US20090124684A1 - Stimulation Of CPT-1 As A Means To Reduce Weight - Google Patents

Stimulation Of CPT-1 As A Means To Reduce Weight Download PDF

Info

Publication number
US20090124684A1
US20090124684A1 US12/266,425 US26642508A US2009124684A1 US 20090124684 A1 US20090124684 A1 US 20090124684A1 US 26642508 A US26642508 A US 26642508A US 2009124684 A1 US2009124684 A1 US 2009124684A1
Authority
US
United States
Prior art keywords
cpt
fatty acid
activity
malonyl
coa
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/266,425
Inventor
Jagan N. Thupari
Leslie E. Landree
Gabrielle Ronnett
Francis P. Kuhajda
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
School of Medicine of Johns Hopkins University
Original Assignee
School of Medicine of Johns Hopkins University
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 School of Medicine of Johns Hopkins University filed Critical School of Medicine of Johns Hopkins University
Priority to US12/266,425 priority Critical patent/US20090124684A1/en
Publication of US20090124684A1 publication Critical patent/US20090124684A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE JOHNS HOPKINS UNIVERSITY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • A23L33/17Amino acids, peptides or proteins
    • A23L33/175Amino acids
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/40Complete food formulations for specific consumer groups or specific purposes, e.g. infant formula
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/205Amine addition salts of organic acids; Inner quaternary ammonium salts, e.g. betaine, carnitine
    • 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
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • 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
    • A61K31/365Lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • This invention is directed to a method for development of therapeutics that selectively enhance fatty acid oxidation, increase energy production, and reduce adiposity while preserving lean mass, through the pharmacological stimulation of CPT-1 activity.
  • C75 Cerulenin treatment of MCF-7 human breast cancer cells in vitro significantly inhibits fatty acid oxidation, probably through increased levels of malonyl-CoA (Loftus, et al. (2000) Science, 288:2379-2381).
  • C75 is a member of a family of ⁇ -methylene- ⁇ -butyrolactones which are known inhibitors of fatty acid synthase (FAS) (Kuhajda, et al. (2000) Proc. Natl. Acad Sci USA, 97:3450-3454).
  • FAS fatty acid synthase
  • C75 reduces the expression of hypothalamic neuropeptide-Y (NPY) leading to reversible inanition (Loftus, et al, 2000).
  • NPY hypothalamic neuropeptide-Y
  • NPY hypothalamic neuropeptide-Y
  • ob/ob mice there was profound loss of fat in the liver and peripheral tissues despite the increased levels of hepatic malonyl-CoA (Loftus, et al., 2000).
  • Malonyl-CoA is a potent inhibitor of fatty acid oxidation through its action as an inhibitor of carnitine-palmitoyl-transferase-1 (CPT-1) (Witters, et al. (1992) J. Biol. Chem., 267:2864-2867).
  • CPT-1 enables the entry of long-chain acyl-CoA's into the mitochondria for fatty acid oxidation.
  • FAS inhibitors genetically and diet-induced obese mice undergo a selective and significant loss of adipose tissue despite the high levels of malonyl-CoA induced by FAS inhibition.
  • malonyl-CoA is a potent inhibitor of fatty acid oxidation through its inhibition of carnitine palmitoyltransferase-1 (CPT-1, E.C. 2.3.1.21), the rapid and selective loss of adipose tissue was surprising. High systemic levels of malonyl-CoA would be expected to inhibit fatty acid oxidation leading instead to a selective loss of lean mass during C75 induced inanition.
  • R is a substitute selected from the group consisting of:
  • this invention provides a method for stabilizing weight comprising chronic administration of an agent that stimulates CPT-1 activity in an amount that does not significantly inhibit FAS.
  • the agent is administered in an amount sufficient to increase fatty acid oxidation.
  • the agent is administered in an amount sufficient to antagonize malonyl CoA inhibition of CPT-1.
  • the agent is administered in an amount sufficient to increase malonyl CoA level.
  • upon administration of the agent malonyl CoA level is not substantially increased. Substantial increase in malonyl CoA level as contemplated herein is equivalent to about one-half the K i for malonyl CoA inhibition of CPT-1.
  • this invention provides a method of screening for agents that induce weight loss, comprising determining whether a candidate weight loss agent stimulates CPT-1 activity; and selecting an agent that stimulates CPT-1 activity.
  • this method further comprises determining whether the candidate weight loss agent is an antagonist of malonyl CoA inhibition of CPT-1, and candidate weight loss agents are selected that obviate malonyl CoA inhibition of CPT-1.
  • this invention provides a therapeutic composition comprising an agent that stimulates CPT-1 activity and L-carnitine.
  • the therapeutic composition comprises an antagonist of malonyl CoA inhibition of CPT-1.
  • this invention provides a nutritional composition comprising nutritionally sufficient amounts of fats, carbohydrates and amino acids, said composition further comprising an antagonist of malonyl CoA inhibition of CPT-1 and L-carnitine.
  • the nutritional composition is adapted for parenteral administration.
  • FIG. 1 shows the effect of C75 on fatty acid oxidation in MCF-7 cells, compared to the effect of Etomoxir.
  • FIG. 2 shows concentration dependent stimulation of CPT-1 activity by C75 and inhibition by malonyl CoA.
  • FIG. 3 shows reversible stimulation of CPT-1 by C75.
  • FIG. 4 shows stimulation of CPT-1 by various C75 analogs.
  • FIG. 5 shows concentration dependent enhancement of cellular ATP levels by C75 in MCF-7 cells.
  • FIG. 6 shows concentration dependent stimulation of fatty acid oxidation by C75 in mouse adipocytes.
  • FIG. 7 shows concentration dependent enhancement of cellular ATP levels by C75 in mouse adipocytes.
  • FIG. 8 shows respiratory exchange ratio (RER) measured by indirect calorimetry for mice in the absence (A) and presence (B, C) of C75.
  • CPT-1 carnitine palmitoyltransferase-1
  • the peripheral (non-CNS) weight loss effect of C-75 is at least in part due to CPT-1 stimulation and increased fatty acid oxidation with concomitant fatty acid synthesis inhibition.
  • C75 is the prototype agent for stimulation of CPT-1; reference to C75 hereinafter includes other suitable agents which stimulate CPT-1 activity, except where indicated otherwise by context.
  • Suitable agents which stimulate CPT-1 activity include a variety of gamma-butyrolactones which can be readily identified by testing the effect on CPT-1 activity of gamma-substituted-alpha-methylene-butyrolactones, such as those described in International Patent Publication WO 2004/006835, incorporated herein by reference, substituted thiotetronic acids, such as those described in International Patent Publication WO 2004/005277, incorporated herein by reference, and substituted thiophene diones described in U.S. Provisional Patent Application 60/574,639, incorporated herein by reference.
  • C-75 abolishes the inhibitory effect of malonyl-CoA on CPT-1 activity.
  • C75 exhibits kinetic features of a slow-binding inhibitor with purified FAS (1), its interaction with CPT-1 appears rapid and competitive.
  • the stimulatory effect of C75 upon fatty acid oxidation may be due to either its direct stimulation of CPT-1 activity, its interference of malonyl-CoA inhibition of CPT-1, or both.
  • the effects of C75 are not restricted to murine CPT-1, as human CPT-1 was similarly affected.
