WO2010065883A2 - Method of treating an overweight or obese subject - Google Patents

Method of treating an overweight or obese subject Download PDF

Info

Publication number
WO2010065883A2
WO2010065883A2 PCT/US2009/066816 US2009066816W WO2010065883A2 WO 2010065883 A2 WO2010065883 A2 WO 2010065883A2 US 2009066816 W US2009066816 W US 2009066816W WO 2010065883 A2 WO2010065883 A2 WO 2010065883A2
Authority
WO
WIPO (PCT)
Prior art keywords
patient
metap2 inhibitor
administering
metap2
inhibitor
Prior art date
Application number
PCT/US2009/066816
Other languages
French (fr)
Inventor
Thomas E. Hughes
James E. Vath
Original Assignee
Zafgen Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zafgen Corporation filed Critical Zafgen Corporation
Priority to US13/133,060 priority Critical patent/US8642650B2/en
Publication of WO2010065883A2 publication Critical patent/WO2010065883A2/en
Priority to US13/568,800 priority patent/US8367721B2/en
Priority to US14/141,910 priority patent/US20150209320A1/en
Priority to US15/492,294 priority patent/US20180064677A1/en
Priority to US16/113,436 priority patent/US20190224156A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/336Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having three-membered rings, e.g. oxirane, fumagillin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4015Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having oxo groups directly attached to the heterocyclic ring, e.g. piracetam, ethosuximide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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

