[go: up one dir, main page]

WO2025017193A1 - Procédés de traitement de la cardiomyopathie induite par une inflexibilité métabolique - Google Patents

Procédés de traitement de la cardiomyopathie induite par une inflexibilité métabolique Download PDF

Info

Publication number
WO2025017193A1
WO2025017193A1 PCT/EP2024/070602 EP2024070602W WO2025017193A1 WO 2025017193 A1 WO2025017193 A1 WO 2025017193A1 EP 2024070602 W EP2024070602 W EP 2024070602W WO 2025017193 A1 WO2025017193 A1 WO 2025017193A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
subject
prodrug
solvate
pharmaceutically acceptable
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.)
Pending
Application number
PCT/EP2024/070602
Other languages
English (en)
Inventor
Helena Edlund
Stefan NORLIN
Madelene ERICSSON
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.)
Betagenon AB
Original Assignee
Betagenon AB
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 Betagenon AB filed Critical Betagenon AB
Publication of WO2025017193A1 publication Critical patent/WO2025017193A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/433Thidiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure

Definitions

  • the present disclosure relates generally to methods of treating, preventing, or delaying the onset of heart diseases such as cardiomyopathy and heart failure by administering a compound provided herein.
  • the present disclosure further relates generally to methods of improving cardiac efficiency using a compound provided herein.
  • the present disclosure also relates generally to methods of inhibiting the activity of a pyruvate dehydrogenase kinase (PDK) using a compound provided herein.
  • PDK pyruvate dehydrogenase kinase
  • AMP-activated protein kinase is a key regulator of energy balance and is activated during times of metabolic stress that reduce the cellular pool of adenosine triphosphate (ATP; fasting, hypoxia, ischemia) or increase ATP consumption (physical activity/muscle contraction).
  • ATP adenosine triphosphate
  • ATP fasting, hypoxia, ischemia
  • ATP consumption physical activity/muscle contraction
  • AMPK is involved in a number of signaling pathways regulating energy metabolism and expenditure. Many of the clinical benefits of exercise or calorie restriction are mediated by increased AMPK activity. As such, it has become an attractive therapeutic target in a broad spectrum of diseases, including cardiometabolic disorders such as obesity and diabetes.
  • AMPK activators The pleiotropic effects of AMPK activation have sparked significant interest in the development of AMPK activators in a broad range of therapeutic applications, including metabolic diseases such as diabetes and obesity, cardiovascular and renal diseases, inflammatory diseases, and aging.
  • a number of these activators interact with the allosteric GUXJ ⁇ DQG ⁇ PHWDEROLWH ⁇ $'D0 ⁇ VLWH ⁇ D ⁇ VLWH ⁇ IRUPHG ⁇ E ⁇ WKH ⁇ LQWHUDFWLRQ ⁇ EHWZHHQ ⁇ DQG ⁇ VXEXQLWV ⁇ and which has an as yet unknown physiological function. Binding at this site allosterically activates AMPK and protects against dephosphorylation.
  • activators for the ADaM site varies between different AMPK trimers, with many activators VKRZLQJ ⁇ D ⁇ KLJKHU ⁇ DFWLYLW ⁇ IRU ⁇ FRQWDLQLQJ ⁇ FRPSOH[HV ⁇ FRPSDUHG ⁇ WR ⁇ $V ⁇ LV ⁇ WKH ⁇ predominant isoform expressed in skeletal muscle and liver, two of the primary target organs LQ ⁇ PHWDEROLF ⁇ GLVHDVHV ⁇ $03. ⁇ DFWLYDWRUV ⁇ WKDW ⁇ DUH ⁇ VHOHFWLYH ⁇ IRU ⁇ FRPSOH[HV ⁇ FRQWDLQLQJ ⁇ WKH ⁇ subunit are not ideally suited to treat metabolic diseases.
  • Cardiometabolic disorders encompass a range of conditions such as hypertension, dyslipidemia, obesity, and diabetes. Each individual's response to treatment can vary significantly due to genetic, lifestyle, and environmental factors. For an individual, these disorders are still progressive conditions leading to significant morbidity and mortality. As western societies continue to age, these conditions are a growing societal burden. An urgent and rising unmet medical need exists for therapies and interventions that can impact in the incidence and progression of cardiometabolic disorders, including interventions that can mimic the clinical benefit of exercise and calorie restriction.
  • provided herein are methods and compositions for treating and/or preventing heart diseases in a subject (e.g., human patient) by administering a compound of formula (I): (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • a subject e.g., human patient
  • methods of enhancing cardiac efficiency by administering to a subject (e.g., human) a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the subject may be a patient or healthy subject.
  • the compound of formula (I) is also referred to as 4-chloro-N-[2-[(4- chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide.
  • the compound of formula (I) is capable of restoring cardiac metabolic flexibility in the heart of a subject (e.g., patient) that suffers from various heart diseases or complications.
  • the compound of formula (I) is an inhibitor of pyruvate dehydrogenase kinase (PDK) such as PDK4 and is capable of increasing glucose oxidation in the heart.
  • PDK pyruvate dehydrogenase kinase
  • the compound of formula (I) can be used to treat or prevent various heart diseases and complications, including cardiomyopathy, heart failure, and atrial fibrillation (AF).
  • the subject e.g., patient
  • the subject is diabetic.
  • the subject e.g., patient
  • the subject is not diabetic.
  • the compound of formula (I) can be used to treat or ameliorate metabolic syndrome, which involves a collection of risk factors that may include high blood sugar, increased blood pressure, excess body fat, high triglyceride or LDL cholesterol levels, insulin resistance, and/or low HDL cholesterol levels.
  • metabolic syndrome involves a collection of risk factors that may include high blood sugar, increased blood pressure, excess body fat, high triglyceride or LDL cholesterol levels, insulin resistance, and/or low HDL cholesterol levels.
  • the disclosure provides methods of treating metabolic syndrome in a human subject in need thereof by administering a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the subject may be a human subject that suffers from a heart disease as set forth herein.
  • the subject may be a human subject that is potentially susceptible to developing a disease or condition associated with metabolic syndrome.
  • the human subject may be pre-diabetic.
  • the human subject has a combination of high blood pressure and high cholesterol and hence is susceptible to heart attack or stroke.
  • a method of treating cardiomyopathy in a subject in need thereof comprising administering to the subject an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the cardiomyopathy is associated with metabolic inflexibility.
  • the cardiomyopathy is dilated cardiomyopathy.
  • the cardiomyopathy is hypertrophic cardiomyopathy. In some embodiments, the cardiomyopathy is restrictive cardiomyopathy. In some embodiments, administration of the compound of formula (I) a pharmaceutically acceptable salt, solvate, or prodrug thereof reduces hypertension in the subject (e.g., patient). In some embodiments, administration of the compound of formula (I) lowers systolic blood pressure in the subject (e.g., patient). In other embodiments. administration of the compound of formula (I) lowers diastolic blood pressure in the subject (e.g., patient). In some embodiments, the subject (e.g., patient) is diabetic. In other embodiments, the subject (e.g., patient) is not diabetic.
  • the subject suffers from diabetic cardiomyopathy.
  • the cardiomyopathy is induced by diabetes.
  • the subject e.g., patient
  • the subject is a diabetic and has Type 1 diabetes.
  • the subject e.g., patient
  • the Type 2 diabetes is severe insulin resistant diabetes.
  • the subject e.g., patient
  • DMD Duchenne muscular dystrophy
  • the subject (e.g., patient) suffering with DMD is from about 5 years old to about 18 years ole or from about 7 years old to about 12 years old.
  • the subject e.g., patient
  • the glucocorticoid-induced cardiomyopathy is anabolic steroid-induced cardiomyopathy.
  • the glucocorticoid- induced cardiomyopathy is endogenous (Cushing’s syndrome) corticosteroid-induced cardiomyopathy.
  • the cardiomyopathy is dilated cardiomyopathy. In some embodiments, the cardiomyopathy is hypertrophic cardiomyopathy. In some embodiments, the cardiomyopathy is restrictive cardiomyopathy. In some embodiments, the cardiomyopathy is diabetic cardiomyopathy. [0016] In some aspects, provided herein is a method of treating heart failure in a subject in need thereof (e.g., patient), comprising administering to the subject an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In some embodiments, the subject’s heart failure is associated with metabolic inflexibility. In some embodiments, heart failure is heart failure with preserved ejection fraction (HFpEF).
  • HFpEF preserved ejection fraction
  • heart failure is heart failure with reduced ejection fraction (HFrEF).
  • the subject e.g., patient also suffers from diabetic cardiomyopathy. In some such embodiments, the diabetic cardiomyopathy of the subject is attenuated.
  • the subject e.g., patient
  • the subject is hyperglycemic. In other embodiments, the subject (e.g., patient) is not hyperglycemic.
  • a method of treating AF in a subject in need thereof comprising administering to the subject an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • administration of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof halts the progression or initiation of AF in a subject (e.g., patient) susceptible to heart disease.
  • a method of improving cardiac efficiency in a subject in need thereof comprising administering to the subject a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • the subject suffers from a heart disease.
  • the subject is diabetic. In other embodiments, the subject is not diabetic.
  • the subject is a human.
  • the compound of formula (I) can be administered as a salt.
  • the salt is an alkali metal salt.
  • the salt is a sodium salt.
  • the alkali metal salts of 4- chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide are of the formula: , wherein X + represents the alkali metal (e.g. lithium, rubidium, cesium, sodium, or potassium) cation.
  • X + represents the alkali metal (e.g. lithium, rubidium, cesium, sodium, or potassium) cation.
  • the compound administered in accordance with the disclosure is a prodrug of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5- yl]benzamide
