, and hydroxypropylethylcellulose, and mixtures thereof. Examples of suitable polyalkylene glycols include, but are not limited to, polyethylene glycol. Examples of suitable thermoplastic polyalkylene oxides include, but are not. limited to, fx>Sy(ethylerie oxide). .Examples of suitable acrylic polymers include, but are not limited to, potassium methacrylatedi vinylbenzene copolymer, polymethylmethacrylate, high-molecular weight crossl inked acrylic acid homopolymers and copolymers such as those commercially available from Noveon Chemicals under the tradename CARBOPOL tM . Examples of suitable hydrocolloids include, but are not limited to, alginates, agar, guar gum, locust bean gum, kappa carrageenan, iota carrageenan, tara, gum arabic, tragacanth, pectin, xanthan gum, gel I an gum, maltodextrin, galactomannan, pusstulan, laminarin, sclerogiucan, gum arabic, inulin, pectin, gelatin, whelan, rhamsan, zooglan, methylan, chitin, cyclodextrin, chitosan, and mixtures thereof. Examples of suitable clays include, but are not limited to, smectites such as bentonite, kaolin, and laponite; magnesium trisilicate; magnesium aluminum silicate; and mixtures thereof. Examples of suitable gelling starches include, but are not limited to, acid hydrolyzed starches, swelling starches such as sodium starch glycolate and derivatives thereof and mixtures thereof. Examples of suitable swelling cross-linked polymers include, but are not limited to, cross-linked polyvinyl pyrrol idone, cross-linked agar, and cross-linked carboxymethylcellu!ose sodium, and mixtures thereof.

[0055] The carrier may contain one or more suitable excipients for ihe formulation of tablets. Examples of suitable excipients include, but are not limited to, fillers, adsorbents, binders, disintegrants, lubricants, giidanis, release-modifying excipients, superdisintegrants, antioxidants, and mixtures thereof.

[0056] Suitable binders include, but are not limited to, dry binders such as polyvinyl pyrroli done and hydroxypropylmethylcellulose; wet binders such as water-soluble polymers, including hydrocolloids such as acacia, alginates, agar, guar gum, locust bean, carrageenan, carboxymethyicellulose, tara, gum arabic, tragacanth. pectin, xanthan. gellan, gelatin, maltodextrin, galactomannao, pusstulan, laminarin, scleroglucari., inuHn, whelan, rhamsan, zooglan, methylan. chitin, cyclodextrin. chiiosan, polyvinyl pyrrolidone, cellulosics, sucrose, and starches; and mixtures thereof. Suitable disintegrants include, but are not limited to, sodium starch gl col ate, cross-linked poly vin l pyrrolidone, cross-linked carboxymethyleellulose, starches, macrocrystalline cellulose, and mixtures thereof.

|0057| Suitable lubricants include, but are not limited to. long chain fatty acids and their salts, such as magnesium stearate and stearic acid, talc, glycerides waxes, and mixtures thereof. Suitable gli.dan.ts include, but are not limited to, colloidal silicon dioxide. Suitable release-modifying excipients include, but are not limited to, insoluble edible materials, pH-dependent polymers, and mixtures thereof,

