WO2008016730A2 - Compositions and methods for reducing cellular fat - Google Patents

Compositions and methods for reducing cellular fat Download PDF

Info

Publication number
WO2008016730A2
WO2008016730A2 PCT/US2007/067086 US2007067086W WO2008016730A2 WO 2008016730 A2 WO2008016730 A2 WO 2008016730A2 US 2007067086 W US2007067086 W US 2007067086W WO 2008016730 A2 WO2008016730 A2 WO 2008016730A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
fat
pgc
fatty acid
reducing cellular
Prior art date
Application number
PCT/US2007/067086
Other languages
French (fr)
Other versions
WO2008016730A3 (en
Inventor
Sarah S. Bacus
Original Assignee
Targeted Molecular Diagnostics, Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Targeted Molecular Diagnostics, Llc filed Critical Targeted Molecular Diagnostics, Llc
Publication of WO2008016730A2 publication Critical patent/WO2008016730A2/en
Publication of WO2008016730A3 publication Critical patent/WO2008016730A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid

Abstract

Methods are disclosed for the treatment of cells, such as adipocytes, to reduce their lipid content.

Description

TITLE COMPOSITIONS AND METHODS FOR REDUCING CELLULAR FAT
BACKGROUND
[0001] The heart has a tremendous capacity for ATP generation which allows it to function as an efficient pump throughout the life of the organism. The adult myocardium uses either fatty acid (FA) and/or glucose oxidation as its main energy sources. Under normal conditions, the adult heart derives most of its energy through oxidation of fatty acids in mitochondria.
[0002] Cells of the myocardium have the ability to switch between carbohydrate glycolysis and the Krebs cycle and to fat fuel sources so that ATP production is maintained at a constant rate under diverse physiological and dietary conditions. This metabolic and fuel selection flexibility is important for normal cardiac function. Although cardiac energy conversion capacity and metabolic flux is modulated at many levels, one important mechanism of regulation occurs at the level of gene expression. The expression of genes involved in multiple energy transduction pathways is dynamically regulated in response to developmental, physiological, and pathophysiological cues.
[0003] The genes involved in these key energy metabolic pathways are transcriptionally regulated by members of the nuclear receptor superfamily, specifically the fatty acid-activated peroxisome proliferator-activated receptors (PPARs) and the nuclear receptor coactivator, PPAR7 coactivator- lα (PGC- Ice), as well as the estrogen receptor-related protein ERRα, ERRβ and ERR7 and their activators PGR-I and PERC. The dynamic regulation of the cardiac PPAR/PGC-1 complex in accordance with physiological and pathophysiological states is described in more detail below.
[0004] PGC- lα is a PPAR7 coactivator, linked to adaptive thermo genesis in brown adipose. Two structurally related proteins, PGC-IB and PARC, have been cloned and appear to be involved in regulating energy metabolic pathways. The tissue-specific and inducible nature of PGC-I a expression suggests its involvement in the dynamic regulation of cellular energy yielding metabolic processes, including mitochondrial biogenesis and oxidation, hepatic gluconeogenesis, and skeletal muscle glucose uptake. PGC- lα is selectively expressed in highly oxidative tissues such as heart, skeletal muscle, brown adipose, and liver, hi the heart PGC- lα expression increases sharply at birth. This coincides with a perinatal shift from glucose metabolism to fat oxidation. PGC-I a activity and expression levels are also known to be induced by cold exposure, fasting, and exercise; stimuli known to promote oxidative metabolism. Forced expression of PGC-I in cardiac myocytes in culture induces expression of nuclear and mitochondrial genes involved in multiple mitochondrial energy-transduction/energy-production pathways, increases cellular mitochondrial number, and stimulates coupled respiration. Signaling pathways associated with these stimuli, including p38 MAP kinase, β-adrenergic/cAMP, nitric oxide, AMP kinase, and Ca2-calmodulin kinase, activate PGC- lα and its downstream target genes either by increasing PGC- \a expression or its transactivation function.
[0005] These metabolic and structural changes can result in dilated cardiomyopathy and diastolic dysfunction in the heart. Interestingly, mitochondrial proliferation is reversible and the cardiomyopathy can be rescued upon reduction in transgene expression. This suggests that, in addition to serving as an activator of cellular fatty acid metabolism through PPARs, PGC- \a is linked to the mitochondrial biogenesis. Therefore, PGC-Io: appears to serve as a master modulator of oxidative energy metabolism and responds to changes in the cellular energy status.
[0006] Evidence is emerging that the estrogen-related receptor (ERR) family of orphan Nuclear Receptors function as PGC-I -activated regulators of cardiac and skeletal muscle energy metabolism. There are three members of the ERR family: ERRα, ERRβ, and ERRγ. ERRo; and ERRγ expression is elevated in adult tissues that rely primarily on mitochondrial oxidative metabolism for ATP production, such as heart and slow twitch skeletal muscle. ERRα expression dramatically increases in heart after birth, in parallel with the global upregulation of enzymes involved in cellular fatty acid uptake and mitochondrial oxidation. Recently, ERRa and ERRγ were identified as novel partners for the PGC-I family of coactivators. This functional relationship between ERR isoforms and PGC-Io; have stimulated interest in the role of ERRs in energy metabolism.
[0007] Deletion of the ERRα gene reveals a tissue-specific role for ERRo; in constitutive regulation of lipid metabolism. White adipose mass is decreased in ERRα — mice coincident with decreased adipocyte size and lipid synthesis rates. In contrast, ERRα likely plays a role in lipid catabolism in heart, consistent with its functional interaction with PGC- lα. ERRα— '- mice, which do not display an overt cardiac phenotype, exhibit a compensatory increase in cardiac PGC- lα and ERRγ expression. These results suggest that ERR isoforms contribute to constitutive expression of fatty acid metabolic genes in heart. However, the metabolic effects of changes in gene expression remain unknown.