  • C75 also increased ATP levels in both the human and murine cells.
  • C75 The effect of C75 on fatty acid metabolism in vivo mirrored the alterations seen on a cellular level.
  • C75 and its family of ⁇ -methylene- ⁇ -butyrolactones appear to act as competitive agonists of CPT-1.
  • This agonist activity of C75 appears to overcome inhibitory effects of malonyl CoA on the same enzyme.
  • the increased fatty acid oxidation induced by C75 is an important mechanism accounting for marked reduction in adiposity seen during C75 treatment of mice.
  • this invention describes a method to develop therapeutics that selectively enhance fatty acid oxidation, increase energy production, and reduce adiposity while preserving lean mass, through the pharmacological stimulation of CPT-1 activity.
  • compositions containing C75 and/or other agents that stimulate CPT-1, and methods of administering such agents are within the skill of the art, particularly in view of the description in U.S. Pat. No. 5,981,575, the text of which is incorporated herein by reference.
  • CPT-1 stimulating agents to increase energy production by administering the agents contemporaneously with fatty acids or compounds containing fatty acid residues is also within the skill of the art, particularly in view of the nutritional compositions disclosed in U.S. Pat. No. 4,434,160, the text of which is incorporated herein by reference.
  • Cerulenin an FAS inhibitor, increases malonyl-CoA amount in MCF-7 cells (3).
  • cerulenin causes inhibition of fatty acid oxidation through the malonyl-CoA inhibition of CPT-1 (Thupari, et al. (2001) Biochem. Biophys. Res. Comm., 285:217-223).
  • C75 treatment of MCF-7 cells resulted in a >5-fold increase in malonyl-CoA through C75 inhibition of fatty acid synthase (FAS) (3).
  • FAS fatty acid synthase
  • Human breast cancer cell line MCF-7 was obtained from the American Type Culture Collection. 1 ⁇ 10 6 MCF-7 cells were plated in T-25 flasks in triplicate and incubated overnight at 37° C. Drugs were then added as indicated diluted from 5 mg/ml stock in DMSO. After 2 hours, medium with drugs was removed and cells were preincubated for 30 min. with 1.5 ml of the following buffer: 114 mM NaCl, 4.7 mM KCl, 1.2 mM KH 2 PO 4 , 1.2 mM MgSO 4 , glucose 11 mM.
  • assay buffer containing: 114 mM NaCl, 4.7 mM KCl, 1.2 mM KH 2 PO 4 , 1.2 mM MgSO 4 , glucose 11 mM, 2.5 mM palmitate (containing with 10 ⁇ Ci of [1- 14 C]palmitate) bound to albumin, 0.8 mM L-carnitine, and cells were incubated at 37° C. for 2 h. Following the incubation, 400 ⁇ l of benzethonium hydrochloride was added to the center well to collect released 14 CO 2 . Immediately, the reaction was stopped by adding 500 ⁇ l of 7% perchloric acid to the cells.
  • C75 treatment increased rather than decreased fatty acid oxidation in MCF-7 cells. This implies a direct effect of C75 upon carnitine palmitoyltransferase-1 (CPT-1).
  • CPT-1 activity was assayed in MCF-7 cells by the following published procedure: MCF-7 cells were plated in DMEM with 10% fetal bovine serum at 10 6 cells in 24 well plates in triplicate. Following overnight incubation at 37° C., the medium was removed and replaced with 700 ⁇ l of assay medium consisting of: 50 mM imidazole, 70 mM KCl, 80 mM sucrose, 1 mM EGTA, 2 mM MgCl 2 , 1 mM DTT, 1 mM KCN, 1 mM ATP, 0.1% fatty acid free bovine serum albumin, 70 ⁇ M palmitoyl-CoA, 0.25 ⁇ Ci [methyl- 14 C]L-carnitine, 40 ⁇ g digitonin with or without 20 ⁇ M malonyl-CoA.
  • MCF-7 cells were treated with C75 at 10 or 20 ⁇ g/mL for 1 hr before CPT-1 activity was assayed.
  • the assay was performed with the C75 and malonyl-CoA concentrations indicated (“M” indicates malonyl-CoA at 20 uM).
  • the level of malonyl-CoA inhibition of the CPT-1 activity is consistent with the activity of the liver isoform of CPT-1 in MCF-7 cells.
  • the K i of malonyl-CoA for the liver isoform of CPT-1 is ⁇ 7 ⁇ M while the K i for the muscle isoform of CPT-1 is 0.07 ⁇ M.
  • MCF-7 cells express predominantly the liver isoform of CPT-1 (consistent with the immunoblot analysis).
  • MCF-7 cells were untreated (left bar) or treated with C75 at 20 ⁇ g/ml for one hour before CPT-1 activity was measured (middle and right bars).
  • C75 was removed from the assay buffer and replaced with buffer (middle bar) or malonyl-CoA 20 ⁇ M was added (left & right bars).
  • 3T3-L1 adipocytes were used in assays similar to those performed with the MCF-7 cells.
  • 3T3-L1 cells were obtained from the American Type Culture Collection, and cells were cultured in DMEM with high glucose (4.5 g/l) (Gibco 12100-046) with 10% fetal calf serum and Biotin (Sigma B-4639) 0.008 g/L. Differentiation was initiated three days after the cells were confluent, when the standard culture medium was replaced with differentiation medium.
  • the differentiation medium contained standard culture medium to which the following were added to achieve the final concentrations: methylisobutylxanthine (MIX) 0.5 mM, dexamethasone (DEX) 1 ⁇ M, and insulin 10 ⁇ g/ml. After 48 hrs, the differentiation medium was replaced with post-differentiation medium which contained insulin at the above concentration, without MIX and DEX. The differentiated cells were ready to be used for experiments in 7-10 days.
  • MIX methylisobutylxanthine
  • DEX dexamethasone
  • C75 increased CPT-1 activity and fatty acid metabolism in the NIH 3T3-L1 cells differentiated into adipocytes.
  • CPT-1 activity, fatty acid oxidation, and total cellular ATP were measured as described in Examples 2, 1, and 4.
  • C75 at doses of 20 ⁇ g/g ml or greater significantly increased fatty acid oxidation (see FIG.
  • C75 induces a profound and rapid stimulation of fatty acid oxidation in vivo.
  • Mice were maintained in the Oxymax calorimeter (Oxymax Equal Flow System®, Columbus Instruments, Columbus, Ohio). Oxygen consumption and CO 2 production was measured in up to four mice simultaneously using the indirect calorimeter. Measurements were recorded every 15 minutes over the entire course of the experiments.
  • the respiratory exchange ratio (RER) was calculated by the Oxymax® software version 5.9. RER is defined as the ratio of CO 2 to O 2 at any given time irrespective if equilibrium was reached. Mice were maintained on water and mouse chow ad libitum. In the control mice ( FIG.
  • C75 treatment led to a rapid, profound increase in fatty acid oxidation as measured by RER.
  • C75 treated animals are able to significantly reduce adipose mass and reverse fatty liver, by allowing fatty acid oxidation to occur despite the high levels of malonyl-CoA generated during FAS inhibition in vivo.
  • FAS activity was measured by monitoring the malonyl-CoA dependent oxidation of NADPH spectrophotometrically at OD 340 in 96-well plates, as described in International Patent Publication WO 2004/005277.
  • the IC 50 for the compounds against FAS was determined by plotting the change in OD 340 against time for each inhibitor concentration tested and determining the rate of change by linear regression. The concentration of a particular compound yielding 50% inhibition of the rate for FAS in the absence of the compound is the IC 50 .
  • Stimulation of CPT-1 activity was measured as described in Example 2, except the cells were preincubated with the compound for 2 hours at the concentrations indicated in the table.
  • Stimulation of Fatty acid oxidation was determined as described in Example 1 for cells preincubated with the compounds at the concentrations indicated in the table.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pediatric Medicine (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Child & Adolescent Psychology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

