WO2007024717A2 - Agents for preventing and treating disorders involving modulation of the ryr receptors - Google Patents

Agents for preventing and treating disorders involving modulation of the ryr receptors Download PDF

Info

Publication number
WO2007024717A2
WO2007024717A2 PCT/US2006/032405 US2006032405W WO2007024717A2 WO 2007024717 A2 WO2007024717 A2 WO 2007024717A2 US 2006032405 W US2006032405 W US 2006032405W WO 2007024717 A2 WO2007024717 A2 WO 2007024717A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
aryl
cycloalkyl
heterocyclyl
group
Prior art date
Application number
PCT/US2006/032405
Other languages
French (fr)
Other versions
WO2007024717A3 (en
Inventor
Andrew Robert Marks
Donald W. Landry
Shixian Deng
Zhen Zhuang Cheng
Stephan E. Lehnart
Original Assignee
The Trustees Of Columbia University In The City Of New York
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
Priority to AU2006283534A priority Critical patent/AU2006283534C1/en
Priority to JP2008528020A priority patent/JP5342877B2/en
Application filed by The Trustees Of Columbia University In The City Of New York filed Critical The Trustees Of Columbia University In The City Of New York
Priority to BRPI0615097-7A priority patent/BRPI0615097B1/en
Priority to PL06801887T priority patent/PL1928850T3/en
Priority to KR1020127003439A priority patent/KR101456342B1/en
Priority to ES06801887T priority patent/ES2421159T3/en
Priority to CA2620183A priority patent/CA2620183C/en
Priority to RS20130294A priority patent/RS52852B/en
Priority to CN2006800395739A priority patent/CN101500576B/en
Priority to EA200800665A priority patent/EA014941B1/en
Priority to NZ566822A priority patent/NZ566822A/en
Priority to EP06801887.8A priority patent/EP1928850B1/en
Priority to DK06801887.8T priority patent/DK1928850T3/en
Priority to SI200631612T priority patent/SI1928850T1/en
Publication of WO2007024717A2 publication Critical patent/WO2007024717A2/en
Priority to PCT/US2007/009715 priority patent/WO2007127145A2/en
Priority to TW096119821A priority patent/TW200815381A/en
Priority to PCT/US2007/012936 priority patent/WO2008060332A2/en
Priority to ARP070102392A priority patent/AR061174A1/en
Priority to PCT/US2007/012969 priority patent/WO2007143112A2/en
Priority to ARP070102391A priority patent/AR061173A1/en
Priority to PCT/US2007/018147 priority patent/WO2008021439A2/en
Priority to PCT/US2007/018138 priority patent/WO2008021432A2/en
Priority to IL189675A priority patent/IL189675A/en
Priority to TNP2008000078A priority patent/TNSN08078A1/en
Priority to NO20081421A priority patent/NO20081421L/en
Priority to HK08113342.0A priority patent/HK1119705A1/en
Publication of WO2007024717A3 publication Critical patent/WO2007024717A3/en
Priority to US12/938,098 priority patent/US8710045B2/en
Priority to HRP20130669TT priority patent/HRP20130669T1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D281/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D281/02Seven-membered rings
    • C07D281/04Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D281/08Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D281/10Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D291/00Heterocyclic compounds containing rings having nitrogen, oxygen and sulfur atoms as the only ring hetero atoms
    • C07D291/08Heterocyclic compounds containing rings having nitrogen, oxygen and sulfur atoms as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/554Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one sulfur as ring hetero atoms, e.g. clothiapine, diltiazem
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/12Antidiuretics, e.g. drugs for diabetes insipidus
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • This invention relates to compounds and their use to treat and prevent disorders and diseases associated with the RyR receptors that regulate calcium channel functioning in cells. More particularly, the invention discloses compounds that are related to 1,4-benzothiazepines and are useful to treat cardiac and skeletal muscular disorders. The invention also discloses pharmaceutical compositions comprising the compounds and articles of manufacture comprising the pharmaceutical compositions.
  • the sarcoplasmic reticulum is a structure in cells that functions, among other things, as a specialized intracellular calcium (Ca 2+ ) store. Channels in the SR called ryanodine receptors (RyRs) open and close to regulate the release of Ca 2+ from the SR into the intracellular cytoplasm of the cell. Release of Ca 2+ into the cytoplasm from the SR increases cytoplasmic Ca 2+ concentration.
  • Open probability (Po) of the RyR receptor refers to the likelihood that the RyR channel is open at any given moment, and therefore capable of releasing Ca 2+ into the cytoplasm from the SR.
  • RyRl is found predominantly in skeletal muscle as well as other tissues
  • RyR2 is found predominantly in the heart as well as other tissues
  • RyR3 is found in the brain as well as other tissues.
  • the RyR channels are formed by four RyR polypeptides in association with four FK506 binding proteins (FKBPs), specifically FKBP 12 (calstabinl) and FKBP12.6 (calstabin2).
  • FKBPs FK506 binding proteins
  • Calstabinl binds to RyRl
  • calstabin2 binds to RyR2
  • calstabinl binds to RyR3.
  • the FKBP proteins (calstabinl and calstabin2) bind to the RyR channel (one molecule per RyR subunit), stabilize RyR-channel functioning, and facilitate coupled gating between neighboring RyR channels, thereby preventing abnormal activation of the channel during the channel's closed state.
  • protein kinase A (PKA) also binds to the cytoplasmic surface of the RyR receptors. PICA phosphorylation of the RyR receptors causes partial dissociation of calstabins from RyRs. Dissociation of calstabin from RyR causes increased open probability of RyR 3 and therefore increased Ca 2+ release from the SR into the intracellular cytoplasm.
  • Ca 2+ release from the SR in skeletal muscle cells and heart cells is a key physiological mechanism that controls muscle performance, because increased concentration of Ca 2+ in the intracellular cytoplasm causes contraction of the muscle.
  • Excitation-contraction (EC) coupling in skeletal muscles involves electrical depolarization of the plasma membrane in the transverse tubule (T-tubule), which activates voltage-gated L-type Ca 2+ channels (LTCCs).
  • LTCCs trigger Ca 2+ release from the SR through physical interaction with RyRl .
  • the resulting increase in cytoplasmic Ca concentration induces actin-myosin interaction and muscle contraction.
  • intracellular Ca 2+ is pumped back into the SR via SR Ca 2+ -ATPase pumps (SERCAs), which is regulated by phospholamban (PLB) depending on the muscle fiber type.
  • SERCAs SR Ca 2+ -ATPase pumps
  • RyR2 is the major Ca 2+ - release channel required for EC coupling and muscle contraction.
  • depolarization of the cardiac-muscle cell membrane during phase zero of the action potential activates voltage-gated Ca 2+ channels.
  • Ca 2+ influx through the open voltage-gated channels in turn initiates Ca 2+ release from the SR via RyR2.
  • This process is known as Ca 2+ -induced Ca 2+ release.
  • the RyR2-mediated, Ca 2+ -induced Ca 2+ release then activates the contractile proteins in the cardiac cell, resulting in cardiac muscle contraction.
  • [Oil] Phosphorylation of cardiac RyR2 by PKA is an important part of the "fight or flight” response that increases cardiac EC coupling gain by augmenting the amount of Ca 2+ released for a given trigger.
  • This signaling pathway provides a mechanism by which activation of the sympathetic nervous system, in response to stress, results in increased cardiac output.
  • PKA phosphorylation of RyR2 increases the open probability of the channel by dissociating calstabin2 (FKBP 12.6) from the channel complex. This, in turn, increases the sensitivity of RyR2 to Ca 2+ -dependent activation.
  • heart failure remains an important cause of mortality in Western countries.
  • An important hallmark of heart failure is reduced myocardial contractility.
  • contractile abnormalities result, in part, from alterations in the signaling pathway that allows the cardiac action potential to trigger Ca 2+ release via RyR2 channels and muscle contraction.
  • the amplitude of the whole- cell Ca 2+ transient is decreased and the duration prolonged.
  • Atrial fibrillation is the most common cardiac arrhythmia in humans, and represents a major cause of morbidity and mortality.
  • Structural and electrical remodeling including shortening of atrial refractoriness, loss of rate-related adaptation of refractoriness, and shortening of the wavelength of re-entrant wavelets - accompany sustained tachycardia. This remodeling is likely important in the development, maintenance and progression of atrial fibrillation. Studies suggest that calcium handling plays a role in electrical remodeling in atrial fibrillation.
  • CPVT Catecholarninergic polymorphic ventricular tachycardia
  • IQ subjects with CPVT physical exertion and/or stress induce bidirectional and/or polymorphic ventricular tachycardias that lead to SCD even in the absence of detectable structural heart disease.
  • CPVT is predominantly inherited in an autosomal-dominant fashion. Individuals with CPVT have ventricular arrhythmias when subjected to exercise, but do not develop arrhythmias at rest. Studies have identified mutations in the human RyR2 gene, on chromosome Iq42-q43, in individuals with CPVT.
  • Failing hearts are characterized by a maladaptive response that includes chronic hyperadrenergic stimulation, hi heart failure, chronic beta-adrenergic stimulation is associated with the activation of beta-adrenergic receptors in the heart, which, through coupling with G-proteins, activate adenylyl cyclase and thereby increase intracellular cAMP concentration.
  • CAMP activates cAMP-dependent PKA, which has been shown to induce hyperphosphorylation of RyR2.
  • chronic heart failure is a chronic hyperadrenergic state that results in several pathologic consequences, including PKA hyperphosphorylation of RyR2.
  • a subpopulation of RyR2 that are particularly "leaky” can release SR Ca 2+ during the resting phase of the cardiac cycle, diastole. This results in depolarizations of the cardiomyocyte membrane known as delayed after-depolarizations (DADs), which are known to trigger fatal ventricular cardiac arrhythmias.
  • DADs delayed after-depolarizations
  • Cardiac arrhythmias are known to be associated with diastolic SR Ca 2+ leaks in patients with CPVT mutations in their RyR2 and otherwise structurally-normal hearts. In these cases, the most common mechanism for induction and maintenance of ventricular tachycardia is abnormal automaticity.
  • One form of abnormal automaticity known as triggered arrhythmia, is associated with aberrant release of SR Ca 2+ , which initiates DADs.
  • DADs are abnormal depolarizations in cardiomyocytes that occur after repolarization of a cardiac action potential.
  • the molecular basis for the abnormal SR Ca 2+ release that results in DADs has not been fully elucidated. However, DADs are known to be blocked by ryanodine, providing evidence that RyR2 plays a key role in the pathogenesis of this aberrant Ca 2+ release.
  • U.S. Patent No. 6,489,125 discusses JTV-519 (4-[3-(4-benzylpiperidin-l- yl)propionyl] -7-methoxy-2,3 ,4,5-tetrahydro- 1 ,4-benzothiazepine monohydrochloride; also known as k201 or ICP-Calstan 100), a 1,4-benzothiazepine, as a new modulator of RyR calcium-ion channels.
  • co-pending U.S. Patent Application No. 09/288,606 discusses a method for regulating contraction of a subject's heart by administering a compound that regulates PKA phosphorylation of an RyR2 receptor and specifically decreases PKA phosphorylation.
  • Co-pending U.S. Patent Application No. 10/608,723 also discusses a method for treating and prophylaxis for atrial tachyarrhythmia and exercise and stress-induced arrhythmias by administration of an agent which inhibits PKA phosphorylation of RyR2.
  • the present invention generally provides compounds that may be classified as 1,4-benzothiazepines and sometimes are referred to herein as "RyCaIs.”
  • R 4 is selected from the group consisting of H, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
  • R 6 is selected from the group consisting Of-ORi 5 , -NHNRi 5 Ri 6 , -NHOH, -NRi 5 Ri 6 , -CH 2 X, acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
  • R 7 is selected from the group consisting of -ORi 5 , -NR) 5 Ri 6 , -NHNRi 5 Ri 6 , -NHOH, -CH 2 X, alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
  • R 8 and Rg independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and
  • Rn, Ri 2 , R 13 , and R 14 independently are selected from the group consisting of H, OH, NH 2 , - NHNH 2 , -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
  • R 15 and Ri 6 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH 2 , alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted; and optionally R15 and Ri 6 together with the N to which they are bonded may form a heterocycle which may be substituted; the nitrogen in the benzothiazepine ring may optionally be a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solv
  • the present invention provides compounds of Formula I, as described above, with the proviso that the compound is not S24 or S68.
  • the present invention provides compounds of Formula I-a:
  • R 6 is selected from the group consisting of -ORi 5 , -NHNR] 5 R 16 , -NHOH, -NRi 5 R 16 , -CH 2 X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted;
  • R 7 is selected from the group consisting of H, -OR 15 , -NRi 5 R 16 , -NHNRi 5 R 16 , -NHOH, - CH 2 X, alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
  • R 8 and Rg independently are selected from the group consisting of -OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
  • m is O, 1, 2, 3, or 4;
  • Rn, R 12 , Ri 3 , and R 14 independently are selected from the group consisting of H, OH, NH 2 , - NHNH 2 , -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
  • R 15 and R 16 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH 2 , alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; and optionally Ri 5 and Rj 6 together with the N to which they are bonded may form a heterocycle which may be substituted or unsubstituted; the nitrogen in the benzothiazepine ring may be optionally a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes
  • R 2 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds formula of I-c:
  • each R, R 7 , q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention provides compounds of formula I-c, wherein R 7 is selected from the group consisting of -OH, -NR 15 Ri 6 , alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
  • the present invention provides compounds of formula of I- d:
  • R 7 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-d, wherein R 7 is selected from the group consisting of -OH, -NRisRie, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted.
  • the present invention provides compounds of formula of I-e:
  • each R, R 5 , q and n is as defined compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention provides compounds of formula I-e, wherein R 5 is selected from the group consisting of-NRisRie, -NHOH, -OR 15 , -CH 2 X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
  • the present invention provides compounds of formula I-e, wherein R 5 is an alkyl substituted by at least one labeling group, such as a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group.
  • a fluorescent labeling group can be selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade BlueTM, Pacific Blue, Marina Blue, Oregon Green, 4 ! ,6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
  • DAPI 6-Diamidino-2-phenylindole
  • the present invention provides compounds of formula of I-f:
  • R 5 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-f, wherein R 5 is selected from the group consisting Of-NR 15 R 16 , -NHOH, -ORi 5 , -CH 2 X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
  • the present invention provides compounds of formula of I-g: wherein W is S or O; each R, R 15 , R 16 , q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention provides compounds of formula I-g, wherein R 15 and R 16 independently are selected from the group consisting of H, OH, NH 2 , alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted; and optionally R 15 and R 16 together with the N to which they are bonded may form a heterocycle which may be substituted.
  • the present invention provides compounds of formula I-g, wherein W is O or S.
  • R 1S , R 16 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-h, wherein R 15 and R 16 independently are selected from the group consisting of H, OH, NH 2 , alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl 'may be substituted; and optionally R15 and Ri 6 together with the N to which they are bonded may form a heterocycle which may be substituted.
  • the present invention provides compounds of formula I-g, wherein W is O or S. [058] In a further embodiment, the present invention provides compounds of formula of I-i:
  • Rn is selected from the group consisting of-NRisRi ⁇ , -NHNRi S R 16 , -NHOH, -OR 15 , -CH 2 X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; each R, q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention provides compounds of formula I-i, wherein R 17 is -NR 15 R 16 , and -OR 15 .
  • R 17 is -OH, -OMe, -NEt, -NHEt, -NHPh, -NH 2 , or -NHCH 2 pyridyl.
  • R 17 is selected from the group consisting Of-NR 15 R 16 , -NHNRi 5 R 16 , -NHOH, -ORi 5 , -CH 2 X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; n is as defined in compounds of formula I-a; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-j, wherein Ri 7 is -NR] 5 Ri 6 or -ORj 5 .
  • Rn is -OH, -OMe, -NEt, - NHEt, -NHPh, -NH 2 , or -NHCH 2 pyridyl.
  • R' is H or OMe
  • R" is H.
  • m is O
  • Rig is CpC 4 alkyl, such as Me, Et, propyl, and butyl.
  • m is 2, and Ris is pyrrolidine, piperidine, piperazine, or morpholine.
  • m is 3, 4, 5, 5, 7, or 8
  • R 18 is a fluorescent labeling group selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade BlueTM, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
  • the present invention provides compounds of formula of 1-1:
  • R 6 and n are as defined in compounds of formula I-a; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula 1-1, wherein R 6 is selected from the group consisting Of-NR 15 R 16 , -NHNR 15 Ri ⁇ , -ORi 5 , -NHOH, - CH 2 X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
  • R 6 is -NRi 5 R 16 such as -NHPh, pyrrolidine, piperidine, piperazine, morpholine, and the like. In some other cases, R 6 is alkoxyl, such as -O-tBu.
  • the present invention provides compounds of formula I-m:
  • R 8 , R 9 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-m, wherein Rg and R9 are independently alkyl, aryl, -OH, alkoxyl, or alkylamino.
  • Rg and R9 are independently alkyl, aryl, -OH, alkoxyl, or alkylamino.
  • Rs is C 1 -C 4 alkyl such as Me, Et, propyl and butyl; and R 9 is aryl such as phenyl.
  • the compound is selected from the group consisting of Sl, S2, S3, S4, S5, S6, S7, S9, SIl, S12, S13, S14, S19, S20, S22, S23, S25, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, 848, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl
  • the compounds of the invention may optionally comprise a labeling group, such as a fluorescent, bioluminescent, chemiluminescent, colorimetric or radioactive labeling group.
  • a labeling group such as a fluorescent, bioluminescent, chemiluminescent, colorimetric or radioactive labeling group.
  • Suitable fluorescent labeling groups include, but are not limited to, bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade BlueTM, Pacific Blue, Marina Blue, Oregon Green, 4', 6-Diamidino-2- ⁇ henylindole (DAPI) , indopyra dyes, lucifer yellow, propidium iodide, porphyrins, and variants and derivatives thereof.
  • DAPI 6-Diamidino-2- ⁇ henylindole
  • indopyra dyes lucifer yellow
  • propidium iodide porphyrins, and
  • the present invention also provides methods for the synthesis of compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, and salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention further provides a method of treating or preventing various disorders and diseases in a subject that are associated with RyR receptors, such as muscular and cardiac disorders, comprising administering to the subject an amount of a compound effective to prevent or treat a disorder or disease associated with the RyR receptors, wherein the compound is of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or salts, hydrates, solvates, complexes and pro-drugs thereof.
  • a method of preventing or treating a leak in a RyR2 receptor in a subject including administering to the subject an amount of a compound effective to prevent or treat a leak in the RyR2 receptor, wherein the compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the subject is, for example, an in vitro system (e.g., cultured cells or tissues) or in vivo system (e.g., animal or human).
  • the present invention provides a method of modulating the binding of RyR and FKBP in a subject, including administering to the subject an amount of a compound effective to modulate the level of RyR-bound FKBP, wherein the compound is of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the subject is, for example, an in vitro system (e.g., cultured cells or tissues) or in vivo system ⁇ e.g., animal or human).
  • the present invention also provides articles of manufacture for treating and preventing disorders and diseases associated with the RyR receptors, such as muscular and cardiac disorders, in a subject.
  • the articles of manufacture comprise a pharmaceutical composition of one or more of the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the articles of manufacture are packaged with indications for various disorders that the pharmaceutical compositions are capable of treating and/or preventing.
  • Figure 1 embodiments A, B, C, and D are, respectively, (A) immunoblots of PKA phosphorylated RyR2 in the presence of FKBP12.6 and increasing JTV-519 concentrations; (B) immunoblots of PICA phosphorylated RyR2 in the presence of 0.5 nM S36; (C) a graph of current through plasma membrane, voltage dependent L-type Ca 2+ channels which are completely blocked by nifedipine but not by S36 in isolated mouse cardiomyocytes; and (D) a graph of the voltage-dependence of L-type Ca 2+ current in channels in the presence of JTV- 519 and S36.
  • FIG. 2 embodiments A, B, C, and D demonstrate the prevention of exercise- induced ventricular arrhythmias by JTV-519 in haploinsufficient calstabin (FKBP12.6) +/ ⁇ mice.
  • Embodiment A are representative telemetric electrocardiograms (ECGs) of an untreated calstabin2 (FKBP 12.6) +/ ⁇ mouse (left), a JTV-519-treated calstabin2 (FICBPl 2.6) +/ ⁇ mouse (middle), and a calstabin2 (FKBP12.6) ⁇ / ⁇ mouse (right).
  • ECGs telemetric electrocardiograms
  • Embodiment B are telemetry recordings of a sustained polymorphic ventricular tachycardia (sVT) in (upper) an untreated haploinsufficient calstabin2 (FKBP 12.6) +/ ⁇ mouse and (lower) a JTV-519-treated calstabin2 (FKBP12.6) +/ ⁇ mouse, each subjected to exercise testing immediately followed by injection with 0.5 mg epinephrine per kilogram of body weight.
  • Embodiment C are graphs showing the numbers of mice with cardiac death (left), sustained VTs (middle), and nonsustained VTs (right) in experimental groups of mice subjected to exercise testing and injection with 0.5 mg/kg epinephrine.
  • Embodiment D provides graphs comparing the dose dependence of pharmacological effects of JTV-519 and S36 in regard to sudden cardiac death (left), sustained VTs (middle), and nonsustained VTs (right).
  • Figure 3 is a graph showing fractional shortening (FS) of the left ventricle assessed by M-mode echocardiography 2 weeks post-myocardial infarction in placebo versus S36-treated mice. S36 treated mice show a significant improvement in FS in the 100 nM and 200 nM groups as compared to placebo.
  • Figure 4 is a graph showing heart weight to body weight (HW/BW) ratios and pressure-volume loops quantifications (dP/dt) one week post-myocardial infarction of placebo and S36-treated mice.
  • S36 treatment results in a beneficial reduction of the HW/BW ratio and increased velocity of pressure development in S36 as compared to placebo treated mice.
  • Figure 5 is a graph summarizing EC 50 values of JTV-519 and a series of Rycal compounds indicating several compounds with a higher biologic activity as evidenced by significantly lower EC 50 values compared to JTV-519.
  • Figure 6, embodiments A, B, and C are, respectively, (A) single-channel current traces of unphosphorylated RyR2-P2328S and unphosphorylated RyR2-WT treated with S36; (B) single-channel current traces of phosphorylated RyR2-P2328S and unphosphorylated RyR2-P2328S treated with S36; and (C) immunoblot analysis of calstabin-2 binding of RyR2-P2328S in the presence or absence of PKA and S36.
  • Figure 7, embodiments A and B are, respectively, (A) an immunoblot of RyR2 immunoprecipitated with an antibody against RyR2, and immunoblots of RyR2 PKA phosphorylation at Ser-2809 and calstabin2; and (B) a bar graph quantifying the relative amount of PKA phosphorylated RyR2 at Ser-2808 (corresponding to human Ser-2809) bound to RyR2 in wild-type (contol) and calstabin2-def ⁇ cient (FKBPl 2.6 '1' ) mice.
  • Figure 8 embodiments A, B, and C are, respectively, bar graphs of (A) quantitative in vivo M-mode echocardiograms comparing ejection fraction (EF) before and following sham operation or permanent left anterior descending (LAD) coronary artery ligation in wild- type and RyR2-S2808A knockin mice; (B) in vivo pressure-volume loop quantification of maximal pressure change over time (dP/dt) in wild-type and RyR2-S2808A knockin mice after sham operation or permanent left anterior descending coronary artery (LAD) ligation; and (C) quantitative M-mode echocardiographic assessment of end-systolic diameter (ESD) in wildtype and RyR2-S2808A knockin mice after sham operation or permanent left anterior descending coronary artery (LAD) ligation.
  • EF ejection fraction
  • LAD permanent left anterior descending coronary artery
  • FIG. 9 embodiments A, B, C, and D demonstrate the effect of JTV-519 on calstabii ⁇ affinity to RyR2 in haploinsufficient calstabin2 (FKBPl 2.6) +/ ⁇ mice after exercise.
  • Embodiment A is immunoblots of equivalent amounts of RyR2 immunoprecipitated with an antibody against RyR2 (top).
  • Embodiment B are bar graphs showing the amount of PKA phosphorylation of RyR2 at Ser-2809 and the amount of calstabin2 bound to RyR2 from control animals and animals after exercise immediately followed by injection with 0.5 mg/kg epinephrine.
  • Embodiment C are single-channel tracings of RyR2 channels isolated from hearts of haploinsufficient calstabin2 +/ ⁇ and calstabin2 ⁇ ⁇ deficient mice immediately following exercise testing and injection of 0.5 mg epinephrine per kg of body weight, both untreated (top) and after treatment (middle and bottom) with JTV-519.
  • Average open probality (Po), open time (To) and average closed time (Tc) are as indicated; and closed state is indicated by 'c'.
  • the dotted lines indicate subconductance levels for partial RyR2 openings.
  • Embodiment D is a bar graph summarizing average open probabilities of single RyR2 channels from haploinsufficient calstabin2 +/ ⁇ and calstabin2 "/ ⁇ deficient mice after exercise with and without JTV-519 treatment. "*" indicates significance level P ⁇ 0.05. Numbers in the bars indicate the number of single channels measured.
  • FIG. 10 3 embodiments A, B, C, D, E, and F demonstrate normalized RyR2 channel gating and increased calstabin2 binding to RyR2 channels after treatment with JTV-519.
  • Embodiment A are immunoblots of immunoprecipitated wild- type RyR2 (RyR2-WT) channels phosphorylated by PKA in the absence or presence of the inhibitor peptide PKIs -24 and incubated with calstabin2 (250 nM) at the indicated JTV-519 concentrations; the immunoblots show the amount of RyR2 (top) and the amount of calstabin2 (bottom) associated with immunoprecipitated RyR2 after incubation with or without the indicated concentrations of JTV-519.
  • Embodiment B is immunoblots of RyR2-S2809D, which mimics constitutive PKA phosphorylation of RyR2, analyzed as in Embodiment A.
  • Embodiment C are binding curves of 35 S-radiolabeled calstabin2 to unphosphorylated or PKA phosphorylated RyR2 or to RyR2-S2809D in the presence or absence of JTV-519, documenting differences in the calstabin2 binding affinity for RyR2.
  • Embodiments D, E, and F are single channel tracings (left) and amplitude histograms (right) of PKA-phosphorylated RyR2s (embodiments E and F) or unphosphorylated (embodiment D in presence of PKA inhibitor PKI 4 ) incubated with calstabin2 (250 nM) with (embodiment F) or without (embodiments D and E) JTV-519 (1 ⁇ M).
  • Single-channel tracings are shown at 150 nM [Ca 2+ ] which mimics the diastolic or resting phase in the heart; channel openings are upward, the dash indicates the full level of channel opening (4 pA), and "c" indicates the closed state of the channels.
  • the amplitude histograms have amplitude represented on the x axis, and events on the y axis indicates the number of channel openings.
  • Embodiments A, B, C, D, E, and F demonstrate that RyRl channel function is increased and normalized in mdx mice treated with JTV-519.
  • Embodiments A and B are, respectively, a single channel current trace and an amplitude histogram of RyRl from soleus muscle of a control (wild-type) mouse under resting conditions.
  • Embodiments C and D are, respectively, a single channel current trace and an amplitude histogram of RyRl from soleus muscle of an mdx mouse.
  • Embodiment E and F are, respectively, a single channel current trace and an amplitude histogram of RyRl from soleus muscle of an mdx mouse treated with JTV-519.
  • Figure 12 embodiments A and B, are, respectively, immunoblots of RyRl, RyRl- pSer 2843 , and RyRl -associated calstabinl in mdx mice and wild-type mice; and bar graphs of the relative amounts of RyRl-pSer 2843 and calstabinl in mdx and wild-type mice.
  • FIG. 13 embodiments A 3 B, and C, demonstrate that a SR Ca 2+ leak is detectable in the skeletal muscles of animals with heart failure.
  • Embodiments A and B are fluorescence line scan images of Ca 2+ sparks in myof ⁇ bers from, respectively, sham and postmyocardial infarction (PMI) rats.
  • Embodiment C provides bar graphs summarizing the amplitude, rise time, FDHM, and FWHM of the Ca 2+ sparks for the sham (open symbols) and PMI (closed symbols) rats.
  • FIG 14, embodiments A and B demonstrate that treatment of wild-type mice with JTV-519 improves soleus muscle fatigue times.
  • Embodiment A provides maximal tetanic force fatigue time tracings for wild-type and calstab ⁇ " ' " mice, treated with JTV-519 or placebo as indicated.
  • Embodiment B are bar graphs summarizing the mean time to fatigue for wild-type and calstabin2 " " mice, treated with JTV-519 or placebo as indicated.
  • FIG. 15 illustrates that the beneficial effects of JTV-519 treatment on skeletal muscle function depend on calstabinl and not calstabin2 binding to RyRl.
  • Embodiment A provides bar graphs of PKA phosphorylation of RyRl at Ser-2844 in mice, which corresponds to Ser-2843 in humans.
  • Embodiment B are bar graphs of the amount of calstabinl bound to RyRl from wild-type and calstabin2 "A mice treated with JTV- 519 or placebo.
  • Embodiments A, B, C, and D demonstrate that JTV-519 normalizes abnormal or leaky RyRl channel function in vivo.
  • Embodiments A and C are single channel current traces of wild-type mice with heart failure treated with placebo (A) and JTV-519 (C).
  • Embodiments B and D are amplitude histograms for wild-type mice with heart failure treated with placebo (B) and JTV-519 (D).
  • FIG. 17 Figure 17, embodiments A and B, demonstrate that JTV-519 increases calstabin binding to PKA phosphorylated RyR.
  • Embodiment A are immunoblots of calstabinl incubated and associated with RyRl and calstabin2 incubated and associated with RyR2 at increasing concentrations of JTV-519.
  • Embodiment B provides graphs summarizing the proportion of calstabinl and calstabin2 bound to RyRl and RyR2, as indicated.
  • FIG. 18 Figure 18, embodiments A, B, C, and D, demonstrate that PKA phosphorylation of Ser-2843 increases the open probability and gating kinetics of RyRl channels.
  • Embodiment A provides single channel current traces and corresponding histogram of wild-type RyRl.
  • Embodiment B provides single channel current traces and corresponding histogram of wild- type RyRl that is PKA phosphorylated.
  • Embodiment C provides single channel current traces and corresponding histogram of RyRl -Ser-2843 A.
  • Embodiment D provides single channel current traces and corresponding histogram of RyRl-Ser-2843D.
  • FIG. 19 demonstrate the PKA hyperphosphorylation and calstabinl deficiency of RyRl channels after sustained exercise.
  • Embodiment A are immunoblots of RyRl, RyRl -pSer 2844 , RyRl -pSer 2849 , and calstabinl for control and swim mice following an exercise regime.
  • Embodiment B is a bar graph summarizing the relative amounts of the indicated compounds following the exercise regime.
  • FIG. 20 illustrates that RyRl PKA phosphorylation increases after exposure to increasing durations of sustained exercise.
  • Embodiment A provides immunoblots of RyRl and RyRl- pSer 2844 following increasing durations of sustained exercise.
  • Embodiment B is a graph showing the relative PKA phosphorlation of RyRl for varying durations of exercise.
  • FIG. 21 demonstrate that RyRl PKA phosphorylation increases with muscle fatigue.
  • Embodiments A and B are, respectively, fatigue time tracings and a bar graph showing mean fatigue times for rat soleus muscle of heart failure and control subjects.
  • Embodiment C is a graph of PKA phosphorylation versus fatigue time.
  • Figure 22 are trichrome and hematoxylin-eosin stains of cross-sections of the mouse M. extensor digitorum longus, and demonstrates myofiber degeneration consistent with dystrophic remodeling following sustained exercise.
  • Figure 23 shows a sample hERG current trace before (control) and after application of ARM036 at 100 ⁇ M. Also shown is the voltage pulse protocol used to evoke the hERG currents.
  • Figure 24 shows a typical time course of the effect of ARM036 on hERG current amplitude. Application of 10 ⁇ M ARM036 is indicated by the horizontal bar.
  • Figure 25 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM036 at various concentrations.
  • Figure 26 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM036-Na at various concentrations.
  • Figure 27 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM047 at various concentrations.
  • Figure 28 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM048 at various concentrations.
  • Figure 29 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM050 at various concentrations.
  • Figure 30 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM057 at various concentrations.
  • Figure 31 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM064 at various concentrations.