[0008] Gene expression profiling in cardiac myocytes that overexpress ERRo; are being used to identify cardiac ERRα target genes. ERRo; activates genes involved in energy production pathways, including cellular fatty acid uptake (LPL, CD36/FAT, H-FABP, FACS-I), β-oxidation (MCAD, VLCAD, LCHAD), and mitochondrial electron transport/oxidative phosphorylation (cytochrome c, COXIV, COXVIII, NADH ubiquinone dehydrogenase, flavoprotein-ubiquinone oxidorcductase, ATP synthase β). ERRα also increases palmitate oxidation rates in cardiac myocytes. Activation of β-oxidation enzymes genes by ERRα involves the PPARo; signaling pathway. ERRo; directly activates PPARo; gene expression, and ERRα-mediated regulation of MCAD and M-CPT I is abolished in cells derived from PPARα— ;- mice. ERRα is also now known to be involved in the PGC-lα regulation of mitochondrial biogenesis. It is known to mediate PGC-lα activation of the NRF pathway through regulation of the Gapba gene, which encodes a subunit of the NRF-2 complex and directly activates genes involved in mitochondrial oxidative metabolism at the level of transcription. ERRα with its coactivator PGC-lα activates the MCAD, cytochrome c, and ATP synthase β gene promoters. Collectively, these results identify ERRα as a regulator of cardiac oxidative energy metabolism through its involvement in the PGC- 1 regulatory circuit. However, the precise biological roles of ERRs in heart have not been identified.
[0009] The nuclear receptor ERRγ (estrogen related receptor gamma) is highly expressed in heart, skeletal muscle, kidney, and brain, as well as in the developing nervous system. The expression of the coactivators PGC-lα and PGC- Iβ in mammalian cells potently augmented transcriptional activation by ERRγ. The constitutive activation function 2 (AF-2) of the orphan receptor is important for the synergistic enhancement. Functional receptor truncation analysis has been used to identify an additional amino-terminal activation function, specific for the ERRγ2 isoform and PGC- Ice. In vitro experiments showed a direct interaction of ERRγ with both coactivators. These findings are consistent with the hypothesis that distinct regulatory functions for PGC-lα and PGC-1/3 as tissue-specific coactivators for ERRγ. Nevertheless, more studies are needed to further define these functions.
[0010] Cardiac-specific overexpression of PGC-I in transgenic mice results in uncontrolled mitochondrial proliferation in cardiac myocytes leading to loss of sarcomeric structure and a dilated cardiomyopathy. Thus, PGC-I is an important regulatory molecule in the control of cardiac mitochondrial number and function in response to energy demands.
[0011] Most, if not all of these regulatory pathways involve phosphorylation of intermediates in a signaling pathway. Inhibition of phosphorylation, such as by the action of various kinase inhibitors, affects these signaling pathways causing alterations in fatty acid metabolism which can cause organ toxicity, including cardiotoxicity. Many new anti-cancer drugs are kinase inhibitors and are accompanied by toxicity. Thus, methods are needed for identifying whether drugs may be accompanied by toxic effects and whether the toxic effects are likely to occur in a patient. Methods are also needed for avoiding toxic effects of these inhibitors while maintaining their potency against the phosphorylated receptor targets.
SUMMARY
[0012] Methods are disclosed for diagnosing whether toxicity, especially cardiotoxicity, is likely to occur in a patient selected for treatment with a variety of drugs, such as tyrosine kinase inhibitors or crbB inhibitors. Methods are also disclosed for evaluating whether a candidate drug is likely to have a toxic or cardiotoxic affect. In one method lipids, such as triglycerides and cholesterol, can be analyzed to determine whether a fatty acid oxidation disorder is present, hi another method enzymes responsible for the observed fatty acid oxidation, such as MCAD, can be determined. With respect to lipid levels it is thought that in normal cells AMP-activated protein kinase activation can lead to a characteristic reduction in the level of lipids and a corresponding increase in glycolytic and shorter carbon chain intermediates, for example of C2 to C6 carbon intermediates. Any statistically significant deviation from the characteristic lipid reduction in normal cells can be considered, for purposes of this disclosure, a fatty acid oxidation disorder. Similarly, with respect to the enzymes involved in these metabolic pathways, any statistically significant change, relative to normal cells, in the amount of activity or levels of these enzymes as measured by Western, Northern, PCR or other techniques, can be considered, for purposes of this disclosure, a fatty acid oxidation disorder. The diagnosis of a fatty acid oxidation disorder can be used to predict an increased risk of toxicity and possibly as a contra-indicator for the use of the drug. Alternatively, in the event a drug is used in a patient having a fatty acid oxidation disorder the methods can be used to indicate the need to closely follow cardiac function in the patient. Alternatively glucose uptake can be measured by known methods, such as by positron emission tomography. In situations where glucose uptake is not diminished or is not diminished to the same extent as in normal noncancerous cells upon administration of a tyrosine kinase inhibitor drug, then the drug treatment is likely to be toxic to the noncancerous cells. Alternatively, if ATP levels decrease more than in normal noncancerous cells upon exposure to a tyrosine kinase inhibitor, then the tyrosine kinase inhibitor is predicted to be toxic.
[0013] Another method for predicting whether cardiotoxicity in a patient selected for treatment with a drug, such as tyrosine kinase inhibitor, especially an erbB inhibitor, is to assess the TNFα levels in the patient, either in the tumor or blood or both. The level of TNFα can be used to predict whether a patient is likely to have an adverse event related to cardiotoxicity resulting from drug, particularly Herceptin, therapy.