This invention provides methods and compositions for inducing weight loss and maintaining optimum weight comprising administering an agent that stimulates carnitine palmitoyl transferase-1 (CPT-1) activity to the patient in need, including human patients. These methods do not require inhibition of fatty acid synthesis. In particular, this invention provides methods for development of therapeutics that selectively enhance fatty acid oxidation, increase energy production, and reduce adiposity while preserving lean mass, through the pharmacological stimulation of CPT-1 activity. In a preferred mode, the agent is administered in an amount sufficient to increase fatty acid oxidation. In another preferred mode, the agent is administered in an amount sufficient to antagonize malonyl CoA inhibition of CPT-1. In yet another preferred mode, the agent is administered in an amount sufficient to increase malonyl CoA level.

Description

    BACKGROUND
  • 1. Field of the Invention
  • This invention is directed to a method for development of therapeutics that selectively enhance fatty acid oxidation, increase energy production, and reduce adiposity while preserving lean mass, through the pharmacological stimulation of CPT-1 activity.
  • 2. Review of Related Art
  • Cerulenin treatment of MCF-7 human breast cancer cells in vitro significantly inhibits fatty acid oxidation, probably through increased levels of malonyl-CoA (Loftus, et al. (2000) Science, 288:2379-2381). C75 is a member of a family of α-methylene-γ-butyrolactones which are known inhibitors of fatty acid synthase (FAS) (Kuhajda, et al. (2000) Proc. Natl. Acad Sci USA, 97:3450-3454). Treatment of mice with C75 leads to inhibition of hepatic fatty acid synthesis and high levels of malonyl-CoA (Loftus, et al. (2000); Pizer, et al. (2000) Cancer Res., 60:213-218). In the brain, C75 reduces the expression of hypothalamic neuropeptide-Y (NPY) leading to reversible inanition (Loftus, et al, 2000). During in vivo treatment of ob/ob mice with C75 there was profound loss of fat in the liver and peripheral tissues despite the increased levels of hepatic malonyl-CoA (Loftus, et al., 2000).
  • Malonyl-CoA is a potent inhibitor of fatty acid oxidation through its action as an inhibitor of carnitine-palmitoyl-transferase-1 (CPT-1) (Witters, et al. (1992) J. Biol. Chem., 267:2864-2867). CPT-1 enables the entry of long-chain acyl-CoA's into the mitochondria for fatty acid oxidation. When treated with FAS inhibitors, genetically and diet-induced obese mice undergo a selective and significant loss of adipose tissue despite the high levels of malonyl-CoA induced by FAS inhibition. Since malonyl-CoA is a potent inhibitor of fatty acid oxidation through its inhibition of carnitine palmitoyltransferase-1 (CPT-1, E.C. 2.3.1.21), the rapid and selective loss of adipose tissue was surprising. High systemic levels of malonyl-CoA would be expected to inhibit fatty acid oxidation leading instead to a selective loss of lean mass during C75 induced inanition.
  • SUMMARY OF THE INVENTION
  • It is an object of this invention to provide methods and compositions for inducing weight loss and maintaining optimum weight which do not require inhibition of fatty acid synthesis. This and other objects are met by one or more of the following embodiments.
  • In one embodiment, this invention provides a method of inducing weight loss comprising administering an agent that stimulates carnitine palmitoyl transferase-1 (CPT-1) activity to the patient in need, including human patients. In a preferred mode, the agent is administered in an amount sufficient to increase fatty acid oxidation. In another preferred mode, the agent is administered in an amount sufficient to antagonize malonyl CoA inhibition of CPT-1. In yet another preferred mode, the agent is administered in an amount sufficient to increase malonyl CoA level. In still another preferred mode, upon administration of the agent, malonyl CoA level is not substantially increased. Substantial increase in malonyl CoA level as contemplated herein is equivalent to about one-half the Ki for malonyl CoA inhibition of CPT-1. In yet another preferred mode, the agent which stimulates CPT-1 activity also inhibits fatty acid synthase (FAS). In an alternative mode, FAS is not significantly inhibited. Insignificant inhibition as contemplated herein is less that 15%, preferably less than 10%, and more preferably less than 5% inhibition. Methods for assay of FAS activity are disclosed in U.S. Pat. No. 5,981,575, incorporated herein by reference. In preferred modes of the above embodiments, the agent which stimulates CPT-1 activity is not a compound of formula:
  • Figure US20090124684A1-20090514-C00001
  • wherein R is a substitute selected from the group consisting of:
  • (a) saturated linear or branched alkyl groups having 3-18 carbon atoms,
  • (b) unsaturated linear or branched alkyl groups having 3-18 carbon atoms,
  • Figure US20090124684A1-20090514-C00002
  • wherein:
    • R1 and R2, the same or different, are H, CH3, C2H5, C3H7, C4H9, CF3, OCH3, F, Cl, or Br;
    • R3 is H, CH3, C2H5, C2H5, C4H9, COOH, COOCH3, COOC2H5, COOC2H5, or COOC4H9;
    • R4 is H, CH3, C2H5, C3H7, or C4H9;
    • X is NH, S, or O;
    • Z is CH2, O, NH, or S;
    • i is 1 to 5;
    • j is 0 to 10;
    • k is 1 to 10;
    • m is 1-13; and
    • n is 1 to 15.
  • In another embodiment, this invention provides a method for stabilizing weight comprising chronic administration of an agent that stimulates CPT-1 activity in an amount that does not significantly inhibit FAS. In a preferred mode, the agent is administered in an amount sufficient to increase fatty acid oxidation. In another preferred mode, the agent is administered in an amount sufficient to antagonize malonyl CoA inhibition of CPT-1. In yet another preferred mode, the agent is administered in an amount sufficient to increase malonyl CoA level. In still another preferred mode, upon administration of the agent, malonyl CoA level is not substantially increased. Substantial increase in malonyl CoA level as contemplated herein is equivalent to about one-half the Ki for malonyl CoA inhibition of CPT-1.
  • In still another embodiment, this invention provides a method of screening for agents that induce weight loss, comprising determining whether a candidate weight loss agent stimulates CPT-1 activity; and selecting an agent that stimulates CPT-1 activity. Preferably, this method further comprises determining whether the candidate weight loss agent is an antagonist of malonyl CoA inhibition of CPT-1, and candidate weight loss agents are selected that obviate malonyl CoA inhibition of CPT-1.
  • In yet another embodiment, this invention provides a therapeutic composition comprising an agent that stimulates CPT-1 activity and L-carnitine. Preferably, the therapeutic composition comprises an antagonist of malonyl CoA inhibition of CPT-1.
  • In still another embodiment, this invention provides a nutritional composition comprising nutritionally sufficient amounts of fats, carbohydrates and amino acids, said composition further comprising an antagonist of malonyl CoA inhibition of CPT-1 and L-carnitine. In one mode, the nutritional composition is adapted for parenteral administration.
  • To investigate the mechanism of action leading to the paradoxical reduction of fatty liver in the setting of high hepatic levels of malonyl-CoA during C75 treatment, the effect of C75 on CPT-1 activity was studied. Surprisingly, C75 and related compounds concomitantly stimulated CPT-1 activity and fatty acid oxidation in vitro while inhibiting FAS. In addition to its overall allosteric activation of CPT-1, C75 abrogated the inhibitory effect of malonyl-CoA on CPT-1 activity in vitro. As a consequence of increased fatty acid oxidation, C75 increased cellular ATP levels.