Definitions

  • Obesity is a complex medical disorder of appetite regulation and metabolism resulting in excessive accumulation of adipose tissue mass.
  • BMI body mass index
  • obesity is a world-wide public health concern that is associated with cardiovascular disease, diabetes, certain cancers, respiratory complications, osteoarthritis, gallbladder disease, decreased life expectancy, and work disability.
  • the primary goals of obesity therapy are to reduce excess body weight, improve or prevent obesity-related morbidity and mortality, and maintain long-term weight loss.
  • Treatment modalities typically include lifestyle management, pharmacotherapy, and surgery. Treatment decisions are made based on severity of obesity, seriousness of associated medical conditions, patient risk status, and patient expectations. Notable improvements in cardiovascular risk and the incidence of diabetes have been observed with weight loss of 5-10% of body weight, supporting clinical guidelines for the treatment of obesity that recommend a target threshold of 10% reduction in body weight from baseline values.
  • prescription anti-obesity medications are typically considered for selected patients at increased medical risk because of their weight and for whom lifestyle modifications (diet restriction, physical activity, and behavior therapy) alone have failed to produce durable weight loss, approved drugs have had unsatisfactory efficacy for severely obese subjects, leading to only -3-5% reduction in body weight after a year of treatment.
  • Bariatric surgery may be considered as a weight loss intervention for patients at or exceeding a BMI of 40 kg/m 2 . Patients with a BMI > 35 kg/m 2 and an associated serious medical condition are also candidates for this treatment option.
  • postoperative complications commonly result from bariatric surgical procedures, including bleeding, embolism or thrombosis, wound complications, deep infections, pulmonary complications, and gastrointestinal obstruction; reoperation during the postoperative period is sometimes necessary to address these complications. Rates of reoperation or conversion surgery beyond the postoperative period depend on the type of bariatric procedure, and in one study ranged from 17% to 31%.
  • Intestinal absorptive abnormalities such as micronutrient deficiency and protein- calorie malnutrition
  • bypass procedures requiring lifelong nutrient supplementation.
  • Major and serious adverse outcomes associated with bariatric surgery are common, observed in approximately 4 percent of procedures performed (including death in 0.3 to 2 percent of all patients receiving laparoscopic banding or bypass surgeries, respectively)
  • MetAP2 encodes a protein that functions at least in part by enzymatically removing the amino terminal methionine residue from certain newly translated proteins such as glyceraldehyde-3- phosphate dehydrogenase (Warder et al. (2008) J Proteome Res 7:4807). Increased expression of the MetAP2 gene has been historically associated with various forms of cancer. Molecules inhibiting the enzymatic activity of MetAP2 have been identified and have been explored for their utility in the treatment of various tumor types (Wang et al. (2003) Cancer Res. 63:7861) and infectious diseases such as microsporidiosis, leishmaniasis, and malaria (Zhang et al. (2002) J. Biomed. Sci.
  • MetAP2 inhibitors may be useful as well for patients with excess adiposity and conditions related to adiposity including type 2 diabetes, hepatic steatosis, and cardiovascular disease (via e.g. ameliorating insulin resistance, reducing hepatic lipid content, and reducing cardiac workload). Methods of treating obese subjects that are more effective than e.g. dieting alone are clearly needed.
  • This disclosure generally relates to methods of treating an overweight or obese subject or patient that include non-parenterally administering a pharmaceutically effective amount of a MetAP2 inhibitor to a patient in need thereof, e.g., a human or a companion animal such as a cat or a dog.
  • a method of treating obesity in a patient in need thereof comprising non-parenterally administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • Such methods may result in, for example, a lower systemic exposure to said MetAP2 inhibitor as compared to a patient parenterally administered the same of amount of the MetAP2 inhibitor.
  • Contemplated pharmaceutically effective amounts may not substantially modulate or suppress angiogenesis.
  • non-parenteral administration may result in decreased body fat and a substantial maintenance of muscle mass in said patient.
  • Such methods may enhance fat oxidation compared to a patient on a restricted food intake diet alone.
  • non-parenterally administration may include oral, buccal, sublingual, transdermal, rectal, nasal administration, or administration via inhalation.
  • Contemplated MetAP2 inhibitors include substantially irreversible inhibitors, such as e.g., fumagillin, fumagillol or fumagillin ketone derivative, siRNA, shRNA, an antibody, or a antisense compound, or, e.g., O-(4-dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof.
  • a contemplated MetAP2 inhibitor may be substantially reversible inhibitor.
  • Also provided herein is a method for controlling or preventing hepatic steatosis in an obese patient being treated for obesity, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient, and/or a method for improving liver function in an obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a method of improving exercise capacity in a patient in need thereof includes administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a method of reducing weight of a patient in a patient in need thereof comprises administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient wherein the metabolic rate of the patient is not substantially reduced as compared to the metabolic rate of a diet only patient on an energy restricted diet alone. Also provided herein is a method of restoring normal metabolic action in an obese patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a method of decreasing body fat in an overweight or obese patient in need thereof comprises administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient resulting in body fat reduction, and wherein said patient substantially maintains muscle mass during the body fat reduction, for example a patient may retain substantially more muscle mass as compared to body fat reduction in a patient using an energy restricted diet alone.
  • a method of activating brown fat function and/or increasing brown fat tissue mass in a patient in need thereof comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient, and/or a method of restoring and/or maintaining thyroid hormone concentrations in an obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • the disclosed methods may include a pharmaceutically effective amount of a
  • MetAP2 inhibitor that does not substantially modulate or suppress angiogenesis in a treated patient.
  • a patient has a lower systemic exposure to said MetAP2 inhibitor as compared to a patient parenterally administered the same amount of the MetAP2 inhibitor.
  • MetAP2 inhibitors may be administered non-parenterally, orally, buccally or sublingually, topically, rectally, or transdermally in at least some disclosed methods.
  • a MetAP2 inhibitor may be administered subcutaneously or intravenously.
  • administration of a MetAP2 inhibitor may occur at least daily, or every other day, or at least weekly.
  • Also contemplated herein is a method for reducing the amount or frequency of administering supplemental insulin in a patient suffering from type 2 diabetes, comprising a pharmaceutically effective amount of a MetAP2 inhibitor.
  • a method for improving surgical outcome in an obese patient in need thereof, by reducing weight of said patient comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient before non-acute surgery, thereby reducing liver and/or abdominal fat in said patient and improving surgical outcome.
  • Such surgeries may include e.g., bariatric surgery, cardiovascular surgery, abdominal surgery, or orthopedic surgery.
  • a method of maintaining a specified weight in a formerly obese patient comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • Disclosed methods may further comprise co-administering an additional weight loss agent and/or may further comprise administering a food restricted diet to a patient.
  • a disclosed method may further comprise assessing the ketone body production level in a patient; and optionally adjusting the amount administered; thereby optimizing the therapeutic efficacy of said MetAP2 inhibitor.
  • a patient may incur greater than or about equal to a 20% weight loss after about 6 months of said administration.
  • a method for treating obesity in a patient in need thereof comprising administering about 0.005 to about 0.04 mg/kg of a MetAP2 inhibitor selected from O-(4-dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof, for example, an oxalate salt, to said patient.
  • This MetAP2 inhibitor may be administered at least daily, or 1, 2, 3 or 4 times a week.
  • MetAP2 may be administered parenterally, e.g., intravenously, or non-parenterally.
  • Upon administration of the MetAP2 inhibitor e.g. daily, or 1, 2, 3, 4, 5, 6 or 7 times a week, for about 6 months, may result in at least a 20% weight loss or more of the patient's original weight.
  • FIG. 1 is a bar graph showing TXN(104) in B 16F10 cell lysate after chymotryptic digest.
  • FIG. 2 is a bar graph showing N-acetyl TXN(I -4) in B 16F10 cell lysate after chymotriptic digest.
  • FIG. 3 is a photograph of a gel showing that fumagillin significantly increased
  • FIG. 4 is a photograph of a gel showing that EC50 of fumagillin to increase
  • TXN levels is ⁇ 10 nM.
  • FIG. 5A is a bar graph showing the concentration of beta-hydroxybutyrate in mice fed a high fat diet (vehicle) or mice fed a high-fat diet plus oral administration of fumagillin for ten days at the concentrations indicated.
  • FIG. 5B is a bar graph showing the concentration of nonesterified fatty acid concentrations under the conditions described for FIG. 5A.
  • FIG. 5C is a bar graph showing the concentration of beta-hydroxybutyrate in mice fed a high fat diet (vehicle) or mice fed a high-fat diet plus oral administration of fumagillin for 250 days at the concentrations indicated.
  • FIG. 6 is a line graph showing the body weight over time of high-fat diet- fed
  • FIG. 7 shows age-matched lean mice maintained on normal mouse chow diet
  • mice fed a high fat diet-fed and treated with 1 mg/kg fumagillin (right) or vehicle (middle) for 250 days.
  • FIG. 8 contains two bar graphs showing a comparison of retroperitoneal fat pad weight in grams (left graph) and as a percentage of total body weight (right graph) for mice fed a high-fat diet (diet-induced obesity (DIO), left bar), mice fed a high-fat diet plus 1 mg/kg fumagillin (DIO + ZGN-201, middle bar), and age-matched lean mice (right bar).
  • DIO diet-induced obesity
  • DIO + ZGN-201, middle bar mice fed a high-fat diet plus 1 mg/kg fumagillin
  • FIG. 8 contains two bar graphs showing a comparison of retroperitoneal fat pad weight in grams (left graph) and as a percentage of total body weight (right graph) for mice fed a high-fat diet (diet-induced obesity (DIO), left bar), mice fed a high-fat diet plus 1 mg/kg fumagillin (DIO + ZGN-201, middle bar), and age-matched lean mice (right bar).
  • mice 9 contains three bar graphs showing concentrations of glucose (top left graph), insulin (top right graph) and glucose-insulin complex (bottom graph) in mice fed a high-fat diet (diet- induced obesity (DIO), left bar), mice fed a high-fat diet plus 1 mg/kg fumagillin (DIO + ZGN-201, middle bar), and age-matched lean mice (right bar).
  • a high-fat diet diet- induced obesity (DIO), left bar
  • mice fed a high-fat diet plus 1 mg/kg fumagillin DIO + ZGN-201, middle bar
  • age-matched lean mice right bar
  • FIG. 10 shows two line graphs displaying body weight (left graph) and food intake (right graph) of mice fed a high fat diet-fed C57BL/6 mice and treated with 1 mg/kg of the MetAP2 inhibitor fumagillin (squares) or vehicle (diamonds) for 14 days.
  • FIG. 11 is a bar graph showing the percentage change in fat tissue and lean tissue in mice conditioned on a high fat (HF) diet, mice conditioned on a high-fat diet and treated with fumagillin (ZGN-201) administered by oral gavage at doses of 0.1 or 0.3 mg/kg daily, or mice fed on a low-fat diet for 28 days.
  • HF high fat
  • ZGN-201 fumagillin
  • FIG. 12 contains two histological images showing a comparison of retroperitoneal fat pad brown adipose tissue content for mice fed a high-fat diet (diet-induced obesity (DIO)) (left image), and mice fed a high-fat diet plus 1 mg/kg fumagillin (DIO + ZGN- 201 (right image).
  • DIO diet-induced obesity
  • FIG. 13 depicts the effect of 7 day administration of various MetAP2 inhibitors on body weight in DIO mice.
  • Obesity and being overweight refer to an excess of fat in proportion to lean body mass. Excess fat accumulation is associated with increase in size (hypertrophy) as well as number (hyperplasia) of adipose tissue cells. Obesity is variously measured in terms of absolute weight, weight:height ratio, degree of excess body fat, distribution of subcutaneous fat, and societal and esthetic norms. A common measure of body fat is Body Mass Index
  • BMI body mass index
  • an overweight adult has a BMI of 25 kg/m 2 to 29.9 kg/m 2
  • an obese adult has a BMI of 30 kg/m or greater.
  • a BMI of 40 kg/m or greater is indicative of morbid obesity or extreme obesity.
  • the definitions of overweight and obese take into account age and gender effects on body fat.
  • BMI does not account for the fact that excess adipose can occur selectively in different parts of the body, and development of adipose tissue can be more dangerous to health in some parts of the body rather than in other parts of the body.
  • "central obesity" typically associated with an "apple- shaped" body, results from excess adiposity especially in the abdominal region, including belly fat and visceral fat, and carries higher risk of comorbidity than "peripheral obesity", which is typically associated with a "pear-shaped” body resulting from excess adiposity especially on the hips.
  • Measurement of waist/hip circumference ratio (WHR) can be used as an indicator of central obesity.
  • a minimum WHR indicative of central obesity has been variously set, and a centrally obese adult typically has a WHR of about 0.85 or greater if female and about 0.9 or greater if male.
  • Methods of determining whether a subject is overweight or obese that account for the ratio of excess adipose tissue to lean body mass may involve obtaining a body composition of the subject.
  • Body composition can be obtained by measuring the thickness of subcutaneous fat in multiple places on the body, such as the abdominal area, the subscapular region, arms, buttocks and thighs. These measurements are then used to estimate total body fat with a margin of error of approximately four percentage points.
  • Another method is bioelectrical impedance analysis (BIA), which uses the resistance of electrical flow through the body to estimate body fat.
  • Another method is using a large tank of water to measure body buoyancy. Increased body fat will result in greater buoyancy, while greater muscle mass will result in a tendency to sink.
  • DEXA fan-beam dual energy X-ray absorptiometry
  • body composition particularly total body fat and/or regional fat mass, to be determined non-invasively.
  • MetAP2 that enhance the level and function of thioredoxin and/or over-rides the inhibitory effects of hyperinsulinemia related at least in part to insulin- stimulation and/or over-rides the inhibitory effects of high fat diet induced NADPH oxidase activity.
  • a coordinated action can be induced which leads to a physiological reduction in body adiposity through increased loss of fat tissue-associated triglyceride, enhanced local generation of 3,5,3'-triiodothyronine active thyroid hormone with corresponding enhanced activity of brown adipose tissue and its sensitivity to physiological stimuli, increased metabolism of free fatty acids by the liver with increased ketone body formation, and reduced food intake.
  • liver PKA function may be suppressed secondary to elevated NADPH oxidase expression.
  • Ketone body production and utilization are typically suppressed in an obese patient, potentially reducing hepatic satiety signals and increasing food consumption.
  • a MetAP2 inhibitor leads to inhibition of thioredoxin amino-terminal methionine processing and increases steady-state thioredoxin levels, reactivating protein kinase A (PKA) function, reactivating adipose tissue lipase activity and/or stimulating production and/or activity of the rate-limiting enzyme of beta-hydroxybutyrate production (3- hydroxymethyl glutaryl CoA synthase), leading to elevated ketone body production, as illustrated in e.g. Fig. 5.
  • PKA protein kinase A
  • the coordinated and physiologic induction of anti-obesity activities mediated by the methods of the present invention may lead to a healthy reduction in tissue levels of triglyceride, diacylglycerol, and other fat-related mediators and oxidants, and can result in a new steady state situation that favors lean body composition and increased whole body energy metabolism.
  • MetAP2 inhibitors leads to fat tissue being converted to ketone bodies and burned as fuel, unlike existing therapies (including e.g., calorie or energy restricted diets) that target central control of food intake and that may carry adverse side effects (e.g. adverse neurological side effects).
  • therapeutically effective doses contemplated herein will not typically induce any anti- angiogenic action.
  • MetAP2 inhibitors include e.g., calorie or energy restricted diets
  • MetAP2 inhibitors refer to a class of molecules that inhibit the activity of
  • MetAP2 e.g., the ability of MetAP2 to cleave the N-terminal methionine residue of newly synthesized proteins to produce the active form of the protein, or the ability of MetAP2 to regulate protein synthesis by protecting the subunit of eukaryotic initiation factor-2 (eIF2) from phosphorylation.
  • eIF2 eukaryotic initiation factor-2
  • Exemplary MetAP2 inhibitors may include irreversible inhibitors that covalently bind to MetAP2.
  • irreversible inhibitors include fumagillin, fumagillol, fumagillin ketone, Fumagillin refers to all stereoisomers, and can be represented by the following structure:
  • Fumagillol refers to all stereoisomers of the following structure:
  • Fumagillin ketone refers to all stereoisomers of the following structure, and can be represented by the following structure:
  • MetAP2 inhibitors such as O-(4- dimethylaminoethoxycinnamoyl)fumagillol (CKD-732, also referred to herein as Compound A), O-(3,4,5- trimethoxycinnamoyl)fumagillol, O-(4-chlorocinnamoyl)fumagillol; O-(4- aminocinnamoyl)fumagillol; O-(4-dimethylaminoethoxycinnamoyl)fumagillol; O-(4- methoxycinnamoyl)fumagillol; O-(4-dimethylaminocinnamoyl)fumagillol; O-(4- hydroxycinnamoyl)fumagillol; O-(3,4-dimethoxy
  • Fumagillin, and some derivatives thereof have a carboxylic acid moiety and can be administered in the form of the free acid.
  • contemplated herein are pharmaceutically acceptable salts of fumagillin, fumagillol, and derivatives thereof.
  • Pharmaceutically acceptable salts illustratively include those that can be made using the following bases: ammonia, L-arginine, benethamine, benzathene, betaine, bismuth, calcium hydroxide, choline, deanol, diethanolamine, diethylarnine, 2-(diethylamino)ethanol, ethylenediamine, N-methylglucarnine, hydrabamine, 1 H-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)morpholine, piperazine, potassium hydroxide, l-(2- hydroxyethyl)pyrrolidine, sodium hydroxide, triethanolamine, zinc hydroxide, diclyclohexlamine, or any other electron pair donor (as described in Handbook of
  • Contemplated pharmaceutically acceptable salts may include hydrochloric acid, bromic acid, sulfuric acid, phosphoric acid, nitric acid, formic acid, acetic acid, trifluoroacetic acid, oxalic acid, fumaric acid, tartaric acid, maleic acid, methanesulfonic acid, benzenesulfonic acid or para-toluenesulfonic acid.
  • Esters of the present invention may be prepared by reacting fumagillin or fumagillol with the appropriate acid under standard esterification conditions described in the literature (Houben-Weyl 4th Ed. 1952, Methods of Organic Synthesis). Suitable fumagillin esters include ethyl methanoate, ethyl ethanoate, ethyl propanoate, propyl methanoate, propyl ethanoate, and methyl butanoate.
  • contemplated irreversible inhibitors of MetAP2 may include a siRNA, shRNA, an antibody or an antisense compound of MetAP2.
  • MetAP2 inhibitors are provided in the following references, each of which is hereby incorporated by reference: Olson et al. (US 7,084,108 and WO 2002/042295), Olson et al. (US 6,548,477; US 7,037,890; US 7,084,108; US 7,268,111; and WO 2002/042295), Olson et al. (WO 2005/066197), Hong et al. (US 6,040,337)., Hong et al. (US 6,063,812 and WO 1999/059986), Lee et al. (WO 2006/080591), Kishimoto et al.
  • contemplated MetAP2 inhibitors do not include fumagillin, fumagillol, fumagillin ketone, CKD-732/Compound A, and/or TNP-470.
  • contemplated MetAP2 inhibitors may include:
  • a method of treating obesity in a patient in need thereof comprising non-parenterally administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a contemplated pharmaceutically effective amount of a MetAP2 as described below does not substantially modulate or suppress angiogenesis, but is still effective as MetAP2 inhibitor.
  • angiogenesis is known to persons skilled in the art, and refers to the process of new blood vessel formation, and is essential for the exponential growth of solid tumors and tumor metastasis.
  • a method of treating obesity in a patient in need thereof comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient, wherein substantially no loss of new blood vessels in fat deposits or other tissue compartments occur as compared to a patient being treated for obesity using an energy restricted diet alone.
  • fumagillin-class molecules irreversibly inhibit enzymatic activity of MetAP2, leading to N-terminal acetylation and stabilization of these proteins at doses considerably lower than those required to suppress angiogenesis or tumor growth in vivo.
  • the long-lasting covalent inhibition of MetAP2 enzymatic activity driven by such MetAP2 inhibitors may be responsible for the segregation of angiogenic effects from metabolic responses mediated by increased thioredoxin and/or glyceraldehyde-3- phosphate levels in vivo.
  • anti-tumor effects driven by angiogenesis inhibition may require a more thorough starvation of the tumor by heavily restricting blood supply, which requires high doses.
  • Metabolic effects may require a minor and incomplete perturbation of the system which occurs at lower doses and without any obvious direct effect on blood vessels.
  • Treated patients used the disclosed methods may have a lower systemic exposure to said MetAP2 inhibitor as compared to a patient parenterally administered the same of amount of the MetAP2 inhibitor.
  • the disclosed methods may result in less accumulation in the reproductive tract (e.g. testis) of a patient, for example, as compared to the same amount of MetAP2 inhibitor subcutaneously administered.
  • Disclosed methods of treating obesity may result in decreased body fat and a substantial maintenance of muscle mass in said patient.
  • fat oxidation is enhanced in a patient as compared to a patient on a restricted food intake diet alone.
  • a method of decreasing body fat in an overweight or obese patient in need thereof comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient resulting in body fat reduction, and wherein said patient substantially maintains muscle mass during the body fat reduction.
  • Such a patient may retain substantially more muscle mass as compared to body fat reduction in a patient using an energy restricted diet alone.
  • disclosed methods upon administration of said MetAP2 inhibitor e.g. daily or weekly, for about 3, 4, 5 or 6 months or more may result in at least a 5%, 10%, 20%, or 30%, or more weight loss based on the patient's original weight.
  • weight loss following treatment with therapeutically effective doses of MetAP2 inhibitors may substantially cease once a patient attains a normal body composition. Without being limited to an theory, this may be due to reliance of the mechanism on re-establishing tone of adrenergic signal transduction in tissues such as fat, liver, and/or skeletal muscle.
  • a method of maintaining a specified weight in a formerly obese patient comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a method for controlling or preventing hepatic steatosis in an obese patient being treated for obesity comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a method for improving liver function in an obese patient comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a method of restoring normal metabolic action in an obese patient in need thereof comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a method of reducing weight of a patient in a patient in need thereof comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient wherein the metabolic rate of the patient is not substantially reduced as compared to the metabolic rate of a diet only patient on an energy restricted diet alone.
  • a method of restoring and/or maintaining thyroid hormone concentrations in an obese patient comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • a method of improving exercise capacity in a patient in need thereof comprises administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • Also provided herein is a method of activating brown fat function in a patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
  • Contemplated herein is a method of reducing the amount or frequency of administering supplemental insulin in a patient suffering from type 2 diabetes, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. Such treatment may be directed to an obese or non-obese patient.
  • a method for improving surgical outcome in an obese patient in need thereof by reducing weight of said patient comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient before non-acute surgery, thereby reducing liver and/or abdominal fat in said patient and improving surgical outcome.
  • Such surgeries may include bariatric surgery, cardiovascular surgery, abdominal surgery, or orthopedic surgery.
  • a subject can further have an overweight- or obesity-related co-morbidities, i.e., diseases and other adverse health conditions associated with, exacerbated by, or precipitated by being overweight or obese.
  • methods provided herein may further include administering at least one other agent that is directed to treatment of these overweight- or obesity- related conditions.
  • Contemplated other agents include those administered to treat type 2 diabetes such as sulfonylureas (e.g., chlorpropamide, glipizide, glyburide, glimepiride); meglitinides (e.g., repaglinide and nateglinide); biguanides (e.g., metformin); thiazolidinediones (rosiglitazone, troglitazone, and pioglitazone); glucagon-like 1 peptide mimetics (e.g.
  • exenatide and liraglutide sodium-glucose cotransporter inhibitors (e.g., dapagliflozin), renin inhibitors, and alpha-glucosidase inhibitors (e.g., acarbose and meglitol), and/or those administered to treat cardiac disorders and conditions, such hypertension, dyslipidemia, ischemic heart disease, cardiomyopathy, cardiac infarction, stroke, venous thromboembolic disease and pulmonary hypertension, which have been linked to overweight or obesity, for example, chlorthalidone; hydrochlorothiazide; indapamide, metolazone; loop diuretics (e.g., bumetanide, ethacrynic acid, furosemide, lasix, torsemide); potassium- sparing agents (e.g., amiloride hydrochloride, spironolactone, and triamterene); peripheral agents (e.g., reserpine); central al
  • agents administered to treat ischemic heart disease include statins, nitrates (e.g., Isosorbide Dinitrate and Isosorbide Mononitrate), beta-blockers, and calcium channel antagonists, agents administered to treat cardiomyopathy including inotropic agents (e.g., Digoxin), diuretics (e.g., Furosemide), ACE inhibitors, calcium antagonists, anti- arrhythmic agents (e.g., Sotolol, Amiodarone and Disopyramide), and beta-blockers, agents administered to treat cardiac infarction including ACE inhibitors, Angiotensin II receptor blockers, direct vasodilators, beta blockers, anti- arrhythmic agents and thrombolytic agents (e.g., Alteplase, Retaplase, Tenecteplase, Anistreplase, and Urokinase), agents administered to treat strokes including statins, nitrates (e.g., Isosorbide Dinitrate and Is
  • agents administered to treat sleep apnea include Modafinil and amphetamines
  • agents administered to treat nonalcoholic fatty liver disease include antioxidants (e.g., Vitamins E and C), insulin sensitizers (Metformin, Pioglitazone, Rosiglitazone, and Betaine), hepatoprotectants, and lipid-lowering agents
  • agentsadministered to treat osteoarthritis of weight-bearing joints include Acetaminophen, non-steroidal anti-inflammatory agents (e.g., Ibuprofen, Etodolac, Oxaprozin, Naproxen, Diclofenac, and Nabumetone), COX-2 inhibitors (e.g., Celecoxib), steroids, supplements (e.g.,
  • agents administered to treat Prader-Willi Syndrome include human growth hormone (HGH), somatropin, and weight loss agents (e.g., Orlistat, Sibutramine, Methamphetamine, Ionamin, Phentermine, Bupropion, Diethylpropion, Phendimetrazine, Benzphetermine, and Topamax), agents administered to treat polycystic ovary syndrome include insulin-sensitizers, combinations of synthetic estrogen and progesterone, Spironolactone, Eflornithine, and Clomiphene, agents administered to treat erectile dysfunction include phosphodiesterase inhibitors (e.g., Tadalafil, Sildenafil citrate, and Vardenafil), prostaglandin E analogs (e.g., Alprostadil), alkaloids (e.g., Yohimbine), and testosterone, agents administered to treat infertility include Clomiphene,
  • Tranylcypromine Selegiline, Rasagiline, Nialamide, Iproniazid, Iproclozide, Toloxatone, Linezolid, Dienolide kavapyrone desmethoxyyangonin, and Dextroamphetamine
  • psychostimulants e.g., Amphetamine, Methamphetamine, Methylphenidate, and Arecoline
  • antipsychotics e.g., Butyrophenones, Phenothiazines, Thioxanthenes, Clozapine, Olanzapine, Risperidone, Quetiapine, Ziprasidone, Amisulpride, Paliperidone, Symbyax, Tetrabenazine, and Cannabidiol
  • mood stabilizers e.g., Lithium carbonate, Valproic acid, Divalproex sodium, Sodium valproate, Lamotrigine, Carbamazepine, Gabapent
  • contemplated methods may further comprising assessing one or more indices of on-going weight loss, e.g. the ketone body production level in a patient; and optionally adjusting the amount administered; thereby optimizing the therapeutic efficacy of said MetAP2 inhibitor.
  • a patient may have a lower systemic exposure (e.g. at least about 2, 3, 5, 10, 20, or at least about 30% less systemic exposure) to the non-parenterally administered (e.g. oral administration) of a MetAP2 inhibitor as compared to a patient parenterally administered (e.g. subcutaneously) the same dose of the MetAP2 inhibitor.
  • non-parenterally administered (e.g. orally administered) MetAP2 inhibitors e.g. an irreversible inhibitor
  • MetAP2 inhibitors may bind less to MetAP2 as compared to subcutaneously administered MetAP2 inhibitors.
  • Contemplated non-parenteral administration includes oral, buccal, transdermal
  • topical e.g. by a dermal patch
  • topical inhalation, or sublingual administration, or e.g., ocular, pulmonary, nasal, rectal or vaginal administration.
  • angiogenesis e.g. a daily dosage of a MetAP2 inhibitor
  • methods that include administering doses of MetAP2 inhibitors that are effective for weight loss, but are significantly smaller doses than that necessary to modulate and/or suppress angiogenesis (which may typically require about 12.5mg/kg to about 50mg/kg or more).
  • contemplated dosage of a MetAP2 inhibitor in the methods described herein may include administering about 25 mg/day, about 10 mg/day, about 5 mg/day, about 3 mg/day, about 2 mg/day, about 1 mg/day, about 0.75 mg/day, about 0.5 mg/day, about 0.1 mg/day, about 0.05 mg/day, or about 0.01 mg/day.
  • an effective amount of the drug for weight loss in a patient may be about 0.0001 mg/kg to about 25 mg/kg of body weight per day.
  • a contemplated dosage may from about 0.001 to 10 mg/kg of body weight per day, about 0.001 mg/kg to 1 mg/kg of body weight per day, about 0.001 mg/kg to 0.1 mg/kg of body weight per day or about 0.005 to about 0.04 mg/kg or about 0.005 to about 0.049 mg/kg of body weight a day.
  • a MetAP2 inhibitor such as disclosed herein (e.g. O-(4- dimethlyaminoethoxycinnamoyl)fumagillol), may be administered about 0.005 to about 0.04 mg/kg of a patient.
  • a method for treating obesity in a patient in need thereof comprising administering, parenterally (e.g. intravenously) or non-parenterally, about 0.005 to about 0.04 mg/kg of a MetAP2 inhibitor selected from O-(4- dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof (for example, an oxalate salt), to said patient.
  • a MetAP2 inhibitor selected from O-(4- dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof (for example, an oxalate salt)
  • Such a method upon administration of said MetAP2 inhibitor e.g. daily or weekly, for about 3, 4 , 5 or 6 months or more may result in at least a 10%, 20%, 30%, or 40% or more weight loss based on the patient's original weight.
  • Contemplated methods may include administration of a composition comprising a MetAP2 inhibitor, for example, hourly, twice hourly, every three to four hours, daily, twice daily, 1, 2, 3 or 4 times a week, every three to four days, every week, or once every two weeks depending on half-life and clearance rate of the particular composition or inhibitor.
  • a composition comprising a MetAP2 inhibitor, for example, hourly, twice hourly, every three to four hours, daily, twice daily, 1, 2, 3 or 4 times a week, every three to four days, every week, or once every two weeks depending on half-life and clearance rate of the particular composition or inhibitor.
  • Treatment can be continued for as long or as short a period as desired.
  • the compositions may be administered on a regimen of, for example, one to four or more times per day.
  • a suitable treatment period can be, for example, at least about one week, at least about two weeks, at least about one month, at least about six months, at least about 1 year, or indefinitely.
  • a treatment period can terminate when a desired result, for example a weight loss target, is achieved. For example, when about loss of about 20% body weight, about 30% body weight or more has been achieved.
  • a treatment regimen can include a corrective phase, during which a MetAP2 inhibitor dose sufficient to provide reduction of excess adiposity is administered, followed by a maintenance phase, during which a lower MetAP2 inhibitor dose sufficient to prevent re-development of excess adiposity is administered.
  • MetAP2 inhibitors can be formulated with conventional excipients to prepare an inhalable composition in the form of a fine powder or atomizable liquid.
  • MetAP2 inhibitors can be formulated with conventional excipients, for example, in the form of eye drops or an ocular implant.
  • excipients useful in eye drops are viscosifying or gelling agents, to minimize loss by lacrimation through improved retention in the eye.
  • Liquid dosage forms for oral or other administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents 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, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents such as, for example, water or other solvents, solubil
  • Dosage forms for topical or transdermal administration of an inventive pharmaceutical composition may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants, or patches.
  • the active agent is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • cutaneous routes of administration are achieved with aqueous drops, a mist, an emulsion, or a cream.
  • Transdermal patches may have the added advantage of providing controlled delivery of the active ingredients to the body.
  • dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • injectable preparations are also contemplated herein, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • compositions for rectal or vaginal administration may be suppositories which can be prepared by mixing a MetAP2 inhibitor with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active agent(s).
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active agent(s).
  • contemplated formulations can be administered by release from a lumen of an endoscope after the endoscope has been inserted into a rectum of a subject.
  • Oral dosage forms such as capsules, tablets, pills, powders, and granules, may be prepared using any suitable process known to the art.
  • a MetAP2 inhibitor may be mixed with enteric materials and compressed into tablets.
  • formulations of the invention are incorporated into chewable tablets, crushable tablets, tablets that dissolve rapidly within the mouth, or mouth wash..
  • Example 1 Fumagillin increases levels of Thioredoxin-1 (TXN) in a dose-dependent manner
  • B 16F10 cells were treated with increasing concentrations of fumagillin.
  • N- terminal chymotryptic fragments of TXN were measured by liquid chromatography/mass spectrometry (LC/MS).
  • Fumagillin significantly increased the cellular level of TXN containing an intact N-terminal methionine [TXN(I -4)] and caused significant accumulation of N-acetyl TXN(I -4), consistent with a role for fumagillin as a MetAP2 inhibitor. See FIGs. 1-2.
  • the EC50 of fumagillin to increase Met-TXN acetylated Met-TXN levels is ⁇ 10 nM.
  • Similar results were seen in Cos-1 and 293T cells that were transfected with
  • Example 2 Oral administration of fumagillin reduces body fat in mammals
  • Oral administration of the MetAP2 inhibitor fumagillin causes an increase in circulating ketone bodies, as measured by concentration of beta hydroxybutyrate, which indicates an increase in the breakdown of stored fat.
  • C57BL/6 mice conditioned on a high fat diet also referred to as a diet-induced obese diet or DIO
  • ZGN- 201 fumagillin administered by oral gavage at doses of 0.3 or 3 mg/kg daily for 10 days resulting in an increase in beta hydroxybutyrate (FIG. 5A) concentrations, without an increase in free fatty acid concentration (FIG. 5B) as well as a reduction in body weight by 7 to 15 percent (data not shown).
  • mice treated daily with 1 mg/kg fumagillin maintained a weight that was 25% less than that of age-matched mice maintained on normal mouse chow diet (lean mice, FIG. 6) for the duration of the experiment.
  • the size and appearance of epididymal fat, peri-renal fat and liver in fumagillin-treated mice was more similar to that of age-matched lean mice than that of untreated mice on a high fat diet (FIG 7).
  • Retroperitoneal fat pad weight in fumagillin-treated mice was also more similar to that of age-matched lean mice than that of untreated mice on a high fat diet (FIG. 8).
  • Adipose tissue in mice fed a high-fat diet show distinctive crown-like structures not seen in lean mice. These crown-like structures were also absent from the adipose tissue of fumagillin-treated mice. Additionally, adipocyte size in fumagillin-treated mice was more similar to that of age-matched lean mice than that of untreated mice on a high fat diet. [0087] Fasted glucose and insulin levels, as well as insulin-glucose product, in fumagillin-treated mice was more similar to that of age-matched lean mice than that of untreated mice on a high fat diet (FIG. 9), indicating that fumagillin normalizes sensitivity to insulin in mice on a high fat diet.
  • Example 3 Fumagillin reduces food intake during the peak of weight loss
  • Weight loss following treatment with therapeutically relevant doses of MetAP2 inhibitors stops once animals attain a normal body composition. Once weight loss reaches a nadir, fumagillin-treated animals enter a third phase in which their food intake returns to normal levels despite maintaining a normal body weight and improved metabolic control evidenced by reduced insulin and glucose concentrations. This third phase can be maintained indefinitely provided the drug is continued (see FIG. 10), or presumably can be maintained in practice through other complementary pharmacologic, dietary, or lifestyle interventions.
  • Example 4 Oral administration of fumagillin results in fat loss with minimal change in lean tissue
  • C57BL/6 mice were conditioned on a low-fat diet (LF), a high fat diet (HF), or a high-fat diet and treated with fumagillin (ZGN-201) administered by oral gavage at doses of 0.1 or 0.3 mg/kg for 28 days. Either dose of fumagillin caused a reduction in body weight down to the level of low fat fed mice. To determine whether the weight loss resulted from loss of fat or loss of lean tissue, the percentage change in fat tissue and lean tissue was determined for LF mice, HF mice, HF mice treated with 0.1 mg/kg fumagillin, and HF mice treated with 0.3 mg/kg fumagillin. As shown in FIG.
  • HF mice treated with either dose of fumagillin experienced a 40-50% reduction in fat (compared to an increase in fat in both the LF and HF control mice), but only a 5-10% reduction in lean tissue, indicating that fumagillin-induced weight loss results primarily from loss of fat, rather than loss of lean tissue.
  • Example 5 Orally administered compounds having MetAP-2 inhibitory cores cause weight loss in diet-induced obese mice
  • mice in this study were not genetically obese, but prior to and during the study, obesity was induced by a high-fat diet. Twelve week-old C57BL/6NTac mice, maintained on a 60% fat diet prior to and during the study, were separated into seven groups, eight animals per group. Average body weight of the mice was approximately 47 g at the start of the study.
  • Each of six groups was orally administered 1.0 mg/kg of a compound (fumagillin and compounds B. C, D, E, F and G as disclosed herein), in 10% gelucire in deoinised water. Each of these six groups was administered a different compound. One group was orally administered fumagillin at 1.0 mg/kg (group 201) in 10% gelucire in deoinised water, and one group was administered 10% gelucire in deoinised water (vehicle). Mice received administrations once a day for 7 days.
  • mice Twelve week-old C57BL/6NTac mice, maintained on a 60% fat diet prior to and during the study, were separated into seven groups, eight animals per group. Average body weight of the mice was approximately 47 g at the start of the study.
  • mice were orally administered 100.0 mg/kg of a compound H, in 10% gelucire in deoinised water. Mice received administrations once a day for 7 days. After 7 days, the average weight loss of the mice was 13.2%