  • the prodrug is has the following structure: , or a salt thereof, wherein R 1 is selected from the group consisting of -C(O)-C 2 H 4 -CO 2 H and - PO 3 H 2 , or a salt or solvate thereof.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient) at a dose of from about 100 mg to about 1,000 mg. All references to dosage amounts discussed herein refer to the free acid (protonated) form.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily at dose of from about 200 mg to about 1,000 mg.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily at dose of from about 400 mg to about 800 mg.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily at dose of from about 100 mg to about 300 mg. In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily at dose of about 100 mg, about 200 mg, about 300 mg, about 400 mg or about 500 mg.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady VWDWH ⁇ EORRG ⁇ SODVPD ⁇ FRQFHQWUDWLRQ ⁇ RI ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ , ⁇ RI ⁇ IURP ⁇ DERXW ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ J ⁇ P/ ⁇
  • compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof is administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady state blood plasma FRQFHQWUDWLRQ ⁇ RI ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ , ⁇ RI ⁇ IURP ⁇ DERXW ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ J ⁇ P
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady state AUC 0-24 of the compound of formula (I) of from about ⁇ K ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ K ⁇ J ⁇ P/ ⁇ ,Q ⁇ VRPH ⁇
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady state AUC 0-24 of the compound of formula (I) of from about ⁇ K ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ K ⁇ J ⁇ P/ ⁇ ,Q ⁇ VRPH ⁇ HPERGLPHQWV ⁇
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady state AUC 0-24 of the compound of formula (I) of from about ⁇ K ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ K ⁇ J ⁇ P/ ⁇ ,Q ⁇ VRPH ⁇ HPERGLPHQWV ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ , ⁇ or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady state AUC 0-24 of the compound of formula (I) of from about ⁇ K ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ ⁇ K ⁇ J ⁇ P/ ⁇ [0026]
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered at doses that do not increase glycogen content in the heart. In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered at doses that reduce glycogen content in the heart. [0028] In any of the preceding embodiments, the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered in combination with one or more therapeutic agents.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof is administered in combination with a sodium-glucose transport protein 2 (SGLT2) inhibitor.
  • SGLT2 inhibitor is dapaglifozin.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof is administered in an oral dosage form.
  • the oral dosage form is a capsule. In other embodiments, the oral dosage form is a tablet.
  • a method of improving fitness or cardiac performance in a subject comprising administering to the subject a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • the subject is a healthy subject.
  • the subject suffers from or is susceptible to a disease, For instance, in some embodiments, the subject is pre-diabetic.
  • the subject manifests systems associated with early stages of heart failure.
  • the subject has high cholesterol levels.
  • the subject can improve cardiac performance or increase fitness levels by taking a once daily dose of between about 50 mg to about 400 mg of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof. In some embodiments, the subject can improve cardiac performance or increase fitness levels by taking a once daily dose of between about 50 mg to about 200 mg of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof. In some embodiments, the subject can improve cardiac performance or increase fitness levels by taking a once daily dose of between about 50 mg to about 100 mg of a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof in a therapy, including any of the methods described herein.
  • a pharmaceutically acceptable salt, solvate, or prodrug thereof for treating and/or preventing heart diseases, including for example, treating and/or preventing heart diseases and complications, including cardiomyopathy, heart failure, and atrial fibrillation.
  • a pharmaceutically acceptable salt, solvate, or prodrug thereof for enhancing or improving cardiac efficiency, which may include restoring metabolic flexibility and glucose oxidation.
  • PDK pyruvate dehydrogenase kinase
  • a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof for the manufacture of a medicament.
  • the medicament is for any of the methods described herein.
  • the use of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, in the manufacture of a medicament for treating and/or preventing heart diseases including for example, treating and/or preventing heart diseases and complications, including cardiomyopathy, heart failure, and atrial fibrillation.
  • the use of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, in the manufacture of a medicament for treating or ameliorating metabolic syndrome which involves a collection of risk factors that may include high blood sugar, increased blood pressure, excess body fat, high triglyceride or LDL cholesterol levels, insulin resistance, and/or low HDL cholesterol levels.
  • FIG. 2 depicts the stimulation in skeletal muscle glucose uptake after administering the compound of formula (I) to STZ mice.
  • FIG. 2(a) Timeline in days for STZ-treatment and FDG-PET scanning.
  • FIG. 2(b) Representative PET/CT images of FDG uptake in an untreated mouse (upper panel), and a mouse treated with the compound of formula (I) (lower panel).
  • FIG. 3 depicts how the compound of formula (I) dose-dependently averts hyperglycemia in db/db mice. Fasted glucose (FIG. 3(a)) and insulin (FIG.
  • FIG. 4 depicts the induction of mitochondrial uncoupling by the compound of formula (I).
  • FIG. 4(a) OCR vs ECAR plot from last baseline measurement (measurement 3 in panel FIG. 4(a) and FIG. 4(b)).
  • FIG. 4(d) Mitochondrial function parameters calculated from OCR data in panel FIG. 4(a). Data are presented as mean ⁇ SEM.
  • FIG. 5 GHSLFWV ⁇ WKH ⁇ SUHVHUYDWLRQ ⁇ RI ⁇ -FHOO ⁇ PDVV ⁇ DQG ⁇ H[SUHVVLRQ ⁇ RI ⁇ -cell markers in db/db mice following administration of the compound of formula (I).
  • FIG. 5(b) Quantification of islet cell area.
  • FIG. 6 depicts the mitigation of hyperglycemia-induced islet gene expression changes in ex vivo cultured islets by the compound of formula (I).
  • FIG. 6(a) MA plots showing differentially expressed genes between mouse islets cultured at 22mM (G22) vs ⁇ P0 ⁇ * ⁇ JOXFRVH ⁇ DQG ⁇ P0 ⁇ JOXFRVH ⁇ 0 ⁇ RI ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ , ⁇ * ⁇ & ⁇ , ⁇ vs 11mM glucose.
  • FIG. 6(a) MA plots showing differentially expressed genes between mouse islets cultured at 22mM (G22) vs ⁇ P0 ⁇ * ⁇ JOXFRVH ⁇ DQG ⁇ P0 ⁇ JOXFRVH ⁇ 0 ⁇ RI ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ , ⁇ * ⁇ & ⁇ , ⁇ vs 11mM glucose.
  • FIG. 7 depicts the mitigation of the effect of chronic hyperglycemia on GSIS, mTORC1, and AMPK signaling in INS-1E cells by the compound of formula (I).
  • Protein expression ratios of Phosphorylated (P-) and total ACC, AMPK, RAPTOR, and S6 in INS- ⁇ ( ⁇ FXOWXUHG ⁇ XQGHU ⁇ P0 ⁇ RU ⁇ P0 ⁇ JOXFRVH ⁇ IRU ⁇ G ⁇ XQWUHDWHG ⁇ DQG ⁇ WUHDWHG ⁇ ZLWK ⁇ 0 of the compound of formula (I) for 4days (FIG. 7(c)) or 2 hours (FIG. 7(d)) (n 4-5/group). Data are presented as mean ⁇ SEM.
  • FIG. 8 depicts immunohistochemical and metabolic analyses of control and STZ mice.
  • FIG. 8(a) Representative insulin (green) and glucagon (red) double immunostaining of pancreases from
  • FIG. 9 depicts standardized uptake values (SUVs) during FDG-PET scanning.
  • FIG. 11(a) depicts PP+ and Som+ cell fraction and total pancreatic proinsulin content in BKS and db/db mice.
  • FIG. 12 depicts PP+ and Som+ cell fraction and total pancreatic proinsulin content in BKS and db/db mice.
  • FIG. 13 depicts the effects of the compound of formula (I) on gene expression signatures in ex vivo cultured mouse and human islets.
  • MA plots showing differentially expressed genes between mouse islets cultured at 22mM (G22) vs 11mM (G11) glucose (FIG. 13(a) ⁇ P0 ⁇ JOXFRVH ⁇ 0 ⁇ RI ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ , ⁇ * ⁇ & ⁇ , ⁇ YV ⁇ P0 ⁇ glucose (FIG.
  • FIG. 13(g) Overrepresentation analysis (ORA) of Molecular signature Hallmark gene sets in human LVOHWV ⁇ FXOWXUHG ⁇ DW ⁇ P0 ⁇ YV ⁇ P0 ⁇ JOXFRVH ⁇ DQG ⁇ P0 ⁇ JOXFRVH ⁇ 0 ⁇ RI ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ formula (I) vs 22mM glucose.
  • ORA Overrepresentation analysis
  • FIG. 14 depicts a representative immunoblot of INS-1E cells cultured under 11m0 ⁇ RU ⁇ P0 ⁇ JOXFRVH ⁇ IRU ⁇ G ⁇ XQWUHDWHG ⁇ DQG ⁇ WUHDWHG ⁇ ZLWK ⁇ 0 ⁇ RI ⁇ WKH ⁇ compound of formula (I) for 4 days (4d) or 2 hours (2h).
  • FIG. 14 depicts a representative immunoblot of INS-1E cells cultured under ⁇ P0 ⁇ RU ⁇ P0 ⁇ JOXFRVH ⁇ IRU ⁇ G ⁇ XQWUHDWHG ⁇ DQG ⁇ WUHDWHG ⁇ ZLWK ⁇ 0 ⁇ RI ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ (I) for 4 days (4d) or 2 hours (2h).
  • FIG. 15 depicts a schematic model for the anti-diabetic effects of the compound of formula (I).
  • a dual AMPK activator and mitochondrial uncoupler compound (I) stimulates glucose uptake and utilization in muscle and averts the glucotoxicity effects of hyperglycePLD ⁇ RQ ⁇ -cell function, in part via reduction of mTORC1 signaling and thus Pdk expression.
  • DETAILED DESCRIPTION [0026] The following description sets forth exemplary compositions, methods, parameters and the like. It should be recognized, however, that such description is not intended as a limitation on the scope of the present disclosure, but is instead provided as a description of exemplary embodiments.
  • Methods of Treatment are methods and compositions for treating and/or preventing heart diseases by administering a compound of formula (I): or a pharmaceutically acceptable salt, solvate, or prodrug thereof. Furthermore, provided herein are methods of enhancing cardiac efficiency by administering to a subject (e.g., patient or healthy subject) a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • a subject e.g., patient or healthy subject
  • a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof e.g., patient or healthy subject
  • reference to administration of a compound of formula (I) will include various salt forms, solvates and prodrugs, as described herein.
  • the compound of formula (I) is capable of reducing metabolic inflexibility in the heart muscle, a phenomenon attributed, in part, to the discovery that the compound of formula (I) inhibits PDK4 and increase glucose oxidation in the heart. As a result, the compound of formula (I) can be used to ameliorate metabolic syndrome and treat various heart conditions such as cardiomyopathy and heart failure.
  • the compound of formula (I) stimulates glucose uptake and glucose utilization in the heart muscle.
  • the compound of formula (I) promotes gene expression profiles favoring glucose oxidation rather than glycogen storage.
  • a compound of formula (I) significantly and in a dose-dependent manner improves cardiovascular function as measured by exercise endurance and cardiac output. Moreover, treatment with a compound of formula (I) improves peripheral perfusion, increases glucose uptake and utilization in skeletal and cardiac muscle without an increase in cardiac glycogen, improves cardiac function on echocardiography without an increase in heart rate, and improves exercise endurance with reduces lactate levels at exhaustion. [0030] The increase in insulin-independent glucose uptake in muscle tissue without an increase in glycogen and the observation of increased energy expenditure in animal models prompted in vitro experiments of mitochondrial function.