  • To test the effect of C75 on fatty acid oxidation in vivo, whole animal calorimetry was utilized to measure the respiratory exchange ratio (RER) in mice treated with C75. Following C75 therapy, the RER dropped within 2 h to the range of 0.7, indicative of fatty acid oxidation. This rate of RER decline was similar to food withdrawal from animals fed ad libitum with mouse chow. These studies indicate that, despite high hepatic levels of malonyl-CoA, C75 treated animals freely oxidized fatty acids.
  • These data suggest that C75 blocks the inhibitory action of malonyl-CoA on CPT-1 activity in vivo leading to a reduction in fatty liver and adipose mass during FAS inhibition. This invention describes a method to develop therapeutics that selectively reduce adiposity while preserving lean mass through the pharmacological stimulation of CPT-1 activity.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the effect of C75 on fatty acid oxidation in MCF-7 cells, compared to the effect of Etomoxir.
  • FIG. 2 shows concentration dependent stimulation of CPT-1 activity by C75 and inhibition by malonyl CoA.
  • FIG. 3 shows reversible stimulation of CPT-1 by C75.
  • FIG. 4 shows stimulation of CPT-1 by various C75 analogs.
  • FIG. 5 shows concentration dependent enhancement of cellular ATP levels by C75 in MCF-7 cells.
  • FIG. 6 shows concentration dependent stimulation of fatty acid oxidation by C75 in mouse adipocytes.
  • FIG. 7 shows concentration dependent enhancement of cellular ATP levels by C75 in mouse adipocytes.
  • FIG. 8 shows respiratory exchange ratio (RER) measured by indirect calorimetry for mice in the absence (A) and presence (B, C) of C75.
  • DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
  • Inhibition of fatty acid synthase (FAS) in vivo has been shown to cause rapid and profound weight loss. Both cerulenin, a natural product, and C-75, a synthetic small-molecule, cause similar weight loss when administered intracerebroventricularly (i.c.v.) to rats. When treated systemically (e.g., intraperitoneally), C-75 causes more profound weight loss, even weight loss greater than starved animals. These data demonstrate a greater peripheral (non-CNS) effect on weight loss for C-75 than for cerulenin.
  • While studying the mechanism of action of this profound peripheral effect of C-75, the inventors have recently found that in addition to inhibition of FAS, C-75 and its family of α-methylene-γ-butyrolactones, directly stimulates carnitine palmitoyltransferase-1 (CPT-1) leading to increased mitochondrial fatty acid oxidation. Cerulenin, in contrast, leads to reduced CPT-1 activity and reduced fatty acid oxidation through generation of high malonyl-CoA levels from FAS inhibition.
  • C75 treatment of MCF-7 cells in vitro stimulated CPT-1 activity from 150-160%. There was also a concomitant increase in fatty acid oxidation. Among the C75 analogs, a carbon chain length of C6-C16 was optimum for CPT-1 stimulatory activity. In the presence of C75, malonyl-CoA is no longer able to inhibit CPT-1 activity, suggesting that in addition to its stimulatory effect, C75 also prevents malonyl-CoA inhibition of CPT-1. There is no detectable covalent interaction between CPT-1 and C75.
  • Thus, the peripheral (non-CNS) weight loss effect of C-75 is at least in part due to CPT-1 stimulation and increased fatty acid oxidation with concomitant fatty acid synthesis inhibition. These data identify a family of α-methylene-γ-butyrolactones as malonate or malonyl-CoA mimetics and CPT-1 as a target for weight loss therapeutics and. More broadly, our data suggest that other malonate or malonyl-CoA mimetics can be designed and synthesized to function as effective weight loss agents.
  • Data demonstrate that C-75 and its family of α-methylene-γ-butyrolactones directly interact with CPT-1 leading to increased CPT-1 enzymatic activity and fatty acid oxidation. The chemical structure of C75 and numerous analogs, as well as methods of synthesizing these analogs, are disclosed in U.S. Pat. No. 5,981,575, which is incorporated herein by reference. The stimulatory effect of C75 is related to the length of the saturated carbon side chain, with the optimum length between 6-18 carbon atoms. With regard to the discussion of the present invention, C75 is the prototype agent for stimulation of CPT-1; reference to C75 hereinafter includes other suitable agents which stimulate CPT-1 activity, except where indicated otherwise by context. Other suitable agents which stimulate CPT-1 activity include a variety of gamma-butyrolactones which can be readily identified by testing the effect on CPT-1 activity of gamma-substituted-alpha-methylene-butyrolactones, such as those described in International Patent Publication WO 2004/006835, incorporated herein by reference, substituted thiotetronic acids, such as those described in International Patent Publication WO 2004/005277, incorporated herein by reference, and substituted thiophene diones described in U.S. Provisional Patent Application 60/574,639, incorporated herein by reference.
  • In addition to its direct effect upon CPT-1, C-75 abolishes the inhibitory effect of malonyl-CoA on CPT-1 activity. Although C75 exhibits kinetic features of a slow-binding inhibitor with purified FAS (1), its interaction with CPT-1 appears rapid and competitive. Thus, the stimulatory effect of C75 upon fatty acid oxidation may be due to either its direct stimulation of CPT-1 activity, its interference of malonyl-CoA inhibition of CPT-1, or both. Interestingly, the effects of C75 are not restricted to murine CPT-1, as human CPT-1 was similarly affected. As a consequence of increased fatty acid oxidation, C75 also increased ATP levels in both the human and murine cells.
  • The effect of C75 on fatty acid metabolism in vivo mirrored the alterations seen on a cellular level. C75 treatment of lean mice led to a profound and rapid increase in fatty acid oxidation, despite the high levels of malonyl-CoA generated by C75 in vivo. Thus, C75 and its family of α-methylene-γ-butyrolactones, appear to act as competitive agonists of CPT-1. This agonist activity of C75 appears to overcome inhibitory effects of malonyl CoA on the same enzyme. The increased fatty acid oxidation induced by C75 is an important mechanism accounting for marked reduction in adiposity seen during C75 treatment of mice.
  • In summary, this invention describes a method to develop therapeutics that selectively enhance fatty acid oxidation, increase energy production, and reduce adiposity while preserving lean mass, through the pharmacological stimulation of CPT-1 activity.
  • Formulation of therapeutic compositions containing C75 and/or other agents that stimulate CPT-1, and methods of administering such agents, are within the skill of the art, particularly in view of the description in U.S. Pat. No. 5,981,575, the text of which is incorporated herein by reference.
  • Use of CPT-1 stimulating agents to increase energy production by administering the agents contemporaneously with fatty acids or compounds containing fatty acid residues is also within the skill of the art, particularly in view of the nutritional compositions disclosed in U.S. Pat. No. 4,434,160, the text of which is incorporated herein by reference.
  • EXAMPLES