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Child & Adolescent Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

METHODS OF TREATING AN OVERWEIGHT OR OBESE SUBJECT
RELATED APPLICATIONS
[0001] This application claims priority to U. S provisional applications U. S. S.N.
61/119,875 filed December 4, 2008, U.S.S.N. 61/119,881 filed December 4, 2008, U.S.S.N. 61/119,884 filed December 4, 2008, U.S.S.N. 61/119,886 filed December 4, 2008, U.S.S.N. 61/119,872 filed December 4, 2008, U.S.S.N. 61/119,877 filed December 4, 2008, U.S.S.N. 61/119,885 filed December 4, 2008, U.S.S.N. 61/119,891 filed December 4, 2008, U.S.S.N. 61/119,888 filed December 4, 2008, U.S.S.N. 61/275,688 filed August 3, 2009, and U.S.S.N. 61/260,194 filed November 11, 2009, each application of which is hereby incorporated by reference.
BACKGROUND [0002] Obesity is a complex medical disorder of appetite regulation and metabolism resulting in excessive accumulation of adipose tissue mass. Typically defined as a body mass index (BMI) of 30 kg/m2 or more, obesity is a world-wide public health concern that is associated with cardiovascular disease, diabetes, certain cancers, respiratory complications, osteoarthritis, gallbladder disease, decreased life expectancy, and work disability. The primary goals of obesity therapy are to reduce excess body weight, improve or prevent obesity- related morbidity and mortality, and maintain long-term weight loss.
[0003] Treatment modalities typically include lifestyle management, pharmacotherapy, and surgery. Treatment decisions are made based on severity of obesity, seriousness of associated medical conditions, patient risk status, and patient expectations. Notable improvements in cardiovascular risk and the incidence of diabetes have been observed with weight loss of 5-10% of body weight, supporting clinical guidelines for the treatment of obesity that recommend a target threshold of 10% reduction in body weight from baseline values. [0004] However, while prescription anti-obesity medications are typically considered for selected patients at increased medical risk because of their weight and for whom lifestyle modifications (diet restriction, physical activity, and behavior therapy) alone have failed to produce durable weight loss, approved drugs have had unsatisfactory efficacy for severely obese subjects, leading to only -3-5% reduction in body weight after a year of treatment.
[0005] Bariatric surgery may be considered as a weight loss intervention for patients at or exceeding a BMI of 40 kg/m2. Patients with a BMI > 35 kg/m2 and an associated serious medical condition are also candidates for this treatment option. Unfortunately, postoperative complications commonly result from bariatric surgical procedures, including bleeding, embolism or thrombosis, wound complications, deep infections, pulmonary complications, and gastrointestinal obstruction; reoperation during the postoperative period is sometimes necessary to address these complications. Rates of reoperation or conversion surgery beyond the postoperative period depend on the type of bariatric procedure, and in one study ranged from 17% to 31%. Intestinal absorptive abnormalities, such as micronutrient deficiency and protein- calorie malnutrition, also are typically seen with bypass procedures, requiring lifelong nutrient supplementation. Major and serious adverse outcomes associated with bariatric surgery are common, observed in approximately 4 percent of procedures performed (including death in 0.3 to 2 percent of all patients receiving laparoscopic banding or bypass surgeries, respectively)
[0006] MetAP2 encodes a protein that functions at least in part by enzymatically removing the amino terminal methionine residue from certain newly translated proteins such as glyceraldehyde-3- phosphate dehydrogenase (Warder et al. (2008) J Proteome Res 7:4807). Increased expression of the MetAP2 gene has been historically associated with various forms of cancer. Molecules inhibiting the enzymatic activity of MetAP2 have been identified and have been explored for their utility in the treatment of various tumor types (Wang et al. (2003) Cancer Res. 63:7861) and infectious diseases such as microsporidiosis, leishmaniasis, and malaria (Zhang et al. (2002) J. Biomed. Sci. 9:34). However, such MetAP2 inhibitors may be useful as well for patients with excess adiposity and conditions related to adiposity including type 2 diabetes, hepatic steatosis, and cardiovascular disease (via e.g. ameliorating insulin resistance, reducing hepatic lipid content, and reducing cardiac workload). Methods of treating obese subjects that are more effective than e.g. dieting alone are clearly needed. SUMMARY
[0007] This disclosure generally relates to methods of treating an overweight or obese subject or patient that include non-parenterally administering a pharmaceutically effective amount of a MetAP2 inhibitor to a patient in need thereof, e.g., a human or a companion animal such as a cat or a dog.
[0008] In one embodiment, a method of treating obesity in a patient in need thereof is provided, comprising non-parenterally administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. Such methods may result in, for example, a lower systemic exposure to said MetAP2 inhibitor as compared to a patient parenterally administered the same of amount of the MetAP2 inhibitor. Contemplated pharmaceutically effective amounts may not substantially modulate or suppress angiogenesis. In exemplary embodiments, non-parenteral administration may result in decreased body fat and a substantial maintenance of muscle mass in said patient. Such methods may enhance fat oxidation compared to a patient on a restricted food intake diet alone. Also provided herein is a method of treating obesity in a patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient, wherein substantially no loss of new blood vessels in fat deposits occur as compared to a patient being treated for obesity using an energy restricted diet alone. In some embodiments, non-parenterally administration may include oral, buccal, sublingual, transdermal, rectal, nasal administration, or administration via inhalation.
[0009] Contemplated MetAP2 inhibitors include substantially irreversible inhibitors, such as e.g., fumagillin, fumagillol or fumagillin ketone derivative, siRNA, shRNA, an antibody, or a antisense compound, or, e.g., O-(4-dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof. In another embodiment, a contemplated MetAP2 inhibitor may be substantially reversible inhibitor.
[0010] Also provided herein is a method for controlling or preventing hepatic steatosis in an obese patient being treated for obesity, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient, and/or a method for improving liver function in an obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. [0011] In an embodiment, a method of improving exercise capacity in a patient in need thereof is provided that includes administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
[0012] A method of reducing weight of a patient in a patient in need thereof is contemplated herein that comprises administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient wherein the metabolic rate of the patient is not substantially reduced as compared to the metabolic rate of a diet only patient on an energy restricted diet alone. Also provided herein is a method of restoring normal metabolic action in an obese patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
[0013] In an embodiment, a method of decreasing body fat in an overweight or obese patient in need thereof is provided that comprises administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient resulting in body fat reduction, and wherein said patient substantially maintains muscle mass during the body fat reduction, for example a patient may retain substantially more muscle mass as compared to body fat reduction in a patient using an energy restricted diet alone.
[0014] A method of activating brown fat function and/or increasing brown fat tissue mass in a patient in need thereof is provided, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient, and/or a method of restoring and/or maintaining thyroid hormone concentrations in an obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
[0015] The disclosed methods may include a pharmaceutically effective amount of a
MetAP2 inhibitor that does not substantially modulate or suppress angiogenesis in a treated patient. In some embodiments of the disclosed methods, a patient has a lower systemic exposure to said MetAP2 inhibitor as compared to a patient parenterally administered the same amount of the MetAP2 inhibitor.
[0016] MetAP2 inhibitors may be administered non-parenterally, orally, buccally or sublingually, topically, rectally, or transdermally in at least some disclosed methods. In some disclosed methods, a MetAP2 inhibitor may be administered subcutaneously or intravenously. In some embodiments, administration of a MetAP2 inhibitor may occur at least daily, or every other day, or at least weekly.
[0017] Also contemplated herein is a method for reducing the amount or frequency of administering supplemental insulin in a patient suffering from type 2 diabetes, comprising a pharmaceutically effective amount of a MetAP2 inhibitor.
[0018] A method for improving surgical outcome in an obese patient in need thereof, by reducing weight of said patient is provided herein, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient before non-acute surgery, thereby reducing liver and/or abdominal fat in said patient and improving surgical outcome. Such surgeries may include e.g., bariatric surgery, cardiovascular surgery, abdominal surgery, or orthopedic surgery. Also provided herein is a method of maintaining a specified weight in a formerly obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
[0019] Disclosed methods may further comprise co-administering an additional weight loss agent and/or may further comprise administering a food restricted diet to a patient. In another embodiment, a disclosed method may further comprise assessing the ketone body production level in a patient; and optionally adjusting the amount administered; thereby optimizing the therapeutic efficacy of said MetAP2 inhibitor. In at least some disclosed methods, a patient may incur greater than or about equal to a 20% weight loss after about 6 months of said administration.
[0020] In another embodiment, a method for treating obesity in a patient in need thereof is provided, comprising administering about 0.005 to about 0.04 mg/kg of a MetAP2 inhibitor selected from O-(4-dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof, for example, an oxalate salt, to said patient. This MetAP2 inhibitor may be administered at least daily, or 1, 2, 3 or 4 times a week. In some embodiments, MetAP2 may be administered parenterally, e.g., intravenously, or non-parenterally. Upon administration of the MetAP2 inhibitor e.g. daily, or 1, 2, 3, 4, 5, 6 or 7 times a week, for about 6 months, may result in at least a 20% weight loss or more of the patient's original weight. BRIEF DESCRIPTION OF FIGURES
[0021] FIG. 1 is a bar graph showing TXN(104) in B 16F10 cell lysate after chymotryptic digest.
[0022] FIG. 2 is a bar graph showing N-acetyl TXN(I -4) in B 16F10 cell lysate after chymotriptic digest.
[0023] FIG. 3 is a photograph of a gel showing that fumagillin significantly increased
TXN in both Cos-1 and 293T cells.
[0024] FIG. 4 is a photograph of a gel showing that EC50 of fumagillin to increase
TXN levels is < 10 nM.
[0025] FIG. 5A is a bar graph showing the concentration of beta-hydroxybutyrate in mice fed a high fat diet (vehicle) or mice fed a high-fat diet plus oral administration of fumagillin for ten days at the concentrations indicated. FIG. 5B is a bar graph showing the concentration of nonesterified fatty acid concentrations under the conditions described for FIG. 5A. FIG. 5C is a bar graph showing the concentration of beta-hydroxybutyrate in mice fed a high fat diet (vehicle) or mice fed a high-fat diet plus oral administration of fumagillin for 250 days at the concentrations indicated.
[0026] FIG. 6 is a line graph showing the body weight over time of high-fat diet- fed
C57BL/6 mice treated with 1 mg/kg fumagillin (squares) or vehicle (diamonds) for 250 days. Age-matched lean C57BL/6 mice maintained on normal mouse chow diet are shown for comparison (triangles).
[0027] FIG. 7 shows age-matched lean mice maintained on normal mouse chow diet
(left) or mice fed a high fat diet-fed and treated with 1 mg/kg fumagillin (right) or vehicle (middle) for 250 days.
[0028] FIG. 8 contains two bar graphs showing a comparison of retroperitoneal fat pad weight in grams (left graph) and as a percentage of total body weight (right graph) for mice fed a high-fat diet (diet-induced obesity (DIO), left bar), mice fed a high-fat diet plus 1 mg/kg fumagillin (DIO + ZGN-201, middle bar), and age-matched lean mice (right bar). [0029] FIG. 9 contains three bar graphs showing concentrations of glucose (top left graph), insulin (top right graph) and glucose-insulin complex (bottom graph) in mice fed a high-fat diet (diet- induced obesity (DIO), left bar), mice fed a high-fat diet plus 1 mg/kg fumagillin (DIO + ZGN-201, middle bar), and age-matched lean mice (right bar).
[0030] FIG. 10 shows two line graphs displaying body weight (left graph) and food intake (right graph) of mice fed a high fat diet-fed C57BL/6 mice and treated with 1 mg/kg of the MetAP2 inhibitor fumagillin (squares) or vehicle (diamonds) for 14 days.
[0031] FIG. 11 is a bar graph showing the percentage change in fat tissue and lean tissue in mice conditioned on a high fat (HF) diet, mice conditioned on a high-fat diet and treated with fumagillin (ZGN-201) administered by oral gavage at doses of 0.1 or 0.3 mg/kg daily, or mice fed on a low-fat diet for 28 days. For each of "Fat Tissue Change" and "Lean Tissue Change," bars from left to right are: LF, HF, HF + 0.3, and HF + 0.1.
[0032] FIG. 12 contains two histological images showing a comparison of retroperitoneal fat pad brown adipose tissue content for mice fed a high-fat diet (diet-induced obesity (DIO)) (left image), and mice fed a high-fat diet plus 1 mg/kg fumagillin (DIO + ZGN- 201 (right image).
[0033] FIG. 13 depicts the effect of 7 day administration of various MetAP2 inhibitors on body weight in DIO mice.
DETAILED DESCRIPTION
Overview
[0034] Obesity and being overweight refer to an excess of fat in proportion to lean body mass. Excess fat accumulation is associated with increase in size (hypertrophy) as well as number (hyperplasia) of adipose tissue cells. Obesity is variously measured in terms of absolute weight, weight:height ratio, degree of excess body fat, distribution of subcutaneous fat, and societal and esthetic norms. A common measure of body fat is Body Mass Index
(BMI). The BMI refers to the ratio of body weight (expressed in kilograms) to the square of height (expressed in meters). Body mass index may be accurately calculated using the formulas: SI units: BMI=weight(kg)/(height2(m2), or US units: BMI= (weight(lb)*703)/(height2(in2).
[0035] In accordance with the U.S. Centers for Disease Control and Prevention (CDC), an overweight adult has a BMI of 25 kg/m2 to 29.9 kg/m2, and an obese adult has a BMI of 30 kg/m or greater. A BMI of 40 kg/m or greater is indicative of morbid obesity or extreme obesity. For children, the definitions of overweight and obese take into account age and gender effects on body fat.
[0036] BMI does not account for the fact that excess adipose can occur selectively in different parts of the body, and development of adipose tissue can be more dangerous to health in some parts of the body rather than in other parts of the body. For example, "central obesity", typically associated with an "apple- shaped" body, results from excess adiposity especially in the abdominal region, including belly fat and visceral fat, and carries higher risk of comorbidity than "peripheral obesity", which is typically associated with a "pear-shaped" body resulting from excess adiposity especially on the hips. Measurement of waist/hip circumference ratio (WHR) can be used as an indicator of central obesity. A minimum WHR indicative of central obesity has been variously set, and a centrally obese adult typically has a WHR of about 0.85 or greater if female and about 0.9 or greater if male.
[0037] Methods of determining whether a subject is overweight or obese that account for the ratio of excess adipose tissue to lean body mass may involve obtaining a body composition of the subject. Body composition can be obtained by measuring the thickness of subcutaneous fat in multiple places on the body, such as the abdominal area, the subscapular region, arms, buttocks and thighs. These measurements are then used to estimate total body fat with a margin of error of approximately four percentage points. Another method is bioelectrical impedance analysis (BIA), which uses the resistance of electrical flow through the body to estimate body fat. Another method is using a large tank of water to measure body buoyancy. Increased body fat will result in greater buoyancy, while greater muscle mass will result in a tendency to sink. Another method is fan-beam dual energy X-ray absorptiometry (DEXA). DEXA allows body composition, particularly total body fat and/or regional fat mass, to be determined non-invasively. [0038] Without being limited by any particular theory or mechanism of action, it is believed that fat oxidation and lipolysis are stimulated through treatment with inhibitors of MetAP2 that enhance the level and function of thioredoxin and/or over-rides the inhibitory effects of hyperinsulinemia related at least in part to insulin- stimulation and/or over-rides the inhibitory effects of high fat diet induced NADPH oxidase activity. A coordinated action can be induced which leads to a physiological reduction in body adiposity through increased loss of fat tissue-associated triglyceride, enhanced local generation of 3,5,3'-triiodothyronine active thyroid hormone with corresponding enhanced activity of brown adipose tissue and its sensitivity to physiological stimuli, increased metabolism of free fatty acids by the liver with increased ketone body formation, and reduced food intake. These effects are evident at doses of a MetAP2 inhibitor that do not substantially modulate angiogenesis.
[0039] In obese and/or hyperinsulinemic patients, liver PKA function may be suppressed secondary to elevated NADPH oxidase expression. Ketone body production and utilization are typically suppressed in an obese patient, potentially reducing hepatic satiety signals and increasing food consumption. However, administration of a MetAP2 inhibitor, without being limited by an theory, leads to inhibition of thioredoxin amino-terminal methionine processing and increases steady-state thioredoxin levels, reactivating protein kinase A (PKA) function, reactivating adipose tissue lipase activity and/or stimulating production and/or activity of the rate-limiting enzyme of beta-hydroxybutyrate production (3- hydroxymethyl glutaryl CoA synthase), leading to elevated ketone body production, as illustrated in e.g. Fig. 5.
[0040] The coordinated and physiologic induction of anti-obesity activities mediated by the methods of the present invention may lead to a healthy reduction in tissue levels of triglyceride, diacylglycerol, and other fat-related mediators and oxidants, and can result in a new steady state situation that favors lean body composition and increased whole body energy metabolism. Without being bound by any theory, it is believed that the mechanistic cascade activated by MetAP2 inhibitors leads to fat tissue being converted to ketone bodies and burned as fuel, unlike existing therapies (including e.g., calorie or energy restricted diets) that target central control of food intake and that may carry adverse side effects (e.g. adverse neurological side effects). Further, therapeutically effective doses contemplated herein will not typically induce any anti- angiogenic action. MetAP2 inhibitors
[0041] MetAP2 inhibitors refer to a class of molecules that inhibit the activity of
MetAP2, e.g., the ability of MetAP2 to cleave the N-terminal methionine residue of newly synthesized proteins to produce the active form of the protein, or the ability of MetAP2 to regulate protein synthesis by protecting the subunit of eukaryotic initiation factor-2 (eIF2) from phosphorylation.
[0042] Exemplary MetAP2 inhibitors may include irreversible inhibitors that covalently bind to MetAP2. For example, such irreversible inhibitors include fumagillin, fumagillol, fumagillin ketone, Fumagillin refers to all stereoisomers, and can be represented by the following structure:
Figure imgf000011_0001
[0043] Fumagillol refers to all stereoisomers of the following structure:
Figure imgf000011_0002
[0044] Fumagillin ketone refers to all stereoisomers of the following structure, and can be represented by the following structure:
Figure imgf000012_0001
[0045] Derivatives and analogs of fumagillin, and pharmaceutically acceptable salts thereof are contemplated herein as irreversible MetAP2 inhibitors, such as O-(4- dimethylaminoethoxycinnamoyl)fumagillol (CKD-732, also referred to herein as Compound A), O-(3,4,5- trimethoxycinnamoyl)fumagillol, O-(4-chlorocinnamoyl)fumagillol; O-(4- aminocinnamoyl)fumagillol; O-(4-dimethylaminoethoxycinnamoyl)fumagillol; O-(4- methoxycinnamoyl)fumagillol; O-(4-dimethylaminocinnamoyl)fumagillol; O-(4- hydroxycinnamoyl)fumagillol; O-(3,4-dimethoxycinnamoyl)fumagillol; O-(3,4- methylenedioxycinnamoyl)fumagillol; O-(3,4,5-trimethoxycinnamoyl)fumagillol; O-(4- nitrocinnamoyl)fumagillol; O-(3,4-dimethoxy-6-aminocinnamoyl)fumagillol; O-(4-acetoxy- 3,5-dimethoxycinnamoyl)fumagillol; O-(4-ethylaminocinnamoyl)fumagillol; O-(4- ethylaminoethoxycinnamoyl)fumagillol; O-(3-dimethylaminomethyl-4- methoxycinnamoyl)fumagillol; O-(4-trifluoromethylcinnamoyl)fumagillol; O-(3,4-dimethoxy- 6-nitrocinnamoyl)fumagillol; O-(4-acetoxycinnamoyl)fumagillol; O-(4- cyanocinnamoyl)fumagillol; 4-(4-methoxycinnamoyl)oxy-2-( 1 ,2-epoxy- 1 ,5-dimethyl-4- hexenyl)-3-methoxy— 1-chloromethyl-l-cyclohexanol; O-(3,4,5- trimethoxycinnamoyl)fumagillol; O-(4-dimethylaminocinnamoyl)fumagillol; O-(3,4,5- trimethoxycinnamoyl)oxy-2-( 1 ,2-epoxy- 1 ,5-dimethyl-4-hexenyl)-3-m- ethoxy- 1 -chloromethyl- 1-cyclohexanol; O-(4-dimethylaminocinnamoyl)oxy-2-(l,2-epoxy-l,5-dimethyl-4-hexenyl)-3- me- thoxy- 1-chloromethyl-l-cyclohexanol; O-(3,5-dimethoxy-4-hydroxycinnamoyl)fumagillol or O-(chloracetyl-carbamoyl) fumagillol(TNP-470). [0046] Fumagillin, and some derivatives thereof, have a carboxylic acid moiety and can be administered in the form of the free acid. Alternatively, contemplated herein are pharmaceutically acceptable salts of fumagillin, fumagillol, and derivatives thereof. Pharmaceutically acceptable salts illustratively include those that can be made using the following bases: ammonia, L-arginine, benethamine, benzathene, betaine, bismuth, calcium hydroxide, choline, deanol, diethanolamine, diethylarnine, 2-(diethylamino)ethanol, ethylenediamine, N-methylglucarnine, hydrabamine, 1 H-imidazole, lysine, magnesium hydroxide, 4-(2-hydroxyethyl)morpholine, piperazine, potassium hydroxide, l-(2- hydroxyethyl)pyrrolidine, sodium hydroxide, triethanolamine, zinc hydroxide, diclyclohexlamine, or any other electron pair donor (as described in Handbook of
Pharmaceutical Salts, Stan & Wermuth, VHCA and Wiley, Uchsenfurt- Hohestadt Germany, 2002). Contemplated pharmaceutically acceptable salts may include hydrochloric acid, bromic acid, sulfuric acid, phosphoric acid, nitric acid, formic acid, acetic acid, trifluoroacetic acid, oxalic acid, fumaric acid, tartaric acid, maleic acid, methanesulfonic acid, benzenesulfonic acid or para-toluenesulfonic acid.
[0047] Esters of the present invention may be prepared by reacting fumagillin or fumagillol with the appropriate acid under standard esterification conditions described in the literature (Houben-Weyl 4th Ed. 1952, Methods of Organic Synthesis). Suitable fumagillin esters include ethyl methanoate, ethyl ethanoate, ethyl propanoate, propyl methanoate, propyl ethanoate, and methyl butanoate.
[0048] In another embodiment, contemplated irreversible inhibitors of MetAP2 may include a siRNA, shRNA, an antibody or an antisense compound of MetAP2.
[0049] Further examples of MetAP2 inhibitors, are provided in the following references, each of which is hereby incorporated by reference: Olson et al. (US 7,084,108 and WO 2002/042295), Olson et al. (US 6,548,477; US 7,037,890; US 7,084,108; US 7,268,111; and WO 2002/042295), Olson et al. (WO 2005/066197), Hong et al. (US 6,040,337)., Hong et al. (US 6,063,812 and WO 1999/059986), Lee et al. (WO 2006/080591), Kishimoto et al. (US 5,166,172; US 5,698,586; US 5,164,410; and 5,180,738), Kishimoto et al. (US 5,180,735), Kishimoto et al. (US 5,288,722), Kishimoto et al. (US 5,204,345), Kishimoto et al. (US 5,422,363), Liu et al. (US 6,207,704; US 6,566,541; and WO 1998/056372), Craig et al. (WO 1999/057097), Craig et al. (US 6,242,494), BaMaung et al. (US 7,030,262), Comess et al. (WO 2004/033419), Comess et al. (US 2004/0157836), Comess et al. (US 2004/0167128), Henkin et al. (WO 2002/083065), Craig et al. (US 6,887,863), Craig et al. (US 2002/0002152), Sheppard et al. (2004, Bioorganic & Medicinal Chemistry Letters 14:865-868), Wang et al. (2003, Cancer Research 63:7861-7869), Wang et al. (2007, Bioorganic & Medicinal Chemistry Letters 17:2817-2822), Kawai et al. (2006, Bioorganic & Medicinal Chemistry Letters 16:3574-3577), Henkin et al. (WO 2002/026782), Nan et al. (US 2005/0113420), Luo et al. (2003, J. Med. Chem., 46:2632-2640), Vedantham et al. (2008, J. Comb. Chem., 10:195-203), Wang et al. (2008, J. Med. Chem., XXXX, Vol. xxx, No. xx), Ma et al. (2007, BMC Structural Biology, 7:84) and Huang et al. (2007, J. Med. Chem., 50:5735-5742), Evdokimov et al. (2007, PROTEINS: Structure, Function, and Bioinformatics, 66:538-546), Garrabrant et al. (2004, Angiogenesis 7:91-96), Kim et al. (2004, Cancer Research, 64:2984-2987), Towbin et al. (2003, The Journal of Biological Chemistry, 278(52):52964-52971), Marino Jr. (U.S. patent number 7,304,082), Kallender et al. (U.S. patent application number 2004/0192914), and Kallender et al. (U.S. patent application numbers 2003/0220371 and 2005/0004116). In some embodiments, contemplated MetAP2 inhibitors do not include fumagillin, fumagillol, fumagillin ketone, CKD-732/Compound A, and/or TNP-470.
[0050] For example, contemplated MetAP2 inhibitors may include:
Figure imgf000014_0001
Figure imgf000015_0001
Methods [0051] A method of treating obesity in a patient in need thereof is provided herein, comprising non-parenterally administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. In some embodiments, a contemplated pharmaceutically effective amount of a MetAP2 as described below, does not substantially modulate or suppress angiogenesis, but is still effective as MetAP2 inhibitor. The term "angiogenesis" is known to persons skilled in the art, and refers to the process of new blood vessel formation, and is essential for the exponential growth of solid tumors and tumor metastasis. For example, provided herein is a method of treating obesity in a patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient, wherein substantially no loss of new blood vessels in fat deposits or other tissue compartments occur as compared to a patient being treated for obesity using an energy restricted diet alone.
[0052] For example, fumagillin-class molecules irreversibly inhibit enzymatic activity of MetAP2, leading to N-terminal acetylation and stabilization of these proteins at doses considerably lower than those required to suppress angiogenesis or tumor growth in vivo. Without being limited to any theory, the long-lasting covalent inhibition of MetAP2 enzymatic activity driven by such MetAP2 inhibitors may be responsible for the segregation of angiogenic effects from metabolic responses mediated by increased thioredoxin and/or glyceraldehyde-3- phosphate levels in vivo. Alternatively, anti-tumor effects driven by angiogenesis inhibition may require a more thorough starvation of the tumor by heavily restricting blood supply, which requires high doses. Metabolic effects, however, may require a minor and incomplete perturbation of the system which occurs at lower doses and without any obvious direct effect on blood vessels.