  • Figure 32 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM074 at various concentrations.
  • Figure 33 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM075 at various concentrations.
  • Figure 34 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM076 at various concentrations.
  • Figure 35 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM077 at various concentrations.
  • Figure 36 is a concentration-response graph showing the percent inhibition of hERG current after application of ARMlOl at various concentrations.
  • Figure 37 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM 102 at various concentrations.
  • Figure 38 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM 103 at various concentrations.
  • Figure 39 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM 104 at various concentrations.
  • Figure 40 is a concentration-response graph showing the percent inhibition of hERG current after application of ARMl 06 at various concentrations.
  • Figure 41 is a concentration-response graph showing the percent inhibition of hERG current after application of ARMl 07 at various concentrations.
  • Figure 42 is a concentration-response graph showing the percent inhibition of hERG current after application of S26 at various concentrations.
  • Figure 43 is a concentration-response graph showing the percent inhibition of hERG current after application of JTV-519 ("ARMOXX”) at various concentrations.
  • RDCaI compounds refers to compounds of the general Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1, 1-m or II as provided by the invention, and herein referred to as "compound(s) of the invention".
  • the compounds of the invention are referred using a numerical naming system, with compound numbers 1 to 123 provided herein. These numbered compounds are referred to using either the prefix “S” or the prefix “ARM.” Thus, the first numbered compound is referred to either as “Sl” or “ARMOOl”, the second numbered compound is referred to as either “S2” or “ARM002”, the third numbered compound is referred to as either “S3” or “ARM003”, and so on.
  • S and the “ARM” nomenclature systems are used interchangeably throughout the specification, the drawings, and the claims.
  • alkyl refers to a linear or branched, saturated hydrocarbon having from 1 to 6 carbon atoms.
  • Representative alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, hexyl, isohexyl, and neohexyl.
  • C 1 -C 4 alkyl refers to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 4 carbon atoms, such as methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, and isobutyl.
  • alkenyl refers to a linear or branched hydrocarbon having from 2 to 6 carbon atoms and having at least one carbon-carbon double bond. In one embodiment, the alkenyl has one or two double bonds.
  • the alkenyl moiety may exist in the E or Z conformation and the compounds of the present invention include both conformations.
  • alkynyl refers to a linear or branched hydrocarbon having from 2 to 6 carbon atoms and having at least one carbon-carbon triple bond.
  • aryl refers to an aromatic group containing 1 to 3 aromatic rings, either fused or linked.
  • cyclic group as used herein includes a cycloalkyl group and a heterocyclic group.
  • cycloalkyl group refers to a three- to seven-membered saturated or partially unsaturated carbon ring. Any suitable ring position of the cycloalkyl group may be covalently linked to the defined chemical structure.
  • exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl,
  • heterocyclic group or “heterocyclic” or “heterocyclyl” or “heterocyclo” as used herein refers to fully saturated, or partially or fully unsaturated, including aromatic (i.e., “heteroaryl") cyclic groups (for example, 4 to 7 membered monocyclic, 7 to 11 membered bicyclic, or 10 to 16 membered tricyclic ring systems) which have at least one heteroatom in at least one carbon atom-containing ring.
  • Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3, or 4 heteroatoms selected from nitrogen atoms, oxygen atoms and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized.
  • the heterocyclic group may be attached to the remainder of the molecule at any heteroatom or carbon atom of the ring or ring system.
  • heterocyclic groups include, but are not limited to, azepanyl, azetidinyl, aziridinyl, dioxolanyl, furanyl, furazanyl, homo piperazinyl, imidazolidinyl, imidazolinyl, isothiazolyl, isoxazolyl, morpholinyl, oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, pyrirm ' dinyl, phenanthridinyl, phenanthrolinyl, piperazinyl, piperidinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidin
  • bicyclic heterocyclic groups include indolyl, isoindolyl, benzothiazolyl, benzoxazolyl, benzoxadiazolyl, benzothienyl, quinuclidinyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuryl, benzofurazanyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl (such as furo[2,3-c]pyridinyl, furo[3,2-b]pyridinyl] or furo[2,3-b]pyridinyl), dihydroisoindolyl, dihydroquinazolinyl (such as 3,4-dihydro
  • phenyl refers to a substituted or unsubstituted phenyl group.
  • alkyl alkenyl
  • alkynyl alkynyl
  • aryl aryl
  • phenyl cyclic group
  • cycloalkyl heterocyclyl
  • heterocyclo heterocyclo
  • heterocycle may further be optionally substituted with one or more substituents.
  • groups such as alkyl, cycloalkyl, alkenyl, alkynyl, cycloalkenyl, alkylaryl, heteroaryl, heterocycle and aryl can themselves be optionally substituted.
  • Exemplary substituents may further optionally include at least one labeling group, such, as a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group.
  • a fluorescent labeling group can be selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade BlueTM, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
  • DAPI 6-Diamidino-2-phenylindole
  • ARMl 18 of the present invention contains a labeling group BODIPY, which is a family of fluorophores based on the 4,4-difluoro-4-bora-3a,4a-diaza-,s'-indacene moiety.
  • BODIPY is a family of fluorophores based on the 4,4-difluoro-4-bora-3a,4a-diaza-,s'-indacene moiety.
  • fluorescent label moieties and fluorescence techniques see, e.g., Handbook of Fluorescent Probes and Research Chemicals, by Richard P. Haughland, Sixth Edition, Molecular Probes, (1996), which is hereby incorporated by reference in its entirety.
  • One of skill in the art can readily select a suitable labeling group, and conjugate such a labeling group to any of the compounds of the invention, without undue experimentation.
  • quaternary nitrogen refers to a tetravalent positively charged nitrogen atom including, for example, the positively charged nitrogen in a tetraalkylammonium group (e.g., tetramethylammonium, N-methylpyridinium), the positively charged nitrogen in protonated ammonium species (e.g., trimethyl-hydroammonium, N-liydropyridinium), the positively charged nitrogen in amine N-oxides (e.g., N-methyl-morpholine-N-oxide, pyridine-N-oxide), and the positively charged nitrogen in an N-amino-ammonium group (e.g., N-aminopyridinium).
  • a tetraalkylammonium group e.g., tetramethylammonium, N-methylpyridinium
  • protonated ammonium species e.g., trimethyl-hydroammonium, N-liydropyridinium
  • the nitrogen in the benzothiazepine ring of compounds of the present invention may optionally be a quaternary nitrogen.
  • Non-limiting examples include ARM-113 and ARM-119.
  • Compounds of the present invention may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present invention.
  • prodrug denotes a compound that, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield compounds of the present invention.
  • All stereoisomers of the compounds of the present invention are contemplated within the scope of this invention.
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers (e.g., as a pure or substantially pure optical isomer having a specified activity), or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the cbiral centers of the present invention may have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • racemic forms can be resolved by physical methods, such as, for example, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography.
  • the individual optical isomers can be obtained from the racemates by any suitable method, including without limitation, conventional methods, such as, for example, salt formation with an optically active acid followed by crystallization.
  • Compounds of the present invention are, subsequent to their preparation, preferably isolated and purified to obtain a composition containing an amount by weight equal to or greater than 99% of the compound ("substantially pure” compound), which is then used or formulated as described herein. Such “substantially pure” compounds of the present invention are also contemplated herein as part of the present invention.
  • the present invention provides compounds that are capable of treating and preventing disorders and diseases associated with the RyR receptors that regulate calcium channel functioning in cells. More particularly, the present invention provides compounds that are capable of treating or preventing a leak in RyR channels.
  • “Disorders and diseases associated with the RyR receptors” means disorders and diseases that can be treated and/or prevented by modulating the RyR receptors that regulate calcium channel functioning in cells.
  • “Disorders and diseases associated with the RyR receptors” include, without limitation, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome.
  • Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise- induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
  • Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof.
  • CPVT catecholaminergic polymorphic ventricular tachycardia
  • Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence.
  • Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss.
  • the present invention provides compounds of Formula I:
  • R 1 is selected from the group consisting of H, oxo, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
  • R 4 is selected from the group consisting of H, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
  • R 6 is selected from the group consisting of -ORi 5 , -NHNRi 5 Ri 6 , -NHOH, -NR 15 Ri 6 , -CH 2 X, acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
  • R 7 is selected from the group consisting Of-ORj 5 , -NRi 5 Ri 6 , -NHNR 15 Ri 6 , -NHOH, -CH 2 X, alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
  • R 8 and R 9 independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
  • R 11 , R 12 , R 13 , and Ri 4 independently are selected from the group consisting of H, OH, NH 2 , - NHNH 2 , -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl maybe optionally substituted;
  • Ri 5 and R 16 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH 2 , alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted; and optionally Ri 5 and Ri 6 together with the N to which they are bonded may form a heterocycle which may be substituted; the nitrogen in the benzothiazepine ring may optionally be a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solv
  • R 2 is not H, -C(O)CHOH 2 , or
  • the present invention provides compounds of Formula I, as described above, with the proviso that said compound is not S24 or S68.
  • the present invention provides compounds of Formula I-a:
  • R 5 is selected from the group consisting of-NRi 5 Ri 6 , -NHNRisRi ⁇ , -NHOH, -OR 15 , - -CO 2 R 15 , -CH 2 X, acyl, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
  • R 6 is selected from the group consisting of -ORi 5 , -NHNRi 5 Ri 6 , -NHOH, -NR 15 R 16 , -CH 2 X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
  • R 7 is selected from the group consisting of H, -OR 15 , -NR 15 Ri 6 , -NHNRi 5 Ri 6 , -NHOH, - CH 2 X, alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
  • R 8 and R9 independently are selected from the group consisting of -OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
  • m is 0, 1, 2, 3, or 4;
  • Rn, R 12 , Ri 3 j and R ⁇ independently are selected from the group consisting of H, OH, NH 2 , - NHNH 2 , -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
  • R 15 and R 16 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH 2 , alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted; and optionally R 15 and R 16 together with the N to which they are bonded may form a heterocycle which may be substituted or unsubstituted; the nitrogen in the benzothiazepine ring may be optionally a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and
  • R 2 is not H, -C(O)CHOH 2 , or
  • the present invention provides compounds of formula I-b:
  • R 2 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds formula of I-c: wherein each R, R 7 , q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention provides compounds of formula I-c, wherein R 7 is selected from the group consisting of -OH, -NR ⁇ R ⁇ , alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
  • the present invention provides compounds of formula of I- d:
  • R 7 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-d, wherein R 7 is selected from the group consisting of -OH, -NR 15 R1 6 , alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted.
  • the present invention provides compounds of formula of I-e:
  • each R, R 5 , q and n is as defined compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention provides compounds of formula I-e, wherein R 5 is selected from the group consisting Of-NRi 5 R 16 , -NHOH, -OR 15 , -CH 2 X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
  • the present invention provides compounds of formula I-e, wherein R 5 is an alkyl substituted by at least one labeling group, such as a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group.
  • R 5 is an alkyl substituted by at least one labeling group, such as a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group.
  • a fluorescent labeling group can be selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade BlueTM, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
  • the present invention provides compounds of formula of I-f:
  • R 5 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' and R" are
  • the present invention provides compounds of formula I-f, wherein R 5 is selected from the group consisting Of-NR 15 R 16 , -NHOH, -OR 1S , -CH 2 X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
  • the present invention provides compounds of formula of I-g:
  • W is S or O; each R, R 15 , R 16 , q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention provides compounds of formula I-g, wherein R 15 and Ri 6 independently are selected from the group consisting of H, OH, NH 2 , alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylallcyl may be substituted; and optionally R15 and R 16 together with the N to which they are bonded may form a heterocycle which may be substituted.
  • the present invention provides compounds of formula I-g, wherein W is O or S.
  • the present invention provides compounds of formula of I-h:
  • Ri5, R 16 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-h, wherein R 15 and Ri 6 independently are selected from the group consisting of H, OH, NH 2 , alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted; and optionally R 15 and R 16 together with the N to which they are bonded may form a heterocycle which may be substituted.
  • the present invention provides compounds of formula I-g, wherein W is O or S.
  • the present invention provides compounds of formula of I-i:
  • R 17 is selected from the group consisting Of-NR 15 R 16 , -NHNRisRi ⁇ , -NHOH, -OR 1 S, -CH 2 X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; each R, q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • the present invention provides compounds of formula I-i, wherein R 17 is -NR I5 R I6 , and -OR 15 .
  • Ri 7 is -OH, -OMe, -NEt, -NHEt 3 -NHPh, -NH 2 , or -NHCH 2 pyridyl.
  • R 17 is selected from the group consisting Of-NRi 5 R 16 , -NHNRi 5 R 16 , -NHOH, -OR 15 , -CH 2 X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; n is as defined in compounds of formula I-a; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-j, wherein Ri 7 is -NR 15 Ri 6 Or-ORi 5 .
  • Ri 7 is -OH, -OMe, -NEt, - NHEt, -NHPh, -NH 2 , or -NHCH 2 pyridyl.
  • R' is H or OMe
  • R" is H.
  • m is O
  • Ris is Ci-C 4 alkyl, such as Me, Et, propyl, and butyl.
  • m is 2, and Rj 8 is pyrrolidine, piperidine, piperazine, or morpholine.
  • Ri 8 is a fluorescent labeling group selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade BlueTM, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
  • the present invention provides compounds of formula of 1-1:
  • R 6 and n are as defined in compounds of formula I-a; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe 3 and R" is H.
  • the present invention provides compounds of formula 1-1, wherein R 6 is selected from the group consisting of -NRisR-ie, -ORi 5 , -NHOH, - CH 2 X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted.
  • R 6 is -NRi 5 R 16 such as -NHPh, pyrrolidine, piperidine, piperazine, morpholine, and the like. In some other cases, R 6 is alkoxyl, such as -O-tBu.
  • R 9 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
  • R' is H or OMe
  • R" is H.
  • the present invention provides compounds of formula I-m, wherein R 8 and R 9 are independently alkyl, aryl, -OH, alkoxyl, or alkylamino.
  • R 8 is Ci-C 4 alkyl such as Me, Et, propyl and butyl; and R 9 is aryl such as phenyl.
  • Examples of such compounds include, without limitation, Sl, S2, S3, S4, S5, S6, S7, S9, SIl, S12, S13, S14, S19, S20, S22, S23, S25, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S
  • R is at positions 2, 3, or 5 on the benzene ring.
  • each R is independently selected from the group consisting of H, halogen, -OH, -NH 2 , -NO 2 , -CN, -N 3 , -SO 3 H, acyl, alkyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero- )arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of
  • each R is independently selected from the group consisting of H, halogen, -OH, -NH 2 , -NO 2 , -CN, -N 3 , -SO 3 H, acyl, alkyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero- )aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero- )arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , -SH, nitro, ox
  • R 5 is selected from the group consisting of -NR 16 , NHNHR 16 , NHOH, -ORi 5 , CONH 2 NHR 16 , CONRi 6 , CH 2 X, acyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (
  • the present invention provides compounds of Formula II: In another embodiment, the present invention provides compounds of Formula II:
  • the compounds of the invention reduce the open probability of RyR by increasing the affinity of FKBP12 (calstabinl) and FKBP12.6 (calstabin2) for, respectively PKA- phosphorylated RyRl and PKA-phosphorylated RyR2.
  • the compounds of the invention normalize gating of mutant RyR channels, including CPVT-associated mutant RyR2 channels, by increasing FKBP12 (calstabinl) and FKBP12.6 (calstabin2) binding affinity.
  • the compounds of the invention revent disorders and conditions involving modulation of the RyR receptors, particularly the RyRl and RyR2 receptors.
  • disorders and conditions include, without limitation, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome.
  • Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise- induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
  • Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof.
  • Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence.
  • Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss.
  • the compounds of the invention treat these disorders and conditions by increasing FKBP 12 (calstabinl)-RyRl binding affinity and increasing FKBP12.6 (calstabin2)-RyR2 binding affinity.
  • the present invention provides a method for limiting or preventing a decrease in the level of RyR-bound FKBP (calstabin) in cells of a subject.
  • RyR includes RyRl, RyR2, and RyR3. Additionally, FKBP includes both FKBP12 (calstabinl) and FKBP12.6 (calstabin2).
  • RyR-bound FKBP therefore refers to RyRl-bound FKBP12 (calstabinl), RyR2-bound FKBP 12.6 (calstabk ⁇ ), and RyR3-bound FKBP 12 (calstabinl).
  • RyR also includes a “RyR protein” and a “RyR analogue.”
  • a 11 RyR analogue is a functional variant of the RyR protein, having RyR biological activity, that has 60% or greater amino-acid-sequence homology with the RyR protein.
  • the RyR of the present invention are unphosphorylated, phosphorylated (e.g., by PKA), or hyperphosphorylated (e.g., by PKA).
  • RyR biological activity refers to the activity of a protein or peptide that demonstrates an ability to associate physically with, or bind with, FKBP 12 (calstabinl) in the case of RyRl and RyR3, and FKBP12.6 (calstabin2) in the case of RyR2 (i.e., binding of approximately two fold or, approximately five fold, above the background binding of a negative control), under the conditions of the assays described herein.
  • FKBP includes both an "FKBP protein” and an “FKBP analogue,” whether it be FKBP12 (calstabinl) or FKBP12.6 (calstabin2).
  • protein shall include a protein, protein domain, polypeptide, or peptide, and any fragment thereof.
  • An "FKBP analogue” is a functional variant of the FKBP protein, having FKBP biological activity, that has 60% or greater amino-acid-sequence homology with the FKBP protein, whether it be FKBP 12 (calstabinl) or FKBP12.6 (calstabin2).
  • FKBP biological activity refers to the activity of a protein or peptide that demonstrates an ability to associate physically with, or bind with, unphosphorylated or non-hyperphosphorylated RyR2 (i.e., binding of approximately two fold, or approximately five fold, above the background binding of a negative control), under the conditions of the assays described herein.
  • RyR-bound FKBP includes a molecule of an FKBP 12 (calstabinl) protein that is bound to an RyRl protein subunit or a tetramer of FKBP 12 that is in association with a tetramer of RyRl, a molecule of FKBP12.6 (calstabin2) protein that is bound to an RyR2 protein subunit or a tetramer of FKBP 12.6 that is in association with a tetramer of RyR2, and a molecule of an FKBP 12 (calstabinl) protein that is bound to an RyR3 protein subunit or a tetramer of FKBP12 that is in association with a tetramer of RyR3. Therefore, "RyR-bound FKBP” refers to "RyRl-bound FKBP12," "RyR2-bound FKBP
  • a "decrease” or “disorder” in the level of RyR-bound FKBP in cells of a subject refers to a detectable decrease, diminution or reduction in the level of RyR-bound FKBP in cells of the subject.
  • Such a decrease is limited or prevented in cells of a subject when the decrease is in any way halted, hindered, impeded, obstructed or reduced by the administration of compounds of the invention, such that the level of RyR-bound FKBP in cells of the subject is higher than it would otherwise be in the absence of the administered compound.
  • the level of RyR-bound FKBP in a subject is detected by standard assays and techniques, including those readily determined from the known art (e.g., immunological techniques, hybridization analysis, immunoprecipitation, Western-blot analysis, fluorescence imaging techniques and/or radiation detection, etc.), as well as any assays and detection methods disclosed herein.
  • standard assays and techniques including those readily determined from the known art (e.g., immunological techniques, hybridization analysis, immunoprecipitation, Western-blot analysis, fluorescence imaging techniques and/or radiation detection, etc.), as well as any assays and detection methods disclosed herein.
  • protein is isolated and purified from cells of a subject using standard methods known in the art, including, without limitation, extraction from the cells (e.g., with a detergent that solubilizes the protein) where necessary, followed by affinity purification on a column, chromatography (e.g., FTLC and HPLC), immunoprecipitation (with an antibody), and precipitation (e.g., with isopropanol and a reagent such as Trizol). Isolation and purification of the protein is followed by electrophoresis (e.g., on an SDS-polyacrylamide gel).
  • a decrease in the level of RyR-bound FKBP in a subject, or the limiting or prevention thereof, is determined by comparing the amount of RyR-bound FKBP detected prior to the administration of JTV-519 or a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, (in accordance with methods described below) with the amount detected a suitable time after administration of the compound.
  • a decrease in the level of RyR-bound FKBP in cells of a subject is limited or prevented, for example, by inhibiting dissociation of FKBP and RyR in cells of the subject; by increasing binding between FKBP and RyR in cells of the subject; or by stabilizing the RyR-FICBP complex in cells of a subject.
  • inhibiting dissociation includes blocking, decreasing, inhibiting, limiting or preventing the physical dissociation or separation of an FKBP subunit from an RyR molecule in cells of the subject, and blocking, decreasing, inhibiting, limiting or preventing the physical dissociation or separation of an RyR molecule from an FKBP subunit in cells of the subject.
  • the term "increasing binding” includes enhancing, increasing, or improving the ability of phosphorylated RyR to associate physically with FKBP (e.g., binding of approximately two fold or, approximately five fold, above the background binding of a negative control) in cells of the subject and enhancing, increasing or improving the ability of FKBP to associate physically with phosphorylated RyR (e.g., binding of approximately two fold, or, approximately five fold, above the background binding of a negative control) in cells of the subject.
  • a decrease in the level of RyR-bound FKBP in cells of a subject is limited or prevented by directly decreasing the level of phosphorylated RyR in cells of the subject or by indirectly decreasing the level of phosphorylated RyR in the cells (e.g., by targeting an enzyme (such as PKA) or another endogenous molecule that regulates or modulates the functions or levels of phosphorylated RyR in the cells).
  • the level of phosphorylated RyR in the cells is decreased by at least 10% in the method of the present invention. In another embodiment, the level of phosphorylated RyR is decreased by at least 20%.
  • the subject of the present invention are in vitro and in vivo systems, including, without limitation, isolated or cultured cells or tissues, non-cell in vitro assay systems and an animal (e.g., an amphibian, a bird, a fish, a mammal, a marsupial, a human, a domestic animal (such as a cat, dog, monkey, mouse or rat) or a commercial animal (such as a cow or Pig))-
  • an animal e.g., an amphibian, a bird, a fish, a mammal, a marsupial, a human, a domestic animal (such as a cat, dog, monkey, mouse or rat) or a commercial animal (such as a cow or Pig)
  • the cells of a subject include striated muscle cells.
  • a striated muscle is a muscle in which the repeating units (sarcomeres) of the contractile myofibrils are arranged in registry throughout the cell, resulting in transverse or oblique striations that are observed at the level of a light microscope.
  • Examples of striated muscle cells include, without limitation, voluntary (skeletal) muscle cells and cardiac muscle cells.
  • the cell used in the method of the present invention is a human cardiac muscle cell.
  • the term "cardiac muscle cell” includes cardiac muscle fibers, such as those found in the myocardium of the heart.
  • Cardiac muscle fibers are composed of chains of contiguous heart- muscle cells, or cardiomyocytes, joined end to end at intercalated disks. These disks possess two kinds of cell junctions: expanded desmosomes extending along their transverse portions, and gap junctions, the largest of which lie along their longitudinal portions.
  • a decrease in the level of RyR-bound FKBP is limited or prevented in cells of a subject by administering the compounds of the invention to the subject; this would also permit contact between cells of the subject and the compounds of the invention.
  • the compounds of the invention are modulators of calcium-ion channels. In addition to regulating Ca 2+ levels in myocardial cells, the compounds of the invention modulate the Na + current and the inward-rectifier K + current in cells, such as guinea pig ventricular cells, and inhibits the delayed-rectifier K + current in cells, such as guinea pig atrial cells.
  • the compounds of the invention are formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo.
  • the present invention provides a pharmaceutical composition comprising compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I- g, I-h, I-i, I-j, I-k, 1-1 or I-m, in admixture with a pharmaceutically acceptable diluent and/or carrier.
  • the pharmaceutically-acceptable carrier must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof.
  • the pharmaceutically-acceptable carrier employed herein is selected from various organic or inorganic materials that are used as materials for pha ⁇ naceutical formulations and which are incorporated as analgesic agents, buffers, binders, disintegrants, diluents, emulsifiers, excipients, extenders, glidants, solubilizers, stabilizers, suspending agents, tonicity agents, vehicles and viscosity-increasing agents.
  • pharmaceutical additives such as antioxidants, aromatics, colorants, flavor-improving agents, preservatives, and sweeteners, are also added.
  • acceptable pharmaceutical carriers include carboxymethyl cellulose, crystalline cellulose, glycerin, gum arabic, lactose, magnesium stearate, methyl cellulose, powders, saline, sodium alginate, sucrose, starch, talc and water, among others.
  • the pha ⁇ naceutical formulations of the present invention are prepared by methods well-known in the pharmaceutical arts.
  • the compounds of Formula I, I-a, I-b, I- c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m are brought into association with a carrier and/or diluent, as a suspension or solution.
  • a carrier and/or diluent as a suspension or solution.
  • one or more accessory ingredients ⁇ e.g., buffers, flavoring agents, surface active agents, and the like.
  • the choice of carrier is determined by the solubility and chemical nature of the compounds, chosen route of administration and standard pharmaceutical practice.
  • the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m are administered to a subject by contacting target cells ⁇ e.g., cardiac muscle cells) in vivo in the subject with the compounds.
  • target cells ⁇ e.g., cardiac muscle cells
  • the compounds are contacted with ⁇ e.g., introduced into) cells of the subject using known techniques utilized for the introduction and administration of proteins, nucleic acids and other drugs.
  • Examples of methods for contacting the cells with ⁇ i.e., treating the cells with) the compounds of the invention include, without limitation, absorption, electroporation, immersion, injection, introduction, liposome delivery, transfection, transfusion, vectors and other drug-delivery vehicles and methods.
  • the target cells are localized to a particular portion of a subject, it is desirable to introduce the compounds of the invention directly to the cells, by injection or by some other means ⁇ e.g., by introducing the compounds into the blood or another body fluid).
  • the target cells are contained in tissue of a subject and are detected by standard detection methods readily determined from the known art, examples of which include, without limitation, immunological techniques ⁇ e.g., immunoliistochemical staining), fluorescence imaging techniques, and microscopic techniques.
  • the compounds of the present invention are administered to a human or animal subject by known procedures including, without limitation, oral administration, sublingual or buccal administration, parenteral administration, transdermal administration, via inhalation or intranasally, vaginally, rectally, and intramuscularly.
  • the compounds of the invention are administered parenterally, by epifascial, intracapsular, intracranial, intracutaneous, intrathecal, intramuscular, intraorbital, intraperitoneal, intraspinal, intrasternal, intravascular, intravenous, parenchymatous, subcutaneous or sublingual injection, or by way of catheter.
  • the agent is adiminstered to the subject by way of delivery to the subject's muscles including, but not limited to, the subject's cardiac muscles.
  • the agent is administered to the subject by way of targeted delivery to cardiac muscle cells via a catheter inserted into the subject's heart.
  • a formulation of the compounds of the invention may be presented as capsules, tablets, powders, granules, or as a suspension or solution.
  • the fo ⁇ nulation has conventional additives, such as lactose, mannitol, cornstarch or potato starch.
  • the formulation also is presented with binders, such as crystalline cellulose, cellulose derivatives, acacia, cornstarch or gelatins.
  • the formulation is presented with disintegrators, such as cornstarch, potato starch or sodium carboxymethylcellulose.
  • the formulation also is presented with dibasic calcium phosphate anhydrous or sodium starch glycolate.
  • lubricants such as talc or magnesium stearate.
  • the compounds of the invention are combined with a sterile aqueous solution that is isotonic with the blood of the subject.
  • a sterile aqueous solution that is isotonic with the blood of the subject.
  • a formulation is prepared by dissolving a solid active ingredient in water containing physiologically-compatible substances, such as sodium chloride, glycine and the like, and having a buffered pH compatible with physiological conditions, so as to produce an aqueous solution, then rendering said solution sterile.
  • physiologically-compatible substances such as sodium chloride, glycine and the like
  • the formulation is presented in unit or multi-dose containers, such as sealed ampoules or vials.
  • the formulation is delivered by any mode of injection, including, without limitation, epifascial, intracapsular, intracranial, intracutaneous, intrathecal, intramuscular, intraorbital, intraperitoneal, intraspinal, intrasternal, intravascular, intravenous, parenchymatous, subcutaneous, or sublingual or by way of catheter into the subject's heart.
  • the compounds of the invention are combined with skin penetration enhancers, such as propylene glycol, polyethylene glycol, isopropanol, ethanol, oleic acid, N-methylpyrrolidone and the like, which increase the permeability of the skin to the compounds of the invention and permit the compounds to penetrate through the skin and into the bloodstream.
  • skin penetration enhancers such as propylene glycol, polyethylene glycol, isopropanol, ethanol, oleic acid, N-methylpyrrolidone and the like, which increase the permeability of the skin to the compounds of the invention and permit the compounds to penetrate through the skin and into the bloodstream.
  • the compound/enhancer compositions also may be further combined with a polymeric substance, such as ethylcellulose, hydroxypropyl cellulose, ethylene/vinylacetate, polyvinyl pyrrolidone, and the like, to provide the composition in gel form, which are dissolved in a solvent, such as methylene chloride, evaporated to the desired viscosity and then applied to backing material to provide a patch.
  • a polymeric substance such as ethylcellulose, hydroxypropyl cellulose, ethylene/vinylacetate, polyvinyl pyrrolidone, and the like
  • the composition is in unit dose form such as a tablet, capsule or single-dose vial.
  • Suitable unit doses i.e., therapeutically effective amounts, can be determined during clinical trials designed appropriately for each of the conditions for which administration of a chosen compound is indicated and will, of course, vary depending on the desired clinical endpoint.
  • the present invention also provides articles of manufacture for treating and preventing disorders, such as cardiac disorders, in a subject.
  • the articles of manufacture comprise a pharmaceutical composition of one or more of the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, as described herein.
  • the articles of manufacture are packaged with indications for various disorders that the pharmaceutical compositions are capable of treating and/or preventing.
  • the articles of manufacture comprise a unit dose of a compound disclosed herein that is capable of treating or preventing a muscular disorder, and an indication that the unit dose is capable of treating or preventing a certain disorder, for example an arrhythmia.
  • the compounds of Formula I, I- a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m are administered to the subject (or are contacted with cells of the subject) in an amount effective to limit or prevent a decrease in the level of RyR-bound FKBP in the subject, particularly in cells of the subject.
  • a suitable amount of the compounds of the invention effective to limit or prevent a decrease in the level of RyR-bound FKBP in the subject ranges from about 0.01 mg/kg/day to about 20 mg/kg/day, and/or is an amount sufficient to achieve plasma levels ranging from about 300 ng/ml to about 1000 ng/ml.
  • the amount of compounds from the invention ranges from about 10 mg/kg/day to about 20 mg/kg/day. In another embodiment, from about 0.01 mg/kg/day to about 10 mg/kg/day is administered.
  • from about 0.01 mg/kg/day to about 5 mg/kg/day is administered, in another embodiment, from about 0.05 mg/kg/day to about 5 mg/kg/day is administered, hi another, preferred embodiment, from about 0.05 mg/kg/day to about 1 mg/kg/day is administered.
  • the present invention provides a new range of therapeutic treatments for patients with various disorders involving modulation of the RyR receptors, particularly skeletal muscular disorders (RyRl), cardiac (RyR2) disorders, and cognitive (RyR3) disorders.
  • RyRl skeletal muscular disorders
  • RV2 cardiac
  • RyR3 cognitive disorders
  • the subject has not yet developed a disorder, such as cardiac disorders (e.g., exercise-induced cardiac arrhythmia).
  • a disorder such as cardiac disorders (e.g., exercise-induced cardiac arrhythmia).
  • the subject is in need of treatment for a disorder, including a cardiac disorder.
  • Various disorders that the compounds of the invention treat or prevent include, but are not limited to, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome.
  • Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
  • Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof.
  • Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence.
  • Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss.
  • One skilled in the art will recognize still other diseases, including but not limited to muscular and cardiac disorders, that the compounds of the invention can be useful to treat, in accordance with the information provided herein.
  • the amount of compounds of the invention effective to limit or prevent a decrease in the level of RyR2 -bound FKBP12.6 in the subject is an amount effective to prevent exercise- induced cardiac arrhythmia in the subject.
  • Cardiac arrhythmia is a disturbance of the electrical activity of the heart that manifests as an abnormality in heart rate or heart rhythm.