  • In order to facilitate a more complete understanding of the invention, a number of Examples are provided below. However, the scope of the invention is not limited to specific embodiments disclosed in these Examples, which are for purposes of illustration only.
  • Example 1 Paradoxical Effects of a Fatty Acid Synthase Inhibitor
  • Cerulenin, an FAS inhibitor, increases malonyl-CoA amount in MCF-7 cells (3). As a consequence of the massive increase in malonyl-CoA, cerulenin causes inhibition of fatty acid oxidation through the malonyl-CoA inhibition of CPT-1 (Thupari, et al. (2001) Biochem. Biophys. Res. Comm., 285:217-223). Previously, it was shown that C75 treatment of MCF-7 cells resulted in a >5-fold increase in malonyl-CoA through C75 inhibition of fatty acid synthase (FAS) (3). The effect of C-75 on fatty acid oxidation was tested as follows.
  • Human breast cancer cell line MCF-7 was obtained from the American Type Culture Collection. 1×106 MCF-7 cells were plated in T-25 flasks in triplicate and incubated overnight at 37° C. Drugs were then added as indicated diluted from 5 mg/ml stock in DMSO. After 2 hours, medium with drugs was removed and cells were preincubated for 30 min. with 1.5 ml of the following buffer: 114 mM NaCl, 4.7 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, glucose 11 mM. After preincubation, 200 μl of assay buffer was added containing: 114 mM NaCl, 4.7 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, glucose 11 mM, 2.5 mM palmitate (containing with 10 μCi of [1-14C]palmitate) bound to albumin, 0.8 mM L-carnitine, and cells were incubated at 37° C. for 2 h. Following the incubation, 400 μl of benzethonium hydrochloride was added to the center well to collect released 14CO2. Immediately, the reaction was stopped by adding 500 μl of 7% perchloric acid to the cells. The flasks with wells were then incubated for 2 h at 37° C. after which the benzethonium hydrochloride was removed and counted for 14C. Blanks were prepared by adding 500 μl of 7% perchloric acid to the cells prior to the incubation with the assay buffer for 2 h.
  • When cells were treated with C75 2 hours before fatty acid oxidation was measured, C75 treatment resulted in a 156% increase in fatty acid oxidation compared to the control (see FIG. 1; p=0.0012, two-tailed t-test, Prism 3.0). In contrast, Etomoxir, a known inhibitor of fatty acid oxidation and non-competitive inhibitor of CPT-1, decreased fatty acid oxidation to 32% of control (p=0.0006, two-tailed t-test, Prism 3.0). C-75 treatment of MCF-7 cells repeatedly resulted in increased fatty acid oxidation with doses from 5-20 μg/ml.
  • Paradoxically, despite an increase in malonyl-CoA similar to that induced by cerulenin, C75 treatment increased rather than decreased fatty acid oxidation in MCF-7 cells. This implies a direct effect of C75 upon carnitine palmitoyltransferase-1 (CPT-1).
  • Example 2 C75 Stimulates Activity of Human CPT-1
  • CPT-1 activity was assayed in MCF-7 cells by the following published procedure: MCF-7 cells were plated in DMEM with 10% fetal bovine serum at 106 cells in 24 well plates in triplicate. Following overnight incubation at 37° C., the medium was removed and replaced with 700 μl of assay medium consisting of: 50 mM imidazole, 70 mM KCl, 80 mM sucrose, 1 mM EGTA, 2 mM MgCl2, 1 mM DTT, 1 mM KCN, 1 mM ATP, 0.1% fatty acid free bovine serum albumin, 70 μM palmitoyl-CoA, 0.25 μCi [methyl-14C]L-carnitine, 40 μg digitonin with or without 20 μM malonyl-CoA. After incubation for 6 minutes at 37° C., the reaction was stopped by the addition of 500 μl of ice-cold 4 M perchloric acid. Cells were then harvested and centrifuged at 13,000×g for 5 min. The pellet was washed with 500 μl ice cold 2 mM perchloric acid and centrifuged again. The resulting pellet was resuspended in 800 μl dH2O and extracted with 150 μl of butanol. The butanol phase was counted by liquid scintillation and represents the acylcarnitine derivative.
  • MCF-7 cells were treated with C75 at 10 or 20 μg/mL for 1 hr before CPT-1 activity was assayed. The assay was performed with the C75 and malonyl-CoA concentrations indicated (“M” indicates malonyl-CoA at 20 uM). Malonyl-CoA treatment alone caused a 46% reduction in CPT-1 activity similar to the previous experiment (see FIG. 2; p=0.02, two-tailed t-test, Prism 3.0). The level of malonyl-CoA inhibition of the CPT-1 activity is consistent with the activity of the liver isoform of CPT-1 in MCF-7 cells. The Ki of malonyl-CoA for the liver isoform of CPT-1 is −7 μM while the Ki for the muscle isoform of CPT-1 is 0.07 μM. Thus, MCF-7 cells express predominantly the liver isoform of CPT-1 (consistent with the immunoblot analysis).
  • There was no statistically significant difference in CPT-1 activity between cells treated with C75 or C75 and malonyl-CoA (FIG. 2). Thus, in the presence of C75, malonyl-CoA lost its inhibitory effect on CPT-1; conversely, C75 stimulation of CPT-1 occurred regardless of the presence of malonyl-CoA. Thus, in the presence of C75, the normal inhibitory activity of malonyl-CoA is lost. Malonyl-CoA inhibition of CPT-1 activity demonstrated that C75 and related compounds were activating CPT-1 rather than CPT-2 activity which is not inhibitable by malonyl-CoA.
  • In a subsequent experiment (data in FIG. 3), MCF-7 cells were untreated (left bar) or treated with C75 at 20 μg/ml for one hour before CPT-1 activity was measured (middle and right bars). During the 6 minutes of the CPT-1 assay, C75 was removed from the assay buffer and replaced with buffer (middle bar) or malonyl-CoA 20 μM was added (left & right bars). Malonyl-CoA treatment alone during the assay resulted in a −70% inhibition of CPT-1 activity (left bar) (p=0.0045, two-tailed t-test, Prism 3.0). Prior C75 treatment with no C75 in the assay buffer resulted in CPT-1 activity of 158% of control (p=0.028, two-tailed t-test, Prism 3.0), similar to the results when C75 is kept in the assay buffer (see above experiment). However, when C75 is removed from the reaction buffer and malonyl-CoA is replaced, C75 stimulatory activity is lost (right bar). Thus, C-75 does not detectably bind covalently to CPT-1, and it is likely a competitive antagonist with malonyl-CoA. These data also suggest that C-75 interacts with CPT-1 at the malonyl-CoA binding site.
  • Example 3 Structure of Effective CPT-1 Stimulators
  • Analogs of α-methylene-γ-butyrolactones differing only in the length of their saturated carbon ‘tail’ were prepared as described in U.S. Pat. No. 5,981,575, incorporated herein by reference. C75 has an eight-carbon tail, C12 and C16 have tails of 12 and 16 carbons respectively. Cells were treated with C75 and C75 analogs at 20 g/ml 1 hr before CPT-1 activity was measured. Malonyl-CoA was added only to the reaction buffer since the whole cell is impermeable to malonyl-CoA. C75 stimulated CPT-1 activity to 166% of control at a dose of 20 μg/ml (see FIG. 4; p=0.0092, two-tailed t-test, Prism 3.0). C12 analog stimulated to 186% (p=0.0099, two-tailed t-test, Prism 3.0) and C16 analog stimulated to 138% of control (p=0.055, two-tailed t-test, Prism 3.0). Malonyl-CoA, an intracellular competitive inhibitor of CPT-1, reduced CPT-1 activity to 64% of control at 20 μM (p=0.023, two-tailed t-test, Prism 3.0). The optimum carbon chain length for CPT-1 activation is between 6 and 16 carbons.
  • Example 4 Enhanced Fatty Acid Oxidation from CPT-1 Stimulation Produces ATP