|0058J Suitable insoluble edible materials for use as release-modifying excipients include, but are not limited to, water-insoluble polymers and low-melting hydrophobic materials, copolymers thereof ' and mixtures thereof. Examples of suitable water- insoluble polymers include, but are not limited to, efhylceilulose, polyvinyl alcohols, polyvinyl acetate, polycaprolactones, cellulose acetate and its derivatives, acrylates, methacryiat.es, acrylic acid copolymers, copolymers thereof and mixtures thereof. Suitable l w-melting hydrophobic materials include, but are not limited to, fats, fatty acid esters, phospholipids, waxes, and mixtures thereof. Examples of suitable fats include, but are not limited to, hydrogenated vegetable oils such as for example cocoa butter, hydrogenated palm kernel oil, hydrogenated cottonseed oil, hydrogenated sunflower oil, and hydrogenated soybean oil, free fatty acids and their salts, and mixtures thereof. Examples of suitable fatty acid esters include, but are not limited to, sucrose fatty acid esters, mono-, di~, and triglycerides, glyceryl behenate, glyceryl pa!mitostearate, glyceryl moiiosiearate, glyceryl tri stearate, glyceryl trilaur late, glyceryl myiistate, GlycoWax- 932, lauroyl macrogol-32 glycerides, stearoyl macrogol-32 glycerides, and mixtures thereof. Examples of suitable phospholipids include phosphatidyl choline, phosphotidyl serene, phosphotidyl enositoi, phosphotidic acid, and mixtures thereof. Examples of suitable waxes include, but are not limited to, carnauba wax, spermaceti wax, beeswax, candeJilla wax, shellac wax, macrocrystalline wax, and paraffin wax; fat-containing mixtures such as chocolate, and mixtures thereof. Examples of super disintegrates include, but are not limited to, croscarmeilose sodium, sodium starch glycolate and cross- linked povidone (erospovidone). in one embodiment the tablet, core contains up to abou 5 percent by weight of such super disi tegrant.

[0059J Examples of antioxidants include, but are not limited to, tocopherols, ascorbic acid, sodium pyrosulfite, butylhydroxytoluene, buiylated hydroxyanisole, edetic acid, and edetate salts, and mixtures thereof. Examples of preservati ves include, but are not limited to, citric acid, tartaric acid, lactic acid, malic acid, acetic acid, benzoic acid, and sorbic acid, and mixtures thereof. 0060] In one embodiment, the immedi ate release coating has an average thickness of at least 50 microns, such as from about 50 microns to about 2500 microns, e.g., from about 250 microns to about 1000 microns, in embodiment, the immediate release coating is typicall compressed at a density of more than about 0.9 g/cc, as measured by the weight and volume of that specific layer.

1006:1 ] in one embodiment, the immediate release coating contains a first portion and second portion, wherein at least one of the portions contains the second pharmaceutically active agent. In one embodiment, the portions contact each other at a center axis of the tablet. In one embodiment, the first portion includes the first pharmaceutically active agent and the second portion includes the second pharmaceutically active agent

|0062] In one embodiment, the first portion contains the first pharmaceutically active agent and the second portion contains the second pharmaceutically active agent. In one embodinieni, one of the portions contains a third pharmaceutically active agent. In one embodiment one of the portions contains a second immediate release portion of the same pharmaceutically active agent as that contained in the tablet core. |0O63J in one embodiment, the outer coating portion is prepared as a dry blend of materials prior to addition to the coated tablet core. In another embodiment the outer coating portion is included of a dried granulation including the pharmaceutically active agent,

|0064] Formulations with different drug release mechanisms described above could be combined in a final dosage form containing single or multiple units. ' Examples of multiple units include multilayer tablets, capsules containing tablets, beads, or granules in a solid or liquid form. Typical, immediate release formulations include compressed tablets, gels, films, coatings, liquids and particles thai can be encapsulated, for example, in a gelatin capsule, Many methods for preparing coatings, covering or incorporating drugs, are known in the art

100651 The immediate release dosage, unit of the dosage form, i.e., a tablet, a plurality of drug-containing beads, granules or particles, or an outer layer of a coated core dosage form, contains a therapeutically effective quantity of the active agent with conventional pharmaceutical excipients. The immediate release dosage unit may or may not be coated, and may or may not be admixed with the delayed release dosage unit or units (as in an encapsulated mixture of immediate release drug-containing granules, particles or beads and delayed release drug-containing granules or beads).

10066) Extended release formulations are generally prepared as diffusion or osmotic systems, for example, as described in "Remingto— The Science and Practice of Pharmacy *\ 20th. Ed.. Lippiiicott Williams & Wilkins, Baltimore, Md., 2000). A. diffusion system typically consists of one of two types o devices, reservoir and matrix, which are wellknown and described in die art. The matrix devices are generally prepared by compressing the drug with a slowly dissolving polymer carrier into a tablet form. [0067] An immediate release portion can be added to the extended release system by means of either applying an immediate release layer on top of the extended release core; using coating or compression processes or in a multiple unit system such as a capsule containing extended and immediate release beads.