[0053] Treated patients used the disclosed methods may have a lower systemic exposure to said MetAP2 inhibitor as compared to a patient parenterally administered the same of amount of the MetAP2 inhibitor. In an exemplary embodiment, the disclosed methods may result in less accumulation in the reproductive tract (e.g. testis) of a patient, for example, as compared to the same amount of MetAP2 inhibitor subcutaneously administered.
[0054] Disclosed methods of treating obesity e.g by non-parenterally administering a MetAP2 inhibitor, may result in decreased body fat and a substantial maintenance of muscle mass in said patient. In certain embodiments, upon administration, fat oxidation is enhanced in a patient as compared to a patient on a restricted food intake diet alone. For example, provided herein is a method of decreasing body fat in an overweight or obese patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient resulting in body fat reduction, and wherein said patient substantially maintains muscle mass during the body fat reduction. Such a patient may retain substantially more muscle mass as compared to body fat reduction in a patient using an energy restricted diet alone.
[0055] In some embodiments, disclosed methods, upon administration of said MetAP2 inhibitor e.g. daily or weekly, for about 3, 4, 5 or 6 months or more may result in at least a 5%, 10%, 20%, or 30%, or more weight loss based on the patient's original weight. In an embodiment, weight loss following treatment with therapeutically effective doses of MetAP2 inhibitors may substantially cease once a patient attains a normal body composition. Without being limited to an theory, this may be due to reliance of the mechanism on re-establishing tone of adrenergic signal transduction in tissues such as fat, liver, and/or skeletal muscle.
[0056] In an embodiment, provided herein is a method of maintaining a specified weight in a formerly obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. [0057] Also provided herein is a method for controlling or preventing hepatic steatosis in an obese patient being treated for obesity, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. In another embodiment, a method for improving liver function in an obese patient is provided, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. For example, a method of restoring normal metabolic action in an obese patient in need thereof is provided, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. In an embodiment, a method of reducing weight of a patient in a patient in need thereof is provided comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient wherein the metabolic rate of the patient is not substantially reduced as compared to the metabolic rate of a diet only patient on an energy restricted diet alone. In a different embodiment, a method of restoring and/or maintaining thyroid hormone concentrations in an obese patient is provided, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
[0058] In an embodiment, a method of improving exercise capacity in a patient in need thereof is provided that comprises administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
[0059] Also provided herein is a method of activating brown fat function in a patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
[0060] Contemplated herein is a method of reducing the amount or frequency of administering supplemental insulin in a patient suffering from type 2 diabetes, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient. Such treatment may be directed to an obese or non-obese patient.
[0061] In an embodiment, a method for improving surgical outcome in an obese patient in need thereof by reducing weight of said patient is provided comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient before non-acute surgery, thereby reducing liver and/or abdominal fat in said patient and improving surgical outcome. Such surgeries may include bariatric surgery, cardiovascular surgery, abdominal surgery, or orthopedic surgery. [0062] In addition to being overweight or obese, a subject can further have an overweight- or obesity-related co-morbidities, i.e., diseases and other adverse health conditions associated with, exacerbated by, or precipitated by being overweight or obese. Because being overweight or obese is associated with other adverse health conditions or co-morbidities, for example diabetes, administering MetAP2 inhibitors brings a benefit in ameliorating, arresting development of or, in some cases, even eliminating, these overweight- or obesity- related conditions or co-morbidities. In some embodiments, methods provided herein may further include administering at least one other agent that is directed to treatment of these overweight- or obesity- related conditions.
[0063] Contemplated other agents include those administered to treat type 2 diabetes such as sulfonylureas (e.g., chlorpropamide, glipizide, glyburide, glimepiride); meglitinides (e.g., repaglinide and nateglinide); biguanides (e.g., metformin); thiazolidinediones (rosiglitazone, troglitazone, and pioglitazone); glucagon-like 1 peptide mimetics (e.g. exenatide and liraglutide); sodium-glucose cotransporter inhibitors (e.g., dapagliflozin), renin inhibitors, and alpha-glucosidase inhibitors (e.g., acarbose and meglitol), and/or those administered to treat cardiac disorders and conditions, such hypertension, dyslipidemia, ischemic heart disease, cardiomyopathy, cardiac infarction, stroke, venous thromboembolic disease and pulmonary hypertension, which have been linked to overweight or obesity, for example, chlorthalidone; hydrochlorothiazide; indapamide, metolazone; loop diuretics (e.g., bumetanide, ethacrynic acid, furosemide, lasix, torsemide); potassium- sparing agents (e.g., amiloride hydrochloride, spironolactone, and triamterene); peripheral agents (e.g., reserpine); central alpha-agonists (e.g., clonidine hydrochloride, guanabenz acetate, guanfacine hydrochloride, and methyldopa); alpha-blockers (e.g., doxazosin mesylate, prazosin hydrochloride, and terazosin hydrochloride); beta-blockers (e.g., acebutolol, atenolol, betaxolol, nisoprolol fumarate, carteolol hydrochloride, metoprolol tartrate, metoprolol succinate, Nadolol, penbutolol sulfate, pindolol, propranolol hydrochloride, and timolol maleate); combined alpha- and beta-blockers (e.g., carvedilol and labetalol hydrochloride); direct vasodilators (e.g., hydralazine hydrochloride and minoxidil); calcium antagonists (e.g., diltiazem hydrochloride and verapamil hydrochloride); dihydropyridines (e.g., amlodipine besylate, felodipine, isradipine, nicardipine, nifedipine, and nisoldipine); ACE inhibitors (benazepril hydrochloride, captopril, enalapril maleate, fosinopril sodium, lisinopril, moexipril, quinapril hydrochloride, ramipril, trandolapril); angiotensin II receptor blockers (e.g., losartan potassium, valsartan, and Irbesartan); and combinations thereof, as well as statins such as mevastatin, lovastatin, pravastatin, simvastatin, velostatin, dihydrocompactin, fluvastatin, atorvastatin, dalvastatin, carvastatin, crilvastatin, bevastatin, cefvastatin, rosuvastatin, pitavastatin, and glenvastatin., typically for treatment of dyslipidemia.
[0064] Other agents that may be co-administered (e.g. sequentially or simultaneously) include agents administered to treat ischemic heart disease including statins, nitrates (e.g., Isosorbide Dinitrate and Isosorbide Mononitrate), beta-blockers, and calcium channel antagonists, agents administered to treat cardiomyopathy including inotropic agents (e.g., Digoxin), diuretics (e.g., Furosemide), ACE inhibitors, calcium antagonists, anti- arrhythmic agents (e.g., Sotolol, Amiodarone and Disopyramide), and beta-blockers, agents administered to treat cardiac infarction including ACE inhibitors, Angiotensin II receptor blockers, direct vasodilators, beta blockers, anti- arrhythmic agents and thrombolytic agents (e.g., Alteplase, Retaplase, Tenecteplase, Anistreplase, and Urokinase), agents administered to treat strokes including anti-platelet agents (e.g., Aspirin, Clopidogrel, Dipyridamole, and Ticlopidine), anticoagulant agents (e.g., Heparin), and thrombolytic agents, agents administered to treat venous thromboembolic disease including anti-platelet agents, anticoagulant agents, and thrombolytic agents, agents administered to treat pulmonary hypertension include inotropic agents, anticoagulant agents, diuretics, potassium (e.g., K-dur), vasodilators (e.g., Nifedipine and Diltiazem), Bosentan, Epoprostenol, and Sildenafil, agents administered to treat asthma include bronchodilators, anti-inflammatory agents, leukotriene blockers, and anti-Ige agents. Particular asthma agents include Zafirlukast, Flunisolide, Triamcinolone, Beclomethasone, Terbutaline, Fluticasone, Formoterol, Beclomethasone, Salmeterol, Theophylline, and Xopenex, agents administered to treat sleep apnea include Modafinil and amphetamines, agents administered to treat nonalcoholic fatty liver disease include antioxidants (e.g., Vitamins E and C), insulin sensitizers (Metformin, Pioglitazone, Rosiglitazone, and Betaine), hepatoprotectants, and lipid-lowering agents, agentsadministered to treat osteoarthritis of weight-bearing joints include Acetaminophen, non-steroidal anti-inflammatory agents (e.g., Ibuprofen, Etodolac, Oxaprozin, Naproxen, Diclofenac, and Nabumetone), COX-2 inhibitors (e.g., Celecoxib), steroids, supplements (e.g. glucosamine and chondroitin sulfate), and artificial joint fluid, agents administered to treat Prader-Willi Syndrome include human growth hormone (HGH), somatropin, and weight loss agents (e.g., Orlistat, Sibutramine, Methamphetamine, Ionamin, Phentermine, Bupropion, Diethylpropion, Phendimetrazine, Benzphetermine, and Topamax), agents administered to treat polycystic ovary syndrome include insulin-sensitizers, combinations of synthetic estrogen and progesterone, Spironolactone, Eflornithine, and Clomiphene, agents administered to treat erectile dysfunction include phosphodiesterase inhibitors (e.g., Tadalafil, Sildenafil citrate, and Vardenafil), prostaglandin E analogs (e.g., Alprostadil), alkaloids (e.g., Yohimbine), and testosterone, agents administered to treat infertility include Clomiphene, Clomiphene citrate, Bromocriptine, Gonadotropin-releasing Hormone (GnRH), GnRH agonist, GnRH antagonist, Tamoxifen/nolvadex, gonadotropins, Human Chorionic Gonadotropin (HCG), Human Menopausal Gonadotropin (HmG), progesterone, recombinant follicle stimulating hormone (FSH), Urofollitropin, Heparin, Follitropin alfa, and Follitropin beta, agents administered to treat obstetric complications include Bupivacaine hydrochloride, Dinoprostone PGE2, Meperidine HCl, Ferro-folic-500/iberet-folic-500, Meperidine, Methylergonovine maleate, Ropivacaine HCl, Nalbuphine HCl, Oxymorphone HCl, Oxytocin, Dinoprostone, Ritodrine, Scopolamine hydrobromide, Sufentanil citrate, and Oxytocic, agents administered to treat depression include serotonin reuptake inhibitors (e.g., Fluoxetine, Escitalopram, Citalopram, Paroxetine, Sertraline, and Venlafaxine); tricyclic antidepressants (e.g., Amitriptyline, Amoxapine, Clomipramine, Desipramine, Dosulepin hydrochloride, Doxepin, Imipramine, Iprindole, Lofepramine, Nortriptyline, Opipramol, Protriptyline, and Trimipramine); monoamine oxidase inhibitors (e.g., Isocarboxazid, Moclobemide, Phenelzine,
Tranylcypromine, Selegiline, Rasagiline, Nialamide, Iproniazid, Iproclozide, Toloxatone, Linezolid, Dienolide kavapyrone desmethoxyyangonin, and Dextroamphetamine); psychostimulants (e.g., Amphetamine, Methamphetamine, Methylphenidate, and Arecoline); antipsychotics (e.g., Butyrophenones, Phenothiazines, Thioxanthenes, Clozapine, Olanzapine, Risperidone, Quetiapine, Ziprasidone, Amisulpride, Paliperidone, Symbyax, Tetrabenazine, and Cannabidiol); and mood stabilizers (e.g., Lithium carbonate, Valproic acid, Divalproex sodium, Sodium valproate, Lamotrigine, Carbamazepine, Gabapentin, Oxcarbazepine, and Topiramate), agents administered to treat anxiety include serotonin reuptake inhibitors, mood stabilizers, benzodiazepines (e.g., Alprazolam, Clonazepam, Diazepam, and Lorazepam), tricyclic antidepressants, monoamine oxidase inhibitors, and beta-blockers, and other weight loss agents, including serotonin and noradrenergic re-uptake inhibitors; noradrenergic re-uptake inhibitors; selective serotonin re-uptake inhibitors; and intestinal lipase inhibitors. Particular weight loss agents include orlistat, sibutramine, methamphetamine, ionamin, phentermine, bupropion, diethylpropion, phendimetrazine, benzphetermine, and topamax.
[0065] In some embodiments, contemplated methods may further comprising assessing one or more indices of on-going weight loss, e.g. the ketone body production level in a patient; and optionally adjusting the amount administered; thereby optimizing the therapeutic efficacy of said MetAP2 inhibitor.
Administration and Formulation
[0066] Contemplated herein are formulations suitable for non-parenteral administration of MetAP2 inhibitors. For example, in certain embodiments, a patient may have a lower systemic exposure (e.g. at least about 2, 3, 5, 10, 20, or at least about 30% less systemic exposure) to the non-parenterally administered (e.g. oral administration) of a MetAP2 inhibitor as compared to a patient parenterally administered (e.g. subcutaneously) the same dose of the MetAP2 inhibitor. For example, non-parenterally administered (e.g. orally administered) MetAP2 inhibitors (e.g. an irreversible inhibitor) may bind less to MetAP2 as compared to subcutaneously administered MetAP2 inhibitors.
[0067] Contemplated non-parenteral administration includes oral, buccal, transdermal
(e.g. by a dermal patch), topical, inhalation, or sublingual administration, or e.g., ocular, pulmonary, nasal, rectal or vaginal administration.
[0068] In another embodiment, provided herein are effective dosages, e.g. a daily dosage of a MetAP2 inhibitor, that may not substantially modulate or suppress angiogenesis. For example, provided here are methods that include administering doses of MetAP2 inhibitors that are effective for weight loss, but are significantly smaller doses than that necessary to modulate and/or suppress angiogenesis (which may typically require about 12.5mg/kg to about 50mg/kg or more). For example, contemplated dosage of a MetAP2 inhibitor in the methods described herein may include administering about 25 mg/day, about 10 mg/day, about 5 mg/day, about 3 mg/day, about 2 mg/day, about 1 mg/day, about 0.75 mg/day, about 0.5 mg/day, about 0.1 mg/day, about 0.05 mg/day, or about 0.01 mg/day. [0069] For example, an effective amount of the drug for weight loss in a patient may be about 0.0001 mg/kg to about 25 mg/kg of body weight per day. For example, a contemplated dosage may from about 0.001 to 10 mg/kg of body weight per day, about 0.001 mg/kg to 1 mg/kg of body weight per day, about 0.001 mg/kg to 0.1 mg/kg of body weight per day or about 0.005 to about 0.04 mg/kg or about 0.005 to about 0.049 mg/kg of body weight a day. In an embodiment a MetAP2 inhibitor such as disclosed herein (e.g. O-(4- dimethlyaminoethoxycinnamoyl)fumagillol), may be administered about 0.005 to about 0.04 mg/kg of a patient.
[0070] For example, provided herein is a method for treating obesity in a patient in need thereof, comprising administering, parenterally (e.g. intravenously) or non-parenterally, about 0.005 to about 0.04 mg/kg of a MetAP2 inhibitor selected from O-(4- dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof (for example, an oxalate salt), to said patient. Such a method, upon administration of said MetAP2 inhibitor e.g. daily or weekly, for about 3, 4 , 5 or 6 months or more may result in at least a 10%, 20%, 30%, or 40% or more weight loss based on the patient's original weight.
[0071] Contemplated methods may include administration of a composition comprising a MetAP2 inhibitor, for example, hourly, twice hourly, every three to four hours, daily, twice daily, 1, 2, 3 or 4 times a week, every three to four days, every week, or once every two weeks depending on half-life and clearance rate of the particular composition or inhibitor.
[0072] Treatment can be continued for as long or as short a period as desired. The compositions may be administered on a regimen of, for example, one to four or more times per day. A suitable treatment period can be, for example, at least about one week, at least about two weeks, at least about one month, at least about six months, at least about 1 year, or indefinitely. A treatment period can terminate when a desired result, for example a weight loss target, is achieved. For example, when about loss of about 20% body weight, about 30% body weight or more has been achieved. A treatment regimen can include a corrective phase, during which a MetAP2 inhibitor dose sufficient to provide reduction of excess adiposity is administered, followed by a maintenance phase, during which a lower MetAP2 inhibitor dose sufficient to prevent re-development of excess adiposity is administered. [0073] For pulmonary (e.g., intrabronchial) administration, MetAP2 inhibitors can be formulated with conventional excipients to prepare an inhalable composition in the form of a fine powder or atomizable liquid. For ocular administration, MetAP2 inhibitors can be formulated with conventional excipients, for example, in the form of eye drops or an ocular implant. Among excipients useful in eye drops are viscosifying or gelling agents, to minimize loss by lacrimation through improved retention in the eye.
[0074] Liquid dosage forms for oral or other administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active agent(s), the liquid dosage forms may contain inert diluents 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, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the ocular, oral, or other systemically-delivered compositions can also include adjuvants such as wetting agents, and emulsifying and suspending agents.
[0075] Dosage forms for topical or transdermal administration of an inventive pharmaceutical composition may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants, or patches. The active agent is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. For example, cutaneous routes of administration are achieved with aqueous drops, a mist, an emulsion, or a cream.
[0076] Transdermal patches may have the added advantage of providing controlled delivery of the active ingredients to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. [0077] When administered in lower doses, injectable preparations are also contemplated herein, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
[0078] Compositions for rectal or vaginal administration may be suppositories which can be prepared by mixing a MetAP2 inhibitor with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active agent(s). Alternatively, contemplated formulations can be administered by release from a lumen of an endoscope after the endoscope has been inserted into a rectum of a subject.
[0079] Oral dosage forms, such as capsules, tablets, pills, powders, and granules, may be prepared using any suitable process known to the art. For example, a MetAP2 inhibitor may be mixed with enteric materials and compressed into tablets.
[0080] Alternatively, formulations of the invention are incorporated into chewable tablets, crushable tablets, tablets that dissolve rapidly within the mouth, or mouth wash..
EXAMPLES
[0081] The examples which follow are intended in no way to limit the scope of this invention but are provided to illustrate aspects of the disclosed methods. Many other embodiments of this invention will be apparent to one skilled in the art.
Example 1: Fumagillin increases levels of Thioredoxin-1 (TXN) in a dose-dependent manner
[0082] B 16F10 cells were treated with increasing concentrations of fumagillin. N- terminal chymotryptic fragments of TXN were measured by liquid chromatography/mass spectrometry (LC/MS). Fumagillin significantly increased the cellular level of TXN containing an intact N-terminal methionine [TXN(I -4)] and caused significant accumulation of N-acetyl TXN(I -4), consistent with a role for fumagillin as a MetAP2 inhibitor. See FIGs. 1-2. The EC50 of fumagillin to increase Met-TXN acetylated Met-TXN levels is < 10 nM. [0083] Similar results were seen in Cos-1 and 293T cells that were transfected with
FLAG-tagged TXN in the absence (TXN) and presence (TXN + F) of fumagillin. Fumagillin significantly increased the expression on TXN in both Cos-1 and 293T cells. See FIG. 3. EC50 of fumagillin to increase TXN levels is < 10 nM. See FIG. 4.
Example 2: Oral administration of fumagillin reduces body fat in mammals
[0084] Oral administration of the MetAP2 inhibitor fumagillin causes an increase in circulating ketone bodies, as measured by concentration of beta hydroxybutyrate, which indicates an increase in the breakdown of stored fat. C57BL/6 mice conditioned on a high fat diet (also referred to as a diet-induced obese diet or DIO) were treated with fumagillin (ZGN- 201) administered by oral gavage at doses of 0.3 or 3 mg/kg daily for 10 days resulting in an increase in beta hydroxybutyrate (FIG. 5A) concentrations, without an increase in free fatty acid concentration (FIG. 5B) as well as a reduction in body weight by 7 to 15 percent (data not shown). Increased hydroxybutyrate concentration persisted at longer time points, as was seen in C57BL/6 mice conditioned on a high fat diet and treated with fumagillin (ZGN-201) as a diet admixture at a dose of 1 mg/kg daily for 250 days (FIG. 5C).
[0085] In addition to showing an increase in beta hydroxybutyrate concentration,
C57BL/6 mice conditioned on a high fat diet and treated with fumagillin (ZGN-201) as a diet admixture (1 mg/kg daily for 250 days) also experienced a reduction in body fat by approximately 40 percent (FIG. 6). Once attaining the maximum weight reduction, mice treated daily with 1 mg/kg fumagillin maintained a weight that was 25% less than that of age- matched mice maintained on normal mouse chow diet (lean mice, FIG. 6) for the duration of the experiment. The size and appearance of epididymal fat, peri-renal fat and liver in fumagillin-treated mice was more similar to that of age-matched lean mice than that of untreated mice on a high fat diet (FIG 7). Retroperitoneal fat pad weight in fumagillin-treated mice was also more similar to that of age-matched lean mice than that of untreated mice on a high fat diet (FIG. 8).
[0086] Adipose tissue in mice fed a high-fat diet show distinctive crown-like structures not seen in lean mice. These crown-like structures were also absent from the adipose tissue of fumagillin-treated mice. Additionally, adipocyte size in fumagillin-treated mice was more similar to that of age-matched lean mice than that of untreated mice on a high fat diet. [0087] Fasted glucose and insulin levels, as well as insulin-glucose product, in fumagillin-treated mice was more similar to that of age-matched lean mice than that of untreated mice on a high fat diet (FIG. 9), indicating that fumagillin normalizes sensitivity to insulin in mice on a high fat diet.
Example 3: Fumagillin reduces food intake during the peak of weight loss
[0088] High fat diet-fed C57BL/6 mice were treated with 1 mg/kg of the MetAP2 inhibitor fumagillin (ZGN-201, squares) or vehicle (diamonds) for 14 days (FIG. 10). Body weights (left panel) were determined daily, while food intake (right panel) was assessed every other day by measuring the food consumed by each cage of three mice over the two day period. Data shown in FIG. 10 are means + SEM, n = 9 animals. As illustrated in FIG. 10, weight loss during treatment with MetAP2 inhibitors follows a sequence of three distinct phases. First, during the initial two to three days of treatment, no overt changes in food intake or body weight are observed. During this time period key changes in MetAP2 substrates occur, including glyceraldehyde-3-phosphate dehydrogenase and thioredoxin (data not shown). Second, the animals enter a period of rapid weight loss that proceeds until essentially all excess fat deposits are consumed. During this second period, fat oxidation is enhanced and food intake is inhibited.
[0089] Weight loss following treatment with therapeutically relevant doses of MetAP2 inhibitors stops once animals attain a normal body composition. Once weight loss reaches a nadir, fumagillin-treated animals enter a third phase in which their food intake returns to normal levels despite maintaining a normal body weight and improved metabolic control evidenced by reduced insulin and glucose concentrations. This third phase can be maintained indefinitely provided the drug is continued (see FIG. 10), or presumably can be maintained in practice through other complementary pharmacologic, dietary, or lifestyle interventions.
Example 4: Oral administration of fumagillin results in fat loss with minimal change in lean tissue
[0090] C57BL/6 mice were conditioned on a low-fat diet (LF), a high fat diet (HF), or a high-fat diet and treated with fumagillin (ZGN-201) administered by oral gavage at doses of 0.1 or 0.3 mg/kg for 28 days. Either dose of fumagillin caused a reduction in body weight down to the level of low fat fed mice. To determine whether the weight loss resulted from loss of fat or loss of lean tissue, the percentage change in fat tissue and lean tissue was determined for LF mice, HF mice, HF mice treated with 0.1 mg/kg fumagillin, and HF mice treated with 0.3 mg/kg fumagillin. As shown in FIG. 11, HF mice treated with either dose of fumagillin experienced a 40-50% reduction in fat (compared to an increase in fat in both the LF and HF control mice), but only a 5-10% reduction in lean tissue, indicating that fumagillin-induced weight loss results primarily from loss of fat, rather than loss of lean tissue.
[0091] Upon histological examination, the studies of treated obese mice show no effect on growth of new blood vessels in growing fat depots and no effect on blood vessel size or density when compared to animals in which weight loss has been induced by simple energy (food) restriction.
Example 5: Orally administered compounds having MetAP-2 inhibitory cores cause weight loss in diet-induced obese mice
[0092] A weight loss study was conducted in obese mice. The mice in this study were not genetically obese, but prior to and during the study, obesity was induced by a high-fat diet. Twelve week-old C57BL/6NTac mice, maintained on a 60% fat diet prior to and during the study, were separated into seven groups, eight animals per group. Average body weight of the mice was approximately 47 g at the start of the study.
[0093] Each of six groups was orally administered 1.0 mg/kg of a compound (fumagillin and compounds B. C, D, E, F and G as disclosed herein), in 10% gelucire in deoinised water. Each of these six groups was administered a different compound. One group was orally administered fumagillin at 1.0 mg/kg (group 201) in 10% gelucire in deoinised water, and one group was administered 10% gelucire in deoinised water (vehicle). Mice received administrations once a day for 7 days.
[0094] Data show that mice administered fumagillin lost the most weight over the course of the 8 days (Fig. 13). Mice in groups B, C, E, and E also lost weight over the course of the 8 days, with mice in group 233 losing the most weight of these four groups (Fig. 13). (Data analyzed by ANCOVA with body weight on Day 1 as covariate follwed by multiple tests against vehicle group: *p<0.05; **p<0.01, ***p<0.001. ) Example 6
[0095] A weight loss study was conducted in obese mice, similar to Example 5.
Twelve week-old C57BL/6NTac mice, maintained on a 60% fat diet prior to and during the study, were separated into seven groups, eight animals per group. Average body weight of the mice was approximately 47 g at the start of the study.
[0096] The group was orally administered 100.0 mg/kg of a compound H, in 10% gelucire in deoinised water. Mice received administrations once a day for 7 days. After 7 days, the average weight loss of the mice was 13.2%
Incorporation by Reference
[0097] References and citations to other documents, such as patents, patent applications, patent publications, journals, books, papers, web contents, have been made throughout this disclosure. All such documents are hereby incorporated herein by reference in their entirety for all purposes.
Equivalents
[0098] Various modifications of the invention and many further embodiments thereof, in addition to those shown and described herein, will become apparent to those skilled in the art from the full contents of this document, including references to the scientific and patent literature cited herein. The subject matter herein contains important information, exemplification and guidance that can be adapted to the practice of this invention in its various embodiments and equivalents thereof.
[0099] What is claimed is :