[0014] Methods are also disclosed for administering drugs that activate AMP activated protein kinase (AMPK), such as certain tyrosine kinase inhibitors, to diminish lipid and fat in patients for cosmetic reasons or weight loss. The method is based on the surprising discovery that activators of AMP activated protein kinase cause a shift in cell metabolism such that lipids are oxidized into smaller carbon intermediates. The metabolic shift results in a surprising reduction in the lipid content of treated cells. Administration of AMP activated protein kinase activators in amounts that are sufficient to activate AMP activated protein kinase can be used to cause cells to loose a portion of their lipid content. Many methods for administering such compounds to cells are known and can be used. Local or systemic administration can be used. Local administration can be by injection, by a skin patch or a salve or lotion.
[0015] A method is also disclosed for administering an AMP activated protein kinase activator to a patient, or including it in a medium for incubation with an organ, in an amount that is sufficient to protect organs such as heart muscle and/or brain cells from the acute distress that would normally result from such trauma as ischemia, cytokine release, glucose deprivation and similar events that cause metabolic tension in such cells and organs where such conditions are diagnosed. Dual kinase inhibitors, particularly tyrosine kinase inhibitors that cause an increase in AMP activated protein kinase activity, can also be used. Preferably, such kinase inhibitors will be specific for their targets as described further in the detailed description. Many methods of administration are known and can be used. For example, the drugs can be included in solutions for perfusing organs or can be administered systemically.
[0016] A method is also disclosed for preserving an organ for transplant. The method involves preparing a preservation solution comprising an AMPK activator and contacting the organ with the preservation solution. The preservation solution can be any known preservation solution to which an AMPK activator is added in a sufficient amount to provide improved protection for the organ.
[0017] Additional features and advantages are described herein, and will be apparent from, the following Detailed Description and the figures.
BRIEF DESCRIPTION OF FIGURES
[0018] Figure 1 is a listing of genes regulated by Herceptin treatment in Au565 cells.
[0019] Figure 2 are photographs of Au-565 cells treated by NDF or Herceptin and stained for lipids.
[0020] Figure 3 are photographs of Au-565 cells treated by GW-2974 and stained for lipids.
[0021] Figure 4 are photographs of primary human cardiac myocytes grown under various conditions and stained for lipids. [0022] Figure 5 is a bar graph illustrating the percentage of human cardiomyocytes testing positive for lipids under various conditions.
[0023] Figure 6 are photographs of MDA-MB-468 cells treated by GW-2974 and intracellular Ca detected by Fluoro-4.
[0024] Figure 7A a photograph of a Western Blot showing the affect of certain tyrosine kinase inhibitors on expression p-eEF2 and p-AMPKα.
[0025] Figure 7B is a photograph of stained cells showing the expression of p- eEF2 in Au565 cells in the presence of various compounds.
[0026] Figure 8 is a photograph of ERRα and MCAD in cardiomyocytes cells with and without treatment by various kinase inhibitors.
[0027] Figure 9 is a bar graph illustrating the growth inhibition of HMCs treated with combinations of different types of erbB inhibitors and TNFα.
[0028] Figure 10 is a western blot of HMCs probed for NF-κB after treatment with either TNFα, GW2974 or Herceptin (or combinations).
DETAILED DESCRIPTION
[0029] In one aspect, the present disclosure is based on the discovery that drugs, such as tyrosine kinase inhibitors, like Herceptin and lapatinib (Tykerb), affect the expression of genes associated with lipid metabolic pathways and dramatically affect the amount of lipid within the cells. Treatment of otherwise normal cells or cells having normal protein tyrosine kinase regulation with the kinase inhibitors of the invention affects fatty acid metabolism by increasing or decreasing the capacity of such cells to oxidize fatty acids. When normal fat cells grown in culture are exposed to kinase inhibitors such as GW2974, GW572016, the lipid stored within those cells rapidly disappears. This observation has also been made in cardiac cells. Such studies can be conducted using Oil red 0 staining for lipids. Thus, treatment with lapatinib (tykerb) and other Herl/Her2 tyrosine kinase inhibitors cause a loss of fat from such cells that is consistent with reduced lipid synthetic rates and/or increased lipid oxidation rates. With other drugs, such as Herceptin, NDF lipid content appears to increase.
[0030] Many kinase inhibitors are also known to be useful as chemotherapeutic agents. In some patients these drugs produce cardiotoxicity. The present disclosure is based on the surprising discovery that cardiotoxicity can be associated with defects in fatty acid metabolism. Thus, patients with certain dysfunctions in fatty acid metabolism or that have high levels of TNFα in blood, and that are undergoing treatment with kinase inhibitors are more likely to suffer from cardiac malfunction such as cardiomyopathy upon treatment with kinase inhibitors such as erbB tyrosine kinase inhibitors, hi addition, it has been discovered that patients having high levels of TNFα, or its downstream survival factor NF-κB, in tumor tissue or serum generally have a better response to Herceptin. This discovery has led to the development of new methods for predicting whether patients will suffer from cardiotoxicity upon treatment with drugs, including kinase inhibitors either alone or in combination with other active agents, that affect phosphorylation states of certain cellular proteins.
[0031] A method is disclosed for analyzing a patient's lipids including triglycerides and cholesterol and/or lipid metabolic enzymes such as, MCAD, among others. The results from such analysis can then be used to predict when cardiotoxicity could result from kinase inhibitor treatment and to provide an early indication that cardiac function should be closely monitored in patients undergoing treatment with drugs, such as kinase inhibitors, including Herceptin, GW572016 or other erbB inhibitors.