  • the compound of formula (I) increased basal oxygen consumption and extracellular acidification rate, suggesting increased glucose utilization by increased flux though the tricarboxylic acid (TCA) cycle and oxidative phosphorylation by mitochondrial uncoupling.
  • TCA tricarboxylic acid
  • mitochondrial uncoupling oxidative phosphorylation by mitochondrial uncoupling.
  • the current data indicate that the compound of formula (I) is a dual AMPK activator and mitochondrial uncoupler.
  • the compound of formula (I) promotes gene expression profiles favoring glucose oxidation in the muscles and heart. Skeletal muscle thioredoxin-inhibiting protein (TXNIP) expression levels are negatively correlated with glucose uptake. As described herein, administration of the compound of formula (I) reduces cardiac TXN1P levels.
  • Txnip mRNA and protein levels were reduced in skeletal muscle of the compound of formula (I) treated compared with hyperglycemic mice (see Example 3, FIG. 2(e), and FIG. 10).
  • skeletal muscle expression of Slc2a4, encoding Glut4 was reduced in STZ mice compared with controls but normalized in the compound of formula (I) treated hyperglycemic mice and that of Slc2a1, encoding Glut1, tended to be increased.
  • Untreated STZ mice showed gradually decreased peak E velocity and increased isovolumetric relaxation time (IVRT), indicating impaired diastolic function, whereas 1 week treatment with the compound of formula (I) reduced IVRT and increased peak E velocity and thus E/A ratio (FIG. 2(g), Table 4).
  • IVRT isovolumetric relaxation time
  • FIG. 2(g) E/A ratio
  • the improved filling of the left ventricle in compound of formula (I) treated mice also resulted in increased stroke volume and cardiac output.
  • cardiac echo data show that peak E velocity is improved and the E/A ratio is improved (FIG. 2(g), Table 4), leading to an improvement in stroke volume (Table 4).
  • this data confirms the impact on cardiomyopathy of the correction in expression of PDK4 and UCP3 and metabolic flexibility when the compound of formula (I) is administered.
  • the compound of formula (I) reduces TXNIP expression levels in LQVXOLQ ⁇ VHFUHWLQJ ⁇ -cells.
  • the disclosure provides PHWKRGV ⁇ RI ⁇ LQGXFLQJ ⁇ -cell rest and preservation.
  • restoring metabolic flexibility is associated with reduced TXNIP expression levels.
  • restoring cardiac metabolic flexibility is associated with inhibition of a PDK protein such as PDK4.
  • restoring metabolic flexibility is associated with increased glycolysis and increased glucose oxidation.
  • the combination of increased metabolic flexibility and increased glucose oxidation results in increased cardiac efficiency. Therefore, the disclosure provides methods of improving or restoring efficiency in a subject by administering a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the subject can be a human patient that suffers from a disease, particularly a heart disease, as set forth herein.
  • the subject can also be a human that does not suffer from any particular diseases.
  • the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be used, for example, to increase cardiac output and improve endurance.
  • the disclosure provides methods or treating or ameliorating metabolic syndrome in a human in need thereof (e.g., human patient), comprising administering a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the subject may be a human subject that suffers from a heart disease as set forth herein.
  • the subject may be a human subject that is potentially susceptible to developing a disease or condition associated with metabolic syndrome.
  • the human subject may be pre-diabetic.
  • the human subject has a combination of high blood pressure and high cholesterol and hence is susceptible to heart attack or stroke.
  • a method of treating a metabolic cardiac disease in a subject e.g., patient.
  • the disclosure provides methods of treating cardiomyopathy in a subject in need thereof (e.g., patient), comprising administering to the subject an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the cardiomyopathy is associated with metabolic inflexibility.
  • the cardiomyopathy is dilated cardiomyopathy.
  • the cardiomyopathy is hypertrophic cardiomyopathy.
  • the cardiomyopathy is restrictive cardiomyopathy.
  • administration of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof reduces hypertension in the subject (e.g., patient). In some embodiments, administration of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, lowers systolic blood pressure in the subject (e.g., patient). In other embodiments. administration of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, lowers diastolic blood pressure in the subject (e.g., patient). In some embodiments, the subject is diabetic. In other embodiments, the subject is not diabetic.
  • the subject suffers from diabetic cardiomyopathy.
  • the cardiomyopathy is induced by diabetes.
  • the subject e.g., patient
  • the subject is a diabetic and has Type 1 diabetes.
  • the subject e.g., patient
  • the Type 2 diabetes is severe insulin resistant diabetes.
  • the diabetic cardiomyopathy is selected from the group consisting of diabetic dilated cardiomyopathy, diabetic hypertrophic cardiomyopathy, diabetic arrhythmogenic cardiomyopathy, diabetic restrictive cardiomyopathy, diabetic left ventricular cardiomyopathy, and diabetic Takotsubo cardiomyopathy.
  • the subject e.g., patient suffers from Duchenne muscular dystrophy (DMD) cardiomyopathy.
  • DMD Duchenne muscular dystrophy
  • the subject e.g., patient suffering with DMD is from about 5 years old to about 18 years ole or from about 7 years old to about 12 years old.
  • the subject e.g., patient suffers from glucocorticoid- induced cardiomyopathy.
  • the glucocorticoid-induced cardiomyopathy is anabolic steroid-induced cardiomyopathy.
  • the glucocorticoid- induced cardiomyopathy is endogenous (Cushing’s syndrome) corticosteroid-induced cardiomyopathy.
  • the subject e.g., patient
  • the cardiomyopathy is arrhythmogenic right ventricular cardiomyopathy.
  • the subject e.g., patient
  • the subject e.g., patient
  • the subject e.g., patient
  • a method of treating heart failure in a subject in need thereof comprising administering to the subject an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the subject is associated with metabolic inflexibility.
  • heart failure is heart failure with preserved ejection fraction (HFpEF).
  • heart failure is heart failure with reduced ejection fraction (HFrEF).
  • the subject e.g., patient
  • the diabetic cardiomyopathy of the subject is attenuated.
  • the subject e.g., patient
  • the subject is hyperglycemic. In other embodiments, the subject (e.g., patient) is not hyperglycemic.
  • a method of preventing or delaying the onset of heart failure in a subject in need thereof comprising administering to the subject a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • heart failure is heart failure with preserved ejection fraction (HFpEF).
  • heart failure is heart failure with reduced ejection fraction (HFrEF).
  • the subject e.g., patient
  • the diabetic cardiomyopathy of the subject is attenuated.
  • a method of ameliorating one or more systems associated with heart failure in a subject in need thereof comprising administering to the subject a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • heart failure is heart failure with preserved ejection fraction (HFpEF).
  • heart failure is heart failure with reduced ejection fraction (HFrEF).
  • the subject e.g., patient
  • the subject also suffers from diabetic cardiomyopathy.
  • the diabetic cardiomyopathy of the subject is attenuated.
  • diastolic blood pressure of the heart failure in the subject is reduced following administration of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the subject e.g., patient
  • the subject also suffers from diabetic cardiomyopathy.
  • the diabetic cardiomyopathy of the subject is attenuated.
  • systolic blood pressure of the heart failure in the subject is reduced following administration of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the subject (e.g., patient) with heart failure is hyperglycemic. In some embodiments, the subject (e.g., patient) with heart failure is not hyperglycemic. In some embodiments, the subject’s heart failure is associated with hyperinsulinemia. In some embodiments, the subject (e.g., patient) with heart failure has diabetic cardiomyopathy. In some embodiments, the subject (e.g., patient) with heart failure has diabetic dilated cardiomyopathy, diabetic hypertrophic cardiomyopathy, diabetic arrhythmogenic cardiomyopathy, diabetic restrictive cardiomyopathy, diabetic left ventricular cardiomyopathy, or diabetic Takotsubo cardiomyopathy.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, to a subject (e.g., patient) with heart failure and diabetic cardiomyopathy reduces the diabetic cardiomyopathy in the subject (e.g., patient).
  • the subject (e.g., patient) with heart failure has diabetes.
  • subject (e.g., patient) patient with heart failure has Type 1 diabetes.
  • the subject (e.g., patient) with heart failure has Type 2 diabetes.
  • the subject (e.g., patient) with heart failure does not have diabetes.
  • the disclosure also provides various other active agents that can be used in combination with a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, to treat heart diseases.
  • the method of treating, preventing, or delaying the onset of heart failure in a subject in need thereof (e.g., patient) further comprises administering to the subject a sodium-glucose transport protein 2 (SGLT2) inhibitor.
  • the SGLT2 inhibitor is empagliflozin or dapaglifozin.
  • the SGLT2 inhibitor is empagliflozin.
  • the SGLT2 inhibitor is dapaglifozin.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, to the subject in need thereof lowers systolic blood pressure in the subject.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, to the subject in need thereof lowers diastolic blood pressure in the subject.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and the SGLT2 inhibitor to the subject in need thereof lowers systolic blood pressure in the subject.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and the SGLT2 inhibitor to the subject in need thereof lowers diastolic blood pressure in the subject.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and empagliflozin to the subject in need thereof lowers systolic blood pressure in the subject.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and empagliflozin to the subject in need thereof lowers diastolic blood pressure in the subject.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and dapaglifozin to the subject in need thereof lowers systolic blood pressure in the subject.