Claims

1. A method of treating obesity in a patient in need thereof, comprising non-parenterally administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
2. The method of claim 1, wherein said pharmaceutically effective amount does not substantially modulate or suppress angiogenesis.
3. The method of claim 1 or 2, wherein said patient has a lower systemic exposure to said MetAP2 inhibitor as compared to a patient parenterally administered the same of amount of the MetAP2 inhibitor.
4. The method of any one of claims 1-3, wherein said MetAP2 inhibitor is administered orally.
5. The method of any one of claims 1-3, wherein said MetAP2 inhibitor is administered buccally or sublingually.
6. The method of any one of claims 1-3, wherein said MetAP2 inhibitor is administered transdermally.
7. The method of any one of claims 1-3, wherein said MetAP2 inhibitor is administered via inhalation.
8. The method of any one of claims 1-3, wherein said MetAP2 inhibitor is administered rectally.
9. The method of any one of claims 1-8, wherein said MetAP2 inhibitor is a substantially irreversible inhibitor.
10. The method of any one of claims 1-9, wherein said MetAP2 inhibitor is selected from the group consisting of a fumagillin, fumagillol or fumagillin ketone derivative, siRNA, shRNA, an antibody, or a antisense compound.
11. The method of any one of claims 1-10, wherein said MetAP2 inhibitor is selected from O-(4-dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof.
12. The method of any one of claims 1-11, wherein said non-parenterally administering results in decreased body fat and a substantial maintenance of muscle mass in said patient.
13. The method of any one of claims 1-12, wherein upon administration, fat oxidation is enhanced as compared to a patient on a restricted food intake diet alone.
14. A method for controlling or preventing hepatic steatosis in an obese patient being treated for obesity, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
15. A method for improving liver function in an obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
16. A method of treating obesity in a patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient, wherein substantially no loss of new blood vessels in fat deposits occur as compared to a patient being treated for obesity using an energy restricted diet alone.
17. A method of improving exercise capacity in a patient in need thereof comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
18. A method of reducing weight of a patient in a patient in need thereof comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient wherein the metabolic rate of the patient is not substantially reduced as compared to the metabolic rate of a diet only patient on an energy restricted diet alone.
19. A method of restoring normal metabolic action in an obese patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
20. A method of decreasing body fat in an overweight or obese patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient resulting in body fat reduction, and wherein said patient substantially maintains muscle mass during the body fat reduction.
21. The method of claim 20, wherein the patient retains substantially more muscle mass as compared to body fat reduction in a patient using an energy restricted diet alone.
22. A method of activating brown fat function and/or increasing brown fat tissue mass in a patient in need thereof, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
23. A method of restoring and/or maintaining thyroid hormone concentrations in an obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
24. The method of any one of claims 14-23, wherein the MetAP2 inhibitor is administered non-parenterally.
25. The method of any one of claims 14-24, wherein said pharmaceutically effective amount does not substantially modulate or suppress angiogenesis.
26. The method of claim 25, wherein said patient has a lower systemic exposure to said MetAP2 inhibitor as compared to a patient parenterally administered the same amount of the MetAP2 inhibitor.
27. The method of any one of claims 14-26, wherein said MetAP2 inhibitor is administered orally.
28. The method of any one of claims 14-26, wherein said MetAP2 inhibitor is administered buccally or sublingually.
29. The method of any one of claims 14-26, wherein said MetAP2 inhibitor is administered topically or transdermally.
30. The method of any one of claims 14-26, wherein said MetAP2 inhibitor is administered via inhalation.
31. The method of any one of claims 14-24, wherein said MetAP2 inhibitor is administered rectally.
32. The method of any one of claims 14-26, wherein said MetAP2 inhibitor is administered subcutaneously or intravenously.
33. The method of any one of claims 14-32, wherein said MetAP2 inhibitor is a substantially irreversible inhibitor.
34. The method of any one of claims 14-33, wherein said MetAP2 inhibitor is selected from the group consisting of a fumagillin, fumagillol or fumagillin ketone derivative, siRNA, shRNA, an antibody, or an antisense compound.
35. The method of any one of claims 15-36, wherein said MetAP2 inhibitor is selected from O-(4-dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof.
36. A method for reducing the amount or frequency of administering supplemental insulin in a patient suffering from type 2 diabetes, comprising a pharmaceutically effective amount of a MetAP2 inhibitor.
37. A method for improving surgical outcome in an obese patient in need thereof by reducing weight of said patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient before non-acute surgery, thereby reducing liver and/or abdominal fat in said patient and improving surgical outcome.
38. The method of claim 37, wherein the surgery is chosen from the group: bariatric surgery, cardiovascular surgery, abdominal surgery, or orthopedic surgery.
39. A method of maintaining a specified weight in a formerly obese patient, comprising administering a pharmaceutically effective amount of a MetAP2 inhibitor to said patient.
40. The method of claim 39, wherein the pharmaceutically effective amount is less as compared to the amount needed to reduce weight or body fat.
41. The method of any one of claims 37-40, wherein said pharmaceutically effective amount does not substantially modulate or suppress angiogenesis.
42. The method of any one of claims 37-41, wherein said MetAP2 inhibitor is a substantially irreversible inhibitor.
43. The method of any one of claims 37-42, wherein said MetAP2 inhibitor is selected from the group consisting of a fumagillin, fumagillol or fumagillin ketone derivative, siRNA, shRNA, an antibody, or an antisense compound.
44. The method of any one of claims 37-43, wherein said MetAP2 inhibitor is selected from O-(4-dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof.
45. The method of any one of claims 1-44, further comprising co-administering an additional weight loss agent.
46. The method of any one of claims 1-45, further comprising administering a food restricted diet to said patient.
47. The method of any one of claims 1-46, further comprising: assessing the ketone body production level in said patient; and optionally adjusting the amount administered; thereby optimizing the therapeutic efficacy of said MetAP2 inhibitor.
48. The method of any one of claims 1-47, wherein said patient incurs greater than or about equal to 20% weight loss after about 6 months of said administration.
49. The method of any one of claims 1-48, wherein said administration occurs at least daily.
50. The method of any one of claims 1-48, wherein said administration occurs at least weekly.
51. A method for treating obesity in a patient in need thereof, comprising administering about 0.005 to about 0.049 mg/kg of a MetAP2 inhibitor selected from O-(4- dimethylaminoethoxycinnamoyl)fumagillol and pharmaceutically acceptable salts thereof, to said patient.
52. The method of claim 51, wherein the MetAP2 inhibitor is an oxalate salt of O-(4- dimethylaminoethoxycinnamoyl)fumagillol.
53. The method of claim 51, wherein said administering occurs daily.
54. The method of claim 51, wherein said administering occurs 1, 2, 3 or 4 times a week.
55. The method of any one of claims 51-54, wherein the O-(4- dimethylaminoethoxycinnamoyl)fumagillol is administered parenterally or non-parenterally.
56. The method of any one of claims 51-55, wherein the O-(4- dimethylaminoethoxycinnamoyl)fumagillol is administered intravenously.
57. The method of any one of claims 51-56, wherein upon administration of said MetAP2 inhibitor daily for about 6 months results in at least a 20% weight loss of the patient's original weight.
PCT/US2009/066816 2008-12-04 2009-12-04 Method of treating an overweight or obese subject WO2010065883A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US13/133,060 US8642650B2 (en) 2008-12-04 2009-12-04 Methods of treating an overweight or obese subject
US13/568,800 US8367721B2 (en) 2008-12-04 2012-08-07 Methods of treating an overweight or obese subject
US14/141,910 US20150209320A1 (en) 2008-12-04 2013-12-27 Methods of treating an overweight or obese subject
US15/492,294 US20180064677A1 (en) 2008-12-04 2017-04-20 Methods of Treating an Overweight or Obese Subject
US16/113,436 US20190224156A1 (en) 2008-12-04 2018-08-27 Methods of Treating an Overweight or Obese Subject