  • As a consequence of increased fatty acid oxidation, ATP was elevated in MCF-7 cells following C75 treatment. 1×105 MCF-7 cells were plated in 96 well plates. Cells were treated with C75 or vehicle. After 2 hours, ATP was measured using a luciferase assay using the ATP Bioluminescence Kit CLS II (Roche) following the manufacturer's protocol. Plates were read by a Perkin Elmer Wallac Victor2 1420 luminometer. C75 treatment at 10 μg/ml and 20 μg/ml both resulted in a significant increase in total cellular ATP (see FIG. 5; p=0.0001; p<0.0001 compared to control, two-tailed t-test, Prism 3.0). Similar results were obtained after 1 hr incubation with C75. Thus, cellular energy production increased as a result of C75 increasing fatty acid oxidation.
  • Example 5 C75 Stimulates Activity of Muscle Form CPT-1
  • To expand the study of effects of C75 on fatty acid metabolism beyond cancer cell lines to normal adipocytes, differentiated (non-transformed) mouse NIH 3T3-L1 adipocytes were used in assays similar to those performed with the MCF-7 cells. 3T3-L1 cells were obtained from the American Type Culture Collection, and cells were cultured in DMEM with high glucose (4.5 g/l) (Gibco 12100-046) with 10% fetal calf serum and Biotin (Sigma B-4639) 0.008 g/L. Differentiation was initiated three days after the cells were confluent, when the standard culture medium was replaced with differentiation medium. The differentiation medium contained standard culture medium to which the following were added to achieve the final concentrations: methylisobutylxanthine (MIX) 0.5 mM, dexamethasone (DEX) 1 μM, and insulin 10 μg/ml. After 48 hrs, the differentiation medium was replaced with post-differentiation medium which contained insulin at the above concentration, without MIX and DEX. The differentiated cells were ready to be used for experiments in 7-10 days.
  • C75 increased CPT-1 activity and fatty acid metabolism in the NIH 3T3-L1 cells differentiated into adipocytes. One week post differentiation, cells were treated with either vehicle control or C75 for 2 hours at doses indicated below. CPT-1 activity, fatty acid oxidation, and total cellular ATP were measured as described in Examples 2, 1, and 4. C75 treatment of 3T3-L1 adipocytes led to a 377% increase in CPT-1 activity above control (p<0.0001, two-tailed t-test, Prism 3.0). As a consequence of increased CPT-1 activity, C75 at doses of 20 μg/g ml or greater, significantly increased fatty acid oxidation (see FIG. 6; 20 μg/ml, p=0.007; <20 μg/ml, p<0.0001; two-tailed t-test, Prism 3.0). Moreover, the increase in fatty acid oxidation led to significantly increased levels of ATP at C75 doses of 20 μg/ml or greater (see FIG. 7; 20 μg/ml, p=0.05; 30 μg/ml, p<0.01; 40 μg/ml, p<0.0001; two-tailed t-test, Prism 3.0). The enhanced fatty acid oxidation induced by C75 is likely responsible for the marked reduction in adipose tissue mass seen with C75 administration in vivo.
  • Example 6 In Vivo Stimulation of CPT-1 Shifts Metabolism to Fatty Acid Oxidation
  • In keeping with the C75 effect on both human and murine CPT-1 and fatty acid metabolism, C75 induces a profound and rapid stimulation of fatty acid oxidation in vivo. Mice were maintained in the Oxymax calorimeter (Oxymax Equal Flow System®, Columbus Instruments, Columbus, Ohio). Oxygen consumption and CO2 production was measured in up to four mice simultaneously using the indirect calorimeter. Measurements were recorded every 15 minutes over the entire course of the experiments. The respiratory exchange ratio (RER) was calculated by the Oxymax® software version 5.9. RER is defined as the ratio of CO2 to O2 at any given time irrespective if equilibrium was reached. Mice were maintained on water and mouse chow ad libitum. In the control mice (FIG. 8A), note the diurnal variation of RER indicating feeding and resting cycles of the animals. An RER of 1 is consistent with oxidation of carbohydrates while 0.7 indicates oxidation of fatty acids. Mice treated with C75 and maintained in the Oxymax calorimeter showed a rapid decrease in the respiratory exchange ratio (RER) to ˜0.7 (FIG. 8B). C75 treatment at 30 mg/kg disrupts the diurnal pattern of the control mice, showing a rapid drop in RER to complete oxidation of fatty acids within about 2 hours. Similarly, C75 treatment at 20 mg/kg shows a similar rate of drop of RER but without the prolonged effect (FIG. 8C). Importantly, the rate of decline of RER was similar to that observed for animals deprived of food (data not shown).
  • Despite the elevated levels of malonyl-CoA generated by C75 in vivo, C75 treatment led to a rapid, profound increase in fatty acid oxidation as measured by RER. Thus, C75 treated animals are able to significantly reduce adipose mass and reverse fatty liver, by allowing fatty acid oxidation to occur despite the high levels of malonyl-CoA generated during FAS inhibition in vivo.
  • Example 7 Enzyme Effector Activity of Various Substituted Gamma-Butyrolactones
  • Various gamma-butyrolactone analogs were prepared and tested for their effect on FAS activity, CPT-1 activity and fatty acid oxidation. The compounds included C-75, a gamma-substituted-alpha-methylene-butyrolactone synthesized as described in U.S. Pat. No. 5,981,575, FAS231 and FAS65, gamma-substituted-alpha-methylene-beta-amido-butyrolactones, synthesized as described in International Patent Publication WO 2004/006835, FAS115, a 5,5-disubstituted thiotetronic ethyl carbonate synthesized as described in International Patent Publication WO 2004/005277, and FAS89B, an 3,3,5,5-tetrasubstitued thiophene dione, synthesized as described in U.S. Provisional Patent Application 60/574,639. These compounds were assayed for inhibitory effect on FAS or stimulatory effect on CPT-1 and fatty acid oxidation, and the results of the assays are shown in the following table.
  • FAS activity was measured by monitoring the malonyl-CoA dependent oxidation of NADPH spectrophotometrically at OD340 in 96-well plates, as described in International Patent Publication WO 2004/005277. The IC50 for the compounds against FAS was determined by plotting the change in OD340 against time for each inhibitor concentration tested and determining the rate of change by linear regression. The concentration of a particular compound yielding 50% inhibition of the rate for FAS in the absence of the compound is the IC50. Stimulation of CPT-1 activity was measured as described in Example 2, except the cells were preincubated with the compound for 2 hours at the concentrations indicated in the table. Stimulation of Fatty acid oxidation was determined as described in Example 1 for cells preincubated with the compounds at the concentrations indicated in the table.
  • TABLE
    Effect of Selected Compounds on Enzyme Activities
    FAS
    Inhibition
    (IC50) CPT-1 Stimulation Fatty Acid Oxidation
    Compound μg/mL μg/mL (% Control) μg/mL (% Control)
    C75 55 20 125 10 400
    Figure US20090124684A1-20090514-C00003
    4.6 20 150 10 140
    Figure US20090124684A1-20090514-C00004
    47.9 10 400 0.625 400
    Figure US20090124684A1-20090514-C00005
    52.1 80 500 40 500
    Figure US20090124684A1-20090514-C00006
    n/a* 20 175 10 175
    *Slow binding assays not yet completed
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious that certain changes and modifications may be practiced within the scope of the appended claims. Modifications of the above-described modes for carrying out the invention that are obvious to persons of skill in medicine, immunology, hybridoma technology, pharmacology, and/or related fields are intended to be within the scope of the following claims.
  • All publications and patent applications mentioned in this specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All such publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.