[0032] The activity of 5-'AMP-activated protein kinase, which has been shown to phosphorylate and inactivate acetyl-CoA carboxylase in other tissues, has been discovered to be significantly increased at the end of ischemia, and remains elevated throughout reperfusion. Accumulation of 5'-AMP during ischemia results in an activation of AMP-activated protein kinase, which phosphorylates and inactivates acetyl-CoA carboxylase during reperfusion. The subsequent decrease in malonyl- CoA levels can result in accelerated fatty acid oxidation rates during reperfusion of ischemic hearts.
[0033] With respect to cardiac toxicity, a variety of fatty acid oxidation disorders are known and are listed below in Table I. If such a disorder is detected in a patient it can provide an indication that kinase inhibitors could be toxic to the heart. TABLE I
Acyl-CoA dehydrogenase deficiencies
Acyl-CoA dehydrogenase, short-chain (SCAD)
Acyl-CoA dehydrogenase, medium-chain (MCAD)
Acyl-CoA dehydrogenase, long-chain (LCAD)
Acyl-CoA dehydrogenase, very long-chain (VLCAD)
2-Enoyl-CoA hydratase deficiency
L-3-Hydroxyacyl-CoA dehydrogenase deficiencies
L-3-Hydroxyacyl-CoA dehydrogenase, short chain (SCHAD)
Trifunctional protein: Long-chain FA (LCHAD)
Alpha subunit (HADHA)
Beta subunit (HADHB)
3-Ketoacyl-CoA thiolase deficiency
3-Ketoacyl-CoA thiolase, Medium chain (MCKAT)
Trifunctional protein α-Methylacyl-CoA racemase (AMACR) deficiency
Carnitine-acylcarnitine translocase deficiency: 3p21
2,4-Dienoyl-CoA reductase deficiency: 8q21
Electron transfer flavoprotein (ETF) deficiency: 15q23
Ichthyosiform erythroderma (NCIE2): CGI58 gene; 3p21
Trifunctional protein deficiencies: Subunits A & B
Tyrosinemia
1° Disorders of Carnitine metabolism
Fatty acid & Carnitine transport pathways
Fatty acid oxidation pathways
Lipid disorders
Mitochondria: Biochemical abnormalities
Peroxisomal disorders
Such disorders can be detected by any suitable method. For example, in certain disorders, fatty acids can be fed to an individual and their metabolism followed. Alternatively, enzyme levels can be determined as in Western blots or mRNA levels for certain gene products can be analyzed, for example. Any detectable decrease provides an indication that a fatty acid oxidation disorder exists and that treatment with a tyrosine kinase inhibitor may be toxic to normal cells and organs.
[0034] In a method, patients who are candidates for treatment with kinase inhibitors can be screened for these diseases to determine whether they are likely to suffer myocardiocyte toxicity. For example, the biological macromolecules can be determined in myocardiocytes grown in culture to determine how the levels of these macromolecules are affected by administration of the candidate drug. In a method human myocardiocytes can be grown in culture and the level of phosphorylated AMP-activated protein kinase can be monitored in the presence of the candidate drug. This can be determined by a Western blot that detects the phosphorylated AMP activated kinase.
[0035] Without limiting the invention, it is believed that under stress conditions such as hypoxia, ischemia, glucose deprivation, and starvation, an increase in the intracellular AMPiATP ratio allosterically activates AMP-activated protein kinase (AMPK), a response designed to maintain cellular energy balance. AMP- activated protein kinase was initially discovered to inhibit preparations of acetyl-CoA carboxylase (ACC) and 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CoA reductase, HMGR). Activation of AMPK is thought to initiate a series of downstream phosphorylation events that switch cells from active ATP consumption (e.g., fatty acid, cholesterol and protein biosynthesis) to ATP production (e.g., fatty acid and glucose oxidation). Stress-induced activation of AMPK is thought to occur following its phosphorylation at threonine 172 on the a. subunit by one or more upstream AMPK kinases (AMPKKs), including calmodulin-dependent kinase kinase β (CAMKK/3), a calcium-activated protein kinase, and LKBl, a serine/threonine kinase encoded by the Peutz-Jegher syndrome tumor suppressor gene. Activation of AMPK in skeletal muscle and heart is believed to lead to the phosphorylation and inhibition of acetyl- CoA carboxylase (ACC), which in turn is thought to reduce the level of malonyl- CoA, itself an inhibitor of carnitine palmitoyltransferase 1 (CPT 1). De-repression of CPT 1 is thought to cause the concomitant increase in /3-oxidation of fatty acid, which is thought to lead to increased mitochondrial production of ATP. Stress-induced activation of AMPK is also thought to inhibit protein synthesis by inhibiting mTOR and directly modulating eEF2, a translation elongation factor known to be associated with cardiac protection. Importantly, alteration in mitochondrial function is thought to lead to cardiomyocyte death by imatinib. Moreover, inhibition of cap-dependent translation via AMPK-mediated TSC2 phosphorylation is thought to be extremely important for cell survival in response to ATP depletion. Increased biosynthesis of, rather than consumption of ATP following AMPK activation may also protect cardiomyocytes against ischemic injury.
[0036] It has been discovered that molecules such as GW2974, a potent small molecule HER2/EGFR tyrosine kinase inhibitor with a similar activity profile to lapatinib, that can activate AMPK and its downstream substrates stimulate fatty acid oxidation, which in turn increases ATP production in HER2-expressing human cardiomyocytes, protecting against apoptosis induced by TNFα, a known cytokine detected in cardiac failure. Conversely, molecules such as trastuzumab that do not activate AMPK result in enhanced cardiomyocyte cell death in response to TNFα. The effects of specific HER2-targeted therapies on AMPK and consequently energy production may predict for the risk associated cardiomyopathy and provide a novel HER2-directed therapeutic strategy to protect myocardium from the killing effects of TNFQ; or other pro-apoptotic stimuli, following acute ischemic injury.