[0068] Delayed release dosage formulations are created by coating a solid dosage form with a film of a polymer which is insoluble in the acid environment of the stomach, but soluble in the neutral environment of small intestines. The delayed release dosage units can be prepared, for example, by coating a drug or a drug-containing composition with a selected coating material . The drug-containing composition may be a tablet for incorporation into a capsule, a tablet for use as an inner core in a "coated core" dosage form, or a plurality of drug-containing beads, particles or granules, for incorporation into either a tablet or capsule.

|0069 ' | A pulsed release dosage form is one that mimics a multiple dosing profile without repeated dosing and typically allows at least a twofold reduction in dosi ng frequency as compared to the drug presented as a conventional dosag form (e.g., as a solution or prompt drug-releasing, conventional solid dosage form). A pulsed release profile is characterized by a time period of no release (lag time) or reduced release followed by rapid drug release.

[0070] Each dosage form contains a therapeutically effective amount of active agent. In one embodiment of dosage forms that mimic a twice daily dosing profile, approximately 30 wt. % to 70 wt. %, preferably 40 wt. % to 60 wt. %., of the total amount of active agent in the dosage form is released in the initial pulse, and., correspondingly approximately 70 wt. % to 3.0 wt %„ preferably 60 wt. % to 40 wt. %, of the total amount of active agent in the dosase form is released in the second pulse. For dosase forms mimickina the twice daily dosing profile, the second pulse is preferably released approximately 3 hours to less than 14 hours, and more preferably approximately 5 hours to 12 hours, following administration. |0071 J Another dosage form contains a compressed tablet or a capsule having a drug- containing immediate release dosage unit, a delayed release dosage unit and an optional second delayed release dosage unit. In this dosage form, the immediate release dosage unit contains a plurality of beads, granules particles that release drug substantially immediately following oral administration to provide an initial dose. The deiayed release dosage unit contains a plurality of coated beads or granules, which release drug approximately 3 hours to 14 hours following oral administration to provide a second dose.

|0072| For purposes of transdermal (e.g., topical ) administration, dilute sterile, aqueous or partially aqueous solutions (usually in about 0.1% to 5% concentration), otherwise similar to the above parenteral solutions, may be prepared,

|0073] Methods of preparing various pharmaceutical compositions with a certain amount of one or more compounds of formula I or other active agents are known, or will be apparent in Sight of this disclosure, to those skilled in this an. For examples of methods of preparing pharmaceutical compositions, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 1 th Edition (1 95). j0074j In addition, in certain embodiments, subject compositions of the present application maybe lyophilized or subjected to another appropriate drying technique such as spray drying. The subject compositions may be administered once, or may be divided into a number of smaller doses to be administered at varying intervals of time, depending in part on the release rate of the compositions and the desired dosage.

|0075] Formulations useful in the methods provided herein include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol 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 a subject composition which may be combined with a carrier material to produce a single dose ma vary depending upon the subject being treated, and the particular mode of administration.

[0076] Methods of preparing these formulations or compositions include the step of bringing into association subject compositions with the carrier and, optionally, one or more accessory ingredients in general , the formulations are prepared by uniformly and intimately bringing into association a subject composition with liquid carriers, or finely di vided solid carriers, or both, and then, if necessary, shaping the product.

[0077] The compounds of formula .1 described herein may be administered in inhalant or aerosol formulations. The inhalant or aerosoi formulations may comprise one or more agents, such as adjuvants, diagnostic agents, imaging agents, or therapeutic agents useful in inhalatio iherapy. The final aerosol formulation may for example contain 0.005-90% w/w, for instance 0.005-50%, 0.005-5% w/w, or 0.0 Ϊ- 1.0% w/w, of medicament relative to the total weight of the formul tion.

[0078] to solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the subject composition is mixed with one or more pharmaceutically acceptable carriers and/or an of the following: (I) fillers or extenders, such as starches, lactose, sucrose, glucose, rnannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethyiceliulose, alginates, gelatin, polyvinyl pyrroiidone, sucrose and or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (?) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium steai ' aie, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. n 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 lactose or milk sugars, as well as .high molecular weight polyethylene glycols and the like.