Applications Claiming Priority (22)

Application Number Priority Date Filing Date Title
US11987508P 2008-12-04 2008-12-04
US11988508P 2008-12-04 2008-12-04
US11988808P 2008-12-04 2008-12-04
US11989108P 2008-12-04 2008-12-04
US11987708P 2008-12-04 2008-12-04
US11988608P 2008-12-04 2008-12-04
US11988408P 2008-12-04 2008-12-04
US11988108P 2008-12-04 2008-12-04
US11987208P 2008-12-04 2008-12-04
US61/119,888 2008-12-04
US61/119,875 2008-12-04
US61/119,885 2008-12-04
US61/119,884 2008-12-04
US61/119,872 2008-12-04
US61/119,891 2008-12-04
US61/119,881 2008-12-04
US61/119,886 2008-12-04
US61/119,877 2008-12-04
US27568809P 2009-08-03 2009-08-03
US61/275,688 2009-08-03
US26019409P 2009-11-11 2009-11-11
US61/260,194 2009-11-11

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US13/133,060 A-371-Of-International US8642650B2 (en) 2008-12-04 2009-12-04 Methods of treating an overweight or obese subject
US13/568,800 Continuation US8367721B2 (en) 2008-12-04 2012-08-07 Methods of treating an overweight or obese subject
US14/141,910 Continuation US20150209320A1 (en) 2008-12-04 2013-12-27 Methods of treating an overweight or obese subject

Publications (1)

Publication Number Publication Date
WO2010065883A2 true WO2010065883A2 (en) 2010-06-10

Family

ID=42233896

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/066816 WO2010065883A2 (en) 2008-12-04 2009-12-04 Method of treating an overweight or obese subject

Country Status (2)

Country Link
US (5) US8642650B2 (en)
WO (1) WO2010065883A2 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012064928A1 (en) * 2010-11-10 2012-05-18 Zafgen Corporation Methods and compositions for treating thyroid hormone related disorders
WO2012064838A1 (en) 2010-11-09 2012-05-18 Zafgen Corporation Crystalline solids of a metap-2 inhibitor and methods of making and using same
WO2012075026A1 (en) * 2010-11-29 2012-06-07 Zafgen Corporation Methods of treating obesity using an effective dose of a metap-2 inhibitor
WO2012075020A1 (en) * 2010-11-29 2012-06-07 Zafgen Corporation Treatment of obesity using non-daily administration of 6 - 0 - (4 - dimethylaminoethoxy) cinnamoyl fumagillol
WO2012074968A1 (en) * 2010-11-29 2012-06-07 Zafgen Corporation Methods of reducing risk of hepatobiliary dysfunction during rapid weight loss with metap-2 inhibitors
WO2012154676A1 (en) * 2011-05-06 2012-11-15 Zafgen Corporation Partially saturated tricyclic compounds and methods of making and using same
US8367721B2 (en) 2008-12-04 2013-02-05 Zafgen, Inc. Methods of treating an overweight or obese subject
WO2013109735A1 (en) * 2012-01-18 2013-07-25 Zafgen, Inc. Tricyclic sulfone compounds and methods of making and using same
WO2013169727A1 (en) * 2012-05-07 2013-11-14 Zafgen, Inc. Polymorphic salt of the oxalate salt of 6 - o - ( 4 - dimethylaminoethoxy) cinnarnoyl fumagillol and methods of making and using same
WO2014039831A1 (en) 2012-09-07 2014-03-13 Takeda Pharmaceutical Company Limited SUBSTITUTED-1,4-DIHYDROPYRAZOLO[4,3-b]INDOLES
JP2014507477A (en) * 2011-03-08 2014-03-27 ザフゲン,インコーポレイテッド Oxaspiro [2.5] octane derivatives and analogs
US8772333B2 (en) 2010-01-08 2014-07-08 Zafgen, Inc. Fumigillol type compounds and methods of making and using same
EP2763671A2 (en) * 2011-10-03 2014-08-13 Zafgen, Inc. Methods of treating age related disorders
US8815309B2 (en) 2010-01-08 2014-08-26 Zafgen, Inc. Methods of treating a subject with benign prostate hyperplasia
US8865746B2 (en) 2008-07-18 2014-10-21 Zafgen, Inc. Methods of treating an overweight or obese subject
JP2015504091A (en) * 2012-01-18 2015-02-05 ザフゲン,インコーポレイテッド Tricyclic sulfonamide compounds and methods for making and using the same
US20150126489A1 (en) * 2012-05-09 2015-05-07 Zafgen, Inc. Fumigillol compounds and methods of making and using same
US9067905B2 (en) 2009-10-09 2015-06-30 Zafgen, Inc. Sulphone compounds and methods of making and using same
US9187494B2 (en) 2011-05-06 2015-11-17 Zafgen, Inc. Aryl-substituted tricyclic sulfonamides as methionyl aminopeptidase 2 modulators
US9242997B2 (en) 2011-05-06 2016-01-26 Zafgen, Inc. Tricyclic pyrazole sulphonamide compunds and methods of making and using same
US9266896B2 (en) 2010-07-22 2016-02-23 Zafgen, Inc. Tricyclic compounds and methods of making and using same
US9321740B2 (en) 2011-01-26 2016-04-26 Zafgen, Inc. Tetrazole compounds and methods of making and using same
WO2016100778A1 (en) 2014-12-19 2016-06-23 Takeda Pharmaceutical Company Limited Fumagillol derivatives
US9434743B2 (en) 2012-03-02 2016-09-06 Takeda Pharmaceutical Company Limited Indazole derivatives
CN104350046B (en) * 2012-05-07 2016-11-30 扎夫根股份有限公司 The polymorphic salt of 6-O-(4-dimethylamino ethoxy) cinnamoyl aspergillus fumigatus cedrol oxalates and preparation and application thereof
US9561209B2 (en) 2012-11-05 2017-02-07 Zafgen, Inc. Methods of treating liver diseases
US9597309B2 (en) 2013-03-14 2017-03-21 Zafgen, Inc. Methods of treating renal disease and other disorders
US9649293B2 (en) 2010-04-07 2017-05-16 Zafgen, Inc. Methods of treating an overweight subject
US9656979B2 (en) 2008-12-04 2017-05-23 Zafgen, Inc. Methods of treating an overweight or obese subject
US9682965B2 (en) 2015-08-11 2017-06-20 Zafgen, Inc. Fumagillol heterocyclic compounds and methods of making and using same
US9839622B2 (en) 2012-05-08 2017-12-12 Zafgen, Inc. Methods of treating hypothalamic obesity
US9868717B2 (en) 2012-11-05 2018-01-16 Zafgen, Inc. Tricyclic sulphonamide compounds and methods of making and using same
US9944613B2 (en) 2015-08-11 2018-04-17 Zafgen, Inc. Fumagillol spirocyclic compounds and fused bicyclic compounds and methods of making and using same
US10174009B2 (en) 2012-11-05 2019-01-08 Zafgen, Inc. Tricyclic sulphonamide compounds and methods of making and using same

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9895449B2 (en) 2010-05-25 2018-02-20 Syndevrx, Inc. Polymer-conjugated MetAP2 inhibitors, and therapeutic methods of use thereof
DK2576638T3 (en) 2010-05-25 2021-03-15 Syndevrx Inc POLYMER CONJUGATED METAP2 INHIBITORS AND THERAPEUTIC PROCEDURES FOR USE
JP6420314B2 (en) 2013-04-10 2018-11-07 シンデブルックス,インコーポレイティド METAP2 inhibitor and method for treating obesity
CA3005450A1 (en) 2015-12-10 2017-06-15 Syndevrx, Inc. Fumagillol derivatives and polymorphs thereof
JP7022066B2 (en) 2016-01-11 2022-02-17 シンデブルックス,インコーポレイティド Treatment of tumors caused by metabolic dysfunction
EP3870231A1 (en) 2018-10-26 2021-09-01 Syndevrx, Inc. Biomarkers of metap2 inhibitors and applications thereof
CN109924485A (en) * 2019-03-29 2019-06-25 华熙生物科技股份有限公司 A kind of slimming health food and preparation method thereof containing hyaluronic acid

Family Cites Families (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5164410A (en) 1988-01-09 1992-11-17 Takeda Chemical Industries, Ltd. Fumagillol derivatives and pharmaceutical compositions thereof
PH26256A (en) 1988-08-12 1992-04-01 Fujisawa Pharmaceutical Co Oxaspiro [2,5] octane derivative
EP0682020A1 (en) 1988-09-01 1995-11-15 Takeda Chemical Industries, Ltd. Fumagillol derivatives useful as angiogenesis inhibitors
US5180738A (en) 1988-09-01 1993-01-19 Takeda Chemical Industries Fumagillol derivatives and pharmaceutical compositions thereof
US5166172A (en) 1988-09-01 1992-11-24 Takeda Chemical Industries, Ltd. Fumagillol derivatives and pharmaceutical compositions thereof
EP0387650B1 (en) 1989-03-06 1993-03-31 Takeda Chemical Industries, Ltd. 6-epifumagillols, production and use thereof
US5288722A (en) 1989-03-06 1994-02-22 Takeda Chemical Industries, Ltd. 6-amino-6-desoxyfumagillols, production and use thereof
US5290807A (en) 1989-08-10 1994-03-01 Children's Medical Center Corporation Method for regressing angiogenesis using o-substituted fumagillol derivatives
US6017954A (en) 1989-08-10 2000-01-25 Children's Medical Center Corp. Method of treating tumors using O-substituted fumagillol derivatives
EP0415294A3 (en) 1989-08-31 1991-06-12 Takeda Chemical Industries, Ltd. Cyclohexanol derivatives, production and use thereof
TW282399B (en) 1990-05-25 1996-08-01 Takeda Pharm Industry Co Ltd
EP0555693B1 (en) 1992-01-30 2001-09-05 Takeda Chemical Industries, Ltd. Method of producing highly watersoluble cyclodextrin complex
EP0602586B1 (en) 1992-12-16 1997-06-04 Takeda Chemical Industries, Ltd. Stable pharmaceutical composition of fumagillol derivatives
NZ304906A (en) 1995-03-27 1999-06-29 Sanofi Sa Use of fumagillol or a fumagillol ester and an optionally saturated alkylcarboxylic or alkyldicarboxylic acid to fight intestinal infections
WO1997013509A1 (en) 1995-10-11 1997-04-17 Fujisawa Pharmaceutical Co., Ltd. Vascular permeation inhibitor
EP0799616A1 (en) 1996-04-01 1997-10-08 Takeda Chemical Industries, Ltd. Oral composition comprising a fumagillol derivative
AU3968597A (en) 1996-08-02 1998-02-25 Children's Medical Center Corporation Method of regulating the female reproductive system through angiogenesis inhibitors
WO1998056372A1 (en) 1997-06-09 1998-12-17 Massachusetts Institute Of Technology TYPE 2 METHIONINE AMINOPEPTIDASE (MetAP2) INHIBITORS AND USES THEROF
DE69824750T2 (en) 1997-10-31 2005-07-07 Children's Medical Center Corp., Boston METHODS FOR REGULATING THE SIZE AND GROWTH OF A BLOODED NORMAL TISSUE
KR100357542B1 (en) 1998-05-15 2002-10-18 주식회사종근당 Fumagillol derivatives and preparation method thereof
KR100357541B1 (en) 1998-05-15 2002-10-18 주식회사종근당 5-Demthoxyfumagillol derivatives and processes for preparing the same
JP2000116337A (en) 1998-10-09 2000-04-25 Nippon Shokuhin Kako Co Ltd Pet food
US6383471B1 (en) 1999-04-06 2002-05-07 Lipocine, Inc. Compositions and methods for improved delivery of ionizable hydrophobic therapeutic agents
PT1177176E (en) 1999-04-28 2006-08-31 Aventis Pharma Gmbh DERIVATIVES OF TRIARILIC ACIDS AS LIGANDS OF PPAR RECEPTORS.
AU2001253418A1 (en) * 2000-04-12 2001-10-30 Smith Kline Beecham Corporation Compounds and methods
US6323228B1 (en) 2000-09-15 2001-11-27 Abbott Laboratories 3-substituted indole angiogenesis inhibitors
JP4212356B2 (en) 2000-11-01 2009-01-21 プラエシス ファーマシューティカルズ インク. Therapeutic agents for modulating angiogenesis and methods of use thereof
US6548477B1 (en) 2000-11-01 2003-04-15 Praecis Pharmaceuticals Inc. Therapeutic agents and methods of use thereof for the modulation of angiogenesis
US20040116495A1 (en) 2001-03-29 2004-06-17 Marino Jr. Joseph P. Compounds and methods
EP1436286B1 (en) 2001-09-27 2009-08-19 Equispharm Co., Ltd. Fumagillol derivatives and preparing method thereof
US6803382B2 (en) 2001-11-09 2004-10-12 Galderma Research & Development, S.N.C. Angiogenesis inhibitors and pharmaceutical and cosmetic use thereof
KR100451485B1 (en) 2002-03-28 2004-10-06 주식회사종근당 Inclusion compounds of fumagillol derivative or its salt, and pharmaceutical compositions comprising the same
US6989392B2 (en) 2002-06-18 2006-01-24 Abbott Laboratories 2-Aminoquinolines as melanin concentrating hormone receptor antagonists
US20040067266A1 (en) 2002-10-07 2004-04-08 Toppo Frank R. Weight loss compound
US20040157836A1 (en) 2002-10-08 2004-08-12 Comess Kenneth M. Sulfonamides having antiangiogenic and anticancer activity
WO2004078113A2 (en) 2003-03-04 2004-09-16 Pharmacia Corporation Treatment and prevention of obesity with cox-2 inhibitors alone or in combination with weight-loss agents
WO2005066197A2 (en) 2003-12-29 2005-07-21 Praecis Pharmaceuticals, Inc. Inhibitors of methionine aminopeptidase-2 and uses thereof
KR100552043B1 (en) 2004-02-28 2006-02-20 주식회사종근당 Composition for obesity treatment comprising fumagillol derivatives
MX2007000142A (en) 2004-06-30 2007-03-26 Combinatorx Inc Methods and reagents for the treatment of metabolic disorders.
US20060045865A1 (en) 2004-08-27 2006-03-02 Spherics, Inc. Controlled regional oral delivery
CN101142210A (en) 2005-01-26 2008-03-12 株式会社钟根堂 Fumagillol derivatives or method for preparation of fumagillol derivatives, and pharmaceutical compositions comprising the same
FR2886855B1 (en) 2005-06-08 2009-07-17 Agronomique Inst Nat Rech USE OF FUMAGILLIN AND ITS DERIVATIVES TO INCREASE BIODAVAILABILITY OF MACROCYLIC LACTONES
US20070078172A1 (en) 2005-06-16 2007-04-05 Jenrin Discovery Mao-b inhibitors useful for treating obesity
WO2009003110A2 (en) 2007-06-26 2008-12-31 Children's Medical Center Corporation Metap-2 inhibitor polymersomes for therapeutic administration
EP2217283A2 (en) 2007-11-28 2010-08-18 Mersana Therapeutics, Inc. Biocompatible biodegradable fumagillin analog conjugates
NZ590732A (en) 2008-07-18 2012-09-28 Zafgen Inc Methods of treating an overweight or obese subject using fumagillin
ES2641471T3 (en) 2008-10-06 2017-11-10 The Johns Hopkins University Quinoline compounds as inhibitors of human angiogenesis, methionine aminopeptidase, and SirT1, and disorders treatment procedures
US8906896B2 (en) 2008-10-24 2014-12-09 Ulrich Bierbach Platinum acridine anti-cancer compounds and methods thereof
WO2010065879A2 (en) 2008-12-04 2010-06-10 Zafgen Corporation Methods of treating an overweight or obese subject
US20120004162A1 (en) 2008-12-04 2012-01-05 Vath James E Methods of Treating an Overweight or Obese Subject
US8642650B2 (en) 2008-12-04 2014-02-04 Zafgen, Inc. Methods of treating an overweight or obese subject
WO2010065881A2 (en) 2008-12-04 2010-06-10 Zafgen Corporation Methods of treating an overweight or obese subject
KR20120083905A (en) 2009-10-09 2012-07-26 자프겐 코포레이션 Sulphone compounds for use in the treatment of obesity
US8815309B2 (en) 2010-01-08 2014-08-26 Zafgen, Inc. Methods of treating a subject with benign prostate hyperplasia
US20130023513A1 (en) 2010-01-12 2013-01-24 Hughes Thomas E Methods and Compositions for Treating Cardiovascular Disorders
WO2011127304A2 (en) 2010-04-07 2011-10-13 Zafgen Corporation Methods of treating an overweight subject
WO2011150338A1 (en) 2010-05-27 2011-12-01 Zafgen Corporation Methods of treating obesity
CN103249735B (en) 2010-07-22 2016-04-06 扎夫根股份有限公司 Tricyclic compound and preparation and application thereof
CA2814413A1 (en) 2010-10-12 2012-04-19 Zafgen, Inc. Sulphonamide compounds and methods of making and using same
MX2013005208A (en) 2010-11-09 2013-08-01 Zafgen Inc Crystalline solids of a metap-2 inhibitor and methods of making and using same.
WO2012064928A1 (en) 2010-11-10 2012-05-18 Zafgen Corporation Methods and compositions for treating thyroid hormone related disorders
US20140011870A1 (en) 2010-11-29 2014-01-09 Zafgen, Inc. Methods of Treating Obesity Using an Effective Dose of a METAP-2 Inhibitor
WO2012074968A1 (en) 2010-11-29 2012-06-07 Zafgen Corporation Methods of reducing risk of hepatobiliary dysfunction during rapid weight loss with metap-2 inhibitors
EP2646016B1 (en) 2010-11-29 2017-05-17 Zafgen, Inc. Treatment if obesity using non-daily administration of 6-0-(4-dimethylaminoethoxy)cinnamoyl fumagillol
BR112013018771A2 (en) 2011-01-26 2019-09-17 Zafgen Inc tetrazole compounds and methods for making and using them
WO2012154679A1 (en) 2011-05-06 2012-11-15 Zafgen Corporation Tricyclic pyrazole sulfonamide compounds and methods of making and using same
CA2835195A1 (en) 2011-05-06 2012-11-15 Zafgen, Inc. Tricyclic sulfonamide compounds and methods of making and using same
US9290472B2 (en) 2011-05-06 2016-03-22 Zafgen, Inc. Partially saturated tricyclic compounds and methods of making and using same
WO2013033430A1 (en) 2011-09-02 2013-03-07 Wake Forest School Of Medicine Targeted delivery and prodrug designs for platinum-acridine anti-cancer compounds and methods thereof