Claims (5)

1. A method of inducing weight loss comprising administering an agent that stimulates carnitine palmitoyl transferase-1 (CPT-1) activity.
2. A method for stabilizing weight comprising chronic administration of an agent that stimulates CPT-1 activity in an amount that does not significantly inhibit FAS.
3. A method of screening for agents that induce weight loss, comprising determining whether a candidate weight loss agent stimulates CPT-1 activity; and selecting an agent that stimulates CPT-1 activity.
4. (canceled)
5. (canceled)
US12/266,425 2002-02-08 2008-11-06 Stimulation Of CPT-1 As A Means To Reduce Weight Abandoned US20090124684A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/266,425 US20090124684A1 (en) 2002-02-08 2008-11-06 Stimulation Of CPT-1 As A Means To Reduce Weight

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US35448002P 2002-02-08 2002-02-08
PCT/US2003/003839 WO2003066043A1 (en) 2002-02-08 2003-02-10 Stimulation of cpt-1 as a means to reduce weight
US10/503,605 US20050143467A1 (en) 2002-02-08 2003-02-10 Stimulation of cpt-1 as a means to reduce weight
US10/917,525 US20050106217A1 (en) 2002-02-08 2004-08-13 Stimulation of CPT-1 as a means to reduce weight
US11/537,968 US7459481B2 (en) 2002-02-08 2006-10-02 Stimulation of CPT-1 as a means to reduce weight
US12/266,425 US20090124684A1 (en) 2002-02-08 2008-11-06 Stimulation Of CPT-1 As A Means To Reduce Weight

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/537,968 Continuation US7459481B2 (en) 2002-02-08 2006-10-02 Stimulation of CPT-1 as a means to reduce weight

Publications (1)

Publication Number Publication Date
US20090124684A1 true US20090124684A1 (en) 2009-05-14

Family

ID=27734382

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/503,605 Abandoned US20050143467A1 (en) 2002-02-08 2003-02-10 Stimulation of cpt-1 as a means to reduce weight
US10/917,525 Abandoned US20050106217A1 (en) 2002-02-08 2004-08-13 Stimulation of CPT-1 as a means to reduce weight
US11/537,968 Expired - Fee Related US7459481B2 (en) 2002-02-08 2006-10-02 Stimulation of CPT-1 as a means to reduce weight
US12/266,425 Abandoned US20090124684A1 (en) 2002-02-08 2008-11-06 Stimulation Of CPT-1 As A Means To Reduce Weight

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US10/503,605 Abandoned US20050143467A1 (en) 2002-02-08 2003-02-10 Stimulation of cpt-1 as a means to reduce weight
US10/917,525 Abandoned US20050106217A1 (en) 2002-02-08 2004-08-13 Stimulation of CPT-1 as a means to reduce weight
US11/537,968 Expired - Fee Related US7459481B2 (en) 2002-02-08 2006-10-02 Stimulation of CPT-1 as a means to reduce weight

Country Status (12)

Country Link
US (4) US20050143467A1 (en)
EP (1) EP1471906A4 (en)
JP (2) JP2005523270A (en)
KR (1) KR20040082417A (en)
CN (1) CN1313089C (en)
AU (2) AU2003215111A1 (en)
BR (1) BR0307421A (en)
CA (1) CA2474884A1 (en)
EA (1) EA007029B1 (en)
IL (1) IL163312A (en)
MX (1) MXPA04007556A (en)
WO (1) WO2003066043A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA007029B1 (en) * 2002-02-08 2006-06-30 Джон Хопкинс Юниверсити Скул Оф Медсин Stimulation of cpt-1 as a means to reduce weight
US7649012B2 (en) * 2002-07-09 2010-01-19 Fasgen, Inc. Compounds, pharmaceutical compositions containing same, and methods of use for same
JP2007529549A (en) * 2004-03-18 2007-10-25 ファスジェン・リミテッド・ライアビリティ・カンパニー Control of eating behavior by changing nerve energy balance
BRPI0510397A (en) * 2004-05-26 2007-11-13 Fasgen Llc compounds, pharmaceutical compositions containing them, and methods of use thereof
ES2342842T3 (en) * 2005-06-06 2010-07-15 F. Hoffmann-La Roche Ag SULFAMIDE DERIVATIVES USED AS INHIBITORS OF THE CARNITI-NA-PALMITOIL-TRANSFER OF LIVER (L-CPT1).
WO2007014248A2 (en) * 2005-07-26 2007-02-01 Johns Hopkins University Method of reducing food intake
EP1760147B1 (en) * 2005-08-29 2008-12-24 F. Hoffmann-La Roche Ag Crystal structures of carnitine palmitoyltransferase-2 (CPT-2) and uses thereof
ES2405259B1 (en) * 2011-11-25 2014-09-29 Centro De Investigación Biomédica En Red Fisiopatología De La Obesidad Y Nutrición (Ciberobn) USE OF THE CHARMER (+) - C75 FOR THE TREATMENT OF OBESITY.
WO2013155528A2 (en) * 2012-04-13 2013-10-17 Fasgen, Inc. Methods for reducing brain inflammation, increasing insulin sensitivity, and reducing ceramide levels