  • an amount of compounds of the invention "effective to prevent exercise- induced cardiac arrhythmia” includes an amount of compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, effective to prevent the development of the clinical impairment or symptoms of the exercise-induced cardiac arrhythmia (e.g., palpitations, fainting, ventricular fibrillation, ventricular tachycardia and sudden cardiac death).
  • the clinical impairment or symptoms of the exercise-induced cardiac arrhythmia e.g., palpitations, fainting, ventricular fibrillation, ventricular tachycardia and sudden cardiac death.
  • the amount of the compounds effective to prevent exercise-induced cardiac arrhythmia in a subject will vary depending upon the particular factors of each case, including the type of exercise-induced cardiac arrhythmia, the subject's weight, the severity of the subject's condition, and the mode of administration of the compounds. This amount is readily determined by the skilled artisan, based upon known procedures, including clinical trials, and methods disclosed herein.
  • the amount of the compounds of the invention effective to prevent the exercise-induced cardiac arrhythmia is an amount effective to prevent exercise-induced sudden cardiac death in the subject.
  • the compounds of the invention prevent exercise-induced cardiac arrhythmia and exercise- induced sudden cardiac death in the subject.
  • the compounds of the invention are also useful in treating a subject who has already experienced clinical symptoms of these various disorders. For example, if the symptoms of the disorder are observed in the subject early enough, the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I- f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, are effective in limiting or preventing a further decrease in the level of RyR-bound FKBP in the subject.
  • the subject of the present invention is a candidate for exercise-induced cardiac disorders, such as exercise-induced cardiac arrhthmia.
  • Exercise-induced cardiac arrhythmia is a heart condition (e.g., a ventricular fibrillation or ventricular tachycardia, including any that leads to sudden cardiac death) that develops during/after a subject has undergone physical exercise.
  • a "candidate" for an exercise-induced cardiac disorder is a subject who is known to be, or is believed to be, or is suspected of being, at risk for developing a cardiac disorder during/after physical exercise.
  • Examples of candidates for exercise-induced cardiac arrhythmia include, without limitation, an animal/person known to have catecholaminergic polymorphic ventricular tachycardia (CPVT); an animal/person suspected of having CPVT; and an animal/person who is known to be, or is believed to be, or is suspected of being at risk for developing cardiac arrhythmia during/after physical exercise, and who is about to exercise, is currently exercising or has just completed exercise.
  • CPVT catecholaminergic polymorphic ventricular tachycardia
  • CPVT catecholaminergic polymorphic ventricular tachycardia
  • an animal/person suspected of having CPVT and an animal/person who is known to be, or is believed to be, or is suspected of being at risk for developing cardiac arrhythmia during/after physical exercise, and who is about to exercise, is currently exercising or has just completed exercise.
  • CPVT is an inherited disorder in individuals with structurally-normal hearts. It is characterized by stress-induced ventricular tachycardia — a lethal ar
  • the subject has been exercising, or is currently exercising, and has developed an exercise-induced disorder.
  • the amount of the compounds of the invention effective to limit or prevent a decrease in the level of RyR-bound FKBP in the subject is an amount of compound effective to treat the exercise-induced disorder in the subject.
  • an amount of compounds of the invention "effective to treat an exercise-induced disorder” includes an amount of a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, effective to alleviate or ameliorate the clinical impairment or symptoms of the exercise- induced disorder ⁇ e.g., in the case of cardiac arrhythmia, palpitations, fainting, ventricular fibrillation, ventricular tachycardia, and sudden cardiac death).
  • the amount of the compounds of the invention effective to treat an exercise-induced disorder in a subject will vary depending upon the particular factors of each case, including the type of exercise- induced disorder, the subject's weight, the severity of the subject's condition, and the mode of administration of the compounds. This amount is readily determined by the skilled artisan, based upon known procedures, including clinical trials, and methods disclosed herein.
  • the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m treat exercise-induced disorders in the subject.
  • the present invention further provides a method for treating exercise-induced disorders in a subject.
  • the method comprises administering the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, to the subject in an amount effective to treat the exercise-induced disorder in the subject.
  • a suitable amount of the compounds effective to treat, for example, exercise-induced cardiac arrhythmia in the subject ranges from about 5 mg/kg/day to about 20 mg/kg/day, and/or is an amount sufficient to achieve plasma levels ranging from about 300 ng/ml to about 1000 ng/ml.
  • the present invention also provides a method for preventing an exercise-induced disorder in a subject.
  • the method comprises administering the compounds of the invention to the subject in an amount effective to prevent the exercise-induced disorder in the subject.
  • a suitable amount of the compounds of the invention effective to prevent the exercise-induced disorder in the subject ranges from about 5 mg/kg/day to about 20 mg/kg/day, and/or is an amount sufficient to achieve plasma levels ranging from about 300 ng/ml to about 1000 ng/ml.
  • the present invention provides a method for preventing exercise-induced disorders in a subject. The method comprises administering the compounds of the invention to the subject in an amount effective to prevent an exercise-induced disorder in the subject.
  • a suitable amount of the compounds of the invention effective to prevent an exercise-induced disorder in the subject ranges from about 5 mg/kg/day to about 20 mg/kg/day, and/or is an amount sufficient to achieve plasma levels ranging from about 300 ng/ml to about 1000 ng/ml.
  • the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m can be used alone, in combination with each other, or in combination with other agents that have cardiovascular activity including, but not limited to, diuretics, anticoagulants, antiplatelet agents, antiarrhythmics, inotropic agents, chronotropic agents, ⁇ and ⁇ blockers, angiotensin inhibitors and vasodilators. Further, such combinations of the compounds of the present invention and other cardiovascular agents are administered separately or in conjunction. In addition, the administration of one element of the combination is prior to, concurrent to or subsequent to the administration of other agent(s).
  • the exercise-induced cardiac arrhythmia in the subject is associated with VT.
  • the VT is CPVT.
  • the subject is a candidate for exercise-induced cardiac arrhythmia, including candidates for exercise-induced sudden cardiac death.
  • the present invention also provides use of the compounds of the invention in a method for limiting or preventing a decrease in the level of RyR-bound FKBP in a subject who is a candidate for a disorder.
  • the present invention also provides use of the compounds of the invention in a method for treating or preventing a muscular disorder in a subject.
  • the present invention provides use of the compounds of the invention in a method for preventing treating or preventing exercise- induced muscular disorders in a subject.
  • the present invention further provides a method for assaying the effects of the compounds of the invention in preventing disorders and diseases associated with the RyR receptors.
  • the method comprises the steps of: (a) obtaining or generating a culture of cells containing RyR; (b) contacting the cells with one or more of the compounds of the invention; (c) exposing the cells to one or more conditions known to increase phosphorylation of RyR in cells; and (d) determining if the one or more compounds of the invention limits or prevents a decrease in the level of RyR-bound FKBP in the cells.
  • a cell “containing RyR” is a cell in which RyR, including RyRl, RyR2, and RyR3, or a derivative or homologue thereof, is naturally expressed or naturally occurs.
  • Conditions known to increase phosphorylation of RyR in cells include, without limitation, PKA.
  • cells are contacted with one or more of the compounds of the invention by any of the standard methods of effecting contact between drugs/agents and cells, including any modes of introduction and administration described herein.
  • the level of RyR-bound FICBP in the cell is measured or detected by known procedures, including any of the methods, molecular procedures and assays known to one of skill in the art or described herein.
  • the one or more compounds of the invention limits or prevents a decrease in the level of RyR-bound FKBP in the cells.
  • RyR including RyRl; RyR2, and RyR3, has been implicated in a number of biological events in cells. For example, it has been shown that RyR2 channels play an important role in EC coupling and contractility in cardiac muscle cells. Therefore, it is clear that preventive drugs designed to limit or prevent a decrease in the level of RyR-bound FKBP in cells, particularly RyR2-bound FICPB 12.6 in cardiac muscle cells, are useful in the regulation of a number of RyR-associated biological events, including EC coupling and contractility. Thus, the one or more compounds of the invention are evaluated for effect on EC coupling and contractility in cells, particularly cardiac muscle cells, and therefore, usefulness for preventing exercise-induced sudden cardiac death.
  • the method of the present invention further comprises the steps of contacting one or more compounds of the invention with a culture of cells containing RyR; and determining if the one or more compounds has an effect on an RyR-associated biological event in the cells.
  • a "RyR-associated biological event” includes a biochemical or physiological process in which RyR levels or activity have been implicated.
  • examples of RyR-associated biological events include, without limitation, EC coupling and contractility in cardiac muscle cells.
  • the one or more compounds are contacted with one or more cells (such as cardiac muscle cells) in vitro.
  • a culture of the cells is incubated with a preparation containing the one or more compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m.
  • the compounds' effect on a RyR-associated biological event then is assessed by any biological assays or methods known in the art, including immunoblotting, single-channel recordings and any others disclosed herein.
  • the present invention is further directed to one or more compounds of Formula I 3 I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, identified by the above-described identification method, as well as a pharmaceutical composition comprising the compound and a pharmaceutically acceptable carrier and/or diluent.
  • the compounds are useful for preventing exercise-induced sudden cardiac death in a subject, and for treating or preventing other RyR-associated conditions.
  • a "RyR-associated condition" is a condition, disease, or disorder in which RyR level or activity has been implicated, and includes an RyR-associated biological event.
  • the RyR-associated condition is treated or prevented in the subject by administering to the subject an amount of the compound effective to treat or prevent the RyR-associated condition in the subject.
  • This amount is readily determined by one skilled in the art.
  • the present invention provides a method for preventing exercise-induced sudden cardiac death in a subject, by administering the one or more compounds of the invention to the subject in an amount effective to prevent the exercise-induced sudden cardiac death in the subject.
  • the present invention also provides an in vivo method for assaying the effectiveness of the compounds of the invention in preventing disorders and diseases associated with the RyR recpetors.
  • the method comprises the steps of: (a) obtaining or generating an animal containing RyR; (b) administering one or more of the compounds of the invention to the animal; (c) exposing the animal to one or more conditions known to increase phosphorylation of RyR in cells; and (d) dete ⁇ nining the extent the compound limits or prevents a decrease in the level of RyR-bound FKBP in the animal.
  • the method further comprises the steps of: (e) administering one or more of the compounds of the invention to an animal containing RyR; and (f) determining the extent of the effect of the compound on a RyR-associated biological event in the animal.
  • a pharmaceutical composition comprising this compound; and a method for preventing exercise-induced sudden cardiac death in a subject, by administering this compound to the subject in an amount effective to prevent the exercise- induced sudden cardiac death in the subject.
  • the diagnostic assays screen for the release of calcium into cells via the RyR channel, using calcium-sensitive fluorescent dyes (e.g., Fluo-3, Fura-2, and the like).
  • Cells are loaded with the fluorescent dye of choice, then stimulated with RyR activators to determine the reduction of the calcium-dependent fluorescent signal (Brillantes, et al, Stabilization of calcium release channel (ryanodine receptor) function by FK506- binding protein. Cell, 77:513-23, 1994; Gillo, et al, Calcium entry during induced differentiation in murine erythroleukemia cells.
  • an assay involves the expression of recombinant RyR channels in a heterologous expression system, such as Sf9, HEK293, or CHO cells.
  • RyR could also be co-expressed with beta-adrenergic receptors. This would permit assessment of the effect of compounds of the invention on RyR activation, in response to addition of beta- adrenergic receptor agonists.
  • the level of PKA phosphorylation of RyR2 which correlates with the degree of heart failure also is assayed and then used to determine the efficacy of the one or more compounds of the invention to block the PKA phosphorylation of the RyR2 channel.
  • Such an assay is based on the use of antibodies that are specific for the RyR2 protein.
  • the RyR2- channel protein is immunoprecipitated and then back-phosphorylated with PKA and [gamma 32 P]-ATP.
  • the amount of radioactive [ 32 P] label that is transferred to the RyR2 protein then is measured using a phosphorimager (Marx, et al, PKA phosphorylation dissociates FKBP 12.6 from the calcium release channel (ryanodine receptor): defective regulation in failing hearts. Cell, 101 :365-76, 2000).
  • Another assay of the compounds of the invention involves use of a phosphoepitope- specif ⁇ c antibody that detects RyRl that is PKA phosphorylated on Ser 2843 or RyR2 that is PKA phosphorylated on Ser 2809. Immunoblotting with such an antibody can be used to assess efficacy of these compounds for therapy for heart failure and cardiac arrhythmias. Additionally, RyR2 S2809A and RyR2 S2809D knock-in mice are used to assess efficacy of therapy for heart failure and cardiac arrhythmias.
  • mice further provide evidence that PKA hyperphosphorylation of RyR2 is a contributing factor in heart failure and cardiac arrhythmias by showing that the RyR2 S2809A mutation inhibits heart failure and arrhythmias, and that the RyR2 S2809D mutation worsens heart failure and arrhythmias.
  • the present invention provides a method of treating heart failure, atrial fibrillation or exercise-induced cardiac arrhythmia, comprising administering to an animal in need thereof, a therapeutically effective amount of a compound selected from the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m.
  • Intracellular Ca 2+ leak is proposed as a principal mediator of depressed muscle performance and dystrophic muscle remodeling.
  • Muscular dystrophies are heterogeneous hereditary diseases characterized by weakness and progressive muscle wasting.
  • dystrophinopathies Duchenne muscular dystrophy (DMD) is one of the most frequent genetic diseases (X-linked; 1 in 3,500 boys) with death usually occurring before age 30 by respiratory and/or cardiac failure in high numbers of patients.
  • Becker muscular dystrophy represents a milder form of the disease associated with a reduction in the amount or expression of a truncated form of the dystrophin protein whereas Duchenne patients have been characterized by complete absence or very low levels of dystrophin.
  • Duchenne and Becker's muscular dystrophy are caused by mutations in the gene encoding the 427-kDa cytoskeletal protein dystrophin.
  • cardiac symptoms are more common than in DMD patients and do not correlate with skeletal muscle symptoms. Since genetic screening will not eliminative DMD due to a high incidence of sporadic cases, an effective therapy is highly desirable.
  • DMD/BMD have been consistently associated with disturbed intracellular calcium metabolism. Because alterations of intracellular Ca 2+ concentrations in DMD myof ⁇ bers are believed to represent a central pathogenic mechanism, development of a therapeutic intervention that prevents intracellular Ca + abnormalities as a cause of skeletal muscle degeneration is highly desirable.
  • the present invention also provides a method of diagnosis of a disease or disorder in a subject, said method comprising: obtaining a cell or tissue sample from the subject; obtaining DNA from the cell or tissue; comparing the DNA from the cell or tissue with a control DNA encoding RyR to determine whether a mutation is present in the DNA from the cell or tissue, the presence of a mutation indicating a disease or disorder.
  • the mutation is a RyR2 mutation on chromosome Iq42-q43.
  • the mutation is one or more CPTV mutations.
  • the mutation may be a mutation that is present in the DNA encoding RyR2 of a SIDS subject.
  • the diagnostic method is used to detect the presence of a disease or disorder in an adult, a child or a fetus.
  • the disease and disorders include, but are not limited to, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome.
  • Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
  • Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof.
  • Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence.
  • Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss.
  • the present invention further provides a method of diagnosis of disorders and diseases in a subject, said method comprising: obtaining cells or tissue sample from the subject; incubating the cells or tissue sample with the compound of the invention under conditions which increase phosphorylation of RyR in cells; determining (a) whether RyR bound to calstabin (i.e.
  • RyRl bound to calstabinl, RyR2 bound to calstabin2, or RyR3 bound to calstabinl) is increased in the cells or tissue compared to RyR bound to calstabin in control cells or tissues said control cells or tissues lacking mutant RyR calcium channels, or (b) whether a decrease in release of calcium occurs in RyR channels compared to a lack of decrease in release of calcium in the control cells; an increase in RyR-bound calstabin in (a) indicating a disorder or disease in the subject or a decrease in release of calcium in RyR channels in (b) compared to the control cells indicating a cardiac disease or disorder in the subject.
  • the diagnostic method is used to detect the presence of a disease or disorder in an adult, a child or a fetus.
  • the disease and disorders include, but are not limited to, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome.
  • Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
  • Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof.
  • Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence.
  • Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss.
  • the present invention further provides a method of diagnosis of a cardiac disorder or disease in a subject, said method comprising: obtaining cardiac cells or tissue sample from the subject; incubating the cardiac cells or tissue sample with the compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, under conditions which increase phosphorylation of RyR2 in cells; determining (a) whether RyR2 bound to calstabin2 is increased in the cells or tissue compared to RyR2 bound to calstabin2 in control cells or tissues said control cells or tissues lacking mutant RyR2 calcium channels, or (b) whether a decrease in release of calcium occurs in RyR2 channels compared to a lack of decrease in release of calcium in the control cells; an increase in RyR2 -bound calstabin2 in (a) indicating a disorder or disease in the subject or a decrease in release of calcium in RyR2 channels in
  • the provided method is used to diagnose CPTV.
  • the provided method also is used to diagnose sudden infant death syndrome (SIDS).
  • SIDS sudden infant death syndrome
  • the provided method additionally is used to diagnose cardiac irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
  • Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise- induced variants thereof.
  • CPVT catecholaminergic polymorphic ventricular tachycardia
  • the compounds of the invention are also useful in diagnostic assays, screening assays and as research tools.
  • the present invention provides, in a further aspect, processes for the preparation of a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, and salts, solvates, hydrates, complexes, and pro-drugs thereof, and pharmaceutically acceptable salts of such pro-drugs.
  • the present invention provides processes for the preparation of compounds selected from the group consisting of Sl, S2, S3, S4, S5, S6, S7, S9, SI l, S12, SB, S14, S19, S20, S22, S23, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102,
  • the solvent is an organic solvent.
  • the organic solvent is methylene chloride (CH 2 Cl 2 ), chloroform (CCl 4 ), formaldehyde (CH 2 O) or methanol (CH 3 OH).
  • the base catalyst is an amine compound.
  • the base catalyst is an alkylamine such as triethylamine (TEA).
  • TAA triethylamine
  • the base catalyst is pyridine.
  • the basic solution is sodium bicarbonate or calcium carbonate.
  • the basic solution is saturated sodium bicarbonate or saturated calcium carbonate.
  • the acidic solution is a sulfuric acid solution, a hydrochloric acid solution, or a nitric acid solution, hi one embodiment, the solution is IN HCl.
  • solvents, organic solvents, base catalysts, basic solutions, and acidic solutions that are used in the embodiments, according to the description herein.
  • the solvents, organic solvents, reactants, catalysts, wash solutions, and so forth are added at appropriate temperatures (e.g. room temperature or about 20°C-25°C, O 0 C, etc.).
  • S68 is available commercially from MicroChemistry Ltd. (Moscow, Russia). See also WO01/55118 for the preparation of S68.
  • S26 is synthesized as an intermediate in the synthesis of S3, S4, S5, and S54, as illustrated in scheme 1 in example 4. Methods for synthesizing S26 are also described in U.S. Patent Applicatin No. 10/680,988.
  • the reactions are carried out as long as needed (e.g., one hour, several hours, overnight, 24 hours, etc.) to obtain the desired or optimal yields of the desired compounds. Often, the reaction mixtures are stirred. The reactions are carried out at appropriate temperatures (e.g. room temperature or about 20°C-25°C, O 0 C, 100 0 C, etc.).
  • Synthon S26 is prepared according to methods described in U.S. Patent App. No. 10/680,988.
  • S3, S4, S5, and S54 are prepared from S26.
  • the product is purified, for example by column chromatography, to yield S3, S4, S5, or S54.
  • the reaction occurs in a solvent, such as an organic solvent like CH 2 Cl 2 , so that a reaction mixture is formed, and the solvent is removed from the reaction mixture before or during purification of the product.
  • a base catalyst such as triethylamine, is used in the synthesis.
  • basic e.g., saturated sodium bicarbonate
  • acidic washes e.g., IN HCl
  • Column chromatography is used to purifying the residue to isolate the desired product.
  • Sl and S2 are prepared from S3 by reaction with EDSTR 1 R 2 , where R is
  • the product is purified, for example by column chromatography, to yield Sl or S2.
  • the reaction occurs in a solvent, such as an organic solvent like CH 2 Cl 2 , so that a reaction mixture is formed, and the solvent is removed from the reaction mixture before or during purification of the product.
  • Column chromatography for example, is used to purifying the residue to isolate the desired product.
  • RCOX an alcohol of formula RCOX
  • a solvent such as an organic solvent like CH 2 Cl 2 , so that a reaction mixture is formed, and the solvent is removed from the reaction mixture before or during purification of the product.
  • a base catalyst such as triethylamine, is used in the synthesis.
  • S40 is formed by reaction with an alcohol of formula RCOX, where R is 4-N 3 ⁇ -OH-C 6 Hs and X is NHS. Column chromatography, for example, is used to purifying the residue to isolate the desired product.
  • SI l and S12 are prepared from S26 by reaction with a compound of formula C 6 H 4 - NCX, wherein X is O (Sl 1) or S (S12).
  • the reaction occurs in a solvent, such as an organic solvent like CH 2 Cl 2 , so that a reaction mixture is formed, and the solvent is removed from the reaction mixture before or during purification of the product.
  • a base catalyst such as triethylamine or pyridine, is used in the synthesis.
  • a base catalyst such as pyridine is used as the solvent in which the reaction takes place, and additional solvent, such as ethyle acetate or another appropriate organic solvent, is added after the reaction occurs.
  • basic e.g., saturated sodium bicarbonate
  • acidic washes e.g., IN HCl
  • Column chromatography is used to purifying the residue to isolate the desired product.
  • the isomers S 13 and S 14 are prepared from S26 by reaction with phenyl methoxyphosphonyl chloride (Ph(MeO)P(O)Cl).
  • a solvent such as an organic solvent, such as methylene chloride.
  • abase catalyst such as triethylamine may be used, for example, by adding it to a reaction mixture formed by mixing the reactants in a solvent. Also, the reaction mixture is washed with basic solution, for example saturated sodium bicarbonate, if necessary.
  • the isomers are separated and purified, for example, using silica gel chromatography.
  • the reaction occurs in the presence of a solvent, such as an organic solvent, such as methylene chloride.
  • a base catalyst such as triethylamine is added to the reaction mixture formed by mixing the reactants in a solvent.
  • base e.g., saturated sodium bicarbonate
  • acid e.g., IN HCl
  • water washes are used to remove unwanted compounds from the reaction mixture, if needed.
  • the intermediate compound is treated with H 2 O 2 .
  • sodium thiosulfate also is used to treat the intermediate.
  • the reaction occurs in the presence of a solvent, such as an organic solvent, such as methanol (CH 3 OH), forming a reaction mixture.
  • a solvent such as an organic solvent, such as methanol (CH 3 OH)
  • CH 3 OH methanol
  • the solvent is removed from the reaction mixture after the reaction takes place and, if desired, the residue is redissolved in another solvent, such as another organic solvent, such as ethyl acetate.
  • the reaction mixture is washed with basic solution (e.g., saturated sodium carbonate) if desired to remove unwanted compounds from the reaction mixture.
  • the reaction mixture is dried (e.g., using sodium sulfate) if it is washed with basic solution.
  • S57 is prepared from S26 and methyl chlorooxoacetate.
  • the reaction occurs in the presence of a solvent, such as an organic solvent, such as methylene chloride.
  • a base catalyst such as pyridine is used as necessary to facilitate or hasten the reaction.
  • the reaction mixture formed by mixing the reactants and a solvent is washed with basic solution ⁇ e.g., saturated sodium bicarbonate), acidic solution ⁇ e.g. HCl), and water. Purification such as silic gel chromatography yields S57.
  • S36 is prepared from S57 by reaction with sodium hydroxide.
  • the reaction takes place in a solvent, such as an organic solvent, such as methanol.
  • the solvent is removed from the reaction mixture formed by mixing the reactants and the solvent, thereby forming a residue.
  • the residue is dissolved in water and washed with another organic solvent, such as ether, to remove unwanted hydrophobic compounds.
  • the aqueous phase from the basic washes is acidified and the product is extracted therefrom using an organic solvent, such as methylene chloride. Further purification is used if necessary.
  • S38 is prepared in a manner similar to S36, except a compound of formula
  • S44 is prepared by treating S36 with thionyl chloride to form crude S36-C1. Excess thionyl chloride, if any, is removed from the reaction mixture. The crude S 36-Cl then is dissolved in a solvent, such as an organic solvent like methylene chloride, and reacted with mono-protected ⁇ e.g., mono-Boc protected) cystamine. A base catalyst such as pyridine is used if desired, and the reaction mixture is quenched with a basic solution ⁇ e.g., saturated sodium bicarbonate). The reaction mixture formed by mixing the cystamine and S36-C1 reactants is purified.
  • a solvent such as an organic solvent like methylene chloride
  • the protecting groups ⁇ e.g., Boc
  • an appropriate acid or base wash ⁇ e.g., trifluoroacetic acid in an organic solvent in the case of the Boc protecting group.
  • the final product then is purified, for example, using chromatography techniques.
  • S57 and S59 are prepared from S36-C1, which is reacted with methanol (S57) or ethylamine (S59).
  • S43 and S45 are prepared from S36-cystamine, which is prepared as disclosed herein.
  • S-36 cystamine is reacted with an NHS activated ester of an appropriate azido compound to yield S43 and S45.
  • the reaction takes place in a solvent, such as an organic solvent.
  • S37 is prepared from S26 by reaction with 4-nitrophenyl chloroforniate (NO 2 C 6 H S OCOCI).
  • the reaction takes place in a solvent and, if desired, abase catalyst such as triethylamine may be used.
  • the reaction mixture formed by mixing the reactants and a solvent is washed with water to remove unwanted hydrophilic compounds.
  • the solvent is removed from the reaction mixture to form a residue, which is purified (e.g., using chromatography techniques) to yield S37.
  • S6, S46-53, S64, S66, and S67 are prepared from S37 by reaction with an amine of formula RNH 2 , wherein NR is NH 2 (S46), NEt 2 (S48), NHCH 2 Ph (S49), NHOH (S51),
  • reaction takes place in the presence of a solvent, such as an organic solvent, such as DMF.
  • a solvent such as an organic solvent, such as DMF.
  • organic solvent such as DMF.
  • only one equivalent of amine is used in the reaction. Purification is accomplished, for example, by SiO 2 column chromatography.
  • S6, S46-53, S64, S66, and S67 also are prepared from S26, via the S26-phosgene intermediate.
  • the S26-phosgene intermediate is formed by reacting S26 with triphosgene. Thereafter, the S26-phosgene is reacted with an amine of formula RNH 2 , where NR is NH 2
  • NRiR 2 is (S55), ⁇ £r o ( . anatomyS5.6,.), Q (S58), -CXJy ""'
  • reaction occurs in a solvent, such as an organic solvent, such as chloroform, thereby forming a reaction mixture.
  • a solvent such as an organic solvent, such as chloroform
  • the solvent is removed from the reaction mixture to form a residue, which is purified, for example, by silica gel column chromatography to yield the final product.
  • S69-75 are prepared from S68, via the S68-phosgene intermediate.
  • S68 is treated with triphosgene to form the intermediate, which in turn is treated with an amine RNH 2 ,
  • R is NH 2 (S70), NEt 2 (S75), NHOH (S74), (S69), (S71),
  • reaction occurs in a solvent, such as an organic solvent, such as chloroform, thereby forming a reaction mixture.
  • a solvent such as an organic solvent, such as chloroform
  • the solvent is removed from the reaction mixture to form a residue, which is purified, for example, by silica gel column chromatography to yield the final product.
  • S76 is prepared from S68 by reaction with methyl chlorooxoacetate.
  • the reaction occurs in the presence of a solvent, such as an organic solvent, such as methylene chloride.
  • a base catalyst such as pyridine is used as necessary to facilitate or hasten the reaction.
  • the reaction mixture formed by mixing the reactants and a solvent is washed with basic solution (e.g., saturated sodium bicarbonate), acidic solution (e.g. HCl), and water. Purification such as silic gel chromatography yields S76.
  • S77 is prepared from S76 by reaction with sodium hydroxide.
  • the reaction takes place in a solvent, such as an organic solvent, such as methanol.
  • the solvent is removed from the reaction mixture formed by mixing the reactants and the solvent, thereby forming a residue.
  • the residue is dissolved in water and washed with another organic solvent, such as ether, to remove unwanted hydrophobic compounds.
  • the aqueous phase from the basic washes is acidified and the product is extracted therefrom using an organic solvent, such as methylene chloride. Further purification is used if necessary.
  • S78-S81 are prepared by treating S77 with thionyl chloride to form crude S77-C1. Excess thionyl chloride, if any, is removed from the reaction mixture. The crude S77-C1 then is dissolved in a solvent, such as an organic solvent like methylene chloride, and reacted with HX, where X is NHEt (S78), NHPh (S79), NH 2 (S80), and NHCH 2 -pyridine (S81). The solvent is removed, and the residue is purified.
  • a solvent such as an organic solvent like methylene chloride
  • S82 is prepared from S68.
  • S68 is reacted with CH 2 CHSO 2 Cl in a manner analogous to the production of S3.
  • the product then is treated with HNRiR 2 in a manner analogous to
  • S83 is prepared from S68.
  • S68 is reacted with RCOCl, wherein R is
  • S84 is prepared from S68 by reaction with benzyl bromide.
  • the reaction takes place in a solvent, such as an organic solvent like methylene chloride.
  • a base catalyst such as triethylamine is added as necessary to catalyze the reaction.
  • the reaction mixture formed by mixing the reactants and the solvent is purified to yield S 84.
  • S85 is prepared from S26.
  • S26 is reacted with di-tert-butyl dicarbonate in a solvent, for example an organic solvent like methylene chloride.
  • a base catalyst such as triethylamine also is used, if necessary.
  • the reaction mixture formed by mixing the reactants and the solvent is washed with saturated sodium bicarbonate solution and the aqueous layer is extracted with an organic solvent. The combined organic layers are dried and concentrated provide S85.
  • S86 is prepared from S85 in a solvent, for example an organic solvent.
  • S85 is treated with BBr 3 to form a reaction mixture.
  • a base catalyst such as triethylamine
  • the reaction is quenched (e.g., in the case of triethylamine, with methanol) aid concentrated. Purification, for example by column chromatography, yields S86.
  • S87 is prepared by reacting S86 with trifluoromethylsulfonyl anhydride. The reaction is carried out in a solvent, such as an organic solvent.
  • a base catalyst such as triethylamine is added if necessary.
  • the reaction mixture formed by mixing the reactants and the solvent is quenched with water, after which the aqueous layer is extracted with an appropriate organic solvent. If desired, the organic layers are dried (e.g., using magnesium sulfate), and the organic layers are concentrated. Purification of the concentrated organic layers yields S87.
  • S88 is prepared from S87 by reaction of morpholine, tris(dibenzylideneacetone)dipalladium(0), 2-(di-tert-butylphosphino)-biphenyl, and potassium phosphate.
  • the reaction mixture is diluted with a solvent, such as methylene chloride or another appropriate organic solvent, and washed with water.
  • aqueous layer formed by washing with water, is extracted with an organic solvent, such as methylene chloride.
  • the organic layers then are dried (e.g., over magnesium sulfate) and concentrated.
  • the residue is purified, for example by silica gel flash chromatography, to yield S88.
  • S89 is prepared from S87 by reaction with benzenethiol and /-Pr 2 NEt in a solvent, such as CH 3 CN or another appropriate organic solvent. After reaction, an organic solvent such as ethyl acetate is added to the reaction mixture. If necessary, the reaction mixture is washed with one or more of acidic (e.g., HCl), basic (e.g. NaOH), and water solutions. After drying (e.g. with Na 2 SO 4 ), the solution is concentrated. Purification, for example by chromatography, yields S89. In an alternative, refluxing S87 with benezethiol in an appropriate solvent such as dioxane with a catalyst such as z-Pr 2 NEt/Pd 2 (dba) 3 /xantphos yields S89.
  • a solvent such as CH 3 CN or another appropriate organic solvent.
  • S90 is prepared from S87 reacted with a base, phenylboronic acid, and a catalyst.
  • the base is K 2 CO 3 and the catalyst is (Pd(Ph 3 P) 4 ).
  • the reaction occurs in a solvent, such as an organic solvent, such as dioxane.
  • the reaction mixture formed by mixing the reactants and the solvent is diluted with a solvent (e.g. methylene chloride), and washed with water to remove unwanted hydrophilic compounds. Concentration and purification of the residue yields S90.
  • a solvent e.g. methylene chloride
  • S92 is prepared from S87 reacted with zinc cyanide.