  • administering the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and dapaglifozin to the subject in need thereof lowers diastolic blood pressure in the subject.
  • a method of improving cardiac efficiency in a subject comprising administering to the subject a compound of formula (I) or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • the subject suffers from a heart disease.
  • the subject has diabetes.
  • the subject e.g., patient
  • Type 1 diabetes In some embodiments, the subject (e.g., patient) has Type 2 diabetes.
  • the subject is a human.
  • the compound of formula (I) behaves as an exercise mimetic in that it increases cardiac function and exercise capacity without promoting glycogen accumulation in heart tissue.
  • the compound of formula (I) promotes mitochondrial uncoupling in myotubes. Mitochondrial uncoupling dissipates the potential-energy gradient across the inner mitochondrial membrane. The potential energy is converted to heat energy rather than being used in oxidative phosphorylation (a process which converts ADP to ATP).
  • administering the compound of formula (I) generates a metabolic demand for glucose that promotes glucose utilization rather than glycogen accumulation in cells.
  • the methods provided herein restore metabolic flexibility in a subject in need thereof (e.g., patient) by averting gene expression changes associated with metabolic inflexibility.
  • the methods provided herein increase the metabolic demand in the subject (e.g., patient).
  • the methods provided herein increase glucose uptake in the subject (e.g., patient).
  • the methods provided herein increase glucose oxidation in the subject (e.g., patient).
  • the methods provided herein decrease glycogen accumulation in the heart.
  • the PDK inhibitor is a PDK1 inhibitor.
  • the PDK inhibitor is a PDK2 inhibitor.
  • the PDK inhibitor is a PDK3 inhibitor.
  • the PDK inhibitor is a PDK4 inhibitor.
  • a method of treating AF in a subject in need thereof comprising administering to the subject an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • administration of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof halts the progression or initiation of AF in a subject (e.g., patient) susceptible to heart disease.
  • the subject e.g., patient
  • the subject e.g., patient
  • the subject e.g., patient
  • the subject e.g., patient
  • the subject has Type 2 diabetes.
  • the subject e.g., patient
  • Fitness Enhancement/Cardiac Performance [0057]
  • a method of improving fitness or cardiac performance in a subject comprising administering to the subject a compound of formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof.
  • the subject is a healthy subject.
  • the subject suffers from or is susceptible to a disease.
  • the subject is pre-diabetic.
  • the subject manifests systems associated with early stages of heart failure.
  • the subject has high cholesterol levels.
  • the subject can improve cardiac performance or increase fitness levels by taking a once daily dose of between about 20 mg to about 500 mg of a compound of formula (I). In some embodiments, the subject can improve cardiac performance or increase fitness levels by taking a once daily dose of between about 50 mg to about 400 mg of a compound of formula (I). In some embodiments, the subject can improve cardiac performance or increase fitness levels by taking a once daily dose of between about 50 mg to about 200 mg of a compound of formula (I). In some embodiments, the subject can improve cardiac performance or increase fitness levels by taking a once daily dose of between about 50 mg to about 100 mg of a compound of formula (I).
  • the subject can improve cardiac performance or increase fitness levels by taking a once daily dose of between about 50 mg to about 100 mg of a compound of formula (I).
  • Treatment Compounds includes exemplary compounds as described in further detail below.
  • the compound used in the methods provided herein may include salts, solvates, or prodrugs thereof.
  • the compound is 4-chloro-N-[2-[(4-chlorophenyl)methyl]- 3-oxo-1,2,4-thiadiazol-5-yl]benzamide, or a salt, solvate or a prodrug thereof.
  • the compound is an alkali metal salt of 4-chloro-N-[2-[(4- chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide.
  • Alkali metals are metals found, along with hydrogen, in group I of the periodic table. The alkali metals are lithium, sodium, potassium, rubidium, cesium, and francium. It will therefore be understood that an “alkali metal salt” is a chemical compound consisting of an assembly of cations of one or more alkali metals and associated anions.
  • an alkali metal salt of 4- chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide refers to a compound comprising alkali metal cations (e.g., lithium, rubidium, cesium, sodium, and potassium) and anions of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5- yl]benzamide.
  • alkali metal salts of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3- oxo-1,2,4-thiadiazol-5-yl]benzamide are as depicted below: .
  • X + represents the alkali metal (e.g. lithium, rubidium, cesium, sodium, or potassium) cation.
  • a “sodium salt” is a chemical compound consisting of an assembly of cations of sodium and associated anions.
  • a sodium salt of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide refers to a compound comprising sodium cations and anions of 4-chloro-N-[2-[(4- chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide.
  • Na + represents the sodium cation.
  • the alkali metal salt of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4- thiadiazol-5-yl]benzamide may dissociate into its anionic and cationic components.
  • a suitable solvent e.g., water
  • the alkali metal salt of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4- thiadiazol-5-yl]benzamide may dissociate into its anionic and cationic components.
  • the depicted structure represents one of the possible tautomeric forms, wherein the actual tautomeric form(s) observed may vary depending on environmental factors such as solvent, temperature, or pH. All tautomeric (and resonance) forms and mixtures thereof are included within the scope of the Invention.
  • alkali metal salts of 4-chloro-N-[2-[(4- chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide are solid under ambient conditions, and thus the scope of the invention includes all amorphous, crystalline, and part crystalline forms thereof.
  • Alkali metal salts of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4- thiadiazol-5-yl]benzamide may be prepared in accordance with techniques that are well known to those skilled in the art.
  • 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3- oxo-1,2,4-thiadiazol-5-yl]benzamide may be reacted with the appropriate alkali metal hydroxide, or an alternative alkali metal base compound. Salt switching techniques may also be used to convert one salt into another salt.
  • Sodium salts of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5- yl]benzamide may be prepared in accordance with techniques that are well known to those skilled in the art.
  • 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4- thiadiazol-5-yl]benzamide may be reacted with sodium hydroxide, or an alternative sodium base compound.
  • Salt switching techniques may also be used to convert one salt into another salt.
  • 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo- 1,2,4-thiadiazol-5-yl]benzamide 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4- thiadiazol-5-yl]benzamide may be prepared in accordance with techniques that are well known to those skilled in the art.
  • 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3- oxo-1,2,4-thiadiazol-5-yl]benzamide may be made in accordance with the techniques described in international patent application WO 2011/004162.
  • the alkali metal salt of 4-chloro-N-[2-[(4- chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide is a sodium or potassium salt of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide.
  • the salt is a sodium salt of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo- 1,2,4-thiadiazol-5-yl]benzamide.
  • the compound used in the methods provided herein is a compound of formula (II): or a salt thereof, wherein R 1 is selected from the group consisting of -C(O)-C 2 H 4 -CO 2 H and - PO 3 H 2 , or a salt or solvate thereof.
  • this compound is able to metabolise in vivo to form 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5- yl] benzamide.
  • this compound is a salt.
  • salts of this compound include: wherein X + represents an alkali metal, alkaline earth metal or quaternary ammonium (e.g. lithium, magnesium, calcium, ammonium, tetramethylammonium and, particularly, sodium and potassium) cation, with appropriate stoichiometric adjustments being made in view of charges of the ions.
  • X + represents an alkali metal (e.g.
  • the treatment compound is administered to a human subject in need thereof in the form of a pharmaceutical formulation, which is also referred to herein as a pharmaceutical dosage form.
  • a pharmaceutical dosage form which is also referred to herein as a pharmaceutical dosage form.
  • the treatment compound is the sole active pharmaceutical ingredient present in the dosage form.
  • treatment compound is present in the dosage form alongside one or more other active pharmaceutical ingredients, or may be administered as part of a combination therapy with one or more other active pharmaceutical ingredients.
  • the treatment compound is provided in the form of particles having a particle size distribution defined by a D90 of less than about 10 ⁇ m (e.g. as measured using laser diffraction).
  • the particles containing the treatment compound may have a particle size distribution defined by a D90 of less than about 10 ⁇ m (e.g. from about 5 ⁇ m to about 10 ⁇ m) (e.g. as measured using laser diffraction).
  • the particle size distribution may alternatively be defined by a D90 of less than about 8 ⁇ m (e.g. from about 5 ⁇ m to about 8 ⁇ m).
  • the particles consisting of the treatment compound may have a particle size distribution defined by a D50 of less than about 6 ⁇ m (e.g. from about 0.5 ⁇ m to about 6 ⁇ m).
  • the particle size distribution of the particles consisting of the treatment compound may further be a defined by a D10 of less than about 2 ⁇ m (e.g. from about 0.2 ⁇ m to about 2 ⁇ m).
  • the particle size distribution parameters mentioned above may be applicable, individually or in combination.
  • the dosage form comprises particles containing the treatment compound, said particles having a particle size distribution defined by a D90 of less than about 10 ⁇ m and a D50 of less than about 6 ⁇ m.
  • said particles may have a particle size distribution defined by a D90 of less than 9 ⁇ m; a D50 of less than 6 ⁇ m or less than 5 ⁇ m; and a D10 of less than 2 ⁇ m or less than 1.5 ⁇ m.
  • the particle size distribution of particles containing the treatment compound may be measured by laser diffraction, using, for example a commercially available particle size analyzer.