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7459481B2 (en) * 2002-02-08 2008-12-02 The Johns Hopkins University School Of Medicine Licensing And Technology Development Stimulation of CPT-1 as a means to reduce weight

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US574639A (en) 1897-01-05 Gold-saving device
DE3026368A1 (en) * 1980-07-11 1982-02-18 Lentia GmbH Chem. u. pharm. Erzeugnisse - Industriebedarf, 8000 München NUTRITIONAL SOLUTION FOR COMPLETE PARENTERAL NUTRITION AND FOR ENHANCED ENERGY PRODUCTION
US5981575A (en) 1996-11-15 1999-11-09 Johns Hopkins University, The Inhibition of fatty acid synthase as a means to reduce adipocyte mass
US5914326A (en) * 1997-08-08 1999-06-22 Ambi Inc. Method for promoting weight and fat loss
WO2000012080A1 (en) * 1998-09-01 2000-03-09 Amway Corporation Diet composition and method of weight management
ATE330598T1 (en) * 1999-11-12 2006-07-15 Univ Johns Hopkins TREATING CANCER BY INCREASE MALONYL-COA LEVELS
AU2001238515A1 (en) * 2000-02-16 2001-08-27 The John Hopkins University School Of Medicine Weight loss induced by reduction in neuropeptide y level
WO2002079501A1 (en) * 2001-03-30 2002-10-10 Trustees Of Boston University Methods and reagents for identifying weight loss promoters and therapeutic uses therefor
US20040161803A1 (en) * 2002-04-01 2004-08-19 Corkey Barbara E. Methods and reagents for identifying weight loss promoters and therpeutic uses therefor
CN101633650A (en) 2002-07-01 2010-01-27 法斯根有限责任公司 Novel compounds, pharmaceutical compositions containing same, and methods of use for same
US7649012B2 (en) 2002-07-09 2010-01-19 Fasgen, Inc. Compounds, pharmaceutical compositions containing same, and methods of use for same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7459481B2 (en) * 2002-02-08 2008-12-02 The Johns Hopkins University School Of Medicine Licensing And Technology Development Stimulation of CPT-1 as a means to reduce weight

Also Published As

Publication number Publication date
KR20040082417A (en) 2004-09-24
WO2003066043A1 (en) 2003-08-14
CN1313089C (en) 2007-05-02
EP1471906A4 (en) 2006-02-01
EP1471906A1 (en) 2004-11-03
CN1638758A (en) 2005-07-13
AU2008249144A1 (en) 2008-12-11
MXPA04007556A (en) 2005-12-05
JP2005523270A (en) 2005-08-04
BR0307421A (en) 2004-12-28
US20070087037A1 (en) 2007-04-19
EA007029B1 (en) 2006-06-30
US20050106217A1 (en) 2005-05-19
AU2003215111A1 (en) 2003-09-02
US7459481B2 (en) 2008-12-02
IL163312A (en) 2011-12-29
JP2010047594A (en) 2010-03-04
US20050143467A1 (en) 2005-06-30
EA200401052A1 (en) 2005-02-24
CA2474884A1 (en) 2003-08-14

Similar Documents

Publication Publication Date Title
US7459481B2 (en) Stimulation of CPT-1 as a means to reduce weight
Bae et al. The old and new biochemistry of polyamines
Winder Energy-sensing and signaling by AMP-activated protein kinase in skeletal muscle
Stoner et al. There’s more to flow-mediated dilation than nitric oxide
Altamimi et al. Cardiac-specific deficiency of the mitochondrial calcium uniporter augments fatty acid oxidation and functional reserve
Ning et al. Propionyl-L-carnitine induces eNOS activation and nitric oxide synthesis in endothelial cells via PI3 and Akt kinases
WO2000062771A1 (en) Uses of phenylglycine derivatives
Schnuck et al. Characterization of the metabolic effect of β-alanine on markers of oxidative metabolism and mitochondrial biogenesis in skeletal muscle
Okamura et al. Effects of atorvastatin, amlodipine, and their combination on vascular dysfunction in insulin-resistant rats
JP5539653B2 (en) Compositions and methods by treatment with tyrosine kinase inhibitors to reduce cellular fat and predict cardiotoxicity
Hall et al. Assessing the physiological concentration and targets of nitric oxide in brain tissue
Yarasheski Managing sarcopenia with progressive resistance exercise training
Nisoli et al. Amino acids and mitochondrial biogenesis
Chen et al. Marked elevation of hypusine formation activity on eukaryotic initiation factor 5A in v-HA-RAS transformed mouse NIH3T3 cells
Ahmad et al. Enhanced expression of endothelial nitric oxide synthase in the myocardium ameliorates the progression of left ventricular hypertrophy in L-arginine treated Wistar-Kyoto rats
Ghigo et al. Retinoic acid‐induced differentiation in a human neuroblastoma cell line is associated with an increase in nitric oxide synthesis
Tawa et al. Impairment by Hypoxia or Hypoxia/Reoxygenation of Nitric Oxide–Mediated Relaxation in Isolated Monkey Coronary Artery: the Role of Intracellular Superoxide
SK16622001A3 (en) Dietary supplement energy-providing to skeletal muscles and protecting the cardiovascular tract
Koh et al. Intracellular fatty acid metabolism in skeletal muscle and insulin resistance
Tanaka et al. Protective effects of (6R)-5, 6, 7, 8-tetrahydro-l-biopterin on local ischemia/reperfusion-induced suppression of reactive hyperemia in rat gingiva
Mokhtar et al. Physical training attenuates phosphocreatine and long-chain acyl-CoA alterations in diabetic rat heart
Hanke et al. Metabolic transformation of rabbit skeletal muscle cells in primary culture in response to low glucose
Wadsworth et al. A 2-Hydroxybutyrate-mediated feedback loop regulates muscular fatigue
Anfossi et al. Glyceryl trinitrate enhances the adenosine‐induced inhibition of platelet responses: A mechanism potentially involved in the in vivo anti‐aggregating effects of organic nitrates
Guillaume Characterization of the neurokinin3-and histamine2-receptors in brown and white adipose tissue

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MA

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE JOHNS HOPKINS UNIVERSITY;REEL/FRAME:047928/0570

Effective date: 20190107