  • the reaction occurs in a solvent, such as an organic solvent like DMF.
  • a catalyst such as Pd(Ph 3 P) 4 also is used to facilitate and hasten the reaction.
  • the reaction mixture formed by mixing the reactants and the solvent, if necessary, is diluted with water and an acidic solution and extacted with an organic solvent.
  • the organic extracts then are washed using a salt solution, dred, filtered, and concentrated. Purification of the residue proceeds, for example, by silica gel column chromatography.
  • S94 is prepared for S86 by reaction with acetic anhydride.
  • the reaction takes place in a solvent, such as an organic solvent like methylene chloride.
  • Triethylamine or another base catalyst is added as necessary. Washing with water, followed by drying (e.g., using sodium sulfate), is used as desired. Purification of the residue yields S94.
  • S95 is prepared from S94 by reaction with anhydrous AlCl 3 , in a solvent if desired.
  • the solvent is an organic solvent like benzene.
  • the reaction mixture is refluxed and cooled on ice. Extraction with an organic solvent, concentration, and purification of the residue yields S95.
  • S96 is prepared from S86 by iodination.
  • S86 is added to a solvent, such as an organic solvent like methanol, with excess NaI and Chloramine-T.
  • the reaction mixture is quenched with Na 2 S 2 O 3 solution. Concentration and purification of the residue yields S96 as a mixture of mono-iodinated and di-iodinated products.
  • S97 is prepared from S86 by reaction with a nitric acid.
  • S86 is protected (e.g., using the Boc protecting groups) and added to concentrated sulfuric acid.
  • Nitric acid is added to the reaction mixture.
  • the reaction mixture is cooled and neutralized (e.g., using Na 2 CO 3 ) to quench the reaction. Organic extraction and subsequent concentration is used to isolate the product. Purification yields S 97.
  • S98 is prepared by hydrogenation of S97.
  • S97 is added to a solution, such as an organic solution like methanol.
  • H 2 gas is bubbled through the solution and Pd/C catalyst or another applicable catalyst is added. Filtration to remove the catalyst and purification yields S97.
  • SlOO is prepared from S98.
  • S98 is dissolved in acid solution, such as aqueous HCl.
  • acid solution such as aqueous HCl.
  • a solution of sodium nitrite, and then NaN 3 in water are added.
  • the reaction mixture is extracted using an organic solvent. If needed, the extract is washed with a basic solution (e.g., saturated sodium bicarbonate) and water.
  • Organic layers from the washing are dried using, for example, anhydrous sodium sulfate, and concentrated to form a residue. The residue is purified to yield SlOO.
  • S99 NaN 3 is substituted with NaBF 4 in a similar manner.
  • SlOl, S102, and S103 may each be prepared from S68.
  • SlOl may be prepared from S68 as follows. Triphosgene is reacted with S68 in the presence of a solvent (such as the organic solvent dichloromethane, CH 2 Cl 2 ) to generate S68- phosgene.
  • a base is also present or added to scavenge acid generated during the reacton. Any suitable base may be used.
  • organic bases such as organic amines like triethylamine, di-isopropylethylamine or pyridine may be used.
  • Inorganic bases such as as sodium bicarbonate may also be used. Then, without the need for purification, the reaction mixture containing the S68-phosgene is treated with 1 -piperonylpiperazine.
  • reaction mixture is washed with one or more of acidic (e.g., HCl), basic (e.g. NaOH), and water solutions.
  • acidic e.g., HCl
  • basic e.g. NaOH
  • S 102 maybe prepared from S68 using the same scheme as for SlOl, with the exception that piperidine is used in the place of piperonylpiperazine.
  • S103 may be prepared from S68 using the same scheme as for SlOl, with the exception that N-Boc 1-piperazine is used in the place of piperonylpiperazine. Also, trifiuoroacetic acid (TFA) is added de-protect the Boc group.
  • TFA trifiuoroacetic acid
  • S 104 may be prepared by reacting S36 with hydrogen peroxide (H 2 O 2 ) in the presence of a solvent (such as MeOH). The solvents are removed (for example under reduced pressure), and the S 104 product can then be purified, for example by re-crystallization.
  • a solvent such as MeOH
  • S 105 may be prepared from S68 as follows. S68 is be reacted with CH 3 O- C(O)C(O)Cl in the presence of a solvent (such as the organic solvent dichloromethane (CH 2 Cl 2 )) and optionally a catalyst (such as pyridine). Preferably, the CH 3 O-C(O)C(O)Cl should be added dropwise. If necessary, the reaction mixture is washed with one or more of acidic (for example HCl), basic (for example NaOH), and water solutions. The solvents are removed and the product may be further purified, for example by SiO 2 column chromatography.
  • a solvent such as the organic solvent dichloromethane (CH 2 Cl 2 )
  • a catalyst such as pyridine
  • the CH 3 O-C(O)C(O)Cl should be added dropwise. If necessary, the reaction mixture is washed with one or more of acidic (for example HCl), basic (for example NaOH), and water solutions. The solvents are removed and
  • S 107 may be prepared from S26 as follows. To a solution of S26 in a solvent (such as MeOH), formaldehyde (CH 2 O) and sodium cyanoborohydride (NaBCNH 3 ) are added and allowed to react. Preferably, the reaction mixture is maintained at around pH 4-5, for example by addition of a few drops of IN HCl. The solvents are then removed, for example under reduced pressure. If necessary, the residue may be dissolved in ethyl acetate and washed with with one or more of a basic solution (for example NaOH), and water. The solvents may be removed, and the product may be further purified, for example using SiO 2 column chromatography.
  • a solvent such as MeOH
  • formaldehyde CH 2 O
  • NaBCNH 3 sodium cyanoborohydride
  • S 108 may be prepared as follows. A mixture of N-benzyloxycarbonyl-glycine (Cbz- GIy,), Diisopropyl-carbodiimide (DIC), and N-hydroxysuccinimide (NHS), are reacted together in a solvent (such as the organic solvent dichloromethane (CH 2 Cl 2 )) for a suitable amount of time. S26 is then added to the mixture and the reaction is allowed to proceed further. If necessary, the reaction mixture is washed with one or more of acidic (for example HCl), basic (for example NaOH), and water solutions. The solvents may then be removed, for example by evaporation. The product may be further purified, for example using SiO 2 column chromatography.
  • a solvent such as the organic solvent dichloromethane (CH 2 Cl 2 )
  • S109 maybe prepared from S108, as follows. S108 in a solvent (such as the organic solvent dichloromethane (CH 2 Cl 2 )) is reacted with HBrZCH 3 CO 2 H. After a suitable amount of time, the reaction mixture is evaporated, for example under reduced pressure. The residue is dissolved in a suitable solvent, such as MeOH, and is treated with propylene oxide. The solvent may then be removed, for example under reduced pressure, to provide crude S 109. The SlOO may be further purified, for example by dissolving in an acidic solution (such as HCl), washing with ethyl acetate, and evaporating.
  • a solvent such as the organic solvent dichloromethane (CH 2 Cl 2 )
  • SIlO may be prepared as follows. A mixture of S26, methyl 1 -bromoacetate and pyridine are reacted in DMF for a suitable amount of time. To this mixture, ethyl aceate is added, and if necessary, the reaction mixture is washed with a basic solution (for example NaHCO 3 ), or water. The product S 110, as an oil, may be purified, for example by SiO 2 column chromatography.
  • Sill may be prepared as follows. A base (such as IN NaOH) is added to Sl 10 in a solvent (such as MeOH), and the mixture is allowed to react for a suitable amount of time. The solvents are then removed, for example under reduced pressure, and the residue may then be dissolved in an aqueous solution, such as water. The aqueous phase may be washed with ethyl acetate and acidified, for example with IN HCl, to pH of around 4. The solvents may then be removed, for example under reduced pressure, to produce crude Si l l. The NaCl may be removed using an alcohol, such as ethanol, to yield pure Sil l as a solid.
  • a base such as IN NaOH
  • MeOH a solvent
  • the solvents may then be removed, for example under reduced pressure, to produce crude Si l l.
  • the NaCl may be removed using an alcohol, such as ethanol, to yield pure Sil l as a solid.
  • Sl 12 maybe prepared as follows: To a mixture of S26 and pyridine in an solvent (such as the organic solvent dichloromethane (CH 2 Cl 2 )), SO 2 Cl 2 is added drop-wise at around O 0 C and reacted for a suitable amount of time. The solvents may be removed, for example under reduced pressure. The residue can be dissolved in a suitable basic solution, such as NaOH. The aqueous solution may then be washed with ethyl acetate, and acidified (for example with IN HCl) to around pH 4. The aqueous phase may be extracted again with ethyl acetate, and the ethyl acetate phase evaporated, for example under reduced pressure, to provide Sl 12, as a powder.
  • an solvent such as the organic solvent dichloromethane (CH 2 Cl 2 )
  • SO 2 Cl 2 is added drop-wise at around O 0 C and reacted for a suitable amount of time.
  • the solvents may be removed, for example under reduced pressure.
  • S 113 may be prepared as follows. S 107 in ethyl acetate is treated with CH 3 I. The mixture is stirred for a suitable amount of time, and the product Sl 13, as white solid, is collected by filtration.
  • Sl 14 maybe prepared as follows.
  • the compound S26, in a solvent such as the organic solvent CH 2 Cl 2 is ideally cooled to around O 0 C.
  • triphosgene is added.
  • a base is also present or added to scavenge acid generated during the reacton.
  • Any suitable base may be used.
  • organic bases such as organic amines like triethylamine, di-isopropylethylamine or pyridine may be used.
  • Inorganic bases such as as sodium bicarbonate may also be used.
  • the reaction is allowed to proceed (ideally around O 0 C) for a suitable amount of time (for example about 1 hour).
  • the resulting S26-phosgene in the reaction mixture may then be treated with N- Boc 1 -piperazine, again ideally at around O 0 C, and the reaction is allowed to proceed (ideally at around 0 0 C) for a suitable amount of time (for example about 1 hour). If necessary, the reaction mixture is washed with one or more of acidic (for example HCl), basic (for example NaOH), and water solutions. The solvents are removed and the product may be further purified, for example by SiO 2 column chromatography.
  • acidic for example HCl
  • basic for example NaOH
  • Sl 15 may be prepared as follows. A mixture of Sl 14 and Lawesson Reagent in toluene is stirred at around 9O 0 C for several hours. The mixture is cooled to room temperature and washed with a suitable base, such as saturated NaHCO 3 . The product SI l 5_may be purified, for example by SiO 2 chromatography.
  • Sl 16 may be prepared as follows. A mixture of Sl 15 and trifluoroacetic acid (TFA) in a suitable solvent solvent (such as the organic solvent dichloromethane (CH 2 Cl 2 )) is stirred at around room temperature for a suitable amount of time (for example, about 2 hours). Evaporation of the solvents, for example under reduced pressure, produces Sl 16.
  • a suitable solvent solvent such as the organic solvent dichloromethane (CH 2 Cl 2 )
  • S 117 may be prepared as follows. A solution of S057 in a suitable solvent (such as the organic solvent dichloromethane (CH 2 Cl 2 )) is cooled to about -78 0 C. To this, IM BBr 3 a suitable solvent (such as the organic solvent dichloromethane (CH 2 Cl 2 )) is added, and the mixture is stirred (still at about 78 0 C) for a suitable amount of time (for example around 3 hours) and then wSed to room temperature. If necessary, the mixture is washed with an acid (such as IN HCl) and/or H 2 O. After removal of the solvents, the product Sl 17 may be purified, for example by SiO 2 column chromatography.
  • a suitable solvent such as the organic solvent dichloromethane (CH 2 Cl 2 )
  • Sl 18 maybe synthesized as follows. S26 in as suitable solvent a suitable solvent (such as the organic solvent dichloromethane (CH 2 Cl 2 )) is treated with BODIPY TMR-X, SE (Molecular Probes Inc.) for a suitable amount of time (for example, around 3 hours). If necessary, the mixture may be washed with an acid (such as 0.01 N HCl) and/or a base (such as NaHCO 3 ). Removal of the solvents, for example under reduced pressure, will yield Sl 18.
  • a suitable solvent such as the organic solvent dichloromethane (CH 2 Cl 2 )
  • BODIPY TMR-X Molecular Probes Inc.
  • Sl 19 may be synthesized as follows. A mixture S 107, H 2 O 2 (for example around 50%), and an alcohol (such as MeOH), is stirred at around room temperature for a suitable amount of time (typically around 2 days). If desired, mass spectrometry may be used to monitor the disappearance of S107 and the formation of the Sl 19 product). The solvents may be removed, for example under reduced pressure, to give Sl 19.
  • S 120 maybe synthesized as follows. A mixture S26, benzyl bromide and Na 2 CO 3 in a solvent (such as DMF), is reacted for a suitable amount of time, preferably overnight. Ethyl acetate is added to the reaction, and then, if necessary, the reaction is washed with a suitable solvent, for example with H 2 O (4x10 ml). The organic phase may be concentrated, for example under reduced pressure, and the residue may be purified, for example by column chromatography to give S 121.
  • a solvent such as DMF
  • S121 may be synthesized as for S120, but using 4-OH-benzyl bromide instead of benzyl bromide.
  • S122 may be synthesized as follows. To a cold solution of compound S26 in a solvent, such as the organic solvent in CH 2 Cl 2 , DIEA is added, and subsequently acetoxyacetyl chloride is added. The reaction is allowed to proceed for a suitable amount of time, and then diluted, (for example with 1.0 AfHCl aqueous solution) and extracted (for example using CH 2 Cl 2 ). The combined organic layers may, if necessary, be washed (for example with H 2 O, brine), dried (for example with Na 2 SO 4 ), filtered, and dried (for example by evaporation).
  • a solvent such as the organic solvent in CH 2 Cl 2
  • the product maybe further purified, for example by chromatography on a silica gel column, and may be eluted with a gradient increasing in polarity from 0 to 50% of petroleum in ethyl acetate. Relevant fractions may then be combined to give the desired product.
  • S 123 maybe synthesized as follows. To a solution of compound S 122 in a solvent (such as MeOH) and THF, preferably at room temperature, is added LiOH. The reaction is allowed to proceed for a suitable amount of time at a suitable temperature (ideally around room temperature, and may then be diluted (for example with with 1.0 MHCl aqueous solution) and extracted (for example with CH 2 Cl 2 ). The combined organic layers may be washed (for example with H 2 O, brine), dried (for example with Na 2 SO 4 ), filtered and dried (for example by evaporation).
  • a solvent such as MeOH
  • THF preferably at room temperature
  • the crude product may be purified, for example by chromatography on a silica gel column, eluted, for example with a gradient increasing in polarity from 0 to 70% petroleum in ethyl acetate. Relevant fractions may then beO combined to give S 123.
  • the compounds used as starting materials for, or generated as intermediates in, the synthesis of the compounds of the invention may themselves have structures encompassed by the formulae of the invention, and/or may themselves be active agents useful in the methods and compositions of the present invention.
  • Such starting materials and intermediates may be useful for, inter alia, treating or preventing various disorders and diseases associated with RyR receptors such as muscular and cardiac disorders, treating or preventing a leak in a RyR2 receptor in a subject, or modulating the binding of RyR and FKBP in a subject.
  • the present invention encompasses any of the starting materials or intermediates disclosed herein that have structures encompassed by Fo ⁇ nula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, and/or which are useful as active agents in the methods and compositions of the present invention.
  • the compound S68 which is useful as a starting material for the synthesis of compounds S69 - S75, maybe used for, inter alia, treating or preventing various disorders and diseases associated with RyR receptors, treating or preventing a leak in a RyR2 receptor, or modulating the binding of RyR and FKBP in a subject.
  • the compound S26 which is useful in the synthesis of many of the compounds described herein (including S3, S4, S5, S7, S9, Sl 1, S12, S13, S14 and other compounds) may be used for, inter alia, treating or preventing various disorders and diseases associated with RyR receptors, treating or preventing a leak in a RyR2 receptor, or modulating the binding of RyR and FKBP in a subject.
  • the compound S25 may also be used for, inter alia, treating or preventing various disorders and diseases associated with RyR receptors, treating or preventing a leak in a RyR2 receptor, or modulating the binding of RyR and FICBP in a subject.
  • the compounds of the present invention are prepared in different forms, such as salts, hydrates, solvates, complexes, pro-drugs or salts of pro-drugs and the invention includes all variant forms of the compounds.
  • compound(s) of the invention means a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, and salts, hydrates, prodrugs and solvates thereof.
  • a "pharmaceutical composition” refers to a mixture of one or more of the compounds described herein, or pharmaceutically acceptable salts, hydrates or pro-drugs thereof, with other chemical components, such as physiologically acceptable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • a "pro-drug” refers to an agent which is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. They are bioavailable, for instance, by oral administration whereas the parent drug is not. The pro-drug also has improved solubility in pharmaceutical compositions over the parent drug.
  • the compound carries protective groups which are split off by hydrolysis in body fluids, e.g., in the bloodstream, thus releasing active compound or is oxidized or reduced in body fluids to release the compound.
  • a compound of the present invention also can be formulated as a pharmaceutically acceptable salt, e.g., acid addition salt, and complexes thereof.
  • a pharmaceutically acceptable salt e.g., acid addition salt, and complexes thereof.
  • the preparation of such salts can facilitate the pharmacological use by altering the physical characteristics of the agent without preventing its physiological effect. Examples of useful alterations in physical properties include, but are not limited to, lowering the melting point to facilitate transmucosal administration and increasing the solubility to facilitate administering higher concentrations of the drug.
  • pharmaceutically acceptable salt means an acid addition salt which is suitable for or compatible with the treatment of a patient or a subject such as a human patient or an animal such as a dog.
  • the te ⁇ n "pharmaceutically acceptable acid addition salt” as used herein means any non-toxic organic or inorganic salt of any base compounds represented by Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or any of their intermediates.
  • Illustrative inorganic acids which form suitable acid addition salts include hydrochloric, hydrobromic, sulfuric and phosphoric acids, as well as metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate.
  • Illustrative organic acids that form suitable acid addition salts include mono-, di-, and tricarboxylic acids such as glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic and salicylic acids, as well as sulfonic acids such as p-toluene sulfonic and methanesulfonic acids.
  • Either mono or di-acid salts can be formed, and such salts exist in either a hydrated, solvated or substantially anhydrous form.
  • the acid addition salts of compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms.
  • the selection of an appropriate salt will be known to one skilled in the art.
  • Other non-pharmaceutically acceptable salts, e.g., oxalates are used, for example, in the isolation of compounds of the invention for laboratory use or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • the compounds of the present invention form hydrates or solvates, which are included in the scope of the claims.
  • the compounds of the present invention exist as regioisomers, configurational isomers, conformers or diasteroisomeric forms all such forms and various mixtures thereof are included in the scope of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m. It is possible to isolate individual isomers using known separation and purification methods, if desired. For example, when a compound of the present invention is a racemate, the racemate can be separated into the (S)-compound and (R)-compound by optical resolution.
  • solvate means a compound of Formula I, I-a, I-b, I-c, I-d, I- e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or a pharmaceutically acceptable salt thereof, wherein molecules of a suitable solvent are incorporated in the crystal lattice.
  • a suitable solvent is physiologically tolerable at the dosage administered. Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a "hydrate.”
  • an "effective amount,” “sufficient amount” or “therapeutically effective amount” of an agent as used herein is that amount sufficient to effect beneficial or desired results, including clinical results and, as such, an “effective amount” depends upon the context in which it is being applied. The response is preventative and/or therapeutic.
  • the term “effective amount” also includes that amount of the compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, which is “therapeutically effective” and which avoids or substantially attenuates undesirable side effects.
  • treatment is an approach for obtaining beneficial or desired results, including clinical results.
  • beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized ⁇ i.e., not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • animal refers to all members of the animal kingdom including, but not limited to, mammals, animals ⁇ e.g., cats, dogs, horses, etc.) and humans.
  • the present invention further provides a composition, comprising radio labeled compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m. Labeling of the compounds is accomplished using one of a variety of different radioactive labels known in the art.
  • the radioactive label of the present invention is, for example, a radioisotope.
  • the radioisotope is any isotope that emits detectable radiation including, without limitation, 35 S, 125 1, 3 H, or 14 C. Radioactivity emitted by the radioisotope can be detected by techniques well known in the art. For example, gamma emission from the radioisotope is detected using gamma imaging techniques, particularly scintigraphic imaging.
  • radio-labeled compounds of the invention are prepared as follows.
  • a compound of the invention may be demethylated at the phenyl ring using BBr 3 .
  • the resulting phenol compound then is re-methylated with a radio-labeled methylating agent (such as 3 H-dimethyl sulfate) in the presence of a base (such as NaH) to provide 3 H-labeled compounds.
  • a radio-labeled methylating agent such as 3 H-dimethyl sulfate
  • a base such as NaH
  • the present invention further provides compounds that may be classified as 1,4- benzothiazepines, including, by way of example and without limitation, Sl, S2, S3, S4, S5, S6, S7, S9, SI l, S12, S13, S14, S19, S20, S22, S23, S25, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S
  • the decrease in the level of RyR-bound FKBP is limited or prevented in the subject by decreasing the level of phosphorylated RyR in the subject.
  • the amount of the agent effective to limit or prevent a decrease in the level of RyR2-bound FKBP12.6 in the subject is an amount of the agent effective to treat or prevent heart failure, atrial fibrillation and/or exercise- induced cardiac arrhythmia in the subject.
  • the amount of the agent effective to limit or prevent a decrease in the level of RyR2-bound FKBP 12.6 in the subject is an amount of the agent effective to prevent exercise-induced sudden cardiac death in the subject.
  • the present invention further provides a method for treating or preventing exercise-induced cardiac arrhythmia in a subject, comprising administering to the subject a 1,4-benzothiazepine compound, as disclosed herein, in an amount effective to treat or prevent exercise-induced cardiac arrhythmia in the subject.
  • the present invention provides a method for preventing exercise-induced sudden cardiac death in a subject, comprising administering to the subject a 1,4-benzothiazepine compound, as disclosed herein, in an amount effective to prevent exercise-induced sudden cardiac death in the subject.
  • the present invention provides a method for treating or preventing atrial fibrillation or heart failure in a subject, comprising administering to the subject a compound, as disclosed herein, in an amount effective to treat or prevent the atrial fibrillation or heart failure in the subject.
  • the compound is selected from the group of compounds consisting of compounds of the formula:
  • n 0, 1, or 2;
  • R is located at one or more positions on the benzene ring; each R is independently selected from the group consisting of H, halogen, -OH, -NH 2 , -NO 2 , -CN, -N 3 , -SO 3 H, acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN
  • Ri is selected from the group consisting of H, oxo, alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl maybe substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
  • R 3 is selected from the group consisting of H , CO 2 Y, CONY, acyl, alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N 3 O, -S-, -CN 3 -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl; and wherein Y is selected from the group consisting of H, alkyl, aryl, cycloalkyl, and heterocyclyl;
  • R 4 is selected from the group consisting of H, alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
  • R 5 is selected from the group consisting of -NR 165 NHNHR 16 , NHOH, -OR 15 , CONH 2 NHRi 6 , CO 2 R 15 , CONR 16 , CH 2 X, acyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
  • Rn is selected from the group consisting Of-OR 1S5 NHNRi 6 , NHOH, -NR 16 , CH 2 X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycl ⁇ alkyl, and (hetero-)cyclyl;
  • R 7 is selected from the group consisting Of-ORi 5 , -NR 165 NHNHR 16 , NHOH, CH 2 X, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
  • R 8 and R 9 independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl; R 10 is selected from the group consisting of NH 2 , OH
  • R n , Ri 2 , R 13 , and Ri 4 independently are selected from the group consisting of H, OH, NH 2 , NHNH 2 , NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted with one or more radicals independently selected from the group consisting of halogen, -N-, -0-, - S-, -CN, -N 3 , nitro, oxo, acyl, alkenyl, alkoxyl, alkyl, alkylamino, amino, aryl, cycloalkyl
  • X is selected from the group consisting of halogen, CN, CO 2 R 15 , CONRi 6 , -NR 16 , -OR 1 S 7 - SO 2 R 7 , and -POR 8 R 9 ;
  • Ri5 and R 16 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, -N-, -0-, -S-, -CN, -N 3 , nitro, oxo, acyl, alkenyl, alkoxyl, alkyl, alkylamino, amino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, and hydroxyl
  • Examples of such compounds include, without limitation, Sl, S2, S3, S4, S5, S6, S7, S9, SIl, S12, S13, S14, S19, S20, S22, S23, S25, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S
  • each R is independently selected from the group consisting of H, halogen, -OH, -NH 2 , -NO 2 , -CN, -N 3 , -SO 3 H, acyl, alkyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero- )aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero- )arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N
  • each R is independently selected from the group consisting of H, halogen, -OH, -NH 2 , -NO 2 , -CN, -N 3 , -SO 3 H, acyl, alkyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero- )arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , -SH, nitro, oxo,
  • R 5 is selected from the group consisting of -NR 16 , NHNHR 16 , NHOH 5 -ORi 5 , CONH 2 NHRi 6 , CONR] 65 CH 2 X, acyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N 3 , nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl
  • S36 is more potent at increasing the binding of FKBP12.6 and RyR2 than JTV-519 and does not block the L-type Ca2+ channel (Ic a ,L) or HERG K + channel (I ⁇ r )-
  • PKA phosphorylated RyR2 is generated as follows: cardiac SR membrane preparations (5 ⁇ l, 50 ⁇ g) are added to a total of 100 ⁇ l of kinase buffer (8 mM MgCl 2 , 10 mM EGTA, 50 rnM Tris-PIPES, pH 6.8) containing 100 ⁇ M MgATP and 40 units of PICA, and incubated at room temperature.
  • Immunoblots are developed using an anti-FKBP antibody (1 :3,000 dilution). The amount of rebinding is quantified using densitometry of Western blots and is compared to the amount of FKBP associated with RyR in non-phosphorylated SR. ECso's for the compounds are determined by generating FKBP binding data using concentrations of compounds ranging from 0.5-1000 nM.
  • currents through L-type Ca 2+ channels in isolated mouse cardiomyocytes are recorded using whole-cell patch clamp recording conditions with Ba 2+ as the charge carrier.
  • the extracellular solution contains (in mM): N-methyl-D-glucamine, 125; BaCl 2 , 20; CsCl, 5; MgCl 2 , 1; HEPES, 10; glucose, 5; pH 7.4 (HCl).
  • the intracellular solution contains (in mM): CsCl, 60; CaCl 2 , 1; EGTA, 11; MgCl 2 , 1; K 2 ATP, 5; HEPES, 10; aspartic acid, 50; pH 7.4 (CsOH).
  • L - Drugs are applied by a local solution changer and reach the cell membrane within 1 s.
  • the effects of nifedipine and S36 are tested with 20 ms long voltage-clamp steps to +10 or +20 mV (peak of current- voltage relation for each individual cell) from holding potentials of -80 mV or -40 mV.
  • the voltage-dependence of L-type Ca 2+ current blocked by JTV-519 (1 ⁇ M) and S36 (l ⁇ M) are measured and presented.
  • embodiments A, B, C, and D S36 prevents exercise- induced sudden cardiac death at lower plasma levels compared with JTV-519.
  • embodiment A are shown representative ECGs of an untreated FKBP12.6 +/ ⁇ mouse and JTV519-treated FICBP12.6 +/" and FKBP12.6 "7" mice. Mice are treated with 0.5 mg JTV- 519/per kilogram of body weight per hour for 7 days with an implanted osmotic mini-pump. JTV-519 has no effect on resting heart rate or other ECG parameters such as heart rate (HR).
  • HR heart rate
  • embodiment B sustained polymorphic ventricular tachycardia recorded by telemetry in an untreated FKBP12.6 +/ mouse (upper tracing) subjected to exercise testing, immediately followed by injection with 0.5 mg epinephrine per kilogram of body weight.
  • Representative telemetry ECG recording of a JTV-519-treated FKBP12.6 +/ ⁇ mouse following the same protocol is shown in the bottom tracing
  • hi embodiment C are shown numbers of mice with cardiac death (left), sustained VTs (>10 beats, middle), and nonsustained VTs (3 to 10 arrhythmogenic beats, right) in experimental groups of mice subjected to exercise testing and injection with 0.5 mg/kg epinephrine.
  • embodiment D the dose-dependence of pharmacological effects of JTV-519 and S36 is shown.
  • Plasma levels of 1 ⁇ M JTV519 prevent cardiac arrhythmias and sudden cardiac death in FKBPl 2.6 +/ ⁇ mice.
  • Plasma levels of 1 ⁇ M and 0.02 ⁇ M S36 also prevent cardiac arrhythmias and sudden cardiac death in FKBP12.6 +/" mice.
  • S36 prevents the development of acute heart failure post-myocardial infarction.
  • Placebo-treated or S36 (100 nM or 200 nM plasma concentrations)-treated mice are subjected to permanent ligation of the left anterior descending coronary artery resulting in myocardial infarction.
  • S36 significantly improves fractional shortening assessed by M-mode echocardiography 2 weeks post-myocardial infarction, compared with placebo.
  • S36 improves cardiac function in chronic heart failure post-myocardial infarction.
  • Wild-type mice are subjected to permanent ligation of the left anterior descending coronary artery resulting in myocardial infarction.
  • Seven days following myocardial infartion mice are treated with S36 (plasma concentration 200 nM) or placebo.
  • Heart weight to body weight (HW/BW) ratios and pressure-volume loops quantifications (dP/dt, slope of the maximum derivative of change in systolic pressure over time) show reverse remodeling and improved cardiac contractility in S36-treated mice compared with placebo.
  • Figure 5 is a summary graph of EC50 values of JTV-519 and compounds S1-S67 disclosed herein.
  • the FKBP12.6 rebinding assay described above is used to determine the amount of FKBP12.6 binding to PKA-phosphorylated RyR2 at various concentrations (0.5 - 1000 nM) of the compounds shown.
  • EC 50 values are calculated using Michaelis-Menten curve fitting.
  • embodiments A, B, and C S36 normalizes CPVT- associated RyR2-P2328S channel function and structure.
  • embodiment A are shown representative single-channel current traces of vmphosphorylated RyR2-P2328S and PKA- phosphorylated RyR2-WT treated with S36 showing no influence of JTV-519 on baseline channel function.
  • PKA phosphorylated RyR2-P2328S as shown in embodiment B, S36 normalizes the single channel closed state to levels approaching those seen in embodiment A, reducing the open probability from 14.4% to 0.3% following administration of 0.1 ⁇ mol/L S36.
  • embodiments A and B show channel openings >1 pA at a higher resolution.
  • Embodiment C shows immunoblot analysis of calstabin-2 binding of RyR2- P2328S in the presence or absence of PKA and 0.1 ⁇ mol/L S36, as indicated.
  • RyR2-P2328S is imrnunoprecipitated and in vitro PKA was phosphorylated as described above.
  • embodiments A and B mice in which cardiac RyR2 cannot be PKA phosphorylated (RyR2-S2808A knockin mice) have improved cardiac function following myocardial infarction.
  • Shown in embodiment A is the quantification of M-mode echocardiograms showing improved ejection fraction in RyR2-S2808A knockin mice compared with wildtype 28 days following permanent coronary artery ligation.
  • Shown in embodiments B and C are pressure-volume loop quantifications showing (embodiment B) improved cardiac contractility and decreased cardiac dilation (embodiment C) in RyR2- S2808A knockin mice compared with wildtype following myocardial infarction.
  • Figure 9 embodiments A, B, C, and D demonstrate the effect of JTV-519 on calstabin2 affinity to RyR2 in haploinsufficient calstabin2 (FKBPl 2.6) +/ ⁇ mice after exercise.
  • the average probability that RyR2 channels will be open in calstabin2 +/ ⁇ mice subjected to exercise were significantly increased compared to those of channels from exercised wild type (control; calstabin2 +/+ ) mice, which are predominately closed under conditions that simulate diastole in the heart.
  • JTV-519 increases the binding affinity of calstabin2 to RyR2.
  • JTV-519 treatment of exercised Calstabin2 ⁇ ⁇ deficient mice did not result in channels with a low P 0 , indicating that the presence of calstabir ⁇ is necessary for JTV-519 effects which are documented as binding of calstabin2 to RyR2.
  • FIG 10 embodiments A, B, C, D, E, and F demonstrate, respectively, normalized RyR2 channel gating and increased calstabin2 binding to RyR2 channels after treatment with JTV-519.