  • Oral Dosage Forms [0075]
  • the treatment compound may be provided in the form of a tablet or a capsule.
  • capsules such as soft gelatin capsules may be prepared containing the treatment compound alone, or together with a suitable vehicle, e.g. vegetable oil, fat, etc.
  • hard gelatin capsules may contain the treatment compound alone, or in combination with solid powdered ingredients such as a disaccharide (e.g. lactose or saccharose), an alcohol sugar (e.g. sorbitol or mannitol), a vegetable starch (e.g. potato starch or corn starch), a polysaccharide (e.g. amylopectin or cellulose derivatives), or gelling agent (e.g. gelatin).
  • a disaccharide e.g. lactose or saccharose
  • an alcohol sugar e.g. sorbitol or mannitol
  • a vegetable starch e.g. potato starch or corn starch
  • a polysaccharide e.g. amylopectin or cellulose derivatives
  • gelling agent e.g. gelatin
  • the one or more pharmaceutically acceptable excipients may be selected with due regard to the intended route of administration in accordance with standard pharmaceutical practice.
  • Such pharmaceutically acceptable excipients are preferably chemically inert to the active compound and are preferably have no detrimental side effects or toxicity under the conditions of use.
  • Suitable pharmaceutical formulations may be found in, for example, Remington The Science and Practice of Pharmacy, 19th ed., Mack Printing Company, Easton, Pennsylvania (1995). A brief review of methods of drug delivery may also be found in, e.g., Langer, Science 249, 1527 (1990). [0077]
  • the pharmaceutical dosage forms described herein may act systemically, and may therefore be administered accordingly using suitable techniques known to those skilled in the art.
  • an oral pharmaceutical dosage form comprising from about 100 to about 1000 mg of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4- thiadiazol-5-yl]benzamide, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • an oral pharmaceutical dosage form comprising from about 200 to about 1000 mg of a sodium salt of 4-chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4- thiadiazol-5-yl]benzamide.
  • Dosage forms intended for oral administration may further comprise an enteric coating in order to prevent or minimize dissolution or disintegration in the gastric environment.
  • oral preparations e.g., capsules or tablets coated by an enteric coating may provide targeted release of the treatment compound in the small intestine.
  • the enteric coating may be present on surface of the formulation (e.g., on the surface of a tablet or a capsule), or each of the particles containing the treatment compound may be coated with the enteric coating.
  • the pharmaceutical dosage form used in the method of the invention further comprises an enteric coating.
  • the enteric coating is present on the pharmaceutical dosage form, and in some variations, said coating may be provided as an outer layer on the pharmaceutical dosage form.
  • particles containing the treatment compound may be individually coated with the enteric coating, and said coated particles may be prepared into the pharmaceutical dosage form.
  • the pharmaceutical dosage form contains particles comprising the treatment compound and each particle is coated with the enteric coating.
  • enteric coating refers to a substance (e.g., a polymer) that is incorporated into an oral medication (e.g., applied onto the surface of a tablet, a capsule, particles or pellets) and that inhibits dissolution or disintegration of the medication in the gastric environment.
  • enteric coatings are typically stable at the highly acidic pH found in the stomach, but break down rapidly in the relatively basic pH of the small intestine. Therefore, enteric coatings prevent release of the active ingredient in the medication until it reaches the small intestine.
  • Any enteric coating known to the skilled person may be used in the present invention.
  • Particular enteric coating materials include those which comprise beeswax, shellac, an alkylcellulose polymer resin (e.g. ethylcellulose polymers, carboxymethylethylcellulose, or hydroxypropyl methylcellulose phthalate) or an acrylic polymer resin (e.g.
  • acrylic acid and methacrylic acid copolymers methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, methacrylate copolymers, methacrylic acid copolymer, aminoalkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), poly(methacrylic acid) (anhydride), polymethacrylate, methyl methacrylate copolymer, poly(methyl methacrylate) copolymer, polyacrylamide, poly(methacrylic acid anhydride), and glycidyl methacrylate copolymers), cellulose acetate phthalate and polyvinyl acetate phthalate.
  • the pharmaceutical compositions comprise the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and at least one pharmaceutically acceptable excipient.
  • the at least one pharmaceutically acceptable excipient may be a lubricant, a binder, a filler, a surfactant, a diluent, an anti- adherent, a coating, a flavoring, a colorant, a glidant, a preservative, a sweetener, a disintegrant, an adsorbent, a buffering agent, an antioxidant, a chelating agent, a dissolution enhancer, a dissolution retardant, or a wetting agent.
  • Particular pharmaceutically acceptable excipients include mannitol, PVP (polyvinylpyrrolidone) K30, lactose, saccharose, sorbitol, starch, amylopectin, cellulose derivatives, gelatin, or another suitable ingredients, as well as disintegrating agents and lubricating agents such as sodium lauryl sulfate, Na-docusate, magnesium stearate, calcium stearate, sodium stearyl fumarate and polyethylene glycol waxes.
  • PVP polyvinylpyrrolidone
  • lactose lactose
  • saccharose lactose
  • sorbitol starch
  • amylopectin cellulose derivatives
  • gelatin or another suitable ingredients
  • disintegrating agents and lubricating agents such as sodium lauryl sulfate, Na-docusate, magnesium stearate, calcium stearate, sodium stearyl fumarate and polyethylene glycol waxes.
  • particles containing the treatment compound may be mixed, either together or separately, with mannitol, PVP (polyvinylpyrrolidone) K30 and sodium lauryl sulfate.
  • the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof may be mixed, either together or separately, with one or more of the pharmaceutical excipients (including basic excipients) listed above.
  • Mixtures of the treatment compound and one or more pharmaceutically acceptable excipients may be processed into pellets or granules, or compressed into tablets.
  • pharmaceutical dosage form of the method of the inventions may be a tablet, mini-tablets, blocks, pellets, particles, granules, or a powder for oral administration.
  • Pharmaceutical formulations that may be mentioned include those in which the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, is present in a total amount that is at least 1% (or at least 10%, at least 30% or at least 50%) by weight of the formulation. That is, the weight ratio of the treatment compound to the totality of the components (i.e., the treatment compound and all pharmaceutical excipients, e.g.
  • a “therapeutically effective amount”, an “effective amount” or a “dosage” as used herein refers to an amount of a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, that is sufficient to produce a desired effect, which can be a therapeutic and/or beneficial effect.
  • the effective amount or dosage will vary with the age or general condition of the individual or subject (e.g., a human), the severity of the condition being treated, the particular agents administered, the duration of the treatment, the nature of any concurrent treatment, the pharmaceutically acceptable carrier used, and like factors within the knowledge and expertise of those skilled in the art. [0089]
  • dosage amounts and pharmacokinetic parameters described below relate to treating patients with particular diseases, specifically heart diseases as set forth herein.
  • the dosage amounts and pharmacokinetic parameters also can apply to human subjects taking the drugs for fitness enhancement or cardiac improvement.
  • lower amounts of the compound of formula (I) may be required to improve fitness or cardiac function than when the compound is administered to a subject (e.g., patient) with a heart disease.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient) at a dose of from about 100 mg to about 1,000 mg.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily at dose of from about 200 mg to about 1,000 mg.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily at dose of from about 400 mg to about 800 mg.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily at dose of from about 100 mg to about 300 mg. In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily at dose of about 100 mg, about 200 mg, about 300 mg, about 400 mg or about 500 mg.
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady VWDWH ⁇ EORRG ⁇ SODVPD ⁇ FRQFHQWUDWLRQ ⁇ RI ⁇ WKH ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ , ⁇ RI ⁇ IURP ⁇ DERXW ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ J ⁇ P/ ⁇ ,Q ⁇ VRPH ⁇ HPERGLPHQWV ⁇ FRPSRXQG ⁇ RI ⁇ IRUPXOD ⁇ , ⁇ RU ⁇ D ⁇ SKDUPDFHXWLFDOO ⁇ acceptable salt, solvate, or prodrug thereof is administered orally once daily to a subject or a subject in need thereof (e.g., patient), wherein the administration results in a steady state EORRG ⁇ SODVPD ⁇ FRQFHQWUDWLRQ ⁇ RI ⁇ WKH ⁇ FRPSR
  • the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady state AUC 0-24 of the compound of formula (I) of from about ⁇ K ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ K ⁇ J ⁇ P/ ⁇ ,Q ⁇ VRPH ⁇ HPERGLPHQWV ⁇ the compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject in need thereof (e.g., patient), wherein the administration results in a steady state AUC 0-24 of the compound of formula (I) of from about ⁇ K ⁇ J ⁇ P/ ⁇ WR ⁇ DERXW ⁇ K ⁇ J ⁇ P/ ⁇ ,Q ⁇ VRPH ⁇ HPERGLPHQWV ⁇ WKH ⁇ compound of formula (I) or a pharmaceutically acceptable salt, solvate, or prodrug thereof can be administered orally once daily to a subject
  • Table 1 Exemplary composition of tablet comprising Na salt tablet of formula (I) Component Content per tablet Function Reference to standard Compound of formula (I) 4 24.2 Drug substance N a salt 4 mg* API specification Microcrystalline cellulose 206.59 mg Filler/ Binder Ph. Eur. Lactose monohydrate 413.17 mg Filler Ph. Eur. Sodium starch glycolate 90.00 mg Disintegrant Ph. Eur. Colloidal silicon dioxide 30.00 mg Glidant Ph. Eur. Magnesium stearate 36.00 mg Lubricant Ph.
  • the method of the invention may comprise (e.g., be combined with) further treatment(s) for the same condition.
  • the salt of the invention may be administered in conjunction with one or more other (i.e. different) therapeutic agents that are useful in treating that disease or disorder.
  • Such combination treatments may involve the administration of the salt of the invention to the subject in conjunction (e.g., sequentially or simultaneously) with the different therapeutic agent in the same formulation, or preferably in a separate formulation.