[0037] In addition, tyrosine kinase inhibitors can be used to reduce fat in cells, particularly cells that are otherwise normal or that lack protein tyrosine kinase activity mediated disease. To this end at least a portion of a mammal or tissue can be treated with a kinase inhibitor such that the amount of lipid in the cells is reduced. Any suitable kinase inhibitor can be used. Methods for determining suitable inhibitors are well known. For example, samples of adipocytes can be grown in the presence and absence of a kinase inhibitor and stained with Oil red 0 by known methods to determine whether the kinase inhibitor causes a reduction in stored fat. Those kinase inhibitors that cause an observable reduction in fat storage are suitable for the present invention. Exemplary kinase inhibitors that are suitable for the invention include erbB inhibitors, especially including GW2974, GW572016, and the like. Table II below shows the reduction in lipid content obtained by treatment with GW2974. Au565 cells were grown under normal conditions known in the art and treated for 2 days with GW2974 (25 μM). The cells were collected, washed and sonicated in water (2,000,000 cells in 200 μL of water). The cells were spun down and were tested for acylcarnitines (byproducts of mitochondrial fatty acid oxidation) by MS/MS for intraceullar metabolites.
TABLE II
Acylcarnitines (pmoles of protein)
C18:l C16 C2
Control (Cell pellet) 8.56 4.09 148.54
GW2974 (Cell pellet) 4.1 0.83 258.88
[0038] In a method cells can be treated with suitable kinase inhibitors to reduce lipid storage. The method can include the steps of contacting the cells with a sufficient amount of a suitable tyrosine kinase inhibitor to cause the cell to rid itself of an amount and preferably most or, more preferably, virtually all of its surplus of stored lipids. The cells can be in an in vitro cell culture or can be located in an individual. The method is particularly effective when used on cells that are disease free or free from protein tyrosine kinase activity related diseases.
[0039] Methods are also disclosed for administering a kinase inhibitor, such as a tyrosine kinase inhibitor or dual tyrosine kinase inhibitor, to a patient, such as during heart reperfusion or during a heart attack, in order to counteract the fatty acid oxidation effect and protect the heart muscle and/or brain cells. Such treatments can be used to protect heart cells, brain cells and cells from other tissues and organs from acute distress caused by ischemia, cytokine release, glucose deprivation or other maladies that metabolically stress such cells.
[0040] Preferably the kinase inhibitors are specific in that they cause a shift in metabolic activity and do not affect unrelated targets. The specificity of various kinase inhibitors can be determined by methods described in Fabian et al, A small molecule-kinase interaction map for clinical kinase inhibitors. Nature Biotechnology 23, p. 329 which is incorporated by reference. It is believed that the shift in metabolic activity is brought about through an increase in AMP activated protein kinase activity.
[0041] The active agent can be administered to an individual orally, locally by injection or through a skin patch, a salve or a lotion or can be administered parenterally so long as it reaches the intended target cells in a sufficient amount to exert its lipid reducing effect. For example, it is preferred to administer the AMP activated protein kinase activator locally in a tissue such as adipose tissue that stores lipid to cause a reduction in lipid content. It may be administered systemically to patients in need of treatment for metabolic stress, heart attack, ischemia and the like.
[0042] The AMP activated protein kinase activators can be administered as salts or solvates or as free chemicals, however, it is preferred to administer the inhibitors in the form of a pharmaceutical formulation. The formulation can contain, in addition to the active agent, one or more pharmaceutically acceptable carriers, diluents or excipients.
[0043] The pharmaceutical formulations can be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. Such a unit can contain for example 0.5 mg to 1 g, preferably 70 mg to 700 mg, more preferably 5 mg to 100 mg of active agent depending on the route of administration and the age, weight and condition of the patient. For example, in mice, 100 mg/kg of GW2974 can be administered to preserve the heart during a period of starvation.
[0044] Pharmaceutical formulations can be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route. Such formulations can be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
[0045] Pharmaceutical formulations adapted for oral administration can be in the form of capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil liquid emulsions and in liposomes. [0046] Pharmaceutical formulations for transdermal administration can be presented as discrete patches intended to remain in intimate contact with the skin of the recipient for a prolonged period of time. The active ingredient can be delivered from the patch by iontophoresis by known methods.
[0047] Pharmaceutical formulations for topical administration can be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
[0048] For treatments of the external tissues the formulations can be applied as a topical ointments or creams. When formulated in an ointment, the active agent can be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active agent can also be formulated in a cream with an oil-in-water cream base or a water-in-oil base. Preferably, such ointments will allow the active agent to penetrate the skin and contact target cells and tissues, particularly for the amelioration of fat in fat laiden tissue and organs.
[0049] Pharmaceutical formulations adapted for topical administration in the mouth include lozenges, pastilles and mouth washes.
[0050] Pharmaceutical formulations for administration by inhalation include fine particle dusts or mists which can be generated by means of various types of metered dose pressurised aerosols, nebulizers or insufflators.
[0051] Pharmaceutical formulations for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
[0052] Pharmaceutical formulations for parenteral administration can include aqueous and non-aqueous sterile injection solutions which can further include antioxidants such as tocopherol, buffers, bacteriostats and solutes to make the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which can include suspending agents and thickening agents.
[0053] Formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules and tablets. [0054] Preferred unit dosage formulations are those containing a daily dose or sub-dose, or an appropriate fraction of an active ingredient.