  • the immunoblots in embodiments A and B show the amounts of RyR2 and calstabin2 associated with immunoprecipitated RyR2 after incubation with the indicated concentrations of JTV-519 for, respectively, wild-type RyR2 (RyR2-WT) channels and RyR2-S2809D.
  • the binding curves in embodiment C demonstrate that JTV-519 significantly increases the affinity of calstabin2 for PKA-phosphorylated RyR2 channels.
  • embodiments A, B, C, D, and E RyRl channel functioning is increased and normalized in mdx (dystrophin deficient) mice treated with JTV-519.
  • channel openings are represented as upward deflections; 'c' indicates the closed state; and 4 pA current open amplitude is indicated by dash.
  • Upper traces represent 5 sees and lower traces 500 ms; dotted lines indicate subconductance states.
  • Embodiment A of Figure 11 shows a single channel current trace of RyRl from soleus muscle of control (wild-type) mouse under resting conditions (cytoplasmic Ca 2+ 150 nM). As seen, RyRl is predominantly closed.
  • Embodiment C of Figure 11 shows that RyRl channel function in mdx mouse shows significantly increased open probability, increased average open, and decreased average closed dwell times, To and Tc, respectively. Increased Po in mdx mice is consistent with intracellular Ca 2+ leak.
  • the amplitude histograms in embodiments B, D, and F show multiple subconductance states consistent with calstabinl (FKBP 12) depletion in RyRl from mdx soleus muscle.
  • Embodiment E of Figure 11 shows an mdx mouse treated with 1.0 ⁇ M JTV-519.
  • the RyRl channels JTV-519-treated mouse demonstrates normal activity that is not significantly different from untreated wild- type trace, thereby indicating that JTV-519 can normalize RyRl channel function m mdx mice.
  • FIG. 12 embodiments A and B, demonstrate that mdx skeletal muscle has normal levels of RyRl PKA phosphorylation, but depleted levels of calstabml.
  • the imrnunoblots in embodiment A show that mdx type mice have depeleted levels of calstabml compared to a control (wild-type) mouse
  • the summary bar graphs of embodiment B show that the mdx mouse, nevertheless, has an equivalent level of PKA-phosphorylation. Therefore, it is concluded that calstabml depeletion is a defect that is consistent with the intracellular Ca 2+ leak observed in skeletal muscle cells from mdx mice and myofibers from human mutation carriers. Intracellular SR Ca 2+ leak is likely to contribute to myofiber death and wasting of muscle mass by toxic intracellular Ca 2+ overload and activation of proteases.
  • FIG. 13 Figure 13, embodiments A, B, and C, demonstrates that SR Ca 2+ leak at the subcellular level m skeletal muscles of animals with heart failure is detectable.
  • Life quality and prognosis in heart failure (HF) patients is severely decreased due to skeletal muscle dysfunction (e g , shortness of breath due to diaphragmatic weakness, and exercise intolerance due to limb skeletal muscle fatigue) in addition to depressed cardiac function.
  • Dysregulation of intracellular SR Ca 2+ release is a pathogenic mechanism underlying skeletal muscle dysfunction m HF HF in animals causes significantly accelerates intrinsic skeletal muscle fatigue.
  • Embodiments A and B of Figure 13 are ⁇ F/F fluorescence line scan images of representative examples of Ca 2+ sparks in myofibers from sham and postmyocardial infarction (PMI) rats and corresponding Ca 2+ spark time course.
  • Embodiment C shows the relative distribution of the spatio-temporal properties of the Ca 2+ sparks Charts indicate 25, 50, 75 percentiles, the horizontal lines indicate the range from 1-99% of the distribution.
  • FDHM full duration at 50% peak amplitude; FWHM, full width at 50% peak amplitude.
  • Figure 14 embodiments A and B, demonstrate that treatment of wild-type mice with JTV-519 improved soleus muscle fatigue times compared with placebo. Soleus muscles from JTV519-treated wild-type mice or calstabin2 mice with heart failure from myocardial infarction are more resistant to fatigue (P ⁇ 0.05) compared to placebo-treated mice. After completion of treatment, soleus muscle was dissected and mounted in a tissue bath to assess isolated skeletal muscle function. Representative 50% of maximal tetanic force fatigue time tracings are shown for wild-type and calstabin2 ⁇ mice, treated with JTV519 or placebo, in embodiment A. In embodiment B are shown bar graph summarizing mean time to fatigue.
  • JTV-519 treatment improved skeletal muscle fatigability in heart failure animals in vivo.
  • calstabin ⁇ 1* knockout mice fatigue times were also significantly improved in JTV519 treated mice, suggesting that the beneficial effects on isolated skeletal muscle function depend on calstabinl and not calstabin2 binding to RyRl.
  • calstabinl appears to be the only isoform of functional significance expressed in skeletal muscles.
  • FIG 15, embodiments A and B demonstrates that in a mouse model of post- myocardial infarction heart failure, RyRl in soleus muscle is also PKA-hyperphosphorylated.
  • JTV-519 increased binding of calstabinl to RyRl in soleus muscle, suggesting that JTV-519 improves skeletal muscle fatigability by normalizing calstabinl rebinding to the channel complex.
  • Equivalent amounts of RyRl were immunoprecipitated with an antibody against RyRl .
  • Embodiment A provides bar graphs show the amount of PKA phosphorylation of RyRl at Ser-2844 in mice (corresponding to human Ser-2843).
  • Embodiment B are bar graphs showing the amount of calstabinl bound to RyRl from wild-type or calstabin2 ⁇ ' ⁇ mice treated with JTV519 or placebo. Mice were treated with JTV519 by implantable osmotic minipumps using a dose of 0.5 mg/kg/day. In summary, JTV-519 treatment resulted in a highly significant increase of calstabinl in the RyRl complex in soleus muscles in vivo.
  • FIG. 16 Figure 16, embodiments A, B, C, and D demonstrates that calstabinl rebinding to RyRl by JTV519 normalizes abnormal or leaky RyRl channel function in vivo.
  • embodiments A and C are shown RyRl single-channel tracings at 150 nM cytoplasmic Ca 2+ representing resting conditions in skeletal muscle for wild-type mice treated with placebo and JTV-519.
  • Channel openings are upward, the dash indicates the full level of channel opening (4 pA), the dotted lines indicate subconductance levels, and 'c' indicates the closed state of the channels.
  • amplitude is represented on the x axis, and events indicates the number of channel openings.
  • Po, To and Tc values correspond to the representative traces. Treatment as indicated on top of traces. Inset shows higher resolution of open states.
  • FIG. 17 demonstrate that JTV-519 also increases the binding affinity of calstabinl for RyRl in skeletal muscle in vivo. This probably explains why JTV519 treated mice with heart failure have increased levels of calstabinl bound to RyRl in soleus muscle.
  • equivalent amounts of skeletal RyRl or cardiac RyR2 were immunoprecipitated, PKA phosphorylated and incubated with calstabinl or calstabin2 at increasing concentrations of JTV519, respectively. Sediment represents only calstabin bound to RyR. Immunoblotting showed that >50 nM of JTV519 increased the binding affinity of calstabin for RyR.
  • the graphs of embodiment B further demonstrate that PKA phosphorylation of RyRl reduced the affinity of calstabinl for RyRl (open circles), whereas treatment with JTV519 (filled circles) restored the binding affinity of calstabinl for RyRl to that of non-PKA phosphorylated RyRl (open squares).
  • FIG. 18 Figure 18, embodiment A, B, C, and D demonstrates that Ser-2843 is the unique PKA phosphorylation site in skeletal RyRl channels.
  • A Representative single channel traces of wild-type RyRl
  • B effect of exogenous PICA phosphorylation of RyRl (wt RyRl-P)
  • C PKA does not affect RyRl-S2843A that contains a non-functional PKA phosphorylation site. Since PKA does not increase RyRl-S2843A activity, Ser-2843 appears to constitute the unique PICA phosphorylation site in RyRl channels in skeletal muscle.
  • (D) constitutively phosphorylated RyRl-S2843D mimics exogenous PKA phosphorylation shown in (B) confirming that Ser-2843 is the unique PKA phosphorylation site in skeletal RyRl channels.
  • RyRl single channel recordings in planar lipid bilayers show activity of the channels at 150 nM [Ca 2+ ] cfa (cytosolic side) with 1 mM ATP. Recordings were at 0 mV, closed state of the channels as indicated by 'c', and channel openings are upward deflections. AU point amplitude histograms are shown on the right.
  • FIG. 19 demonstrates the depletion of stabilizing calstabinl and PKA hyperphosphorylation of RyRl channels from sustained exercise.
  • Aerobic exercise can be defined as a form of physical exercise that increases the heart rate and enhances oxygen intake to improve performance. Examples of aerobic exercise are running, cycling, and swimming.
  • mice were challenged by aerobic exercise (forced swimming) for 90 mins twice daily. The animals were accustomed to swimming in preliminary training sessions: day -3 twice 30 mins, day -2 twice 45 mins, day -1 twice 60 mins, day 0 and following twice 90 mins.
  • mice were then exercised for 1, 7, or 21 additional, consecutive days for 90 mins twice daily. Between swimming sessions separated by a 4 hour rest period the mice are kept warm and given food and water. An adjustable-current water pool was used to exercise mice by swimming. An acrylic pool (90 cm long x 45 cm wide x 45 cm deep) filled with water to a depth of 25 cm was used. A current in the pool was generated with a pump. The current speed during the swimming session was at a constant speed of 1 1/min flow rate. The water temperature was maintained at 34°C with an electric heater. Age- and weight-matched mice were used to exclude differences in buoyancy from body fat.
  • RyRl channels are PKA hyperphosphorylated and depleted of the stabilizing calstabinl subunit after 3 weeks of 90 mins swimming twice daily.
  • the immunoprecipitated RyRl macromolecular channel complex shows increased PKA phosphorylation at Ser-2844 (corresponding to human RyRl-Ser-2843) whereas CaMKII phosphorylation at Ser-2849 (corresponding to human RyRl-Ser-2848) is unchanged.
  • Concomitant with increased RyRl -Ser-2844 PKA hyperphosphorylation calstabinl is depleted from the channel complex.
  • FIG. 20 Figure 20, embodiments A and B, demonstrate that PKA phosphorylation increases for increasing durations of sustained exercise.
  • mice were exposed to swimming for 1, 7, or 21 days followed by immediate sacrifice. Longer exposure to sustained exercise results in a significant increase of RyRl PKA hyperphosphorylation beginning at 7 days and saturating at 21 days.
  • embodiment A the immunoprecipitated RyRl channel complex shows significantly and above physiologic levels increased PKA phosphorylation at Ser-2844 (corresponding to human RyRl-Ser-2843) after 7 days of swimming exercise.
  • embodiment B inormalization of RyR2-Ser-2844 phosphorylation within the tetrameric channel complex documents a significant increase in PKA phosphorylation. *, P ⁇ 0.05; **, P ⁇ 0.005.
  • the data of Figure 20 shows that sustained exercise results in significantly increased RyRl phosphorylation by protein kinase A (PICA) which contributes to depletion of the stabilizing calstabinl subunit from the channel complex as the cause of a gain-of-function defect.
  • PICA protein kinase A
  • Figure 21 provides data showing that showing that chronically increased sympathetic stimulation of skeletal muscles, results in RyRl -dependent intracellular Ca 2+ leak and significantly increased muscle fatigue. What is the functional consequence of chronically increased RyRl PKA hyperphosphorylation? As shown in Figure 21 for mice and rats with heart failure from myocardial infarction, chronic RyRl PKA hyperphosphorylation results in increased muscle fatigue.
  • Figure 21 provides data showing that sustained exercise causes RyRl PKA hyperphosphorylation and calstabinl depletion, and Figure 21 shows that the identical defect occurs in disease forms with increased sympathetic activity causing intracellular SR Ca 2+ leak and significantly accelerated skeletal muscle fatigue.
  • Trichrome stain shows packed myofibers of similar cross-sectional dimension in non- exercised control (WT) mice (left). Three weeks swimming results in myofiber degeneration and interstitial collagen deposits with irregular fiber sizes. Hematoxylin-eosin (H&E) stain indicates nuclear changes and myofiber death. These changes are consistent with dystrophic remodeling.
  • the rapid delayed rectifier potassium channel (1(Kr)) is important for repolarization of the cardiac action potential.
  • HERG is the pore-forming subunit of the I(Kr) channel. Suppression of I(Kr) function, for example as a side-effect of a drug or the result of a mutation in hERG, can lead to long-QT (LQT) syndrome, which is associated with increased risk of life-threatening arrhythmias.
  • LQT long-QT
  • the compounds of the present invention exhibit a lower level of hERG blocking activity than JTV-519, as demonstrated in Figures 23- 43. Thus, the compounds of the present invention are expected to be less toxic and/or exhibit fewer side effects than JTV-519.
  • Figures 23 to 26 illustrate the effect of the compound ARM036 (also referred to as S36) and ARM036-Na (a sodium salt of ARM036) on hERG currents.
  • ARM036 also referred to as S36
  • ARM036-Na a sodium salt of ARM036
  • Figure 23 shows a typical hERG voltage-clamp current recording before (control) and after application of ARM036 at lOO ⁇ M.
  • the voltage pulse protocol used to activate the hERG currents is illustrated below the current trace. It can be seen that, following activation by the conditioning prepulse (to +20 mV), partial repolarization (-50 mV test pulse) of the membrane evoked a large, slowly decaying outward tail current.
  • Application of ARM036 minimally reduced the outward tail current in a concentration- and time-dependent manner.
  • Figure 24 shows a typical time course of the effect of ARM036 at 100 mM on the amplitude of the hERG channel current.
  • Figure 25 is a graph showing the concentration-dependence of the effect of ARM036 on the hERG current. Table 1 provides the numerical data that is illustrated graphically in Figure 25. Because the highest concentration of ARM036 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM036.
  • Figure 26 is a graph showing the concentration-dependence of the effect of ARM036- Na on the hERG current. Table 2 provides the numerical data that is illustrated graphically in Figure 26. Because the highest concentration of ARM036-Na tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM036-NA
  • Figure 27 is a graph showing the concentration-dependence of the effect of ARM047 on the hERG current.
  • Table 3 provides the numerical data that is illustrated graphically in Figure 27.
  • the IC 50 value for ARM047 block of the hERG current was 2.496 ⁇ M.
  • Figure 28 is a graph showing the concentration-dependence of the effect of ARM048 on the hERG current. Table 4 provides the numerical data that is illustrated graphically in Figure 28. Because the highest concentration of ARM048 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM048.
  • Figure 29 is a graph showing the concentration-dependence of the effect of ARM050 on the hERG current. Table 5 provides the numerical data that is illustrated graphically in Figure 29. Because the highest concentration of ARM050 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM050.
  • Figure 30 is a graph showing the concentration-dependence of the effect of ARM057 on the hERG current. Table 6 provides the numerical data that is illustrated graphically in Figure 30. Because the highest concentration of ARM057 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM057.
  • Figure 31 is a graph showing the concentration-dependence of the effect of ARM064 on the hERG current. Table 7 provides the numerical data that is illustrated graphically in Figure 31. Because the highest concentration of ARM064 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM064.
  • Figure 32 is a graph showing the concentration-dependence of the effect of ARM074 on the hERG current. Table 8 provides the numerical data that is illustrated graphically in Figure 32. Because the highest concentration of ARM050 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM074.
  • Figure 33 is a graph showing the concentration-dependence of the effect of ARM075 on the hERG current. Table 9 provides the numerical data that is illustrated graphically in Figure 33. Because the highest concentration of ARM075 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM075.
  • Figure 34 is a graph showing the concentration-dependence of the effect of ARM076 on the hERG current.
  • Table 10 provides the numerical data that is illustrated graphically in Figure 34. Because the highest concentration of ARM076 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM076. Table 10
  • Figure 35 is a graph showing the concentration-dependence of the effect of ARM077 on the hERG current. Table 11 provides the numerical data that is illustrated graphically in Figure 35. Because the highest concentration of ARM077 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARM077.
  • Figure 36 is a graph showing the concentration-dependence of the effect of ARMlOl on the hERG current. Table 12 provides the numerical data that is illustrated graphically in Figure 36. Because the highest concentration of ARMlOl tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARMlOl .
  • Figure 37 is a graph showing the concentration-dependence of the effect of ARM102 on the hERG current. Table 13 provides the numerical data that is illustrated graphically in Figure 37. Because the highest concentration of ARM102 tested resulted in less than 50% current inhibition, it was not possible to determine an ICs 0 value for ARMl 02.
  • Figure 38 is a graph showing the concentration-dependence of the effect of ARM103 on the hERG current. Table 14 provides the numerical data that is illustrated graphically in Figure 38. Because the highest concentration of ARM103 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARMl 03.
  • Figure 39 is a graph showing the concentration-dependence of the effect of ARM 104 on the hERG current. Table 15 provides the numerical data that is illustrated graphically in Figure 39. Because the highest concentration of ARM104 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARMl 04.
  • Figure 40 is a graph showing the concentration-dependence of the effect of ARM 106 on the hERG current. Table 16 provides the numerical data that is illustrated graphically in Figure 40. Because the highest concentration of ARM106 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARMl 06.
  • Figure 41 is a graph showing the concentration-dependence of the effect of ARM 107 on the hERG current. Table 17 provides the numerical data that is illustrated graphically in Figure 41. Because the highest concentration of ARM 107 tested resulted in less than 50% current inhibition, it was not possible to determine an IC 50 value for ARMl 07.
  • Figure 42 is a graph showing the concentration-dependence of the effect of S26 on the hERG current.
  • Table 18 provides the numerical data that is illustrated graphically in Figure 42.
  • the IC 50 value for S26 was 7.029 ⁇ M.
  • Figure 43 is a graph showing the concentration-dependence of the effect of JTV-519 (referred to in the figure as "ARMOXX”) on the hERG current.
  • Tablel9 provides the numerical data that is illustrated graphically in Figure 43.
  • the IC 50 value for JTV-519 was 0.463 ⁇ M.
  • E-4031 a known blocker of hERG currents, was used as a positive control.
  • the compounds of the present invention exhibit reduced hERG blocking activity as compared to JTV-519.
  • the compounds of the invention are expected to be less toxic and/or exhibit fewer side effects than JTV-519.
  • Table 20 below provides EC 50 values for compounds Sl -S 107. These EC 50 data were obtained using thee FKBP12.6 rebinding assay described above to determine the amount of FKBP 12.6 binding to PKA-phosphorylated RyR2 at various concentrations (0.5 - 1000 nM) of the compounds shown in Table 20. The EC50 values are calculated using Michaelis- Menten curve fitting. Table 20
  • a highly-efficient assay for high-throughput screening for small molecules is developed by immobilizing FKBP, either FKBP 12 or FKBP12.6 ⁇ e.g., wild-type FKBP12.6 or a fusion protein, such as GST- FKBP 12.6) onto a 96-well plate coated with glutathione, using standard procedures.
  • PKA- phosphorylated ryanodine receptor (RyR) specifically RyRl or RyR3 in the case of FKBP 12 and RyR2 in the case of FKBP12.6, is loaded onto the FKBP-coated plate, and incubated with compounds at various concentrations (10-100 nM) for 30 min.
  • the plate is washed to remove the unbound RyR, and then incubated with anti-RyR antibody (e.g., for 30 min).
  • anti-RyR antibody e.g., for 30 min.
  • the plate is washed again to remove unbound anti-RyR antibody, and then treated with florescent-labeled secondary antibody.
  • the plate is read by an automatic fluorescent plate reader for binding activity.
  • RyR is PKA-phosphorylated in the presence Of 32 P-ATP.
  • Radioactive PKA-phosphorylated RyR is loaded onto an FKBP-coated, 96-well plate, in the presence of JTV-519 analogues and other compounds at various concentrations (10-100 nM) for 30 min. The plate is washed to remove the unbound radiolabeled RyR, and then read by an automatic plate reader. PKA-phosphorylated RyR also is coated to the plate, and incubated with 32 S- labeled FKBP in the presence of the compounds.
  • Cardiac SR membranes are prepared, as previously described (Marx, et al, PKA phosphorylation dissociates FKBP 12.6 from the calcium release channel (ryanodine receptor): defective regulation in failing hearts. Cell, 101:365-76, 2000; Kaftan, et al, Effects of rapamycin on ryanodine receptor/Ca ⁇ 2+ ⁇ -release channels from cardiac muscle. Circ. Res., 78:990-97, 1996). 35 S-labelled FKBP12.6 was generated using the TNTTM Quick Coupled Transcription/Translation system from Promega (Madison, WI). [ 3 H] ryanodine binding is used to quantify RyR2 levels.
  • microsomes 100 ⁇ g of microsomes are diluted in 100 ⁇ l of 10-mM imidazole buffer (pH 6.8), incubated with 250-nM (final concentration) [ 35 S] -FKBP 12.6 at 37°C for 60 min, then quenched with 500 ⁇ l of ice-cold imidazole buffer. Samples are centrifuged at 100,000 g for 10 min and washed three times in imidazole buffer. The amount of bound [ 35 S]-FKBP 12.6 is determined by liquid scintillation counting of the pellet.
  • EXAMPLE 2 - IMMUNOBLOTS Immunoblotting of microsomes (50 ⁇ g) is performed as described, with anti- FKBP12/12.6 (1:1,000), anti-RyR-5029 (1:3,000) (Jayaraman, et al, FK506 binding protein associated with the calcium release channel (ryanodine receptor). /. Biol. Chem., 267:9474- 77, 1992), or anti-phosphoRyR2-P2809 (1:5,000) for 1 h at room temperature (Reiken, et al., Beta-blockers restore calcium release channel function and improve cardiac muscle performance in human heart failure. Circulation, 107:2459-66, 2003).
  • the P2809- phosphoepitope-specific anti-RyR2 antibody is an affinity-purified polyclonal rabbit antibody, custom-made by Zymed Laboratories (San Francisco, CA) using the peptide, CRTRRI-(pS)-QTSQ, which corresponds to RyR2 PKA-phosphorylated at Ser 2809 . After incubation with HRP -labeled anti-rabbit IgG (1 :5,000 dilution; Transduction Laboratories, Lexington, KY), the blots are developed using ECL (Amersham Pharmacia, Piscataway, NJ).
  • Symmetric solutions used for channel recordings are: trans compartment - HEPES, 250 mmol/L; Ba(OH) 2 , 53 mmol/L (in some experiments, Ba(OH) 2 is replaced by Ca(OH) 2 ); pH 7.35; and cis compartment - HEPES, 250 mmol/L; Tris-base, 125 mmol/L; EGTA, 1.0 mmol/L; and CaCl 2 , 0.5 mmol/L; pH 7.35. Unless otherwise indicated, single-channels recordings are made in the presence of 150-nM [Ca 2+ ] and 1.0-mM [Mg 2+ ] in the cis compartment.
  • Synthon S26 was prepared according to methods described in U.S. Patent App. No. 10/680,988.
  • S36-cystamine which is purified by SiO 2 column chromatography in 80% yield.
  • Deprotection of the Boc-group is achieved with trifluoroacetic acid in CH 2 Cl 2 , and the deprotected S36-cystamine is used for the synthesis of S43 and S45 by reaction with NHS-activated ester of azido compounds. Yield is 75% for S43 and 80% for S45.
  • S44 is synthesized as a by-product of the following reaction: S36 (50mg, 0.19mmol) is treated with thionyl chloride (2ml) at room temperature overnight.
  • S76-S81 are synthesized from commercially available S68 as shown in Scheme 12 in 70-95% yield.
  • Synthesis of S94 To a solution of S86 (lmmol) in CH 2 Cl 2 (10 ml) is added at O 0 C acetic anhydride (1.2 mmol) and triethylamine (1.3 mmol). The reaction mixture is stirred at room temperature overnight, then washed with H 2 O. After drying with Na 2 SO 4 , the solvent is evaporated and the product S94 (98% yield by NMR) is used for the next reaction without further purification.
  • Synthesis of S95 To a stirred solution of S84 (0.5 mmol) in benzene (20 ml) is added anhydrous AlCl 3 (0.6 mmol) drop-wise. The reaction mixture is refluxed for 5 hours and poured on to crushed ice (10 g). After extraction and concentration, the product S95 is purified by silica gel column chromatography in 83% yield.
  • ARM103 may be accomplished as shown in Scheme 16. The following are examples of the synthesis.
  • S102 was synthesized from S68 using the same method used to synthesize SlOl, with the exception that piperidine was used in place of 1- piperonylpiperazine. The structure of the product was confirmed by nuclear magnetic resonance (NMR), mass spectroscopy (MS) and/or by elemental analysis.
  • NMR nuclear magnetic resonance
  • MS mass spectroscopy
  • Synthesis of S103 S103 was synthesized from S68 using the same method used to synthesize SlOl, with the exception that N-Boc 1-piperazine was used in place of 1- piperonylpiperazine, and in a subsequent step the Boc group was deprotected using trifluoroacetic acid (TFA). The structure of the product was confirmed by nuclear magnetic resonance (NMR), mass spectroscopy (MS) and/or by elemental analysis.
  • Scheme 17 Synthesis of ARM104
  • Synthesis of S104 maybe accomplished as shown in Scheme 17. The following is an example of the synthesis.
  • a mixture ARM036 (S36) (27 mg, 0.1 mmol), 50% H 2 O 2 (1 ml), and MeOH (3 ml) was stirred at room temperature for 2 days to generate the ARMl 04 product.
  • Mass spectroscopy (MS) was used to monitor the disappearance of ARM036 and the appearance of the product ARM104. The solvents were removed under reduced pressure, and the product was purified by re-crystallization. The final yield was 26 mg of ARM104 at 85% purity.
  • the structure of the final product was dete ⁇ nined by nuclear magnetic resonance (NMR) and/or MS.
  • Synthesis of S105 maybe accomplished as shown in Scheme 18. The following is an example of the synthesis: To a stirred solution of S68 (80 mg, 0.48 mmol) and pyridine (0.1 ml, excess) in CH 2 Cl 2 (50 ml) at O 0 C, CH 3 O-C(O)C(O)Cl (70 mg, 0.58 mmol) was added dropwise. The reaction mixture was stirred at O 0 C for 2 hours and washed with IN HCl, saturated sodium bicarbonate, and water. Removal of the solvents and purification by SiO 2 column chromatography were performed to produce the ARM105 product as a white solid (yield: 95 mg, 94%).
  • Synthesis of S107 maybe accomplished as shown in Scheme 19. The following is an example of the synthesis: To S26 (180 mg, 0.92 mmol) in MeOH (20 ml) was added 30% CH 2 O solution (1.5 ml, excess) and sodium cyanoborohydride (NaBCNH 3 ) (0.4g excess). The reaction mixture was stirred at room temperature, and the pH of the solution was maintained at around pH 4-5 by addition of a few drop of IN HCl. After 3 hours, the solvents were removed under reduced pressure. The residue was dissolved in 20 ml ethyl acetate and washed with H 2 O and saturated NaHCO 3 (2 x 10 ml). The solvents were removed and the ARM107 was purified by SiO 2 column chromatography to give a yield: 170 mg, 93%.
  • Synthesis of S108 maybe accomplished as shown in Scheme 20.
  • Cbz- Gly 129 mg, 0.61 mmol
  • DIC Diisopropyl-carbodiimide
  • NHS N- hydroxysuccinimide
  • Synthesis of S 109 (ARM 109) maybe accomplished as shown in Scheme 21.
  • Synthesis of S112 maybe accomplished as shown in Scheme 24.
  • Synthesis of S113 maybe accomplished as shown in Scheme 25.
  • Synthesis of S114 maybe accomplished as shown in Scheme 25A.
  • S26 (195 mg, lmmol) in CH 2 Cl 2 (50 ml) was cooled to O 0 C.
  • triphosgene 150 mg, 0.5 mmol
  • pyridine 0.5 ml. excess
  • the resulting S26- phosgene in the reaction mixture was treated withN-Boc 1-piperazine (200 mg, 1.1 mmol) at O 0 C.
  • Synthesis of S115 may be accomplished as shown in Scheme 26.
  • Synthesis of S116 maybe accomplished as shown in Scheme 27.
  • Synthesis of S117 may be accomplished as shown in Scheme 28.
  • Synthesis of S118 may be accomplished as shown in Scheme 29.
  • Synthesis of S119 maybe accomplished as shown in Scheme 30.
  • Synthesis of S120 (ARM120) and S121 (ARM121) may be accomplished as shown in Scheme 31.
  • S120 was similarly synthesized, but using 4-OH-benzyl bromide instead of benzyl bromide.
  • the crude product was purified by chromatography on a silica gel column, eluting with a gradient increasing in polarity from 0 to 70% petroleum in ethyl acetate. Relevant fractions were combined to give S123 (244 mg, 100%).
  • a highly-efficient assay for high-throughput screening for small molecules is developed by immobilization of FKBP 12.6 (GST-fusion protein) onto a 96-well plate coated with glutathione.
  • PKA-phosphorylated ryanodine receptor type 2 (RyR2) is loaded onto the FKBP12.6-coated plate, and incubated with JTV-519 analogues at various concentrations (10-100 nM) for 30 min. Thereafter, the plate is washed to remove the unbound RyR2, and then incubated with anti-RyR2 antibody for 30 min. The plate is again washed to remove unbound anti-RyR2 antibody, and then treated with florescent-labeled secondary antibody. The plate is read by an automatic fluorescent plate reader for binding activity.
  • RyR2 is PKA-phosphorylated in the presence of 32P-ATP. Radioactive PKA-phosphorylated RyR2 is loaded onto an FKBP12.6-coated, 96-well plate, in the presence of JTV-519 analogues at various concentrations (10-100 nM) for 30 min. The plate is washed to remove the unbound radiolabeled RyR2, and then read by an automatic plate reader.
  • HEK 293 human embryonic kidney 293
  • HEK 293 cells do not express endogenous hERG.
  • HEK293 cells were transfected with a plasmid containing the hERG cDNA and a neomycin resistance gene. Stable transfectants were selected by culturing the cells in the presence of G418. The selection pressure was maintained by continued culture in the presence of G418.
  • D-MEM/F-12 Dulbecco's Modified Eagle Medium/Nutreint Mizture F-12
  • fetal bovin serum 199U/ml penicillin G sodium, lO ⁇ g/mL streptomycin sulfate and 500 ⁇ g/mL G418.
  • Cells for use in electrophysiology were cultured in 35 mm dishes.
  • Electrophysiological recordings were performed at room temperature (18°C-24°C). Each cell acted as its own control. The effect of ARM0036 was evaluated at two concentrations: 10 and lOO ⁇ M. Each concentration was tested in at least three cells (n > 3). 90 nM Cisapride (commercially available from TOCRIS Bioscience) was used as a positive control for hERG blockade. For recording, cells were transferred to the recording chamber and superfused with vehicle control solution. The patch pipette solution for whole cell recording contained 130 mM potassium aspartate, 5 mM MgC12, 5 mM EGTA, 4 mM ATP and 10 mM HEPES.
  • the pH was adjusted to 7.2 with KOH.
  • the pipette solution was prepared in batches, aliquoted, and stored frozen. A fresh aliquot was thawed and used each day.
  • Patch pipettes were made from glass capillary tubing using a P-97 micropipette puller (Sutter Instruments, Novato, CA). A commercial patch clamp amplifier was used for whole cell recordings. Before digitization, current records were low-pass filtered at one-fifth of the sampling frequency.
  • Onset and steady stae block of hERG current was measured using a pulse pattern with fixed amplitudes (conditioning prepulse: +20 mV for 2 seconds; test pulse: _50 mV for 2 seconds) repeated at 10 second intervals, from a holding potential of -80 mV. Peak tail current was measured during the 2 second step to _50 mV. A steady state was maintained for at least 30 seconds before applying the test compound or the positive control. Peak tail current was monitored until a new steady state was achieved. Test compound concentrations were applied cumulatively in ascending order without washout between applications.
  • each compound was tested at 0.01, 0.1, 1 and 1OmM, with each concentration tested in 2 cells (n > 2). The duration of exposure to each test concentration was 5 minutes. Other aspects of the experimental protocols were essentially similar to those described in Example 6. For some compounds it was not possible to determine the IC 50 because the highest concentration of the test compound used did not block the hERG channel by 50% or more.

Abstract

The present invention provides compounds of Formula I, (I) and salts, hydrates, solvates, complexes, and prodrugs thereof. The present invention further provides methods for synthesizing compounds of Formula I. The invention additionally provides pharmaceutical compositions comprising the compounds of Formula I and methods of using the pharmaceutical compositions of Formula I to treat and prevent disorders and diseases associated with the RyR receptors that regulate calcium channel functioning in cells.