  • administering in conjunction with we include that the respective active ingredients are administered, sequentially or simultaneously, as part of a medical intervention directed towards treatment of the relevant condition.
  • simultaneously we mean that the salt of the invention and the different therapeutic agent are administered alongside one another, either in a single pharmaceutical dosage form comprising both active ingredients or in separate dosage forms administered at the same time.
  • the term “administration in conjunction with” includes that the salt of the invention and the different therapeutic agent are administered either together, or sufficiently closely in time, to enable a beneficial effect for the patient that is greater, over the course of the treatment of the relevant condition, than if either agent is administered alone in the absence of the other component over the same course of treatment. Determination of whether a combination provides a greater beneficial effect in respect of, and over the course of, treatment of a particular condition will depend upon the condition to be treated, but may be achieved routinely by the skilled person.
  • the term “in conjunction with” includes that one or other of the two active ingredients may be administered (optionally repeatedly) prior to, after, and/or at the same time as, administration of the other.
  • the terms “administered simultaneously” and “administered at the same time as” include instances in which the individual doses of the salt of the invention and the different therapeutic agent are administered within 6 hours, 3 hours, 2 hours, 1 hour, 45 minutes, 30 minutes, 20 minutes or 10 minutes) of each other.
  • the other therapeutic agent will be a sodium-glucose transport protein 2 (SGLT2) inhibitor, or a pharmaceutically acceptable salt, solvate or prodrug thereof such that the combination is useful for treating diseases such as type 2 diabetes.
  • the method of the invention involves sequential or simultaneous administration of the sodium salt of 4- chloro-N-[2-[(4-chlorophenyl)methyl]-3-oxo-1,2,4-thiadiazol-5-yl]benzamide and the SGLT2 inhibitor.
  • a sodium-glucose transport protein 2 inhibitor is a substance or agent that elicits a decrease in one or more functions of sodium- glucose transport protein 2, and by “decrease in the functions of sodium-glucose transport protein 2” we include the cessation of one or more functions of sodium-glucose transport protein 2, or a reduction in the rate of a particular function.
  • a particular function that may be fully or partially inhibited is the ability of sodium-glucose transport protein 2 to act as a glucose transporter.
  • the sodium-glucose transport protein 2 inhibitor is a gliflozin.
  • Gliflozins are a known class of small-molecule sodium-glucose transport protein 2 inhibitors. Hawley et al. (Diabetes, 2016, 65, 2784–2794) and Villani et al. (Molecular Metabolism, 2016, 5, 1048–1056) have recently discussed the possible mechanisms of action of certain gliflozins.
  • gliflozins which may be mentioned include dapagliflozin, canagliflozin, empagliflozin, ipragliflozin, tofogliflozin, sergliflozin (such as sergliflozin etabonate), remogliflozin (such as remogliflozin etabonate), ertugliflozin and sotagliflozin.
  • the sodium-glucose transport protein 2 inhibitor is dapagliflozin. [0104]
  • the sodium-glucose transport protein 2 inhibitor is a pharmaceutically acceptable salt of a gliflozin.
  • the further active ingredient may be a pharmaceutically acceptable salt of dapagliflozin, canagliflozin, empagliflozin, ipragliflozin, tofogliflozin, sergliflozin (such as sergliflozin etabonate), remogliflozin (such as remogliflozin etabonate), ertugliflozin or sotagliflozin.
  • the sodium-glucose transport protein 2 inhibitor is a solvate of a gliflozin.
  • the further active ingredient may be a solvate of dapagliflozin, canagliflozin, empagliflozin, ipragliflozin, tofogliflozin, sergliflozin (such as sergliflozin etabonate), remogliflozin (such as remogliflozin etabonate), ertugliflozin or sotagliflozin.
  • the sodium-glucose transport protein 2 inhibitor is a prodrug of a gliflozin.
  • the further active ingredient may be a prodrug of dapagliflozin, canagliflozin, empagliflozin, ipragliflozin, tofogliflozin, sergliflozin (such as sergliflozin etabonate), remogliflozin (such as remogliflozin etabonate), ertugliflozin or sotagliflozin.
  • dapagliflozin canagliflozin, empagliflozin, ipragliflozin, tofogliflozin, sergliflozin (such as sergliflozin etabonate), remogliflozin (such as remogliflozin etabonate), ertugliflozin or sotagliflozin.
  • the methods of the invention may also have the advantage that the dose-efficient methods using the salt of the invention may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g. higher oral bioavailability and/or lower clearance) than over other therapies known in the prior art, whether for use in the above-stated indications or otherwise.
  • methods of the invention may have the advantage that they are more efficacious and/or exhibit advantageous properties in vivo such as fewer side effects as a result of the dose-efficient characteristics of the salt of the invention.
  • a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof in a therapy, including any of the methods described herein.
  • the use of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof for treating and/or preventing heart diseases, including for example, treating and/or preventing heart diseases and complications, including cardiomyopathy, heart failure, and atrial fibrillation.
  • a pharmaceutically acceptable salt, solvate, or prodrug thereof for treating cardiomyopathy, or preventing or delaying the onset of cardiomyopathy.
  • a pharmaceutically acceptable salt, solvate, or prodrug thereof for treating heart failure or atrial fibrillation in a subject in need thereof.
  • PDK pyruvate dehydrogenase kinase
  • a compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof for the manufacture of a medicament.
  • the medicament is for any of the methods described herein.
  • the use of the compound of formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, in the manufacture of a medicament for treating or ameliorating metabolic syndrome which involves a collection of risk factors that may include high blood sugar, increased blood pressure, excess body fat, high triglyceride or LDL cholesterol levels, insulin resistance, and/or low HDL cholesterol levels.
  • mice were housed in the certified animal facility, with 12 h light/dark cycle and ad libitum access to the respective diets. Leptin receptor-deficient male BKS.Cg-Dock7m +/+ Leprdb/J (db/db), and C57BLKS/J (BKS) mice were obtained.
  • F1 mice were obtained from breeding male C57BL/6J mice with female CBA/CaCrl. Nine weeks old male F1 mice were treated with multiple low dose streptozotocin (50 mg/kg*day for 5 consecutive days; freshly prepared in 0.1 mM sodium citrate, pH 4.5) to induce diabetes. Mice were ad libitum fed D10001 diet or D10001 formulated with the compound of formula (I) at 0.25 mg/g, 0.5 mg/g and 1 mg/g of the compound of formula (I) (CAS # 1261289-04-6). Cohorts of BKS, db/db and F1 mice were housed in groups of 4-5 mice/cage.
  • mice with apparent health problems such as >10% reduction in body weight or fighting were excluded with no differences between groups.
  • mice were randomly allocated to the cages, based on weight and fasting blood glucose and allocated cage-wise, or if possible individually, to different treatments in order to minimize influence of starting weight and glucose homeostasis.
  • in vitro analyses such as western blot, qPCR, histology and immunohistology work, samples from 5-9 mice/diet were randomly selected. All in vivo analyses were performed between 9 am to 3 pm.
  • mice were sedated with ⁇ 2% isoflurane in oxygen (800 mL/min) and cannulated via the tail vein using a 27G needle and a tailor-made catheter.
  • FDG Fluorodeoxyglucose
  • PET/CT imaging started with a 50 kV, 0.088 mAs, helical CT acquisition reconstructed to images with 0.375x0.375x0.377mm 3 voxel size. PET images were reconstructed to a voxel size (0.4x0.4x0.4), with 4 iterations and 4 subsets using the Tera-Tomo 3D iterative reconstruction, with attenuation, scatter, and randoms-correction. Image analysis and Patlak pharmaco-kinetic calculation was performed with imlook4d software. During the scans, mice were supervised on a temperature-controlled bed, and blood glucose was measured at 10, 20, and 40 minutes after injection, using tail vein blood.
  • a region-of-interest (ROI) consisting of both the left and right gluteus maxims/rectus femoris muscles was defined in coronal views as 10-pixel-diameter and 10-slice cylinders.
  • the myocardial ROI was defined by thresholding a manually delineated search volume in the last PET frame at 40% of the highest voxel.
  • the vena cava ROI was defined as the voxels above 60% of the maximum voxel in the first frame with uptake.
  • An image-derived input function was approximated using the time activity curve from the vena cava ROI. Patlak analysis was performed employing above image-derived input function, determining the irreversible uptake rate K i by linear regression between 16-40 minutes.
  • Patlak K i values were calculated on ROI level for quantification, and on voxel level for illustrations.
  • Echocardiography For echocardiography, mice were sedated using 1.5-2% isoflurane, in 0.8L ⁇ min -1 O 2 (g), and placed on a temperature-controlled table. Chest hair was removed using hair removal cream. Respiration and ECG were monitored during the scan, and anaesthesia adjusted to avoid depression of respiration. Total scan time did not exceed 15 minutes.
  • Stroke volume, cardiac output and wall thicknesses were measured in the parasternal long-axis view using Bmode and M-mode images. Diastolic left ventricle inflow used transmitral doppler in the apical four-chamber view. Off-line analysis was done in a blinded manner. [0117] Mitochondrial respiration analyses. C2C12 myoblasts were in growth media, 10% fetal bovine serum and 20U/ml Penicillin-Streptomycin.
  • C2C12 myoblasts were seeded in poly-L-Lysin coated XF96 plates at 7500 cells/cm 2 , cultivated in growth media for 3 days to 80% confluence, thereafter switched to differentiation media (DM; growth media with FBS replaced by 2% horse serum for 6 days with media changes every 2 days.
  • Respiration assays were performed using a Seahorse XFe96 Extracellular flux analyzer according to the manufacturer’s “Mito-stress” protocol and mitochondrial parameters. Briefly, differentiated myotubes were pre-treated by switching to growth media with 1% horse serum together with the compound of formula (I) (sodium salt formulation) for 4h.
  • Myotubes were then equilibrated for 1h in Seahorse assay medium (1mM Na-Pyruvate, 10mM Glucose, 2mM L-Glutamine) adjusted to pH7.4 and supplemented with the compound of formula (I) as during pretreatment.
  • Measurements of oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) was collected for baseline, followed by sequential DGGLWLRQ ⁇ RI ⁇ 0 ⁇ ROLJRP ⁇ FLQ ⁇ 0 ⁇ )&&3 ⁇ DQG ⁇ 0 ⁇ URWHQRQH ⁇ 0 ⁇ DQWLP ⁇ FLQ ⁇ $ ⁇ Mitochondrial function parameters were calculated according to the manufacturer’s recommendations. [0118] Western Blot analysis.