[0055] It should be understood that in addition to the ingredients particularly mentioned above, the formulations can include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration can include flavoring agents.
[0056] The animal requiring treatment with a compound, salt or solvate of the present invention is usually a mammal, such as a human being.
[0057] Therapeutically effective amounts of the active agent, salt or solvate of the present invention will depend upon a number of factors including, for example, the age and weight of the animal, the severity of the condition requiring treatment, the nature of the formulation, and the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian. However, an effective amount of a compound of the present invention for the treatment of toxicity, will generally be in the range of 0.1 to 500 mg/kg body weight of recipient (mammal) per day and more usually in the range of 1 to 200 mg/kg body weight per day. Thus, for a 70 kg adult mammal, the actual amount per day would usually be from 70 to 700 mg and this amount can be given in a single dose per day or any number of sub-doses per day such that the total daily dose is the same. An effective amount of a salt or solvate of the present invention can be determined as a proportion of the effective amount of the compound per se.
[0058] The compounds of the present invention and their salts and solvates can be employed alone or in combination with other therapeutic agents. Combination therapies according to the present invention thus comprise the administration of at least one AMP activated protein kinase activator of the invention or a pharmaceutically acceptable salt or solvate thereof and at least one other pharmaceutically active agent, such as a cancer therapeutic. Combination actives can be administered together or separately and, when administered separately can be administered simultaneously or sequentially in any order. The amounts of the kinase inhibitor of the invention and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. Example 1
[0059] The following example demonstrates the identification of genes that are affected by treatment of Herceptin in an in vitro cell culture of Au565 cells. Au565 cells were grown under normal conditions and treated with Herceptin or left untreated. Cells were pelleted, snap frozen in liquid nitrogen and analyzed in a microarray using standard conditions. Cy3 and Cy5 labeled cDNA was prepared from RNA isolated from the cell pellets. Genes involved in lipid metabolism are shown in Table III. Genes involved in other pathways that were either upregulated or downregulated are also shown in Figure 1.
TABLE III
Changes in Metabolic Genes by Microarray Analysis of Au565 Cells That Were Untreated or Treated with Herceptin
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000018_0002
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Example 2
[0060] This example demonstrates that adipocytes lose lipid when treated with a small molecule tyrosine kinase inhibitor, GW2974. Figure 2 shows that Au565 cells treated with either an ErbB stimulatory ligand, NDF, or the monoclonal antibody Herceptin, both result in the production of lipids. This is shown by the staining of lipids with oil red (lipids are represented by red dots) against the background counterstaining of the cells (hematoxylin). Figure 3 shows that lipids are present in untreated Au565 cells but are reduced in cells treated with the dual EGFR and ErbB2 inhibitor, GW2974. Figure 4 shows cardiomyocyte cells treated with either GW2974, Herceptin or NDF. Lipids increase in cells treated with Herceptin and NDF (compared with untreated cells) but not decrease in cells treated with GW2974. Figure 5 shows a quantitative measure of lipids in control, Herceptin and GW2974 treated cells.
[0061] Treatment of cells with GW2974 causes a redistribution of intracellular calcium (Figure 6). This can be seen in MDA-MB-468 breast cancer cells where calcium is detected by fluorescently by Fluoro-4. This redistribution of calcium results in the activation and phosphorylation of AMPK. Activated AMPK represses translation by phosphorylation of the translation factor eEF-2 (Figure 7), which inactivates eEF-2 and represses protein synthesis, a known effect of TKIs. Figure 7A shows a western blot of Au565 cells treated with either a stimulatory ligand (EGF) or GW2974 and probed for p-eEF-2. p-eEF-2 is dramatically increased after GW2974 treatment. Figure 7B shows expression of p-eEF-2 by IHC. C225 and Herceptin do not increase p-eEF-2, however TKIs like Iressa, GW2974 and rapamycin do.
[0062] ERRα plays a role in lipid metabolism in cardiac cells, and MCAD is an enzyme that breaks down lipids and fatty acids. Mutations in MCAD is a common genetic disorder, especially in those of northern European descent. Figure 8 shows that in Herceptin treated cells, the level of ERRα diminished slightly. MCAD is expressed in Herceptin treated cells but is completely absent from GW2974 treated cells. EXAMPLE 3
[0063] The following example demonstrates the change in mRNA expression profile of cells treated with GW2974.
[0064] Au565 cells were grown under normal conditions and were untreated or treated with GW2974 (25 μM). Cells were pelleted, snap frozen in liquid nitrogen and subjected to microarray analysis. RNA was isolated using the Agilent Total RNA Isolation Kit. Cy3 and Cy5 labeled cRNA was prepared using the Agilent Low RNA Input Fluorescent Linear Amplification Kit. Labeled cRNAs were hybridized to a G41 10A Human 1A(V2) microarray consisting of 60-mer oligonucleotides representing over 18K well-characterized, full length, human genes. Table IV provides the results in Table form. TABLE IV
TABLE IV A - Ion Channel
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Table IV B - Cardiac regulation
Figure imgf000026_0001
Table IV C - Fatty acid and amino acid metabolism
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
[0065] It should be understood that various changes and modifications to the presently preferred embodiments described herein will be apparent to those skilled in the art. Such changes and modifications can be made without departing from the spirit and scope of the present subject matter and without diminishing its intended advantages. It is therefore intended that such changes and modifications be covered by the appended claims.