[0079J Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsioris, solutions, suspensions, syrups and elixirs. In addition to the subject com positions, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example,, water or other solvents, solubilizing agents and ernuis.ifse.rs, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, corn, peanut, sunflower, soybean, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. 0080) Suspensions, in addition to the subject compositions, may contain suspending agents such as, for example, ethoxy Sated isostearyl alcohols, polyoxyethylene sorbitol, and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar- agar and tragacanth, and mixtures thereof.

[0081 ) Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing a subject composition with one or .more suitable non-irritating carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax, or a salicylate, and which is solid at room tem erature, but liquid at bod temperature and, therefore, will melt in the appropriate body cavity and release the encapsulated corapound(s) and composition^). Formulations which are suitable for vaginal admini tration also include pessaries, tampons, creams, gels, pastes, foams, or spray formulations containing such carriers as are known in the art to be appropriate.

|0082) Dosage forms for transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and inhalants. A subject composition may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required. For transdermal administration, the complexes may include lipophilic and hydrophilic groups to achieve the desired water solubilit and transport properties.

[0083] The ointments, pastes, creams and gels may contain, in addition to subject compositions, other carriers, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Powders and spra s may contain, in addition to a subject composition, exci ients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and po!yamide powder, or mixtures of such substances. Sprays may additionally contain customary propellants, such as chiorofluorohydrocarbons arid volatile unsubstituted hydrocarbons, such as butane and propane.

[0084] Methods of delivering a composition or compositions via a transdermal patch are known in the art. Exemplary patches and methods of patch deliver are described in US Patent Nos. 6,974,588, 6,564,093, 6,312,716, 6,440,454, 6,267,983, 6,239,180, and 6,103,275.

[0085] In another embodiment, a transdermal patch may comprise: a substrate sheet comprising a composite film formed of a resin composition comprising 100 parts by weight of a polyvinyl chlonde-po!yurethane composite and 2-10 parts by weight, of a styrene-ethylene-butylene-styrene copolymer, a first adhesive layer on the one side of the composite film, and a polyalkylene terephthaiaie film adhered to the one side of the composite film by means of the first adhesive layer, a primer layer which comprises a saturated polyester resin and is formed on the surface of the polyalkylene tereph thai ate film; and a second adhesive Iayer comprising a siyrene-diene-styrene block copolymer containing a pharmaceutical agent layered on the primer layer. A method for the manufacture of the above-mentioned substrate sheet comprises preparing the above resin composition molding the resin composition into a composite film by a calendar process, and then adhering a polyalkyiene terephthalate film on one side of the composite Film by means of an adhesive layer thereby forming the substrate sheet, and forming a primer layer comprising a saturated polyester resin on the outer surface of the polyalkyiene terephthalate film.

|0086j Another type of patch comprises incorporating the drug directly in a pharmaceutically acceptable adhesive and laminating the drug-containing adhesive onto a suitable backing member, e.g. a polyester backing membrane. The drug should be present at a concentration which will not affect the adhesive properties, and at the same time deliver the required clinical dose.

|008T| Transdermal patches may be passive or active. Passive transdermal drug delivery systems currently available, such as the nicotine, estrogen and nitroglycerine patches, deliver small -molecule drugs. Many of the newly developed proteins and peptide drugs are too large to be delivered through passive transdermal patches and may be delivered using technology such as electrical assist (iontophoresis) for large-molecule drugs.

|0088) Iontophoresis is a technique employed for enhancing the .flux of ionized substances through membranes by application, of electric current. One example of an iontophoretic membrane is given, in U.S. Pat. No. 5,080,646 to Theeuwes. The principal mechanisms by which iontophoresis enhances molecular transport across the skin are (a) repelling a charged ion from an electrode of the same charge, (b) electroos.mos.is, the convective movement of solvent that occurs through a charged pore in response the preferential passage of counter-ions when an electric Field is applied or (c) increase skin permeability due to application of electrical current. 0089| In some cases, it may be desirable to administer in the form of a kit, it may comprise a container for containin the separate compositions such as a divided bottle or a divided foil packet Typically the kit comprises directions for the administration of the separate components. The kit farm is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.

|0090] An example of such a kit is a so-called blister pack. Blister packs are well known in the packaging industry and are widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively sti f material covered with a foil of a plastic material that may be transparent. 0 9IJ Methods and compositions for the treatment of hyperglycemia. Among other things, herein is provided a method of treating hyperglycemia, comprising administering to a patient in need thereof a therapeutically effective amount of compound of Formula I:

Formula I.