Description

AGENTS FOR PREVENTING AND TREATING DISORDERS INVOLVING MODULATION OF THE RYR RECEPTORS
[001] This invention was made with government support under NIH Grant No. POl HL 67849-01. As such, the United States government may have certain rights in this invention. This application claims priority to U.S. patent application serial number 11/212,413, filed on August 25, 2005.
FIELD OF THE INVENTION
[002] This invention relates to compounds and their use to treat and prevent disorders and diseases associated with the RyR receptors that regulate calcium channel functioning in cells. More particularly, the invention discloses compounds that are related to 1,4-benzothiazepines and are useful to treat cardiac and skeletal muscular disorders. The invention also discloses pharmaceutical compositions comprising the compounds and articles of manufacture comprising the pharmaceutical compositions.
BACKGROUND OF THE INVENTION
[003] The sarcoplasmic reticulum (SR) is a structure in cells that functions, among other things, as a specialized intracellular calcium (Ca2+) store. Channels in the SR called ryanodine receptors (RyRs) open and close to regulate the release of Ca2+ from the SR into the intracellular cytoplasm of the cell. Release of Ca2+ into the cytoplasm from the SR increases cytoplasmic Ca2+ concentration. Open probability (Po) of the RyR receptor refers to the likelihood that the RyR channel is open at any given moment, and therefore capable of releasing Ca2+ into the cytoplasm from the SR.
[004] There are three types of ryanodine receptors, all of which are highly-related Ca2+ channels: RyRl, RyR2, and RyR3. RyRl is found predominantly in skeletal muscle as well as other tissues, RyR2 is found predominantly in the heart as well as other tissues, and RyR3 is found in the brain as well as other tissues. The RyR channels are formed by four RyR polypeptides in association with four FK506 binding proteins (FKBPs), specifically FKBP 12 (calstabinl) and FKBP12.6 (calstabin2). Calstabinl binds to RyRl, calstabin2 binds to RyR2, and calstabinl binds to RyR3. The FKBP proteins (calstabinl and calstabin2) bind to the RyR channel (one molecule per RyR subunit), stabilize RyR-channel functioning, and facilitate coupled gating between neighboring RyR channels, thereby preventing abnormal activation of the channel during the channel's closed state. [005] Besides the calstabin binding proteins, protein kinase A (PKA) also binds to the cytoplasmic surface of the RyR receptors. PICA phosphorylation of the RyR receptors causes partial dissociation of calstabins from RyRs. Dissociation of calstabin from RyR causes increased open probability of RyR3 and therefore increased Ca2+ release from the SR into the intracellular cytoplasm.
[006] Ca2+ release from the SR in skeletal muscle cells and heart cells is a key physiological mechanism that controls muscle performance, because increased concentration of Ca2+ in the intracellular cytoplasm causes contraction of the muscle.
[007] Excitation-contraction (EC) coupling in skeletal muscles involves electrical depolarization of the plasma membrane in the transverse tubule (T-tubule), which activates voltage-gated L-type Ca2+ channels (LTCCs). LTCCs trigger Ca2+ release from the SR through physical interaction with RyRl . The resulting increase in cytoplasmic Ca concentration induces actin-myosin interaction and muscle contraction. To enable relaxation, intracellular Ca2+ is pumped back into the SR via SR Ca2+-ATPase pumps (SERCAs), which is regulated by phospholamban (PLB) depending on the muscle fiber type.
[008] It has been shown that disease forms that result in sustained activation of the sympathetic nervous system and increased plasma catecholamine levels cause maladaptive activation of intracellular stress pathways resulting in destabilization of the RyRl channel closed state and intracellular Ca2+ leak. SR Ca2+ leak via RyRl channels was found to deplete intracellular SR calcium stores, to increase compensatory energy consumption, and to result in significant acceleration of muscle fatigue. The stress-induced muscle defect permanently reduces isolated muscle and in vivo performance particularly in situations of increased demand.
[009] It also has been shown that destabilization of the RyRl closed state occurs under pathologic conditions of increased sympathetic activation and involves depletion of the stabilizing calstabinl (FKBP 12) channel subunit. Proof-of-principle experiments have shown that PKA activation as an end effector of the sympathetic nervous systems increases RyRl PKA phosphorylation at Ser-2843 which decreases the binding affinity of calstabinl to RyRl and increases channel open probability.
[010] In cardiac striated muscle, RyR2 is the major Ca2+- release channel required for EC coupling and muscle contraction. During EC coupling, depolarization of the cardiac-muscle cell membrane during phase zero of the action potential activates voltage-gated Ca2+ channels. Ca2+ influx through the open voltage-gated channels in turn initiates Ca2+ release from the SR via RyR2. This process is known as Ca2+-induced Ca2+ release. The RyR2-mediated, Ca2+-induced Ca2+ release then activates the contractile proteins in the cardiac cell, resulting in cardiac muscle contraction.
[Oil] Phosphorylation of cardiac RyR2 by PKA is an important part of the "fight or flight" response that increases cardiac EC coupling gain by augmenting the amount of Ca2+ released for a given trigger. This signaling pathway provides a mechanism by which activation of the sympathetic nervous system, in response to stress, results in increased cardiac output. PKA phosphorylation of RyR2 increases the open probability of the channel by dissociating calstabin2 (FKBP 12.6) from the channel complex. This, in turn, increases the sensitivity of RyR2 to Ca2+-dependent activation.
[012] Despite advances in treatment, heart failure remains an important cause of mortality in Western countries. An important hallmark of heart failure is reduced myocardial contractility. In heart failure, contractile abnormalities result, in part, from alterations in the signaling pathway that allows the cardiac action potential to trigger Ca2+ release via RyR2 channels and muscle contraction. In particular, in failing hearts, the amplitude of the whole- cell Ca2+ transient is decreased and the duration prolonged.
[013] Cardiac arrhythmia, a common feature of heart failure, results in many of the deaths associated with the disease. Atrial fibrillation (AF) is the most common cardiac arrhythmia in humans, and represents a major cause of morbidity and mortality. Structural and electrical remodeling - including shortening of atrial refractoriness, loss of rate-related adaptation of refractoriness, and shortening of the wavelength of re-entrant wavelets - accompany sustained tachycardia. This remodeling is likely important in the development, maintenance and progression of atrial fibrillation. Studies suggest that calcium handling plays a role in electrical remodeling in atrial fibrillation.
[014] Approximately 50% of all patients with heart disease die from fatal cardiac arrhythmias. In some cases, a ventricular arrhythmia in the heart is rapidly fatal - a phenomenon referred to as "sudden cardiac death" (SCD). Fatal ventricular arrhythmias and SCD also occur in young, otherwise-healthy individuals who are not known to have structural heart disease. In fact, ventricular arrhythmia is the most common cause of sudden death in otherwise-healthy individuals. [015] Catecholarninergic polymorphic ventricular tachycardia (CPVT) is an inherited disorder in individuals with structurally normal hearts. It is characterized by stress-induced ventricular tachycardia - a lethal arrhythmia that causes SCD. IQ subjects with CPVT, physical exertion and/or stress induce bidirectional and/or polymorphic ventricular tachycardias that lead to SCD even in the absence of detectable structural heart disease. CPVT is predominantly inherited in an autosomal-dominant fashion. Individuals with CPVT have ventricular arrhythmias when subjected to exercise, but do not develop arrhythmias at rest. Studies have identified mutations in the human RyR2 gene, on chromosome Iq42-q43, in individuals with CPVT.
[016] Failing hearts (e.g. , in patients with heart failure and in animal models of heart failure) are characterized by a maladaptive response that includes chronic hyperadrenergic stimulation, hi heart failure, chronic beta-adrenergic stimulation is associated with the activation of beta-adrenergic receptors in the heart, which, through coupling with G-proteins, activate adenylyl cyclase and thereby increase intracellular cAMP concentration. CAMP activates cAMP-dependent PKA, which has been shown to induce hyperphosphorylation of RyR2. Thus, chronic heart failure is a chronic hyperadrenergic state that results in several pathologic consequences, including PKA hyperphosphorylation of RyR2.
[017] The PKA hyperphosphorylation of RyR2 has been proposed as a factor contributing to depressed contractile function and arrhythmogenesis in heart failure. Consistent with this hypothesis, PKA hyperphosphorylation of RyR2 in failing hearts has been demonstrated, in vivo, both in animal models and in patients with heart failure undergoing cardiac transplantation.
[018] In failing hearts, the hyperphosphorylation of RyR2 by PKA induces the dissociation of FKBP12.6 (calstabin2) from the RyR2 channel. This causes marked changes in the biophysical properties of the RyR2 channel, including increased open probability (Po) due to an increased sensitivity to Ca2+-dependent activation; destabilization of the channel, resulting in subconductance states; and impaired coupled gating of the channels, resulting in defective EC coupling and cardiac dysfunction. Thus, PKA-hyperphosphorylated RyR2 is very sensitive to low-level Ca2+ stimulation, and this. manifests itself as a diastolic SR Ca2+ leak through the PKA hyperphosphorylated RyR2 channel.
[019] The maladaptive response to stress in heart failure results in depletion of FKBP12.6 from the channel macromolecular complex. This leads to a shift to the left in the sensitivity of RyR2 to Ca2+-induced Ca2+ release, resulting in channels that are more active at low-to- moderate Ca2+ concentrations. Over time, the increased "leak" through RyR2 results in resetting of the SR Ca2+ content to a lower level, which in turn reduces EC coupling gain and contributes to impaired systolic contractility.
[020] Additionally, a subpopulation of RyR2 that are particularly "leaky" can release SR Ca2+ during the resting phase of the cardiac cycle, diastole. This results in depolarizations of the cardiomyocyte membrane known as delayed after-depolarizations (DADs), which are known to trigger fatal ventricular cardiac arrhythmias.
[021] In patients with CPVT mutations in their RyR2 and otherwise structurally-normal hearts, a similar phenomenon is at work. Specifically, it is known that exercise and stress induce the release of catecholamines that activate beta-adrenergic receptors in the heart. Activation of the beta-adrenergic receptors leads to PKA hyperphosphorylation of RyR2 channels. Evidence also suggests that the PKA hyperphosphorylation of RyR2 resulting from beta-adrenergic-receptor activation renders mutated RyR2 channels more likely to open in the relaxation phase of the cardiac cycle, increasing the likelihood of arrhythmias.
[022] Cardiac arrhythmias are known to be associated with diastolic SR Ca2+ leaks in patients with CPVT mutations in their RyR2 and otherwise structurally-normal hearts. In these cases, the most common mechanism for induction and maintenance of ventricular tachycardia is abnormal automaticity. One form of abnormal automaticity, known as triggered arrhythmia, is associated with aberrant release of SR Ca2+, which initiates DADs. DADs are abnormal depolarizations in cardiomyocytes that occur after repolarization of a cardiac action potential. The molecular basis for the abnormal SR Ca2+ release that results in DADs has not been fully elucidated. However, DADs are known to be blocked by ryanodine, providing evidence that RyR2 plays a key role in the pathogenesis of this aberrant Ca2+ release.
[023] U.S. Patent No. 6,489,125 discusses JTV-519 (4-[3-(4-benzylpiperidin-l- yl)propionyl] -7-methoxy-2,3 ,4,5-tetrahydro- 1 ,4-benzothiazepine monohydrochloride; also known as k201 or ICP-Calstan 100), a 1,4-benzothiazepine, as a new modulator of RyR calcium-ion channels.
[024] Co-pending application U.S. Serial No. 10/763,498 discusses RyR2 as a target for treating and preventing heart failure and cardiac arrhythmias, including atrial fibrillation and cardiac arrhythmias that cause exercise-induced sudden cardiac death (SCD). RyR2 channels with 7 different CPVT mutations (e.g., S2246L, R2474S, N4104K, R4497C, P2328S, Q4201R, V4653F) were found to have functional defects that resulted in channels that became leaky (i.e., a calcium leak) when stimulated during exercise. The mechanism for the VT in CPVT has been demonstrated to be the same as the mechanism for VT in heart failure.
[025] It has been shown that exercise-induced arrhythmias and sudden death (in patients with CPVT) result from a reduced affinity of FKBP12.6 (calstabin2) for RyR2. Additionally, it has been demonstrated that exercise activates RyR2 as a result of phosphorylation by adenosine 3', 5'-monophosphate (cAMP)-dependent protein kinase (PKA). Mutant RyR2 channels, which had normal function in planar lipid bilayers under basal conditions, were more sensitive to activation by PKA phosphorylation - exhibiting increased activity (open probability) and prolonged open states, as compared with wild-type channels. In addition, PKA-phosphorylated mutant RyR2 channels were resistant to inhibition by Mg2+, a physiological inhibitor of the channel, and showed reduced binding to FKBP12.6 (aka calstabin2, which stabilizes the channel in the closed state). These findings indicate that, during exercise, when the RyR2 are PKA-phosphorylated, the mutant CPVT channels are more likely to open in the relaxation phase of the cardiac cycle (diastole), increasing the likelihood of arrhythmias triggered by SR Ca2+ leak.
[026] Additionally, co-pending U.S. Patent Application No. 09/288,606 discusses a method for regulating contraction of a subject's heart by administering a compound that regulates PKA phosphorylation of an RyR2 receptor and specifically decreases PKA phosphorylation. Co-pending U.S. Patent Application No. 10/608,723 also discusses a method for treating and prophylaxis for atrial tachyarrhythmia and exercise and stress-induced arrhythmias by administration of an agent which inhibits PKA phosphorylation of RyR2.
SUMMARY OF THE INVENTION
[027] In view of the foregoing, there is a need to identify new agents effective for treating or preventing disorders and diseases associated with the RyR receptors that regulate calcium channel functioning in cells, including skeletal muscular disorders and diseases and especially cardiac disorders and diseases. More particularly, a need remains to identify new compounds that can be used to treat RyR associated disorders by, for example, repairing the leak in RyR channels, and enhancing binding of FKBP proteins (calstabinl and calstabin2) to PKA-phosphorylated RyR, and to mutant RyR that otherwise have reduced affinity for, or do not bind to, FKBP 12 and FKBP 12.6. Embodiments of the invention solve some or all of these needs.
[028] Accordingly, the present invention generally provides compounds that may be classified as 1,4-benzothiazepines and sometimes are referred to herein as "RyCaIs."
[029] The present invention further provides compounds of Formula I:
Figure imgf000009_0001
wherein, n is 0, 1, or 2; q is O, 1, 2, 3, or 4; each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, -O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero- )arylamino; wherein each acyl, -O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be optionally substituted; R1 is selected from the group consisting of H, oxo, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
R2 is selected from the group consisting of H, -C(=O)R5, -C(=S)R6, -SO2R7, -P(=O)R8R9, - (CH2)m-R10, alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl may be optionally substituted;
R3 is selected from the group consisting of H , -CO2Y, -C(=O)NHY, acyl, -O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl maybe optionally substituted; and wherein Y is selected from the group consisting of H, alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl, and wherein each alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
R4 is selected from the group consisting of H, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
R5 is selected from the group consisting of -NR1SRi6, -(CH2)C1NR15R16, -NHNR1SR16, - NHOH, -OR15, -C(=O)NHNRi5R16, -CO2Ri5, -C(=O)NRi5R16, -CH2X, acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl maybe optionally substituted, and wherein q is 1, 2, 3, 4, 5, or 6;
R6 is selected from the group consisting Of-ORi5, -NHNRi5Ri6, -NHOH, -NRi5Ri6, -CH2X, acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
R7 is selected from the group consisting of -ORi5, -NR)5Ri6, -NHNRi5Ri6, -NHOH, -CH2X, alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted; R8 and Rg independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
R1O is selected from the group consisting of -NR15R16, OH, -SO2R11, -NHSO2Rn, C(=O)(R12), NHC=O(R12), -OC=O(Ri2), and -P(=O)R13Ri4;
Rn, Ri2, R13, and R14 independently are selected from the group consisting of H, OH, NH2, - NHNH2, -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
X is selected from the group consisting of halogen, -CN, -CO2Ri5, -C(=O)NR15Ri6, -NRi5Ri6, -ORi5, -SO2R7, and-P(=O)R8R9; and
R15 and Ri6 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted; and optionally R15 and Ri6 together with the N to which they are bonded may form a heterocycle which may be substituted; the nitrogen in the benzothiazepine ring may optionally be a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and prodrugs thereof; provided that when q is O and n is O, then R2 is not H, Et, -C(=0)NH2, (=O)NHPh, - C(=8)NH-nButyl, -C(=O)NHC(=O)CH2C1, -C(=0)H, -C(=0)Me, -C(=O)Et, - C(=0)CH=CH2, -S(=0)2Me, or -S(=O)2Et; further provided that when q is O and n is 1 or 2, then R2 is not -C(=O)Me, -C(=O)Et, - S(=O)2Me, or -S(=O)2Et; further provided that when q is 1, and R is Me, Cl, or F at the 6 position of the benzothiazepene ring, then R2 is not H, Me5 -C(=O)H, -C(=0)Me, -C(O)Et, -C(O)Ph, - S(=O)2Me, or -S(O)2Et; and further provided that when q is 1, n is 0, and R is OCT3, OH, Ci-C3 allcoxyl at the 7 position
of the benzothiazepene ring, then R2 is not H, -
Figure imgf000012_0001
C(=O)CH=CH2, or
[030] In one embodiment, the present invention provides compounds of Formula I, as described above, with the proviso that the compound is not S24 or S68.
[031] In one embodiment, the present invention provides compounds of Formula I-a:
Figure imgf000012_0002
wherein: n is 0, 1, or 2; q is O, 1, 2, 3, or 4; each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted or unsubstituted;
R2 is selected from the group consisting of H, -C=O(R5), -C=S(R6), -SO2R7, -P(=O)R8R9, - (CH2)m-Rio, alkyl, aryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl maybe substituted or unsubstituted;
R5 is selected from the group consisting of-NR15Ri6, -NHNR]5R16, -NHOH, -ORi5, - C(=O)NHNRisRi6, -CO2Ri5, -C(^O)NRi5R16, -CH2X, acyl, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R6 is selected from the group consisting of -ORi5, -NHNR]5R16, -NHOH, -NRi5R16, -CH2X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted;
R7 is selected from the group consisting of H, -OR15, -NRi5R16, -NHNRi5R16, -NHOH, - CH2X, alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R8 and Rg independently are selected from the group consisting of -OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R1O is selected from the group consisting of -NRiSRi6, OH, -SO2Rn, -NHSO2R11, - C(=0)Ri2, -NH(C=O)Ri2, -0(C=O)R12, and -P(=O)RBRi4; m is O, 1, 2, 3, or 4;
Rn, R12, Ri3, and R14 independently are selected from the group consisting of H, OH, NH2, - NHNH2, -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
X is selected from the group consisting of halogen, -CN, -CO2Ri5, -C(=O)NRi5Ri6, -NRi5Ri6, -ORi5, -SO2R7, and -P(=O)R8R9; and
R15 and R16 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; and optionally Ri5 and Rj6 together with the N to which they are bonded may form a heterocycle which may be substituted or unsubstituted; the nitrogen in the benzothiazepine ring may be optionally a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and prodrugs thereof; provided that when q is 0 and n is 0, then R2 is not H, Et, -CC=O)NH2, (=O)NHPh, - C(=S)NH-nButyl, -C(=O)NHC(=O)CH2C1, -C(=O)H, -CC=O)Me, -CC=O)Et, - C(=O)CH=CH2, -S(=O)2Me, or -SC=O)2Et; further provided that when q is O and n is 1 or 2, then R2 is not -C(=O)Me, -C(=O)Et, - SC=O)2Me, or -SC=O)2Et; further provided that when q is 1, and R is Me, Cl, or F at the 6 position of the benzothiazepene ring, then R2 is not H, Me, -C(=O)H, -C(O)Me, -C(O)Et, -C(O)Ph, - SC=O)2Me, or -S(O)2Et; and further provided that when q is 1, n is O, and R is OCT3, OH, C1-C3 alkoxyl at the 7 position
of the benzothiazepene ring, then R2 is not H, -
Figure imgf000014_0001
C(=O)CH=CH2, or
[032] In certain embodiments, the present invention provides compounds of formula I-a, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2C1-C4alkyl, -S(=O)CrC4alkyl, -S-Cr Qalkyl, -OSC=O)2CF3, Ph, -NHCH2Ph, -C(O)Me, -OC(O)Me, morpholinyl and propenyl; and n is O, 1, or 2.
[033] In other embodiments, the present invention provides compounds of formula I-a, wherein R2 is selected from the group consisting of -C=O(Rs), -C=S(R6), -SO2R7, - P(O)R8R9, and -(CH2)m-Ri0.
[034] In yet another embodiment, the present invention provides compounds of formula I-b:
Figure imgf000014_0002
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio maybe substituted or unsubstituted;
R2 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[035] In certain embodiments, the present invention provides compounds of formula I-b, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S C=O)2C1 -Qalkyl, -S(=O)CrC4alkyl, -S-Ci- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[036] In other embodiments, the present invention provides compounds of formula I-b, wherein R2 is selected from the group consisting of -C=O(Rs), -C=S(R6), -SO2R7, - P(=O)R8R9, and -(CH2)m-R10.
[037] In yet another embodiment, the present invention provides compounds formula of I-c:
Figure imgf000015_0001
wherein each R, R7, q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[038] In certain embodiments, the present invention provides compounds of formula I-c, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)d-C4alkyl, -S-C,- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O, I, or 2. [039] In other embodiments, the present invention provides compounds of formula I-c, wherein R7 is selected from the group consisting of -OH, -NR15Ri6, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
[040] In a further embodiment, the present invention provides compounds of formula of I- d:
Figure imgf000016_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R7 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[041] In certain embodiments, the present invention provides compounds of formula I-d, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)Ci-C4alkyl, -S-C1- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -OC(=O)Me, morpholinyl and propenyl; and n is 0, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[042] In other embodiments, the present invention provides compounds of formula I-d, wherein R7 is selected from the group consisting of -OH, -NRisRie, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted.
[043] In one embodiment, the present invention provides compounds of formula of I-e:
Figure imgf000017_0001
wherein each R, R5, q and n is as defined compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[044] In certain embodiments, the present invention provides compounds of formula I-e, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)Ci-C4alkyl, -S-C1- Qalkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O, 1, or 2.
[045] In other embodiments, the present invention provides compounds of formula I-e, wherein R5 is selected from the group consisting of-NRisRie, -NHOH, -OR15, -CH2X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
[046] In some embodiments, the present invention provides compounds of formula I-e, wherein R5 is an alkyl substituted by at least one labeling group, such as a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group. A fluorescent labeling group can be selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4!,6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
[047] In another embodiment, the present invention provides compounds of formula of I-f:
Figure imgf000018_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsύbstituted;
R5 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[048] In certain embodiments, the present invention provides compounds of formula I-f, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3,
Figure imgf000018_0002
-SC=O)C1 -Qalkyl, -S-C1- C4alkyl, -OS(O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[049] In other embodiments, the present invention provides compounds of formula I-f, wherein R5 is selected from the group consisting Of-NR15R16, -NHOH, -ORi5, -CH2X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
[050] In yet another embodiment, the present invention provides compounds of formula of I-g:
Figure imgf000019_0001
wherein W is S or O; each R, R15, R16, q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[051] In certain embodiments, the present invention provides compounds of formula I-g, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe5 -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2C1-C4alkyl, -S(=O)Ci-C4alkyl, -S-C1- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O, 1, or 2.
[052] In other embodiments, the present invention provides compounds of formula I-g, wherein R15 and R16 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted; and optionally R15 and R16 together with the N to which they are bonded may form a heterocycle which may be substituted.
[053] In some embodiments, the present invention provides compounds of formula I-g, wherein W is O or S.
[054] In yet another embodiment, the present invention provides compounds of formula of I-h:
Figure imgf000020_0001
wherein W is S or O; wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R1S, R16 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[055] In certain embodiments, the present invention provides compounds of formula I-h, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SC=O)2C1 -C4alkyl, -S(=O)d-C4alkyl, -S-C1- Qalkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -0C(=0)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[056] In other embodiments, the present invention provides compounds of formula I-h, wherein R15 and R16 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl 'may be substituted; and optionally R15 and Ri6 together with the N to which they are bonded may form a heterocycle which may be substituted.
[057] In some embodiments, the present invention provides compounds of formula I-g, wherein W is O or S. [058] In a further embodiment, the present invention provides compounds of formula of I-i:
Figure imgf000021_0001
wherein Rn is selected from the group consisting of-NRisRiό, -NHNRiSR16, -NHOH, -OR15, -CH2X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; each R, q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[059] In certain embodiments, the present invention provides compounds of formula I-i, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2C1-C4alkyl, -S(=O)Ci-C4alkyl, -S-C1- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O, 1, or 2.
[060] In other embodiments, the present invention provides compounds of formula I-i, wherein R17 is -NR15R16, and -OR15. In certain other embodiments, R17 is -OH, -OMe, -NEt, -NHEt, -NHPh, -NH2, or -NHCH2pyridyl.
[061] In one embodiment, the present invention provides compounds of formula of I-j:
Figure imgf000022_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH5 -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R17 is selected from the group consisting Of-NR15R16, -NHNRi5R16, -NHOH, -ORi5, -CH2X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; n is as defined in compounds of formula I-a; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[062] In certain embodiments, the present invention provides compounds of formula I-j, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)C,-C4alkyl, -S-C1- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -0C(=0)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[063] In other embodiments, the present invention provides compounds of formula I-j, wherein Ri7 is -NR]5Ri 6 or -ORj 5. In certain other embodiments, Rn is -OH, -OMe, -NEt, - NHEt, -NHPh, -NH2, or -NHCH2pyridyl.
[064] In another embodiment, the present invention provides compounds of foπnula I-k:
Figure imgf000023_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R18 is selected from the group consisting Of-NR15R16, -C(=O)NR15R16, -(C=O)OR15, -OR15, alkyl, aryl, cycloalkyl, heterocyclyl, and at one labeling group; wherein each alkyl, aryl, cycloalkyl, and heterocyclyl may be substituted or unsubstituted; wherein p is 1, 2, 3, 4, 5, 6, 7, 8 9, or 10; and n is O, 1, or 2; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[065] In certain embodiments, the present invention provides compounds of formula I-k, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)Ci-C4alkyl, -S-Ci- C4alkyl, -OS(^O)2CF3, Ph, -NHCH2Ph, -C(O)Me, -OC(O)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[066] In other embodiments, the present invention provides compounds of formula I-k, wherein Ris is selected from the group consisting Of-NRi5R16, -(C=O)OR15, -OR15, alkyl, aryl, and at one labeling group; and wherein each alkyl and aryl may be substituted or unsubstituted. In some cases, m is 1, and Ri8 is Ph, C(=0)0Me, C(O)OH, aminoalkyl, NH2, NHOH, or NHCbz. In other cases, m is O, and Rig is CpC4 alkyl, such as Me, Et, propyl, and butyl. In yet other cases, m is 2, and Ris is pyrrolidine, piperidine, piperazine, or morpholine. In some embodiments, m is 3, 4, 5, 5, 7, or 8, and R18 is a fluorescent labeling group selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
[067] In yet another embodiment, the present invention provides compounds of formula of 1-1:
Figure imgf000024_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R6 and n are as defined in compounds of formula I-a; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[068] In certain embodiments, the present invention provides compounds of formula 1-1, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2CrC4alkyl, -S(=O)Ci-C4alkyl, -S-Ci- Qalkyl, -OS(O)2CF3, Ph, -NHCH2Ph, -C(K))Me, -OC(K))Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[069] In other embodiments, the present invention provides compounds of formula 1-1, wherein R6 is selected from the group consisting Of-NR15R16, -NHNR15Ri β, -ORi5, -NHOH, - CH2X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted. In some cases, R6 is -NRi5R16 such as -NHPh, pyrrolidine, piperidine, piperazine, morpholine, and the like. In some other cases, R6 is alkoxyl, such as -O-tBu.
[070] In a further embodiment, the present invention provides compounds of formula I-m:
Figure imgf000025_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)aryltliio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R8, R9 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[071] In certain embodiments, the present invention provides compounds of formula I-m, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Q-C4alkyl, -S(=O)Ci-C4alkyl, -S-C1- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -0C(=0)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[072] In other embodiments, the present invention provides compounds of formula I-m, wherein Rg and R9 are independently alkyl, aryl, -OH, alkoxyl, or alkylamino. In some cases, Rs is C1-C4 alkyl such as Me, Et, propyl and butyl; and R9 is aryl such as phenyl.
[073] In one embodiment, the compound is selected from the group consisting of Sl, S2, S3, S4, S5, S6, S7, S9, SIl, S12, S13, S14, S19, S20, S22, S23, S25, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, 848, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S103, S104, S105, S107, S108, S109, SIlO, Sil l, S112, S113, S114, S115, S116, S117, S118, S119, S120, S121, S122, and S123.
[074] The compounds of the invention may optionally comprise a labeling group, such as a fluorescent, bioluminescent, chemiluminescent, colorimetric or radioactive labeling group. Suitable fluorescent labeling groups include, but are not limited to, bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4', 6-Diamidino-2-ρhenylindole (DAPI) , indopyra dyes, lucifer yellow, propidium iodide, porphyrins, and variants and derivatives thereof. One of skill in the art can readily select a suitable marker or labeling group, and conjugate such a labeling group to any of the compounds of the invention, without undue experimentation.
[075] The present invention also provides methods for the synthesis of compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, and salts, hydrates, solvates, complexes and pro-drugs thereof.
[076] The present invention further provides a method of treating or preventing various disorders and diseases in a subject that are associated with RyR receptors, such as muscular and cardiac disorders, comprising administering to the subject an amount of a compound effective to prevent or treat a disorder or disease associated with the RyR receptors, wherein the compound is of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or salts, hydrates, solvates, complexes and pro-drugs thereof.
[077] Also provided is a method of preventing or treating a leak in a RyR2 receptor in a subject, including administering to the subject an amount of a compound effective to prevent or treat a leak in the RyR2 receptor, wherein the compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or salts, hydrates, solvates, complexes and pro-drugs thereof. The subject is, for example, an in vitro system (e.g., cultured cells or tissues) or in vivo system (e.g., animal or human).
[078] In addition, the present invention provides a method of modulating the binding of RyR and FKBP in a subject, including administering to the subject an amount of a compound effective to modulate the level of RyR-bound FKBP, wherein the compound is of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or salts, hydrates, solvates, complexes and pro-drugs thereof. The subject is, for example, an in vitro system (e.g., cultured cells or tissues) or in vivo system {e.g., animal or human).
[079] The present invention also provides articles of manufacture for treating and preventing disorders and diseases associated with the RyR receptors, such as muscular and cardiac disorders, in a subject. The articles of manufacture comprise a pharmaceutical composition of one or more of the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or salts, hydrates, solvates, complexes and pro-drugs thereof. The articles of manufacture are packaged with indications for various disorders that the pharmaceutical compositions are capable of treating and/or preventing.
[080] Other features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples while indicating various embodiments of the invention are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE FIGURES
[081] Figure 1, embodiments A, B, C, and D are, respectively, (A) immunoblots of PKA phosphorylated RyR2 in the presence of FKBP12.6 and increasing JTV-519 concentrations; (B) immunoblots of PICA phosphorylated RyR2 in the presence of 0.5 nM S36; (C) a graph of current through plasma membrane, voltage dependent L-type Ca2+ channels which are completely blocked by nifedipine but not by S36 in isolated mouse cardiomyocytes; and (D) a graph of the voltage-dependence of L-type Ca2+ current in channels in the presence of JTV- 519 and S36.
[082] Figure 2, embodiments A, B, C, and D demonstrate the prevention of exercise- induced ventricular arrhythmias by JTV-519 in haploinsufficient calstabin (FKBP12.6)+/~ mice. Embodiment A are representative telemetric electrocardiograms (ECGs) of an untreated calstabin2 (FKBP 12.6)+/~ mouse (left), a JTV-519-treated calstabin2 (FICBPl 2.6)+/~ mouse (middle), and a calstabin2 (FKBP12.6)~/~ mouse (right). Embodiment B are telemetry recordings of a sustained polymorphic ventricular tachycardia (sVT) in (upper) an untreated haploinsufficient calstabin2 (FKBP 12.6)+/~ mouse and (lower) a JTV-519-treated calstabin2 (FKBP12.6)+/~ mouse, each subjected to exercise testing immediately followed by injection with 0.5 mg epinephrine per kilogram of body weight. Embodiment C are graphs showing the numbers of mice with cardiac death (left), sustained VTs (middle), and nonsustained VTs (right) in experimental groups of mice subjected to exercise testing and injection with 0.5 mg/kg epinephrine. Embodiment D provides graphs comparing the dose dependence of pharmacological effects of JTV-519 and S36 in regard to sudden cardiac death (left), sustained VTs (middle), and nonsustained VTs (right).