Claims

CLAIMS The invention is claimed as follows:
1. A method for reducing cellular fat in a cell comprising: contacting a cell with a sufficient amount of a AMPK activator to cause a substantial reduction in the fat content of the cell.
2. The method for reducing fat of Claim 1, wherein the AMPK activator is a tyrosine kinase inhibitor.
3. The method for reducing fat of Claim 1, wherein the AMPK activator is an ErbB tyrosine kinase inhibitor.
4. The method for reducing cellular fat of Claim 1, wherein the AMPK activator is administered parenterally.
5. The method for reducing cellular fat of Claim 1, wherein the AMPK activator is administered locally.
6. The method for reducing cellular fat of Claim 1, wherein the AMPK activator is administered locally by injection.
7. The method for reducing cellular fat of Claim 1, wherein the AMPK activator is administered locally by a skin patch.
8. The method for reducing cellular fat of Claim 1, wherein the AMPK activator is administered locally by a salve.
9. The method for reducing cellular fat of Claim 1, wherein the AMPK activator is administered locally in a lotion or by injection or in a systemic therapy.
10. The method for reducing cellular fat of Claim 1, wherein the cell is an adipocyte.
PCT/US2007/067086 2006-08-02 2007-04-20 Compositions and methods for reducing cellular fat WO2008016730A2 (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US82123006P 2006-08-02 2006-08-02
US60/821,230 2006-08-02
US82737206P 2006-09-28 2006-09-28
US60/827,372 2006-09-28
US82834506P 2006-10-05 2006-10-05
US60/828,345 2006-10-05
US86773606P 2006-11-29 2006-11-29
US60/867,736 2006-11-29