R independently represents D, H, methyl, ethyl, C¾CO- or each n is independently 0,1, 2, or 3;

R" independently represents H, D, 33

R independently represents 



a is independently 2,3 or 7;

each b is independentl 3, 5 or 6;

e is independently 1, 2 or 6;

c and d are each independently H, D, -OH, -OD, CpCc-alkyl, - H 2 or -COC¾;

Κ R', R'\ R' each i dependentl are selected from the groups consisti g of H, D, -OH, -OD, methyl, ethyl, -OC¾, -OCD 3 or acetyl.

Me { ods for using compounds of formula I: 0092] The invention also includes methods for treating hyperglycemia, insulin resistance, diabetes mellitus, diabetes insipidus, type i diabetes, type 2 diabetes, microvascular complications, macrovascular complications, lipid disorders, prediabetes, obesity, arrhythmia, myocardial infarction, stroke, neuropathy, renal complications, hypertriglyceridemi , cardiovascular complications, and post prandial hyperglycemia.

METHODS OF MAKING

J0093J Examples of synthetic pathways useful for making compounds of formula I are set forth in example below and generalized in scheme 1 : Scheme-1 :

K 2 co 3

[0094] Step- ! : Synthesis of compound 2:

2 |0095 L1AIH 4 (3.5 eq) was added in portions to a cooled (0 °C) and dry solution of compound I in THF (0.15 ). The reaction mixture was stirred for 20 hours, allowing it to warm to rt. The excess LiAlH* was quenched with water at 0 °C. The mixture was diluted with EtOAc and washed with sat. aq NH4CI (3 X ). The organic phase was dried (MgSO. ) and concentrated. The resulting product (2) was used crude in the next reaction. A small sample was purified by silica gel column chromatography (20%~~ 50% EtOAc in PE) for characterization purposes to provide 2 as a color! ess oil.

(0096 ' | Step-2: Synthesis of compound 4:

|0097J A solution of oxaiyl chloride (4 eq) in DCM. (1 M) was cooled to -78 °C. After dropwise addition of a solution of DMSO (5 eq) in DCM (2 M) over 10 minutes, the reaction mixture was stirred for 40 minutes while being kept below -70 °C. Next, a dry solution of the glucitol 2 intermediate from above reaction in DCM (0.5 M) was added dropwise to the reaction mixture over a 15 minutes period, while keeping the reaction mixture below -70 °C. After stirring the reaction mixture for 2 hours below -65 °C, EhN (12 eq) was added dropwise over a 10 minute period, while keeping the reaction mixture below -65 "C. After addition, the reaction mixture was allowed to warm to -5 °C over 2 hours.

|0098] The Swern reaction mixture was concentrated at a moderate temperature ( -30 °C) with simultaneous coevaporation with toluene (3 > ). The residue was dissolved in MeOH (0.05 M relative to starting compound) and NH4HCO 2 (20 eq) was added. The mixture was cooled to 0 °C and stirred until all NH 4 HCO? had dissolved, Activated 3A molsieves (10 g/romol ) were added and reaction mixture was stirred for 20 minutes, after which NaB¾CN (4 eq) was added. The reaction mixture was kept at 0 °C for one hour after which the cooling source was removed and the reaction was stirred for an additional 20 hours. Alter removal of the mol sieves over a glass microfibre filter, the filtrate was concentrated, dissolved in EtOAc and washed with sat aq NaHCO.». The aqueous phase was back-extracted with EtOAc (3*) and the combined organic layers were dried (MgSO. ) and concentrated. The resulting residue was purified by silica gel column chromatography (20%~ 75% EtOAc in PE) to provide 4 (9.932 g, 18.98 mmol) as a light yello crystalline solid.