[083] Figure 3 is a graph showing fractional shortening (FS) of the left ventricle assessed by M-mode echocardiography 2 weeks post-myocardial infarction in placebo versus S36-treated mice. S36 treated mice show a significant improvement in FS in the 100 nM and 200 nM groups as compared to placebo.
[084] Figure 4 is a graph showing heart weight to body weight (HW/BW) ratios and pressure-volume loops quantifications (dP/dt) one week post-myocardial infarction of placebo and S36-treated mice. S36 treatment results in a beneficial reduction of the HW/BW ratio and increased velocity of pressure development in S36 as compared to placebo treated mice.
[085] Figure 5 is a graph summarizing EC50 values of JTV-519 and a series of Rycal compounds indicating several compounds with a higher biologic activity as evidenced by significantly lower EC50 values compared to JTV-519.
[086] Figure 6, embodiments A, B, and C are, respectively, (A) single-channel current traces of unphosphorylated RyR2-P2328S and unphosphorylated RyR2-WT treated with S36; (B) single-channel current traces of phosphorylated RyR2-P2328S and unphosphorylated RyR2-P2328S treated with S36; and (C) immunoblot analysis of calstabin-2 binding of RyR2-P2328S in the presence or absence of PKA and S36.
[087] Figure 7, embodiments A and B, are, respectively, (A) an immunoblot of RyR2 immunoprecipitated with an antibody against RyR2, and immunoblots of RyR2 PKA phosphorylation at Ser-2809 and calstabin2; and (B) a bar graph quantifying the relative amount of PKA phosphorylated RyR2 at Ser-2808 (corresponding to human Ser-2809) bound to RyR2 in wild-type (contol) and calstabin2-defϊcient (FKBPl 2.6'1') mice.
[088] Figure 8, embodiments A, B, and C, are, respectively, bar graphs of (A) quantitative in vivo M-mode echocardiograms comparing ejection fraction (EF) before and following sham operation or permanent left anterior descending (LAD) coronary artery ligation in wild- type and RyR2-S2808A knockin mice; (B) in vivo pressure-volume loop quantification of maximal pressure change over time (dP/dt) in wild-type and RyR2-S2808A knockin mice after sham operation or permanent left anterior descending coronary artery (LAD) ligation; and (C) quantitative M-mode echocardiographic assessment of end-systolic diameter (ESD) in wildtype and RyR2-S2808A knockin mice after sham operation or permanent left anterior descending coronary artery (LAD) ligation.
[089] Figure 9, embodiments A, B, C, and D demonstrate the effect of JTV-519 on calstabiiώ affinity to RyR2 in haploinsufficient calstabin2 (FKBPl 2.6)+/~ mice after exercise. Embodiment A is immunoblots of equivalent amounts of RyR2 immunoprecipitated with an antibody against RyR2 (top). Embodiment B are bar graphs showing the amount of PKA phosphorylation of RyR2 at Ser-2809 and the amount of calstabin2 bound to RyR2 from control animals and animals after exercise immediately followed by injection with 0.5 mg/kg epinephrine. Embodiment C are single-channel tracings of RyR2 channels isolated from hearts of haploinsufficient calstabin2+/~ and calstabin2^~ deficient mice immediately following exercise testing and injection of 0.5 mg epinephrine per kg of body weight, both untreated (top) and after treatment (middle and bottom) with JTV-519. Average open probality (Po), open time (To) and average closed time (Tc) are as indicated; and closed state is indicated by 'c'. The dotted lines indicate subconductance levels for partial RyR2 openings. Embodiment D is a bar graph summarizing average open probabilities of single RyR2 channels from haploinsufficient calstabin2+/~ and calstabin2"/~ deficient mice after exercise with and without JTV-519 treatment. "*" indicates significance level P < 0.05. Numbers in the bars indicate the number of single channels measured.
[090] Figure 103 embodiments A, B, C, D, E, and F demonstrate normalized RyR2 channel gating and increased calstabin2 binding to RyR2 channels after treatment with JTV-519. Embodiment A are immunoblots of immunoprecipitated wild- type RyR2 (RyR2-WT) channels phosphorylated by PKA in the absence or presence of the inhibitor peptide PKIs-24 and incubated with calstabin2 (250 nM) at the indicated JTV-519 concentrations; the immunoblots show the amount of RyR2 (top) and the amount of calstabin2 (bottom) associated with immunoprecipitated RyR2 after incubation with or without the indicated concentrations of JTV-519. Embodiment B is immunoblots of RyR2-S2809D, which mimics constitutive PKA phosphorylation of RyR2, analyzed as in Embodiment A. Embodiment C are binding curves of 35S-radiolabeled calstabin2 to unphosphorylated or PKA phosphorylated RyR2 or to RyR2-S2809D in the presence or absence of JTV-519, documenting differences in the calstabin2 binding affinity for RyR2. Embodiments D, E, and F are single channel tracings (left) and amplitude histograms (right) of PKA-phosphorylated RyR2s (embodiments E and F) or unphosphorylated (embodiment D in presence of PKA inhibitor PKI4) incubated with calstabin2 (250 nM) with (embodiment F) or without (embodiments D and E) JTV-519 (1 μM). Single-channel tracings are shown at 150 nM [Ca2+] which mimics the diastolic or resting phase in the heart; channel openings are upward, the dash indicates the full level of channel opening (4 pA), and "c" indicates the closed state of the channels. The amplitude histograms have amplitude represented on the x axis, and events on the y axis indicates the number of channel openings.
[091] Figure 11, embodiments A, B, C, D, E, and F demonstrate that RyRl channel function is increased and normalized in mdx mice treated with JTV-519. Embodiments A and B are, respectively, a single channel current trace and an amplitude histogram of RyRl from soleus muscle of a control (wild-type) mouse under resting conditions. Embodiments C and D are, respectively, a single channel current trace and an amplitude histogram of RyRl from soleus muscle of an mdx mouse. Embodiment E and F are, respectively, a single channel current trace and an amplitude histogram of RyRl from soleus muscle of an mdx mouse treated with JTV-519.
[092] Figure 12, embodiments A and B, are, respectively, immunoblots of RyRl, RyRl- pSer2843, and RyRl -associated calstabinl in mdx mice and wild-type mice; and bar graphs of the relative amounts of RyRl-pSer2843 and calstabinl in mdx and wild-type mice.
[093] Figure 13, embodiments A3 B, and C, demonstrate that a SR Ca2+ leak is detectable in the skeletal muscles of animals with heart failure. Embodiments A and B are fluorescence line scan images of Ca2+ sparks in myofϊbers from, respectively, sham and postmyocardial infarction (PMI) rats. Embodiment C provides bar graphs summarizing the amplitude, rise time, FDHM, and FWHM of the Ca2+ sparks for the sham (open symbols) and PMI (closed symbols) rats.
[094] Figure 14, embodiments A and B, demonstrate that treatment of wild-type mice with JTV-519 improves soleus muscle fatigue times. Embodiment A provides maximal tetanic force fatigue time tracings for wild-type and calstab^"'" mice, treated with JTV-519 or placebo as indicated. Embodiment B are bar graphs summarizing the mean time to fatigue for wild-type and calstabin2" " mice, treated with JTV-519 or placebo as indicated.
[095] Figure 15, embodiments A and B, demonstrate that the beneficial effects of JTV-519 treatment on skeletal muscle function depend on calstabinl and not calstabin2 binding to RyRl. Embodiment A provides bar graphs of PKA phosphorylation of RyRl at Ser-2844 in mice, which corresponds to Ser-2843 in humans. Embodiment B are bar graphs of the amount of calstabinl bound to RyRl from wild-type and calstabin2"A mice treated with JTV- 519 or placebo.
[096] Figure 16, embodiments A, B, C, and D demonstrate that JTV-519 normalizes abnormal or leaky RyRl channel function in vivo. Embodiments A and C are single channel current traces of wild-type mice with heart failure treated with placebo (A) and JTV-519 (C). Embodiments B and D are amplitude histograms for wild-type mice with heart failure treated with placebo (B) and JTV-519 (D).
[097] Figure 17, embodiments A and B, demonstrate that JTV-519 increases calstabin binding to PKA phosphorylated RyR. Embodiment A are immunoblots of calstabinl incubated and associated with RyRl and calstabin2 incubated and associated with RyR2 at increasing concentrations of JTV-519. Embodiment B provides graphs summarizing the proportion of calstabinl and calstabin2 bound to RyRl and RyR2, as indicated.
[098] Figure 18, embodiments A, B, C, and D, demonstrate that PKA phosphorylation of Ser-2843 increases the open probability and gating kinetics of RyRl channels. Embodiment A provides single channel current traces and corresponding histogram of wild-type RyRl. Embodiment B provides single channel current traces and corresponding histogram of wild- type RyRl that is PKA phosphorylated. Embodiment C provides single channel current traces and corresponding histogram of RyRl -Ser-2843 A. Embodiment D provides single channel current traces and corresponding histogram of RyRl-Ser-2843D.
[099] Figure 19, embodiments A and B, demonstrate the PKA hyperphosphorylation and calstabinl deficiency of RyRl channels after sustained exercise. Embodiment A are immunoblots of RyRl, RyRl -pSer2844, RyRl -pSer2849, and calstabinl for control and swim mice following an exercise regime. Embodiment B is a bar graph summarizing the relative amounts of the indicated compounds following the exercise regime.
[0100] Figure 20, embodiments A and B, demonstrate that RyRl PKA phosphorylation increases after exposure to increasing durations of sustained exercise. Embodiment A provides immunoblots of RyRl and RyRl- pSer2844 following increasing durations of sustained exercise. Embodiment B is a graph showing the relative PKA phosphorlation of RyRl for varying durations of exercise.
[0101] Figure 21, embodiments A, B, and C, demonstrate that RyRl PKA phosphorylation increases with muscle fatigue. Embodiments A and B are, respectively, fatigue time tracings and a bar graph showing mean fatigue times for rat soleus muscle of heart failure and control subjects. Embodiment C is a graph of PKA phosphorylation versus fatigue time.
[0102] Figure 22 are trichrome and hematoxylin-eosin stains of cross-sections of the mouse M. extensor digitorum longus, and demonstrates myofiber degeneration consistent with dystrophic remodeling following sustained exercise.
[0103] Figure 23 shows a sample hERG current trace before (control) and after application of ARM036 at 100 μM. Also shown is the voltage pulse protocol used to evoke the hERG currents.
[0104] Figure 24 shows a typical time course of the effect of ARM036 on hERG current amplitude. Application of 10 μM ARM036 is indicated by the horizontal bar.
[0105] Figure 25 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM036 at various concentrations.
[0106] Figure 26 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM036-Na at various concentrations.
[0107] Figure 27 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM047 at various concentrations.
[0108] Figure 28 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM048 at various concentrations.
[0109] Figure 29 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM050 at various concentrations.
[0110] Figure 30 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM057 at various concentrations.
[0111] Figure 31 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM064 at various concentrations.
[0112] Figure 32 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM074 at various concentrations.
[0113] Figure 33 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM075 at various concentrations.
[0114] Figure 34 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM076 at various concentrations. [0115] Figure 35 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM077 at various concentrations.
[0116] Figure 36 is a concentration-response graph showing the percent inhibition of hERG current after application of ARMlOl at various concentrations.
[0117] Figure 37 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM 102 at various concentrations.
[0118] Figure 38 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM 103 at various concentrations.
[0119] Figure 39 is a concentration-response graph showing the percent inhibition of hERG current after application of ARM 104 at various concentrations.
[0120] Figure 40 is a concentration-response graph showing the percent inhibition of hERG current after application of ARMl 06 at various concentrations.
[0121] Figure 41 is a concentration-response graph showing the percent inhibition of hERG current after application of ARMl 07 at various concentrations.
[0122] Figure 42 is a concentration-response graph showing the percent inhibition of hERG current after application of S26 at various concentrations.
[0123] Figure 43 is a concentration-response graph showing the percent inhibition of hERG current after application of JTV-519 ("ARMOXX") at various concentrations.
DETAILED DESCRIPTION OF THE INVENTION
[0124] As used herein and in the appended claims, the singular foπns "a," "an," and "the" include plural references unless the content clearly dictates otherwise. Thus, for example, reference to "an agent" includes a plurality of such agents and equivalents thereof known to those skilled in the art, and reference to "the FKBP12.6 polypeptide" is a reference to one or more FKBP12.6 polypeptides (also known as calstabin2) and equivalents thereof known to those skilled in the art, and so forth. AU publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.
[0125] The following are definitions of terms used in the present specification. The initial definition provided for a group or term herein applies to that group or term throughout the present specification individually or as part of another group, unless otherwise indicated. [0126] As used herein, the term "RyCaI compounds" refers to compounds of the general Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1, 1-m or II as provided by the invention, and herein referred to as "compound(s) of the invention".
[0127] The compounds of the invention are referred using a numerical naming system, with compound numbers 1 to 123 provided herein. These numbered compounds are referred to using either the prefix "S" or the prefix "ARM." Thus, the first numbered compound is referred to either as "Sl" or "ARMOOl", the second numbered compound is referred to as either "S2" or "ARM002", the third numbered compound is referred to as either "S3" or "ARM003", and so on. The "S" and the "ARM" nomenclature systems are used interchangeably throughout the specification, the drawings, and the claims.
[0128] The term "alkyl" as used herein refers to a linear or branched, saturated hydrocarbon having from 1 to 6 carbon atoms. Representative alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, hexyl, isohexyl, and neohexyl. The term "C1-C4 alkyl" refers to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 4 carbon atoms, such as methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, and isobutyl.
[0129] The term "alkenyl" as used herein refers to a linear or branched hydrocarbon having from 2 to 6 carbon atoms and having at least one carbon-carbon double bond. In one embodiment, the alkenyl has one or two double bonds. The alkenyl moiety may exist in the E or Z conformation and the compounds of the present invention include both conformations.
[0130] The term "alkynyl" as used herein refers to a linear or branched hydrocarbon having from 2 to 6 carbon atoms and having at least one carbon-carbon triple bond.
[0131] The term "aryl" as used herein refers to an aromatic group containing 1 to 3 aromatic rings, either fused or linked.
[0132] The term "cyclic group" as used herein includes a cycloalkyl group and a heterocyclic group.
[0133] The term "cycloalkyl group" as used herein refers to a three- to seven-membered saturated or partially unsaturated carbon ring. Any suitable ring position of the cycloalkyl group may be covalently linked to the defined chemical structure. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl,
[0134] The term "halogen" as used herein refers to fluorine, chlorine, bromine, and iodine. [0135] The term "heterocyclic group" or "heterocyclic" or "heterocyclyl" or "heterocyclo" as used herein refers to fully saturated, or partially or fully unsaturated, including aromatic (i.e., "heteroaryl") cyclic groups (for example, 4 to 7 membered monocyclic, 7 to 11 membered bicyclic, or 10 to 16 membered tricyclic ring systems) which have at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3, or 4 heteroatoms selected from nitrogen atoms, oxygen atoms and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. The heterocyclic group may be attached to the remainder of the molecule at any heteroatom or carbon atom of the ring or ring system. Exemplary heterocyclic groups include, but are not limited to, azepanyl, azetidinyl, aziridinyl, dioxolanyl, furanyl, furazanyl, homo piperazinyl, imidazolidinyl, imidazolinyl, isothiazolyl, isoxazolyl, morpholinyl, oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, pyrirm'dinyl, phenanthridinyl, phenanthrolinyl, piperazinyl, piperidinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiomorpholinyl, thiophenyl, triazinyl, and triazolyl. Exemplary bicyclic heterocyclic groups include indolyl, isoindolyl, benzothiazolyl, benzoxazolyl, benzoxadiazolyl, benzothienyl, quinuclidinyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuryl, benzofurazanyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl (such as furo[2,3-c]pyridinyl, furo[3,2-b]pyridinyl] or furo[2,3-b]pyridinyl), dihydroisoindolyl, dihydroquinazolinyl (such as 3,4-dihydro-4-oxo- quinazolinyl), triazinylazepinyl, tetrahydroquinolinyl and the like. Exemplary tricyclic heterocyclic groups include carbazolyl, benzidolyl, phenanthrolinyl, acridinyl, phenanthridinyl, xanthenyl and the like.
[0136] The term "phenyl" as used herein refers to a substituted or unsubstituted phenyl group.
[0137] The aforementioned terms "alkyl," "alkenyl," "alkynyl," "aryl," "phenyl," "cyclic group," "cycloalkyl," "heterocyclyl," "heterocyclo," and "heterocycle" may further be optionally substituted with one or more substituents. Exemplary substituents include but are not limited to one or more of the following groups: hydrogen, halogen, CF3, OCF3, cyano, nitro, N3, oxo, cycloalkyl, alkenyl, alkynyl, heterocycle, aryl, alkylaryl, heteroaryl, ORa, SR3, S(K))Re,
Figure imgf000036_0001
P(=O)2ORa, NRbRc, NRbS(=0)2Re, NRbP(=0)2Re,
Figure imgf000036_0002
OC(=O)Ra, OC(=O)NRbRc, NRbC(=0)0Ra, NRdC(=0)NRbRo, NRdSC=O)2NRbR0, NRdP(=0)2NRbRc, NRbC(=0)Ra, or NRbP(=O)2Re, wherein Ra is hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylaryl, heteroaryl, heterocycle, or aryl; Rb, R0 and Rd are independently hydrogen, alkyl, cycloalkyl, alkylaryl, heteroaryl, heterocycle, aryl, or said Rb and R0 together with the N to which they are bonded optionally form a heterocycle; and Re is alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, alkylaryl, heteroaryl, heterocycle, or aryl. In the aforementioned exemplary substitutents, groups such as alkyl, cycloalkyl, alkenyl, alkynyl, cycloalkenyl, alkylaryl, heteroaryl, heterocycle and aryl can themselves be optionally substituted.
[0138] Exemplary substituents may further optionally include at least one labeling group, such, as a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group. A fluorescent labeling group can be selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof. For example, ARMl 18 of the present invention contains a labeling group BODIPY, which is a family of fluorophores based on the 4,4-difluoro-4-bora-3a,4a-diaza-,s'-indacene moiety. For further information on fluorescent label moieties and fluorescence techniques, see, e.g., Handbook of Fluorescent Probes and Research Chemicals, by Richard P. Haughland, Sixth Edition, Molecular Probes, (1996), which is hereby incorporated by reference in its entirety. One of skill in the art can readily select a suitable labeling group, and conjugate such a labeling group to any of the compounds of the invention, without undue experimentation.
[0139] The term "quaternary nitrogen" refers to a tetravalent positively charged nitrogen atom including, for example, the positively charged nitrogen in a tetraalkylammonium group (e.g., tetramethylammonium, N-methylpyridinium), the positively charged nitrogen in protonated ammonium species (e.g., trimethyl-hydroammonium, N-liydropyridinium), the positively charged nitrogen in amine N-oxides (e.g., N-methyl-morpholine-N-oxide, pyridine-N-oxide), and the positively charged nitrogen in an N-amino-ammonium group (e.g., N-aminopyridinium).
[0140] Throughout the specification, unless otherwise noted, the nitrogen in the benzothiazepine ring of compounds of the present invention may optionally be a quaternary nitrogen. Non-limiting examples include ARM-113 and ARM-119. [0141] Compounds of the present invention may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present invention.
[0142] The term "prodrug" as employed herein denotes a compound that, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield compounds of the present invention.
[0143] All stereoisomers of the compounds of the present invention (for example, those which may exist due to asymmetric carbons on various substituents), including enantiomeric forms and diastereomeric forms, are contemplated within the scope of this invention. Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers (e.g., as a pure or substantially pure optical isomer having a specified activity), or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The cbiral centers of the present invention may have the S or R configuration as defined by the IUPAC 1974 Recommendations. The racemic forms can be resolved by physical methods, such as, for example, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography. The individual optical isomers can be obtained from the racemates by any suitable method, including without limitation, conventional methods, such as, for example, salt formation with an optically active acid followed by crystallization.
[0144] Compounds of the present invention are, subsequent to their preparation, preferably isolated and purified to obtain a composition containing an amount by weight equal to or greater than 99% of the compound ("substantially pure" compound), which is then used or formulated as described herein. Such "substantially pure" compounds of the present invention are also contemplated herein as part of the present invention.
[0145] All configurational isomers of the compounds of the present invention are contemplated, either in admixture or in pure or substantially pure form. The definition of compounds of the present invention embraces both cis (Z) and trans (E) alkene isomers, as well as cis and trans isomers of cyclic hydrocarbon or heterocyclic rings.
[0146] Throughout the specifications, groups and substituents thereof may be chosen to provide stable moieties and compounds.
[0147] The present invention provides compounds that are capable of treating and preventing disorders and diseases associated with the RyR receptors that regulate calcium channel functioning in cells. More particularly, the present invention provides compounds that are capable of treating or preventing a leak in RyR channels. "Disorders and diseases associated with the RyR receptors" means disorders and diseases that can be treated and/or prevented by modulating the RyR receptors that regulate calcium channel functioning in cells. "Disorders and diseases associated with the RyR receptors" include, without limitation, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome. Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise- induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure. Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof. Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence. Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss.
Compounds
[0148] In one embodimentm, the present invention provides compounds of Formula I:
Figure imgf000038_0001
wherein, n is 0, I5 or 2; q is O, 1, 2, 3, or 4; each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(O)2CF3, acyl, -O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero- )arylamino; wherein each acyl, -O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be optionally substituted;
R1 is selected from the group consisting of H, oxo, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
R2 is selected from the group consisting of H, -C(=0)R5, -C(=S)R6, -SO2R7, -P(=O)R8R9, - (CH2)U1-R1O, alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl maybe optionally substituted;
R3 is selected from the group consisting of H , -CO2Y, -C(=0)NHY, acyl, -O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted; and wherein Y is selected from the group consisting of H, alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl, and wherein each alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
R4 is selected from the group consisting of H, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be optionally substituted;
R5 is selected from the group consisting Of-NRi5Ri6, -(CH2)qNRi5Ri6, -NHNRi5Ri6, - NHOH, -ORi5, -C(=O)NHNR15Ri6, -CO2R]5, -C(O)NRi5Ri6, -CH2X, acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted, and wherein q is 1 , 2, 3, 4, 5, or 6;
R6 is selected from the group consisting of -ORi5, -NHNRi5Ri6, -NHOH, -NR15Ri6, -CH2X, acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
R7 is selected from the group consisting Of-ORj5, -NRi5Ri6, -NHNR15Ri6, -NHOH, -CH2X, alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
R8 and R9 independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted;
R10 is selected from the group consisting of -NRi5Ri6, OH, -SO2Rn, -NHSO2Rn, C(=O)(R12), NHC=O(Ri2), -OC=O(Ri2), and -P(=O)R13R14;
R11, R12, R13, and Ri4 independently are selected from the group consisting of H, OH, NH2, - NHNH2, -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl maybe optionally substituted;
X is selected from the group consisting of halogen, -CN, -CO2Ri5, -C(=O)NRi5Ri6, -NR15RI β, -OR15, -SO2R7, and -P(=O)R8R9; and
Ri5 and R16 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be optionally substituted; and optionally Ri 5 and Ri6 together with the N to which they are bonded may form a heterocycle which may be substituted; the nitrogen in the benzothiazepine ring may optionally be a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and prodrugs thereof; provided that when q is 0 and n is 0, then R2 is not H, Et, -C(=O)NH2, (O)NHPh, - C(=S)NH-nButyl, -C(O)NHC(O)CH2Cl, -C(=O)H, -C(O)Me, -C(O)Et, - C(O)CH=CH2, -S(=O)2Me, or -S(O)2Et; further provided that when q is O and n is 1 or 2, then R2 is not -C(O)Me, -C(=O)Et, - S(O)2Me5 or -S(=O)2Et; further provided that when q is 1, and R is Me, Cl, or F at the 6 position of the benzothiazepene ring, then R2 is not H, Me, -C(O)H, -C(=O)Me, -C(O)Et, -C(O)Ph, - S(O)2Me, or -S(O)2Et; and further provided that when q is 1, n is O, and R is OCT3, OH, C1-C3 alkoxyl at the 7 position
of the benzothiazepene ring, then R2 is not H, -C(O)CHOH2, or
Figure imgf000041_0001
[0149] In one embodiment, the present invention provides compounds of Formula I, as described above, with the proviso that said compound is not S24 or S68.
[0150] In one embodiment, the present invention provides compounds of Formula I-a:
Figure imgf000041_0002
wherein: n is 0, 1, or 2; q is O, 1, 2, 3, or 4; each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H5 -S(O)2alkyl, -S(O)alkyl, -OS(O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted or unsubstituted;
R2 is selected from the group consisting of H, -C=O(R5), -C=S(R6), -SO2R7, -P(=O)R8R9, - (CH2)m-Rio, alkyl, aryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl maybe substituted or unsubstituted;
R5 is selected from the group consisting of-NRi5Ri6, -NHNRisRiβ, -NHOH, -OR15, -
Figure imgf000042_0001
-CO2R15,
Figure imgf000042_0002
-CH2X, acyl, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R6 is selected from the group consisting of -ORi5, -NHNRi5Ri6, -NHOH, -NR15R16, -CH2X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R7 is selected from the group consisting of H, -OR15, -NR15Ri6, -NHNRi5Ri6, -NHOH, - CH2X, alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R8 and R9 independently are selected from the group consisting of -OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
Rio is selected from the group consisting of -NRi5Ri6, OH, -Sθ2Rπ, -NHSO2R11, - C(=0)R12, -NH(C=O)Ri2, -0(C=O)R12, and -P(=O)R]3Ri4; m is 0, 1, 2, 3, or 4;
Rn, R12, Ri3j and Rμ independently are selected from the group consisting of H, OH, NH2, - NHNH2, -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; X is selected from the group consisting of halogen, -CN, -CO2R15, -C(=O)NRi5Ri6, -NRiSR16, -ORis, -SO2R7, and -P(O)R8R9; and
R15 and R16 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted; and optionally R15 and R16 together with the N to which they are bonded may form a heterocycle which may be substituted or unsubstituted; the nitrogen in the benzothiazepine ring may be optionally a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and prodrugs thereof; provided that when q is O and n is O, then R2 is not H, Et, -C(=O)NH2, (=O)NHPh, - C(=S)NH-nButyl, -C(=O)NHC(=O)CH2C1, -C(O)H, -C(=0)Me, -C(=O)Et, - C(=O)CH=CH2, -Si=O)2Me, or -S(=O)2Et; further provided that when q is O and n is 1 or 2, then R2 is not -C(=O)Me, -C(O)Et, - S(=O)2Me, or -S(O)2Et; further provided that when q is 1 , and R is Me, Cl, or F at the 6 position of the benzothiazepene ring, then R2 is not H, Me, -C(O)H, -C(=0)Me, -C(=O)Et, -C(=O)Ph, - S(=O)2Me, or -S(=O)2Et; and further provided that when q is 1, n is O, and R is OCT3, OH, C1-C3 alkoxyl at the 7 position
of the benzothiazepene ring, then R2 is not H, -C(O)CHOH2, or
Figure imgf000043_0001
[0151] In certain embodiments, the present invention provides compounds of formula I-a, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)CrC4alkyl, -S-Ci- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(O)Me, -OC(O)Me, morpholinyl and propenyl; and n is O, 1, or 2.
[0152] In other embodiments, the present invention provides compounds of foπnula I-a, wherein R2 is selected from the group consisting Of-C=O(R5), -C=S(R6), -SO2R7, - P(O)R8R9, and -(CH2V-Ri0. [0153] In yet another embodiment, the present invention provides compounds of formula I-b:
Figure imgf000044_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R2 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0154] In certain embodiments, the present invention provides compounds of formula I-b, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)C1-C4alkyl, -S-C1- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[0155] In other embodiments, the present invention provides compounds of formula I-b, wherein R2 is selected from the group consisting Of-C=O(R5), -C=S(R6), -SO2R7, - P(^O)R8R9, and -(CH2)m-R10.
[0156] In yet another embodiment, the present invention provides compounds formula of I-c:
Figure imgf000045_0001
wherein each R, R7, q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0157] In certain embodiments, the present invention provides compounds of formula I-c, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -St=O)2C1 -C4alkyl, -S(=O)C1-C4alkyl, -S-C1- C4alkyl, -OS(^O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -0C(=0)Me, morpholinyl and propenyl; and n is 0, 1, or 2.
[0158] In other embodiments, the present invention provides compounds of formula I-c, wherein R7 is selected from the group consisting of -OH, -NR^R^, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
[0159] In a further embodiment, the present invention provides compounds of formula of I- d:
Figure imgf000045_0002
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R7 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0160] In certain embodiments, the present invention provides compounds of formula I-d, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SC=O)2C1 -C4alkyl, -S(=O)CrC4alkyl, -S-Ci- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -0C(=0)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[0161] In other embodiments, the present invention provides compounds of formula I-d, wherein R7 is selected from the group consisting of -OH, -NR15R16, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted.
[0162] In one embodiment, the present invention provides compounds of formula of I-e:
Figure imgf000046_0001
wherein each R, R5, q and n is as defined compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0163] In certain embodiments, the present invention provides compounds of formula I-e, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2CrC4alkyl, -SC=O)C1 -C4alkyl, -S-C1- Qalkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(ND)Me, -OC(=O)Me, morpholinyl and propenyl; and n is 0, 1, or 2.
[0164] In other embodiments, the present invention provides compounds of formula I-e, wherein R5 is selected from the group consisting Of-NRi5R16, -NHOH, -OR15, -CH2X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
[0165] In some embodiments, the present invention provides compounds of formula I-e, wherein R5 is an alkyl substituted by at least one labeling group, such as a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group. A fluorescent labeling group can be selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
[0166] In another embodiment, the present invention provides compounds of formula of I-f:
Figure imgf000047_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R5 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof. [0167] In certain embodiments, the present invention provides compounds of formula I-f, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2CrC4alkyl, -SC=O)C1 -C4alkyl, -S-C1- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -OC(=O)Me, morpholinyl andpropenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[0168] In other embodiments, the present invention provides compounds of formula I-f, wherein R5 is selected from the group consisting Of-NR15R16, -NHOH, -OR1S, -CH2X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
[0169] In yet another embodiment, the present invention provides compounds of formula of I-g:
Figure imgf000048_0001
wherein W is S or O; each R, R15, R16, q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0170] In certain embodiments, the present invention provides compounds of formula I-g, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)C1-C4alkyl, -S-C1- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O, 1, or 2.
[0171] In other embodiments, the present invention provides compounds of formula I-g, wherein R15 and Ri 6 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylallcyl may be substituted; and optionally R15 and R16 together with the N to which they are bonded may form a heterocycle which may be substituted.
[0172] In some embodiments, the present invention provides compounds of formula I-g, wherein W is O or S.
[0173] In yet another embodiment, the present invention provides compounds of formula of I-h:
Figure imgf000049_0001
wherein W is S or O; wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
Ri5, R16 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0174] In certain embodiments, the present invention provides compounds of formula I-h, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(^O)2C1 -Qalkyl, -S(=O)C1-C4alkyl, -S-Cj- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -0C(=0)Me, morpholinyl and propenyl; and n is 0, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[0175] In other embodiments, the present invention provides compounds of formula I-h, wherein R15 and Ri6 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted; and optionally R15 and R16 together with the N to which they are bonded may form a heterocycle which may be substituted.
[0176] In some embodiments, the present invention provides compounds of formula I-g, wherein W is O or S.
[0177] In a further embodiment, the present invention provides compounds of formula of I-i:
Figure imgf000050_0001
wherein R17 is selected from the group consisting Of-NR15R16, -NHNRisRiδ, -NHOH, -OR1S, -CH2X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; each R, q, and n is as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0178] In certain embodiments, the present invention provides compounds of formula I-i, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S^O^Q-Qalkyl, -S(=O)d-C4alkyl, -S-Ci- C4alkyl, -OS(^O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -0C(=0)Me, morpholinyl and propenyl; and n is 0, 1, or 2.