Publications (2)

Publication Number Publication Date
WO2008016730A2 true WO2008016730A2 (en) 2008-02-07
WO2008016730A3 WO2008016730A3 (en) 2008-08-28

Family

ID=38997743

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/067086 WO2008016730A2 (en) 2006-08-02 2007-04-20 Compositions and methods for reducing cellular fat

Country Status (1)

Country Link
WO (1) WO2008016730A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030212034A1 (en) * 2001-06-11 2003-11-13 Winder William W Method of treatment of obesity and paralyzed muscle and ergogenic aids
US20050038068A1 (en) * 2003-05-16 2005-02-17 Iyengar Rajesh R. Thienopyridones as AMPK activators for the treatment of diabetes and obesity
US20060069117A1 (en) * 2002-11-05 2006-03-30 Sylvain Rault Imidazopyridine derivatives, preparation method and pharmaceutical compositions containing same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030212034A1 (en) * 2001-06-11 2003-11-13 Winder William W Method of treatment of obesity and paralyzed muscle and ergogenic aids
US20060069117A1 (en) * 2002-11-05 2006-03-30 Sylvain Rault Imidazopyridine derivatives, preparation method and pharmaceutical compositions containing same
US20050038068A1 (en) * 2003-05-16 2005-02-17 Iyengar Rajesh R. Thienopyridones as AMPK activators for the treatment of diabetes and obesity

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BRANDT ET AL.: 'Mammary glands reconstituted with Neu/ErbB2 transformed HC11 cells provide a novel orthotopic tumor model for testing anti-cancer agents' ONCOGENE vol. 20, 2001, pages 5459 - 5465 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors

Also Published As

Publication number Publication date
WO2008016730A3 (en) 2008-08-28

Similar Documents

Publication Publication Date Title
Zhenyukh et al. High concentration of branched-chain amino acids promotes oxidative stress, inflammation and migration of human peripheral blood mononuclear cells via mTORC1 activation
WO2008016730A2 (en) Compositions and methods for reducing cellular fat
Wang et al. Obesity accelerates ovarian follicle development and follicle loss in rats
JP2013063090A (en) Composition and method for reducing cellular fat and predicting cardiotoxicity by treatment with tyrosine kinase inhibitor
Wang et al. Resveratrol induces brown-like adipocyte formation in white fat through activation of AMP-activated protein kinase (AMPK) α1
Onodera et al. Increased sugar uptake promotes oncogenesis via EPAC/RAP1 and O-GlcNAc pathways
Aliaga et al. Requirement of the MAP kinase cascade for cell cycle progression and differentiation of human intestinal cells
Odaira et al. Mechanisms underpinning AMP-activated protein kinase-related effects on behavior and hippocampal neurogenesis in an animal model of depression
Xie et al. Omentin-1 exerts bone-sparing effect in ovariectomized mice
Kitada et al. Calorie restriction in overweight males ameliorates obesity-related metabolic alterations and cellular adaptations through anti-aging effects, possibly including AMPK and SIRT1 activation
US20100048944A1 (en) Interactions of hedgehog and liver x receptor signaling pathways
Wilson et al. Pyruvate induces mitochondrial biogenesis by a PGC-1 α-independent mechanism
Mejhert et al. Semaphorin 3C is a novel adipokine linked to extracellular matrix composition
Zhong et al. β-hydroxy-β-methylbutyrate (HMB) improves mitochondrial function in myocytes through pathways involving PPARβ/δ and CDK4
Pratchayasakul et al. Estrogen restores brain insulin sensitivity in ovariectomized non-obese rats, but not in ovariectomized obese rats
Zhang et al. Effect of Mitofusin 2 on smooth muscle cells proliferation in hypoxic pulmonary hypertension
Mastrototaro et al. Insulin modulates the Na+/Mg2+ exchanger SLC41A1 and influences Mg2+ efflux from intracellular stores in transgenic HEK293 cells
DeClercq et al. Isomer-specific effects of conjugated linoleic acid on blood pressure, adipocyte size and function
Lv et al. Estrogen deficiency accelerates postmenopausal atherosclerosis by inducing endothelial cell ferroptosis through inhibiting NRF2/GPX4 pathway
US7915255B2 (en) Metabolism-modulating agents and uses therefor
Huang et al. Peroxisomal gene and protein expression increase in response to a high-lipid challenge in human skeletal muscle
Yan et al. WY14643 improves left ventricular myocardial mitochondrial and systolic functions in obese rats under chronic persistent hypoxia via the PPARα pathway
Giguère et al. ADAP1 limits neonatal cardiomyocyte hypertrophy by reducing integrin cell surface expression
Li et al. High-fat feeding in cardiomyocyte-restricted PPARδ knockout mice leads to cardiac overexpression of lipid metabolic genes but fails to rescue cardiac phenotypes
Zhenyukh et al. mononuclear cells via mTORC1 activation, Free Radical Biology and Medicine

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: 07761015

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

NENP Non-entry into the national phase in:

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 07761015

Country of ref document: EP

Kind code of ref document: A2