  • Vastus muscles were isolated from non-fasted mice and crushed in a mortar using a pestle and liquid nitrogen.
  • Vastus muscles and INS-1E cell samples were homogenized in ice cold protein lysis buffer (100mM Tris pH 6.8, 2% SDS), with protease inhibitor cocktail and phosphatase inhibitor cocktail and sonicated. The vastus supernatant was collected after 10 min at 14,000 rpm. Protein concentration was measured using the BCA kit after which samples were diluted in Laemmli buffer and denaturized.
  • RNA from isolated islets was prepared using RNeasy Micro Kit.
  • Total RNA from gastrocnemius muscles and left cardiac ventricles was prepared using RNeasy Fibrous Tissue Mini kit after first crushing the tissue in liquid nitrogen using a pestle.
  • First strand cDNA synthesis was done using SuperScript III according to the manufacturer’s instructions.
  • Primers used for qRT-PCR are listed in Table 3.
  • Expression of Tbp was used for normalization of samples from islets, gastrocnemius muscle and left cardiac ventricle. Normalization by Tbp was validated by Rpl32 in left cardiac ventricle samples and gastrocnemius muscles.
  • Table 3 [0120] Islet isolation and culture.
  • Mouse islets were isolated essentially and cultured for 48h in media (RPMI 1640 medium, 1% fetal bovine serum, 10 mM HEPES, 1 mM sodium S ⁇ UXYDWH ⁇ 0 ⁇ -mercaptoethanol, 50U/ml Pen:Strep) supplemented with 11mM glucose or 22mM glucose +/- ⁇ 0 ⁇ RI ⁇ WKH ⁇ FRPSRXnd of formula (I).
  • Human islets from nondiabetic donors were cultured for 48 h in media (CMRL medium, 10 % fetal bovine serum, 20 U/ml Pen:Strep and 1X GlutaMax) supplemented with 5.5mM glucose or 25mM glucose +/- ⁇ 0 ⁇ of the compound of formula (I).
  • RNA-seq was performed. RNA-seq libraries were prepared from 150ng total RNA using the Illumina TruSeq stranded mRNA Library Kit followed by 100 bp paired- end sequencing on a NovaSeq 6000 Illumina Sequencer. RNA-seq reads were processed. Fastq files with 100-nt paired-end sequenced reads were quality-checked, aligned to the mouse or human genome (GRCm39 or GRCh38. Fragment - gene hits were counted. Subsequent analysis was performed in R using packages DESeq2 and clusterProfiler.
  • INS-1E cells were washed and equilibrated for 1h in UB buffer, transferred to UB buffer containing either 2 or 20mM glucose for 30min, and insulin levels in the supernatant was determined by ELISA. Insulin secretion was normalized to protein content.
  • INS-1E and C2C12 were commercially available and therefore not authenticated following purchase. The cells were free of mycoplasma as determined by PCR.
  • the experimental design was considered to consist of a non- diabetic control group (BKS or Control (i.e. vehicle injected F1 mice), a diabetic control (db/db mice or STZ mice, i.e. STZ injected F1 mice) and compound-of-formula-(I)-treated group(s) (db/db mice or STZ mice kept on a diet formulated with the compound of formula (I)).
  • BKS or Control i.e. vehicle injected F1 mice
  • db/db mice or STZ mice i.e. STZ injected F1 mice
  • compound-of-formula-(I)-treated group(s) db/db mice or STZ mice kept on a diet formulated with the compound of formula (I)
  • the validity of the diabetic models was tested by comparing the control and diabetic groups.
  • the effect of the compound-of-formula-(I) diet was evaluated by comparing the compound-of-formula-(I)-treated group
  • Pancreatic insulin content was 10-20-fold lower in mice treated with the compound of formula (I) compared with non-diabetic control mice (FIGS. 1(c), 8(b)).
  • islet cell area and Ins+ cell fraction were decreased whereas Glu+ islet cell fraction was increased (FIGS. 1(d)-1(f), 8(a), 8(c), 8(d)).
  • the compound of formula (I) rapidly reduced glucose levels in STZ mice also when treatment was initiated first at day 15, i.e. when the mice had developed overt diabetes, without enhancing plasma insulin levels (FIGS. 1(g), 1(h), 8(e)).
  • Pancreatic insulin content was again lower, ⁇ 20-fold, in mice treated with the compound of formula (I) compared with that of control mice (FIGS. 1(i), 8(g)). Similarly, islet cell area and Ins+ islet cell fraction was reduced and Glu+ islet cell fraction increased in untreated mice and STZ mice treated with the compound of formula (I) (FIGS. 1(j)-1(l), 8(f), 8(h), 8(i)). Together these findings show that the compound of formula (I) potently and in an insulin independent manner reverts diabetes in an insulin deficient mouse model of diabetes.
  • Example 2 The compound of formula (I) ameliorates hyperglycemia in STZ mice by stimulating muscle glucose uptake.
  • the compound of formula (I) promoted gene expression changes favoring glucose uptake, oxidative glucose metabolism, and ATP generation in skeletal muscle and heart of STZ mice (FIGS. 2(e), 2(f)).
  • Hyperglycemia has been shown to rapidly increase cardiac expression of Pdk4 and Ucp3, and to provoke metabolic inflexibility and cardiac dysfunction in mice [34].
  • Untreated STZ mice showed gradually decreased peak E velocity and increased isovolumetric relaxation time (IVRT), indicating impaired diastolic function, whereas 1 week treatment with the compound of formula (I) reduced IVRT and increased peak E velocity and thus E/A ratio (FIG. 2(g), Table 4).
  • the improved filling of the left ventricle in mice treated with the compound of formula (I) also resulted in increased stroke volume and cardiac output (Table 4).
  • Table 4 shows echocardiographic measurements of left ventricular dimensions in para-sternal long axis (PLAX) B-mode or M-mode.
  • HR heart rate
  • SV stroke volume
  • CO cardiac output
  • EDV end-diastolic volume
  • ESV end-systolic volume
  • AWd/s anterior wall thickness in diastole/systole
  • LVIDd anterior wall thickness in diastole/systole
  • PWd/s posterior wall thickness in diastole/systole
  • EF ejection fraction
  • FS fractional shortening
  • E/A ratio of E and A peak wave velocity
  • Decel time deceleration time of E wave from peak to projected baseline
  • IVRT isovolumetric relaxation time.
  • db/db mice were treated with a diet formulated with the compound of formula (I) at a concentration of 0.5 or 1.0 mg/g of the compound of formula (I), denoted Cmpd-(I)-(0.5) and Cmpd-(I)-(1.0), for 9w.
  • BKS mice were used as controls.
  • compensatory hyperinsulinemia was evident in db/db mice (FIGS. 3(a), 3(b)).
  • Untreated db/db mice failed to compensate for the ensuing insulin resistance and blood glucose levels rapidly increased as a result of declining insulin levels (FIGS.
  • Txnip, Pdk4 and Ucp3 together with the increased expression of Slc2a4, Peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1alpha (Ppargc1a), a positive regulator of mitochondrial biogenesis and respiration, and Cox8b, a driver of oxidative phosphorylation, in skeletal muscle of db/db mice treated with the compound of formula (I) compared with untreated db/db mice supports this notion (FIG. 3(f)).
  • PPC Peroxisome proliferator-activated receptor-gamma coactivator
  • Example 5 The compound of formula (I) stimulates mitochondrial uncoupling in myotubes.
  • the increase in glucose utilization and reduced glycogen content observed in skeletal muscle and heart of STZ and db/db mice treated with the compound of formula (I) suggests that the compound of formula (I) increases energy expenditure by generating a metabolic demand either via futile cycling and/or mitochondrial uncoupling.
  • Studies of mitochondrial and glycolytic function in intact, differentiated C2C12 myotubes were performed to elucidate a potential uncoupling potential for the compound of formula (I).
  • the compound of formula (I) dose-dependently increased oxygen consumption rate (OCR), a measure of oxidative phosphorylation, in C2C12 myotubes, and the ratio of basal OCR to basal ECAR (extracellular acidification rate) (FIGS. 4(a)-4(c)), indicating that the compound of formula (I) increased the cellular preference for oxidative metabolism.
  • OCR oxygen consumption rate
  • ECAR extracellular acidification rate
  • Example 6 The compound of formula (I) preserves beta-cell function and mass in db/db mice. [0131] Consistent with the described initial compensatory increase in beta-cell mass in db/db mice, islet cell area was increased at 6w of age in db/db compared with BKS mice (FIGS. 5(a), 5(b)).
  • pancreatic insulin and proinsulin content and the pancreatic insulin:proinsulin ratio a measure of the efficiency of proinsulin processing
  • pancreatic insulin and proinsulin content and the pancreatic insulin:proinsulin ratio a measure of the efficiency of proinsulin processing
  • Example 7 The compound of formula (I) alleviates islet gene expression changes provided by acute hyperglycemia.
  • DEGs differentially expressed genes
  • the compound of formula (I) reduced the number of DEGs to 143 upregulated and 208 downregulated (FIGS. 6(a), 13(a)-13(c)).
  • exposure of mouse islets to high glucose resulted in a significant increase in Txnip expression which was attenuated by the compound of formula (I) (FIG. 6(b)).
  • GSEA gene set enrichment analysis
  • RNA seq analysis identified mTORC1 signaling to be upregulated in islets exposed to high (22 mM) glucose, which was prevented by exposure to the compound of formula (I) (FIG. 6(c)).
  • INS-1E cells were cultured at 25 mM glucose for 4 days (d), which impaired GSIS whereas concomitant exposure to the compound of formula (I) during the entire 4 d culture period averted these negative effects (FIG. 7(a)).
  • Acetyl-CoA carboxylase (ACC) and Raptor showed that both long- and short-term exposure of INS1-E cells to the compound of formula (I) at high glucose reduced pS6-Ser240/244 levels but increased pACC-Ser79 and pRaptor-Ser792 levels (FIGS. 7(c), 7(d), 14). Together, these findings provide evidence that the compound of formula (I) preserves beta-cell function under hyperglycemic conditions by antagonizing mTORC1 and preserving AMPK signaling.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des procédés de traitement, de prévention ou de retardement de l'apparition de maladies cardiaques telles que la cardiomyopathie et l'insuffisance cardiaque par administration d'un composé de traitement à un sujet qui en a besoin. La présente divulgation se rapporte en outre de manière générale à des procédés d'amélioration de l'efficacité cardiaque à l'aide du composé de traitement à un sujet qui en a besoin. La présente divulgation se rapporte également de manière générale à des procédés d'inhibition de l'activité d'une pyruvate déshydrogénase kinase (PDK) à l'aide du composé de traitement pour un sujet qui en a besoin. Le composé de traitement est un composé de formule (I), ou un sel, un solvate ou un promédicament pharmaceutiquement acceptable de celui-ci.
PCT/EP2024/070602 2023-07-20 2024-07-19 Procédés de traitement de la cardiomyopathie induite par une inflexibilité métabolique Pending WO2025017193A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363514700P 2023-07-20 2023-07-20
US63/514,700 2023-07-20