{00991 Step-

4 5

[00100} A solution of 4 (.1.06 g, i .91 mmol) in EtOH (50 ml,} was acidified to pB -2 with i aq HC! and purged of oxygen by bubbling argon through the solution for 15 minutes. Pd/C (10 wt , 100 mg) was added and the mixture was exposed to 4 bar of hydrogen for 20 hours. The reaction mixture was filtered over a glass microfiber filter and the filter cake was rinsed successively with MeOH (4x20 niL) and i¾0 (2 20 niL). The combined filtrate was concentrated and coevaporated. with MeOH (3 - 50 ra.L). The residue was purified by column chromatography with aluminum oxide (isocraiic 16:3.7:0.3, w-propanol : H2O.-NH4OH) to provide 5 (290 mg, 1.78 mmol) in 93% yield as a white foam.

{00101} Step-4: Sy nthesi s of compound 7:

5 6

? (00102} A. dry suspension of potassium carbonate (7.2 mmol), 5 (4.8 rnmol) and 1,3- dic oropropane (5.3 romolin DMF was heated to 90 °C for 5 h. The reaction mixture was filtered over a gias mierofibre filter and the filtrate was concentrated. The residue was purified by silicagel column chromatography (MeQH in CHCIj + 1% NH 4 OH) to obtain the ntermediate 7.

(00103} Step-5: Synthesis of compound 9:

(00104} In a B flask the acid 8 (1.2 mmol) & anhydrous K2CO (1.1 mmol) was taken in dry DMF (10 vol) stir at room temperature for 30 min and then cooled to -10 °C, compound 7 was added slowiy drop wise & then allowed to stir at room temperature for 12 h. Reaction was monitored by TLC. On completion of the reaction, the reaction mixture was poured into water (10 mL) and extracted with ethyl acetate (2x5 mi). The combined organic layers were washed with water (2 x 5mL) followed by brine solution (10 m ' L), dried over anhydrous Na2$0 4 nd evaporated under reduced pressure. The crude was purified by column chromatography over 100-200 mesh silica gel to obtain the final product 9.

EXAMPLES

(0 1051 Ϊ he Solubility of the Compound of Formula Ϊ (1-3) in Water was Compared with that of ekosapentaenoic acid (EPA) [001 6} Measurement of the water solubility of the test compounds is accomplished by using methods well known to those skilled in the art. Specifically, to a weighed amount of the test, compound of the example compound Formula I ( 1 -3) distilled water was added in small proportions until a clear solution was obtained. The total volume of the solution is measured. The water solubility is calculated by dividing the weight of the salt, in milligrams (nig), by the volume of the solution, in mi... The water solubility of the compound of formula 1 (1 -3) when measured using the above technique, was determined to be 68.6 mg/ml. Likewise, the water solubility of EPA was found to be <0.2 mg mL. The compound of Formula I (1-3) is therefore, at least 260 times more soluble in water than EPA itself. This is a clear indication of an unexpectedly high degree of bioavailability of the compositions of the invention. Highly water soluble medicinal preparations, when administered orally, result in efficient absorption from the gastrointestinal tract in to systemic circulation. Furthermore, water soluble preparations are especially suitable for parenteral administration.

[00107) The term "sample" refers to a sample of a body fluid, to a sample of separated cel ls or to a sample from a tissue or an organ. Samples of body fluids can be obtained by well known techniques and include, preferably, samples of blood, piasma, serum, or urine, more preferably, samples of blood, plasma or serum.

EQUIVALENTS

[00.1.08] The present disclosure provides among other things compositions and methods for treating hyperglycemia and their complications. While specific embodiments of the subject disclosure have been discussed, the above specification is illustrative and not. .restrictive.

INCORPORATION BY REFERENCE

{001 9j All publications and patents mentioned herein, including those items listed above, are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by- reference, hi case of conflict, the present application, including any definitions herein, will control.