Cited By (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9925166B2 (en) 2008-07-18 2018-03-27 Zafgen, Inc. Methods of treating an overweight or obese subject
US8865746B2 (en) 2008-07-18 2014-10-21 Zafgen, Inc. Methods of treating an overweight or obese subject
US9701651B2 (en) 2008-12-04 2017-07-11 Zafgen, Inc. Methods of treating an overweight or obese subject
US9656979B2 (en) 2008-12-04 2017-05-23 Zafgen, Inc. Methods of treating an overweight or obese subject
US8642650B2 (en) 2008-12-04 2014-02-04 Zafgen, Inc. Methods of treating an overweight or obese subject
US8367721B2 (en) 2008-12-04 2013-02-05 Zafgen, Inc. Methods of treating an overweight or obese subject
US9067905B2 (en) 2009-10-09 2015-06-30 Zafgen, Inc. Sulphone compounds and methods of making and using same
US9067913B2 (en) 2010-01-08 2015-06-30 Zafgen, Inc. Fumigillol type compounds and methods of making and using same
US8772333B2 (en) 2010-01-08 2014-07-08 Zafgen, Inc. Fumigillol type compounds and methods of making and using same
US8815309B2 (en) 2010-01-08 2014-08-26 Zafgen, Inc. Methods of treating a subject with benign prostate hyperplasia
US9649293B2 (en) 2010-04-07 2017-05-16 Zafgen, Inc. Methods of treating an overweight subject
US10406134B2 (en) 2010-04-07 2019-09-10 Zafgen, Inc. Methods of treating an overweight subject
US9266896B2 (en) 2010-07-22 2016-02-23 Zafgen, Inc. Tricyclic compounds and methods of making and using same
US9839623B2 (en) 2010-07-22 2017-12-12 Zafgen, Inc. Tricyclic compounds and methods of making and using same
US8735447B2 (en) 2010-11-09 2014-05-27 Zafgen, Inc. Crystalline solids of a MetAP-2 inhibitor and methods of making and using same
CN103347870A (en) * 2010-11-09 2013-10-09 扎夫根股份有限公司 Crystalline solids of metap-2 inhibitor and method of making and using same
WO2012064838A1 (en) 2010-11-09 2012-05-18 Zafgen Corporation Crystalline solids of a metap-2 inhibitor and methods of making and using same
EA029941B1 (en) * 2010-11-09 2018-06-29 Зафджен, Инк. Pharmaceutical composition based on a crystalline form of a metap-2 inhibitor and method of treating obesity
US9827221B2 (en) 2010-11-09 2017-11-28 Zafgen, Inc. Crystalline solids of a metAP-2 inhibitor and methods of making and using same
KR20140009994A (en) * 2010-11-09 2014-01-23 자프겐 인크. Crystalline solids of a metap-2 inhibitor and methods of making and using same
US8349891B2 (en) 2010-11-09 2013-01-08 Zafgen, Inc. Crystalline solids of a MetAP-2 inhibitor and methods of making and using same
US9371312B2 (en) 2010-11-09 2016-06-21 Zafgen, Inc. Crystalline solids of a MetAP-2 inhibitor and methods of making and using same
WO2012064928A1 (en) * 2010-11-10 2012-05-18 Zafgen Corporation Methods and compositions for treating thyroid hormone related disorders
WO2012075020A1 (en) * 2010-11-29 2012-06-07 Zafgen Corporation Treatment of obesity using non-daily administration of 6 - 0 - (4 - dimethylaminoethoxy) cinnamoyl fumagillol
WO2012075026A1 (en) * 2010-11-29 2012-06-07 Zafgen Corporation Methods of treating obesity using an effective dose of a metap-2 inhibitor
WO2012074968A1 (en) * 2010-11-29 2012-06-07 Zafgen Corporation Methods of reducing risk of hepatobiliary dysfunction during rapid weight loss with metap-2 inhibitors
US8980946B2 (en) 2010-11-29 2015-03-17 Zafgen, Inc. Treatment of obesity using non-daily administration of 6-O-(4-dimethylaminoethoxy) cinnamoyl fumagillol
JP2013543899A (en) * 2010-11-29 2013-12-09 ザフゲン,インコーポレイテッド Treatment of obesity with non-daily administration of 6-O- (4-dimethylaminoethoxy) cinnamoyl fumagillol
US9000035B2 (en) 2010-11-29 2015-04-07 Zafgen, Inc. Treatment of obesity using non-daily administration of 6-O-(4-dimethylaminoethoxy) cinnamoyl fumagillol
CN103379905A (en) * 2010-11-29 2013-10-30 扎夫根股份有限公司 Treatment of obesity using non-daily administration of 6-0-(4-dimethylaminoethoxy) cinnamoyl fumagillol
US9173865B2 (en) 2010-11-29 2015-11-03 Zafgen, Inc. Treatment of obesity using non-daily administration of 6-O-(4-dimethylaminoethoxy) cinnamoyl fumagillol
US9321740B2 (en) 2011-01-26 2016-04-26 Zafgen, Inc. Tetrazole compounds and methods of making and using same
JP2014507477A (en) * 2011-03-08 2014-03-27 ザフゲン,インコーポレイテッド Oxaspiro [2.5] octane derivatives and analogs
US10259796B2 (en) 2011-03-08 2019-04-16 Zafgen, Inc. Oxaspiro[2.5]octane derivatives and analogs
US9328082B2 (en) 2011-03-08 2016-05-03 Zafgen, Inc. Oxaspiro[2.5]octane derivatives and analogs
US9290472B2 (en) 2011-05-06 2016-03-22 Zafgen, Inc. Partially saturated tricyclic compounds and methods of making and using same
CN103764641B (en) * 2011-05-06 2016-10-26 扎夫根股份有限公司 The tricyclic compound of fractional saturation and preparation and application thereof
WO2012154676A1 (en) * 2011-05-06 2012-11-15 Zafgen Corporation Partially saturated tricyclic compounds and methods of making and using same
US9617237B2 (en) 2011-05-06 2017-04-11 Zafgen, Inc. Partially saturated tricyclic compounds and methods of making and using same
US9187494B2 (en) 2011-05-06 2015-11-17 Zafgen, Inc. Aryl-substituted tricyclic sulfonamides as methionyl aminopeptidase 2 modulators
CN103764641A (en) * 2011-05-06 2014-04-30 扎夫根股份有限公司 Partially saturated tricyclic compounds and methods of making and using same
EA025526B1 (en) * 2011-05-06 2017-01-30 Зафджен Инк. Partially saturated tricyclic compounds and methods of making and using same
US9242997B2 (en) 2011-05-06 2016-01-26 Zafgen, Inc. Tricyclic pyrazole sulphonamide compunds and methods of making and using same
JP2014514349A (en) * 2011-05-06 2014-06-19 ザフゲン,インコーポレイテッド Partially saturated tricyclic compounds and methods for their production and use
US9446016B2 (en) 2011-10-03 2016-09-20 Zafgen, Inc. Methods of treating age related disorders
EP2763671A2 (en) * 2011-10-03 2014-08-13 Zafgen, Inc. Methods of treating age related disorders
US9440943B2 (en) 2012-01-18 2016-09-13 Zafgen, Inc. Tricyclic sulfone compounds and methods of making and using same
JP2015504091A (en) * 2012-01-18 2015-02-05 ザフゲン,インコーポレイテッド Tricyclic sulfonamide compounds and methods for making and using the same
JP2015509102A (en) * 2012-01-18 2015-03-26 ザフゲン,インコーポレイテッド Tricyclic sulfone compounds and methods for making and using the same
CN104169268A (en) * 2012-01-18 2014-11-26 扎夫根股份有限公司 Tricyclic sulfone compounds and methods of making and using same
US9359369B2 (en) 2012-01-18 2016-06-07 Zafgen, Inc. Tricyclic sulfonamide compounds and methods of making and using same
WO2013109735A1 (en) * 2012-01-18 2013-07-25 Zafgen, Inc. Tricyclic sulfone compounds and methods of making and using same
US9434743B2 (en) 2012-03-02 2016-09-06 Takeda Pharmaceutical Company Limited Indazole derivatives
CN104350046B (en) * 2012-05-07 2016-11-30 扎夫根股份有限公司 The polymorphic salt of 6-O-(4-dimethylamino ethoxy) cinnamoyl aspergillus fumigatus cedrol oxalates and preparation and application thereof
CN104350046A (en) * 2012-05-07 2015-02-11 扎夫根股份有限公司 Polymorphic salt of the oxalate salt of 6 - o - ( 4 - dimethylaminoethoxy) cinnarnoyl fumagillol and methods of making and using same
WO2013169727A1 (en) * 2012-05-07 2013-11-14 Zafgen, Inc. Polymorphic salt of the oxalate salt of 6 - o - ( 4 - dimethylaminoethoxy) cinnarnoyl fumagillol and methods of making and using same
US9260419B2 (en) 2012-05-07 2016-02-16 Zafgen, Inc. Polymorphic salt of a metap-2 inhibitor and methods of making and using same
US9839622B2 (en) 2012-05-08 2017-12-12 Zafgen, Inc. Methods of treating hypothalamic obesity
US10220015B2 (en) 2012-05-09 2019-03-05 Zafgen, Inc. Fumigillol compounds and methods of making and using same
US20150126489A1 (en) * 2012-05-09 2015-05-07 Zafgen, Inc. Fumigillol compounds and methods of making and using same
US9573918B2 (en) 2012-05-09 2017-02-21 Zafgen, Inc. Fumigillol compounds and methods of making and using same
US9895339B2 (en) 2012-05-09 2018-02-20 Zafgen, Inc. Fumigillol compounds and methods of making and using same
US9475816B2 (en) 2012-09-07 2016-10-25 Takeda Pharmaceutical Company Limited Substituted-1,4-dihydropyrazolo[4,3-b]indoles
WO2014039831A1 (en) 2012-09-07 2014-03-13 Takeda Pharmaceutical Company Limited SUBSTITUTED-1,4-DIHYDROPYRAZOLO[4,3-b]INDOLES
US9868717B2 (en) 2012-11-05 2018-01-16 Zafgen, Inc. Tricyclic sulphonamide compounds and methods of making and using same
US9561209B2 (en) 2012-11-05 2017-02-07 Zafgen, Inc. Methods of treating liver diseases
US10174009B2 (en) 2012-11-05 2019-01-08 Zafgen, Inc. Tricyclic sulphonamide compounds and methods of making and using same
US10064839B2 (en) 2012-11-05 2018-09-04 Zafgen, Inc. Methods of treating liver diseases
US9849106B2 (en) 2013-03-14 2017-12-26 Zafgen, Inc. Methods of treating impaired wound healing
US10231946B2 (en) 2013-03-14 2019-03-19 Zafgen, Inc. Methods of treating ischemic organ damage and other disorders
US9597309B2 (en) 2013-03-14 2017-03-21 Zafgen, Inc. Methods of treating renal disease and other disorders
US9827255B2 (en) 2014-12-19 2017-11-28 Takeda Pharmaceutical Company Limited Fumagillol derivatives
WO2016100778A1 (en) 2014-12-19 2016-06-23 Takeda Pharmaceutical Company Limited Fumagillol derivatives
US10328089B2 (en) 2014-12-19 2019-06-25 Takeda Pharmaceutical Company Limited Fumagillol derivatives
US10023561B2 (en) 2015-08-11 2018-07-17 Zafgen, Inc. Fumagillol heterocyclic compounds and methods of making and using same
US9944613B2 (en) 2015-08-11 2018-04-17 Zafgen, Inc. Fumagillol spirocyclic compounds and fused bicyclic compounds and methods of making and using same
US9682965B2 (en) 2015-08-11 2017-06-20 Zafgen, Inc. Fumagillol heterocyclic compounds and methods of making and using same

Also Published As

Publication number Publication date
US20180064677A1 (en) 2018-03-08
US20190224156A1 (en) 2019-07-25
US20120322867A1 (en) 2012-12-20
US8367721B2 (en) 2013-02-05
US20150209320A1 (en) 2015-07-30
US8642650B2 (en) 2014-02-04
US20120034233A1 (en) 2012-02-09

Similar Documents

Publication Publication Date Title
US20190224156A1 (en) Methods of Treating an Overweight or Obese Subject
US10406134B2 (en) Methods of treating an overweight subject
US9701651B2 (en) Methods of treating an overweight or obese subject
US20120010259A1 (en) Methods of Treating an Overweight or Obese Subject
US20120010290A1 (en) Methods of Treating an Overweight or Obese Subject
US20140011870A1 (en) Methods of Treating Obesity Using an Effective Dose of a METAP-2 Inhibitor
US8980946B2 (en) Treatment of obesity using non-daily administration of 6-O-(4-dimethylaminoethoxy) cinnamoyl fumagillol
US9839622B2 (en) Methods of treating hypothalamic obesity

Legal Events

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

Ref document number: 09831214

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 13133060

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 09831214

Country of ref document: EP

Kind code of ref document: A2