Publications (1)

Publication Number Publication Date
WO2025017193A1 true WO2025017193A1 (fr) 2025-01-23

Family

ID=92106899

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2024/070602 Pending WO2025017193A1 (fr) 2023-07-20 2024-07-19 Procédés de traitement de la cardiomyopathie induite par une inflexibilité métabolique

Country Status (1)

Country Link
WO (1) WO2025017193A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011004162A2 (fr) 2009-07-08 2011-01-13 Betagenon Ab Composés utilisés en tant que médicaments
WO2021074646A1 (fr) * 2019-10-18 2021-04-22 Balticgruppen Bio Ab Nouvelles préparations

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011004162A2 (fr) 2009-07-08 2011-01-13 Betagenon Ab Composés utilisés en tant que médicaments
WO2021074646A1 (fr) * 2019-10-18 2021-04-22 Balticgruppen Bio Ab Nouvelles préparations

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
"Remington The Science and Practice of Pharmacy", vol. 249, 1995, MACK PRINTING COMPANY, article "A brief review of methods of drug delivery may also be found in", pages: 1527
ERICSSON MADELENE ET AL: "AMPK activator O304 improves metabolic and cardiac function, and exercise capacity in aged mice", COMMUNICATIONS BIOLOGY, vol. 4, no. 1, 18 November 2021 (2021-11-18), XP093211016, ISSN: 2399-3642, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8602430/pdf/42003_2021_Article_2837.pdf> [retrieved on 20241002], DOI: 10.1038/s42003-021-02837-0 *
HAWLEY ET AL., DIABETES, vol. 65, 2016, pages 2784 - 2794
NORLIN STEFAN ET AL: "O304 ameliorates hyperglycemia in mice by dually promoting muscle glucose effectiveness and preserving [beta]-cell function", COMMUNICATIONS BIOLOGY, vol. 6, no. 1, 25 August 2023 (2023-08-25), XP093208598, ISSN: 2399-3642, Retrieved from the Internet <URL:https://www.nature.com/articles/s42003-023-05255-6> [retrieved on 20241002], DOI: 10.1038/s42003-023-05255-6 *
PÄR STENEBERG ET AL: "PAN-AMPK activator O304 improves glucose homeostasis and microvascular perfusion in mice and type 2 diabetes patients", JCI INSIGHT, vol. 3, no. 12, 21 June 2018 (2018-06-21), XP055594531, [retrieved on 20241002], DOI: 10.1172/jci.insight.99114 *
VILLANI ET AL., MOLECULAR METABOLISM, vol. 5, 2016, pages 1048 - 1056
WANG HONGCHAO ET AL: "Neuregulin-4 attenuates diabetic cardiomyopathy by regulating autophagy via the AMPK/mTOR signalling pathway", CARDIOVASC DIABETOLOGY, vol. 21, no. 1, 11 October 2022 (2022-10-11), GB, XP093210904, ISSN: 1475-2840, Retrieved from the Internet <URL:https://cardiab.biomedcentral.com//cardiab.biomedcentral.com/counter/pdf/10.1186/s12933-022-01643-0.pdf> [retrieved on 20241002], DOI: 10.1186/s12933-022-01643-0 *
YANG FAN ET AL: "Metformin Inhibits the NLRP3 Inflammasome via AMPK/mTOR-dependent Effects in Diabetic Cardiomyopathy", INTERNATIONAL JOURNAL OF BIOLOGICAL SCIENCES, vol. 15, no. 5, 10 March 2019 (2019-03-10), pages 1010 - 1019, XP093211079, ISSN: 1449-2288, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6535781/pdf/ijbsv15p1010.pdf> [retrieved on 20241002], DOI: 10.7150/ijbs.29680 *

Similar Documents

Publication Publication Date Title
JP2022020624A (ja) 心房拡大又はリモデリングを特徴とする疾患を治療するためのnep阻害剤
JP4938905B2 (ja) 選択的s1p1レセプターアゴニストの投与法
RS59493B2 (sr) Kombinacioni tretman raka
US20170348263A1 (en) Compositions and methods of using modified release solabegron for lower urinary tract symptoms
EP2349280B1 (fr) Procédés de traitement de l hyperuricémie et d états pathologiques associés
JP2017537165A (ja) 下部尿路系症状を対象とした徐放性ソラベグロンの組成物及び使用方法
JP2016535101A (ja) 心臓リモデリング及び他の心疾患の治療
JP2023541766A (ja) 慢性腎疾患の治療のためのジボテンタン及びダパグリフロジンの組み合わせ
JP6180417B2 (ja) 心不全またはニューロン損傷の治療剤製造のための化合物の使用
RS60240B1 (sr) Postupci i kompozicije za lečenje depresije korišćenjem ciklobenzaprina
EP1809289A1 (fr) Utilisation de ranolazine en combinaison avec au moins un agent de remodelage pour inverser un remodelage du ventricule gauche dans le traitement de l&#39;insuffisance cardiaque
WO2025017193A1 (fr) Procédés de traitement de la cardiomyopathie induite par une inflexibilité métabolique
JP2018135278A (ja) 循環器疾患及び/又はミトコンドリア病の改善用医薬
WO2025017194A1 (fr) Procédés de régulation des taux de glycémie à l&#39;aide d&#39;un activateur à double ampk et d&#39;un découpleur mitochondrial
US20130143848A1 (en) Use of megestrol acetate for improving heart function and the treatment of heart insufficiency
CN116570580A (zh) 丙酮酸脱氢酶激酶抑制剂在治疗射血分数保留型心力衰竭中的应用
WO2019067771A1 (fr) Compositions et procédés pour traiter la cardiomyopathie septique
WO2019004465A1 (fr) Composition pharmaceutique contenant du pémafibrate
WO2025027205A1 (fr) Traitement du cancer avec l&#39;inhibiteur 4-chloro-n-(2-(4-chlorobenzyl)-3-oxo-2,3-dihydro-1,2,4-thiadiazol-5-yl)benzamide de pdk
JP7390698B2 (ja) 若年性パジェット病治療薬
EP4058010A1 (fr) Utilisation d&#39;un stabilisant de mastocytes pour le traitement d&#39;une insuffisance cardiaque avec fraction d&#39;éjection préservée
EP2604258A1 (fr) Composition pharmaceutique comprenant du losartan pour le traitement ou la prévention de la toxicité musculaire induite par des médicaments à base de statine
TWI900606B (zh) 用於治療內皮素相關疾病之齊泊騰坦和達格列淨的組合
RU2453313C1 (ru) Фармацевтическая композиция, препятствующая развитию синдрома предвозбуждения
TW202313072A (zh) 檸檬酸鐵之兒科調配物

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: 24748308

Country of ref document: EP

Kind code of ref document: A1