[0179] In other embodiments, the present invention provides compounds of formula I-i, wherein R17 is -NRI5RI6, and -OR15. In certain other embodiments, Ri7 is -OH, -OMe, -NEt, -NHEt3 -NHPh, -NH2, or -NHCH2pyridyl.
[0180] In one embodiment, the present invention provides compounds of formula of I-j:
Figure imgf000051_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R17 is selected from the group consisting Of-NRi5R16, -NHNRi5R16, -NHOH, -OR15, -CH2X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; n is as defined in compounds of formula I-a; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0181] In certain embodiments, the present invention provides compounds of formula I-j, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)Ci-C4alkyl, -S-Q- C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(O)Me, -OC(O)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[0182] In other embodiments, the present invention provides compounds of formula I-j, wherein Ri7 is -NR15Ri6 Or-ORi5. In certain other embodiments, Ri7 is -OH, -OMe, -NEt, - NHEt, -NHPh, -NH2, or -NHCH2pyridyl.
[0183] In another embodiment, the present invention provides compounds of formula I-k:
Figure imgf000052_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R18 is selected from the group consisting Of-NR15R16, -Q=O)NR15R16, -(C=O)OR15, -OR15, alkyl, aryl, cycloalkyl, heterocyclyl, and at one labeling group; wherein each alkyl, aryl, cycloalkyl, and heterocyclyl may be substituted or unsubstituted; wherein p is 1, 2, 3, 4, 5, 6, 7, 8 9, or 10; and n is O, 1, or 2; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0184] In certain embodiments, the present invention provides compounds of formula I-k, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2C1-C4alkyl, -S(=O)Ci-C4alkyl, -S-Ci- Qalkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -0C(=0)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[0185] In other embodiments, the present invention provides compounds of formula I-k, wherein Rj8 is selected from the group consisting Of-NR15Ri6, -(C=O)ORi5, -ORi5, alkyl, aryl, and at one labeling group; and wherein each alkyl and aryl may be substituted or unsubstituted. In some cases, m is 1, and R18 is Ph, C(=0)0Me, C(=O)OH, aminoalkyl, NH2, NHOH, or NHCbz. In other cases, m is O, and Ris is Ci-C4 alkyl, such as Me, Et, propyl, and butyl. In yet other cases, m is 2, and Rj8 is pyrrolidine, piperidine, piperazine, or morpholine. In some embodiments, m is 3, 4, 5, 5, 7, or 8, and Ri8 is a fluorescent labeling group selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
[0186] In yet another embodiment, the present invention provides compounds of formula of 1-1:
Figure imgf000053_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R6 and n are as defined in compounds of formula I-a; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0187] In certain embodiments, the present invention provides compounds of formula 1-1, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)Ci-C4alkyl, -S-C1- Qalkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, -OC(=O)Me, morpholinyl and propenyl; and n is 0, 1 or 3. In some cases, R' is H or OMe3 and R" is H.
[0188] In other embodiments, the present invention provides compounds of formula 1-1, wherein R6 is selected from the group consisting of -NRisR-ie,
Figure imgf000053_0002
-ORi5, -NHOH, - CH2X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted. In some cases, R6 is -NRi5R16 such as -NHPh, pyrrolidine, piperidine, piperazine, morpholine, and the like. In some other cases, R6 is alkoxyl, such as -O-tBu.
[0189] In a further embodiment, the present invention provides compounds of formula I-m:
Figure imgf000054_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN3 -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
Rs, R9 and n are as defined in compounds of formula I-a above; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
[0190] hi certain embodiments, the present invention provides compounds of formula I-m, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci-C4alkyl, -S(=O)Ci-C4alkyl, -S-Q- Qalkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, -0C(=0)Me, morpholinyl and propenyl; and n is O, 1 or 3. In some cases, R' is H or OMe, and R" is H.
[0191] In other embodiments, the present invention provides compounds of formula I-m, wherein R8 and R9 are independently alkyl, aryl, -OH, alkoxyl, or alkylamino. In some cases, R8 is Ci-C4 alkyl such as Me, Et, propyl and butyl; and R9 is aryl such as phenyl.
[0192] The compounds of Formula I3 I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m treat and prevent disorders and diseases associated with the RyR receptors. [0193] Examples of such compounds include, without limitation, Sl, S2, S3, S4, S5, S6, S7, S9, SIl, S12, S13, S14, S19, S20, S22, S23, S25, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S103, S104, S105, S107, S108, S109, SIlO, Si l l, S112, S113, S114, S115, S116, S117, S118, S119, S120, S121, S122, and S123. These compounds have the following structures:
Figure imgf000055_0001
Figure imgf000055_0002
Figure imgf000055_0003
Figure imgf000056_0001
Figure imgf000056_0002
Figure imgf000056_0003
Figure imgf000056_0004
Figure imgf000057_0001
Figure imgf000057_0002
Figure imgf000057_0003
Figure imgf000057_0004
Figure imgf000058_0001
Figure imgf000058_0002
Figure imgf000058_0003
S22
Figure imgf000059_0001
Figure imgf000059_0002
Figure imgf000059_0003
S25
Figure imgf000059_0004
S26
Figure imgf000060_0001
Figure imgf000060_0002
Figure imgf000060_0003
Figure imgf000060_0004
S43
Figure imgf000061_0001
Figure imgf000061_0002
Figure imgf000061_0003
Figure imgf000061_0004
Figure imgf000062_0001
Figure imgf000062_0002
Figure imgf000062_0003
Figure imgf000062_0004
Figure imgf000063_0001
Figure imgf000063_0002
Figure imgf000063_0003
Figure imgf000063_0004
Figure imgf000064_0001
Figure imgf000064_0002
Figure imgf000064_0003
Figure imgf000065_0001
Figure imgf000065_0002
Figure imgf000065_0003
Figure imgf000065_0004
Figure imgf000066_0001
Figure imgf000066_0002
Figure imgf000066_0003
Figure imgf000067_0001
S68
Figure imgf000067_0002
S69
O
Figure imgf000067_0003
Figure imgf000068_0001
S71
Figure imgf000068_0002
S72
Figure imgf000068_0003
Figure imgf000068_0004
S74
Figure imgf000069_0001
Figure imgf000069_0002
S76
Figure imgf000069_0003
Figure imgf000070_0001
S78
Figure imgf000070_0002
S79
Figure imgf000070_0003
Figure imgf000071_0001
S81
Figure imgf000071_0002
S82
Figure imgf000071_0003
S83
Figure imgf000071_0004
Figure imgf000072_0001
Figure imgf000072_0002
Figure imgf000072_0003
Figure imgf000072_0004
Figure imgf000073_0001
S89
Figure imgf000073_0002
Figure imgf000073_0003
S91
Figure imgf000074_0001
S92
Figure imgf000074_0002
S93
Figure imgf000074_0003
S94
Figure imgf000074_0004
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Arm 120 [0194] In one embodiment of the present invention, for compounds of Formula I, if R2 is
C=O(R5) or SO2R7, then R is at positions 2, 3, or 5 on the benzene ring.
[0195] In another embodiment of the invention, for compounds of Formula I, if R2 is C=O(Rs) or SO2R7, then each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -N3, -SO3H, acyl, alkyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero- )arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl.
[0196] In another embodiment of the invention, for compounds of Formula I, if R2 is C=O(R5) or SO2R7, then there are at least two R groups attached to the benzene ring. Furthermore, there are at least two R groups attached to the benzene ring, and both R groups are attached at positions 2, 3, or 5 on the benzene ring. Still furthermore, each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -N3, -SO3H, acyl, alkyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero- )aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero- )arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl.
[0197] In another embodiment of the invention, for compounds of Formula I, if R2 is C=O(R5), then R5 is selected from the group consisting of -NR16, NHNHR16, NHOH, -ORi5, CONH2NHR16, CONRi6, CH2X, acyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl.
[0198] In another embodiment, the present invention provides compounds of Formula II: In another embodiment, the present invention provides compounds of Formula II:
Figure imgf000082_0001
wherein R=OR'", SR'", NR'", alkyl, orhalide and R'" = alkyl, aryl, or H, and wherein R can be at position 6, 7, 8, or 9. Formula II is discussed also in co-pending application 10/680,988, the disclosure of which is incorporated herein in its entirety by reference. Routes of Activity
[0199] The compounds of the invention, such as the compounds of Formula I, I-a, I-b, I-c, I- d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, reduce the open probability of RyR by increasing the affinity of FKBP12 (calstabinl) and FKBP12.6 (calstabin2) for, respectively PKA- phosphorylated RyRl and PKA-phosphorylated RyR2. Moreover, the compounds of the invention normalize gating of mutant RyR channels, including CPVT-associated mutant RyR2 channels, by increasing FKBP12 (calstabinl) and FKBP12.6 (calstabin2) binding affinity. Therefore, the compounds of the invention revent disorders and conditions involving modulation of the RyR receptors, particularly the RyRl and RyR2 receptors. Examples of such disorders and conditions include, without limitation, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome. Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise- induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure. Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof. Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence. Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss. The compounds of the invention treat these disorders and conditions by increasing FKBP 12 (calstabinl)-RyRl binding affinity and increasing FKBP12.6 (calstabin2)-RyR2 binding affinity.
[0200] In accordance with the foregoing, the present invention provides a method for limiting or preventing a decrease in the level of RyR-bound FKBP (calstabin) in cells of a subject. As used herein, "RyR" includes RyRl, RyR2, and RyR3. Additionally, FKBP includes both FKBP12 (calstabinl) and FKBP12.6 (calstabin2). "RyR-bound FKBP" therefore refers to RyRl-bound FKBP12 (calstabinl), RyR2-bound FKBP 12.6 (calstabkώ), and RyR3-bound FKBP 12 (calstabinl).
[0201] As used herein, "RyR" also includes a "RyR protein" and a "RyR analogue." A11RyR analogue" is a functional variant of the RyR protein, having RyR biological activity, that has 60% or greater amino-acid-sequence homology with the RyR protein. The RyR of the present invention are unphosphorylated, phosphorylated (e.g., by PKA), or hyperphosphorylated (e.g., by PKA). As further used herein, the term "RyR biological activity" refers to the activity of a protein or peptide that demonstrates an ability to associate physically with, or bind with, FKBP 12 (calstabinl) in the case of RyRl and RyR3, and FKBP12.6 (calstabin2) in the case of RyR2 (i.e., binding of approximately two fold or, approximately five fold, above the background binding of a negative control), under the conditions of the assays described herein.
[0202] As used herein, "FKBP" includes both an "FKBP protein" and an "FKBP analogue," whether it be FKBP12 (calstabinl) or FKBP12.6 (calstabin2). Unless otherwise indicated herein, "protein" shall include a protein, protein domain, polypeptide, or peptide, and any fragment thereof. An "FKBP analogue" is a functional variant of the FKBP protein, having FKBP biological activity, that has 60% or greater amino-acid-sequence homology with the FKBP protein, whether it be FKBP 12 (calstabinl) or FKBP12.6 (calstabin2). As further used herein, the term "FKBP biological activity" refers to the activity of a protein or peptide that demonstrates an ability to associate physically with, or bind with, unphosphorylated or non-hyperphosphorylated RyR2 (i.e., binding of approximately two fold, or approximately five fold, above the background binding of a negative control), under the conditions of the assays described herein.
[0203] FKBP binds to the RyR channel, one molecule per RyR subunit. Accordingly, as used herein, the term "RyR-bound FKBP" includes a molecule of an FKBP 12 (calstabinl) protein that is bound to an RyRl protein subunit or a tetramer of FKBP 12 that is in association with a tetramer of RyRl, a molecule of FKBP12.6 (calstabin2) protein that is bound to an RyR2 protein subunit or a tetramer of FKBP 12.6 that is in association with a tetramer of RyR2, and a molecule of an FKBP 12 (calstabinl) protein that is bound to an RyR3 protein subunit or a tetramer of FKBP12 that is in association with a tetramer of RyR3. Therefore, "RyR-bound FKBP" refers to "RyRl-bound FKBP12," "RyR2-bound FKBP12.6," and "RyR3-bound FKBP12."
[0204] In accordance with the method of the present invention, a "decrease" or "disorder" in the level of RyR-bound FKBP in cells of a subject refers to a detectable decrease, diminution or reduction in the level of RyR-bound FKBP in cells of the subject. Such a decrease is limited or prevented in cells of a subject when the decrease is in any way halted, hindered, impeded, obstructed or reduced by the administration of compounds of the invention, such that the level of RyR-bound FKBP in cells of the subject is higher than it would otherwise be in the absence of the administered compound.
[0205] The level of RyR-bound FKBP in a subject is detected by standard assays and techniques, including those readily determined from the known art (e.g., immunological techniques, hybridization analysis, immunoprecipitation, Western-blot analysis, fluorescence imaging techniques and/or radiation detection, etc.), as well as any assays and detection methods disclosed herein. For example, protein is isolated and purified from cells of a subject using standard methods known in the art, including, without limitation, extraction from the cells (e.g., with a detergent that solubilizes the protein) where necessary, followed by affinity purification on a column, chromatography (e.g., FTLC and HPLC), immunoprecipitation (with an antibody), and precipitation (e.g., with isopropanol and a reagent such as Trizol). Isolation and purification of the protein is followed by electrophoresis (e.g., on an SDS-polyacrylamide gel). A decrease in the level of RyR-bound FKBP in a subject, or the limiting or prevention thereof, is determined by comparing the amount of RyR-bound FKBP detected prior to the administration of JTV-519 or a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, (in accordance with methods described below) with the amount detected a suitable time after administration of the compound.
[0206] A decrease in the level of RyR-bound FKBP in cells of a subject is limited or prevented, for example, by inhibiting dissociation of FKBP and RyR in cells of the subject; by increasing binding between FKBP and RyR in cells of the subject; or by stabilizing the RyR-FICBP complex in cells of a subject. As used herein, the term "inhibiting dissociation" includes blocking, decreasing, inhibiting, limiting or preventing the physical dissociation or separation of an FKBP subunit from an RyR molecule in cells of the subject, and blocking, decreasing, inhibiting, limiting or preventing the physical dissociation or separation of an RyR molecule from an FKBP subunit in cells of the subject. As further used herein, the term "increasing binding" includes enhancing, increasing, or improving the ability of phosphorylated RyR to associate physically with FKBP (e.g., binding of approximately two fold or, approximately five fold, above the background binding of a negative control) in cells of the subject and enhancing, increasing or improving the ability of FKBP to associate physically with phosphorylated RyR (e.g., binding of approximately two fold, or, approximately five fold, above the background binding of a negative control) in cells of the subject. Additionally, a decrease in the level of RyR-bound FKBP in cells of a subject is limited or prevented by directly decreasing the level of phosphorylated RyR in cells of the subject or by indirectly decreasing the level of phosphorylated RyR in the cells (e.g., by targeting an enzyme (such as PKA) or another endogenous molecule that regulates or modulates the functions or levels of phosphorylated RyR in the cells). In one embodiment, the level of phosphorylated RyR in the cells is decreased by at least 10% in the method of the present invention. In another embodiment, the level of phosphorylated RyR is decreased by at least 20%.
[0207] The subject of the present invention are in vitro and in vivo systems, including, without limitation, isolated or cultured cells or tissues, non-cell in vitro assay systems and an animal (e.g., an amphibian, a bird, a fish, a mammal, a marsupial, a human, a domestic animal (such as a cat, dog, monkey, mouse or rat) or a commercial animal (such as a cow or Pig))-
[0208] The cells of a subject include striated muscle cells. A striated muscle is a muscle in which the repeating units (sarcomeres) of the contractile myofibrils are arranged in registry throughout the cell, resulting in transverse or oblique striations that are observed at the level of a light microscope. Examples of striated muscle cells include, without limitation, voluntary (skeletal) muscle cells and cardiac muscle cells. In one embodiment, the cell used in the method of the present invention is a human cardiac muscle cell. As used herein, the term "cardiac muscle cell" includes cardiac muscle fibers, such as those found in the myocardium of the heart. Cardiac muscle fibers are composed of chains of contiguous heart- muscle cells, or cardiomyocytes, joined end to end at intercalated disks. These disks possess two kinds of cell junctions: expanded desmosomes extending along their transverse portions, and gap junctions, the largest of which lie along their longitudinal portions.
[0209] A decrease in the level of RyR-bound FKBP is limited or prevented in cells of a subject by administering the compounds of the invention to the subject; this would also permit contact between cells of the subject and the compounds of the invention. The compounds of the invention are modulators of calcium-ion channels. In addition to regulating Ca2+ levels in myocardial cells, the compounds of the invention modulate the Na+ current and the inward-rectifier K+ current in cells, such as guinea pig ventricular cells, and inhibits the delayed-rectifier K+ current in cells, such as guinea pig atrial cells.
Pharmaceutical Composition
[0210] The compounds of the invention are formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo. According to another aspect, the present invention provides a pharmaceutical composition comprising compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I- g, I-h, I-i, I-j, I-k, 1-1 or I-m, in admixture with a pharmaceutically acceptable diluent and/or carrier. The pharmaceutically-acceptable carrier must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof. The pharmaceutically-acceptable carrier employed herein is selected from various organic or inorganic materials that are used as materials for phaπnaceutical formulations and which are incorporated as analgesic agents, buffers, binders, disintegrants, diluents, emulsifiers, excipients, extenders, glidants, solubilizers, stabilizers, suspending agents, tonicity agents, vehicles and viscosity-increasing agents. If necessary, pharmaceutical additives, such as antioxidants, aromatics, colorants, flavor-improving agents, preservatives, and sweeteners, are also added. Examples of acceptable pharmaceutical carriers include carboxymethyl cellulose, crystalline cellulose, glycerin, gum arabic, lactose, magnesium stearate, methyl cellulose, powders, saline, sodium alginate, sucrose, starch, talc and water, among others.
[0211] The phaπnaceutical formulations of the present invention are prepared by methods well-known in the pharmaceutical arts. For example, the compounds of Formula I, I-a, I-b, I- c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, are brought into association with a carrier and/or diluent, as a suspension or solution. Optionally, one or more accessory ingredients {e.g., buffers, flavoring agents, surface active agents, and the like) also are added. The choice of carrier is determined by the solubility and chemical nature of the compounds, chosen route of administration and standard pharmaceutical practice.
[0212] The compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, are administered to a subject by contacting target cells {e.g., cardiac muscle cells) in vivo in the subject with the compounds. The compounds are contacted with {e.g., introduced into) cells of the subject using known techniques utilized for the introduction and administration of proteins, nucleic acids and other drugs. Examples of methods for contacting the cells with {i.e., treating the cells with) the compounds of the invention include, without limitation, absorption, electroporation, immersion, injection, introduction, liposome delivery, transfection, transfusion, vectors and other drug-delivery vehicles and methods. When the target cells are localized to a particular portion of a subject, it is desirable to introduce the compounds of the invention directly to the cells, by injection or by some other means {e.g., by introducing the compounds into the blood or another body fluid). The target cells are contained in tissue of a subject and are detected by standard detection methods readily determined from the known art, examples of which include, without limitation, immunological techniques {e.g., immunoliistochemical staining), fluorescence imaging techniques, and microscopic techniques.
[0213] Additionally, the compounds of the present invention are administered to a human or animal subject by known procedures including, without limitation, oral administration, sublingual or buccal administration, parenteral administration, transdermal administration, via inhalation or intranasally, vaginally, rectally, and intramuscularly. The compounds of the invention are administered parenterally, by epifascial, intracapsular, intracranial, intracutaneous, intrathecal, intramuscular, intraorbital, intraperitoneal, intraspinal, intrasternal, intravascular, intravenous, parenchymatous, subcutaneous or sublingual injection, or by way of catheter. In one embodiment, the agent is adiminstered to the subject by way of delivery to the subject's muscles including, but not limited to, the subject's cardiac muscles. In an embodiment, the agent is administered to the subject by way of targeted delivery to cardiac muscle cells via a catheter inserted into the subject's heart.
[0214] For oral administration, a formulation of the compounds of the invention may be presented as capsules, tablets, powders, granules, or as a suspension or solution. The foπnulation has conventional additives, such as lactose, mannitol, cornstarch or potato starch. The formulation also is presented with binders, such as crystalline cellulose, cellulose derivatives, acacia, cornstarch or gelatins. Additionally, the formulation is presented with disintegrators, such as cornstarch, potato starch or sodium carboxymethylcellulose. The formulation also is presented with dibasic calcium phosphate anhydrous or sodium starch glycolate. Finally, the formulation is presented with lubricants, such as talc or magnesium stearate.
[0215] For parenteral administration (i.e., administration by injection through a route other than the alimentary canal), the compounds of the invention are combined with a sterile aqueous solution that is isotonic with the blood of the subject. Such a formulation is prepared by dissolving a solid active ingredient in water containing physiologically-compatible substances, such as sodium chloride, glycine and the like, and having a buffered pH compatible with physiological conditions, so as to produce an aqueous solution, then rendering said solution sterile. The formulation is presented in unit or multi-dose containers, such as sealed ampoules or vials. The formulation is delivered by any mode of injection, including, without limitation, epifascial, intracapsular, intracranial, intracutaneous, intrathecal, intramuscular, intraorbital, intraperitoneal, intraspinal, intrasternal, intravascular, intravenous, parenchymatous, subcutaneous, or sublingual or by way of catheter into the subject's heart.
[0216] For transdermal administration, the compounds of the invention are combined with skin penetration enhancers, such as propylene glycol, polyethylene glycol, isopropanol, ethanol, oleic acid, N-methylpyrrolidone and the like, which increase the permeability of the skin to the compounds of the invention and permit the compounds to penetrate through the skin and into the bloodstream. The compound/enhancer compositions also may be further combined with a polymeric substance, such as ethylcellulose, hydroxypropyl cellulose, ethylene/vinylacetate, polyvinyl pyrrolidone, and the like, to provide the composition in gel form, which are dissolved in a solvent, such as methylene chloride, evaporated to the desired viscosity and then applied to backing material to provide a patch.
[0217] In some embodiments, the composition is in unit dose form such as a tablet, capsule or single-dose vial. Suitable unit doses, i.e., therapeutically effective amounts, can be determined during clinical trials designed appropriately for each of the conditions for which administration of a chosen compound is indicated and will, of course, vary depending on the desired clinical endpoint. The present invention also provides articles of manufacture for treating and preventing disorders, such as cardiac disorders, in a subject. The articles of manufacture comprise a pharmaceutical composition of one or more of the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, as described herein. The articles of manufacture are packaged with indications for various disorders that the pharmaceutical compositions are capable of treating and/or preventing. For example, the articles of manufacture comprise a unit dose of a compound disclosed herein that is capable of treating or preventing a muscular disorder, and an indication that the unit dose is capable of treating or preventing a certain disorder, for example an arrhythmia.
[0218] In accordance with a method of the present invention, the compounds of Formula I, I- a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, are administered to the subject (or are contacted with cells of the subject) in an amount effective to limit or prevent a decrease in the level of RyR-bound FKBP in the subject, particularly in cells of the subject. This amount is readily determined by the skilled artisan, based upon known procedures, including analysis of titration curves established in vivo and methods and assays disclosed herein, hi one embodiment, a suitable amount of the compounds of the invention effective to limit or prevent a decrease in the level of RyR-bound FKBP in the subject ranges from about 0.01 mg/kg/day to about 20 mg/kg/day, and/or is an amount sufficient to achieve plasma levels ranging from about 300 ng/ml to about 1000 ng/ml. In an embodiment, the amount of compounds from the invention ranges from about 10 mg/kg/day to about 20 mg/kg/day. In another embodiment, from about 0.01 mg/kg/day to about 10 mg/kg/day is administered. In another embodiment, from about 0.01 mg/kg/day to about 5 mg/kg/day is administered, In another embodiment, from about 0.05 mg/kg/day to about 5 mg/kg/day is administered, hi another, preferred embodiment, from about 0.05 mg/kg/day to about 1 mg/kg/day is administered.
Uses
[0219] The present invention provides a new range of therapeutic treatments for patients with various disorders involving modulation of the RyR receptors, particularly skeletal muscular disorders (RyRl), cardiac (RyR2) disorders, and cognitive (RyR3) disorders.
[0220] In one embodiment of the present invention, the subject has not yet developed a disorder, such as cardiac disorders (e.g., exercise-induced cardiac arrhythmia). In another embodiment of the present invention, the subject is in need of treatment for a disorder, including a cardiac disorder.
[0221] Various disorders that the compounds of the invention treat or prevent include, but are not limited to, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome. Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure. Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof. Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence. Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss. One skilled in the art will recognize still other diseases, including but not limited to muscular and cardiac disorders, that the compounds of the invention can be useful to treat, in accordance with the information provided herein.
[0222] The amount of compounds of the invention effective to limit or prevent a decrease in the level of RyR2 -bound FKBP12.6 in the subject is an amount effective to prevent exercise- induced cardiac arrhythmia in the subject. Cardiac arrhythmia is a disturbance of the electrical activity of the heart that manifests as an abnormality in heart rate or heart rhythm. As used herein, an amount of compounds of the invention "effective to prevent exercise- induced cardiac arrhythmia" includes an amount of compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, effective to prevent the development of the clinical impairment or symptoms of the exercise-induced cardiac arrhythmia (e.g., palpitations, fainting, ventricular fibrillation, ventricular tachycardia and sudden cardiac death). The amount of the compounds effective to prevent exercise-induced cardiac arrhythmia in a subject will vary depending upon the particular factors of each case, including the type of exercise-induced cardiac arrhythmia, the subject's weight, the severity of the subject's condition, and the mode of administration of the compounds. This amount is readily determined by the skilled artisan, based upon known procedures, including clinical trials, and methods disclosed herein. In one embodiment, the amount of the compounds of the invention effective to prevent the exercise-induced cardiac arrhythmia is an amount effective to prevent exercise-induced sudden cardiac death in the subject. In another embodiment, the compounds of the invention prevent exercise-induced cardiac arrhythmia and exercise- induced sudden cardiac death in the subject.
[0223] Because of its ability to stabilize RyR-bound FKBP and maintain and restore balance in the context of dynamic PKA phosphorylation and dephosphorylation of RyR, the compounds of the invention are also useful in treating a subject who has already experienced clinical symptoms of these various disorders. For example, if the symptoms of the disorder are observed in the subject early enough, the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I- f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, are effective in limiting or preventing a further decrease in the level of RyR-bound FKBP in the subject.
[0224] Additionally, the subject of the present invention is a candidate for exercise-induced cardiac disorders, such as exercise-induced cardiac arrhthmia. Exercise-induced cardiac arrhythmia is a heart condition (e.g., a ventricular fibrillation or ventricular tachycardia, including any that leads to sudden cardiac death) that develops during/after a subject has undergone physical exercise. A "candidate" for an exercise-induced cardiac disorder is a subject who is known to be, or is believed to be, or is suspected of being, at risk for developing a cardiac disorder during/after physical exercise. Examples of candidates for exercise-induced cardiac arrhythmia include, without limitation, an animal/person known to have catecholaminergic polymorphic ventricular tachycardia (CPVT); an animal/person suspected of having CPVT; and an animal/person who is known to be, or is believed to be, or is suspected of being at risk for developing cardiac arrhythmia during/after physical exercise, and who is about to exercise, is currently exercising or has just completed exercise. As discussed above, CPVT is an inherited disorder in individuals with structurally-normal hearts. It is characterized by stress-induced ventricular tachycardia — a lethal arrhythmia that causes sudden cardiac death. In subjects with CPVT, physical exertion and/or stress induce bidirectional and/or polymorphic ventricular tachycardias that lead to sudden cardiac death (SCD) in the absence of detectable structural heart disease. Individuals with CPVT have ventricular arrhythmias when subjected to exercise, but do not develop arrhythmias at rest.
[0225] Accordingly, in still another embodiment of the present invention, the subject has been exercising, or is currently exercising, and has developed an exercise-induced disorder. In this case, the amount of the compounds of the invention effective to limit or prevent a decrease in the level of RyR-bound FKBP in the subject is an amount of compound effective to treat the exercise-induced disorder in the subject. As used herein, an amount of compounds of the invention "effective to treat an exercise-induced disorder" includes an amount of a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, effective to alleviate or ameliorate the clinical impairment or symptoms of the exercise- induced disorder {e.g., in the case of cardiac arrhythmia, palpitations, fainting, ventricular fibrillation, ventricular tachycardia, and sudden cardiac death). The amount of the compounds of the invention effective to treat an exercise-induced disorder in a subject will vary depending upon the particular factors of each case, including the type of exercise- induced disorder, the subject's weight, the severity of the subject's condition, and the mode of administration of the compounds. This amount is readily determined by the skilled artisan, based upon known procedures, including clinical trials, and methods disclosed herein. In one embodiment, the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, treat exercise-induced disorders in the subject.
[0226] The present invention further provides a method for treating exercise-induced disorders in a subject. The method comprises administering the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, to the subject in an amount effective to treat the exercise-induced disorder in the subject. A suitable amount of the compounds effective to treat, for example, exercise-induced cardiac arrhythmia in the subject ranges from about 5 mg/kg/day to about 20 mg/kg/day, and/or is an amount sufficient to achieve plasma levels ranging from about 300 ng/ml to about 1000 ng/ml. The present invention also provides a method for preventing an exercise-induced disorder in a subject. The method comprises administering the compounds of the invention to the subject in an amount effective to prevent the exercise-induced disorder in the subject. A suitable amount of the compounds of the invention effective to prevent the exercise-induced disorder in the subject ranges from about 5 mg/kg/day to about 20 mg/kg/day, and/or is an amount sufficient to achieve plasma levels ranging from about 300 ng/ml to about 1000 ng/ml. Additionally, the present invention provides a method for preventing exercise-induced disorders in a subject. The method comprises administering the compounds of the invention to the subject in an amount effective to prevent an exercise-induced disorder in the subject. A suitable amount of the compounds of the invention effective to prevent an exercise-induced disorder in the subject ranges from about 5 mg/kg/day to about 20 mg/kg/day, and/or is an amount sufficient to achieve plasma levels ranging from about 300 ng/ml to about 1000 ng/ml.
[0227] Additionally, the compounds prevent irregular heartbeat disorders in subjects with heterozygous defects in the FKBP 12.6 gene. [0228] The compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, can be used alone, in combination with each other, or in combination with other agents that have cardiovascular activity including, but not limited to, diuretics, anticoagulants, antiplatelet agents, antiarrhythmics, inotropic agents, chronotropic agents, α and β blockers, angiotensin inhibitors and vasodilators. Further, such combinations of the compounds of the present invention and other cardiovascular agents are administered separately or in conjunction. In addition, the administration of one element of the combination is prior to, concurrent to or subsequent to the administration of other agent(s).
[0229] In various embodiments of the above-described methods, the exercise-induced cardiac arrhythmia in the subject is associated with VT. In some embodiments, the VT is CPVT. In other embodiments of these methods, the subject is a candidate for exercise-induced cardiac arrhythmia, including candidates for exercise-induced sudden cardiac death.
[0230] In view of the foregoing methods, the present invention also provides use of the compounds of the invention in a method for limiting or preventing a decrease in the level of RyR-bound FKBP in a subject who is a candidate for a disorder. The present invention also provides use of the compounds of the invention in a method for treating or preventing a muscular disorder in a subject. Furthermore, the present invention provides use of the compounds of the invention in a method for preventing treating or preventing exercise- induced muscular disorders in a subject.
[0231] Accordingly, the present invention further provides a method for assaying the effects of the compounds of the invention in preventing disorders and diseases associated with the RyR receptors. The method comprises the steps of: (a) obtaining or generating a culture of cells containing RyR; (b) contacting the cells with one or more of the compounds of the invention; (c) exposing the cells to one or more conditions known to increase phosphorylation of RyR in cells; and (d) determining if the one or more compounds of the invention limits or prevents a decrease in the level of RyR-bound FKBP in the cells. As used herein, a cell "containing RyR" is a cell in which RyR, including RyRl, RyR2, and RyR3, or a derivative or homologue thereof, is naturally expressed or naturally occurs. Conditions known to increase phosphorylation of RyR in cells include, without limitation, PKA.
[0232] In the method of the present invention, cells are contacted with one or more of the compounds of the invention by any of the standard methods of effecting contact between drugs/agents and cells, including any modes of introduction and administration described herein. The level of RyR-bound FICBP in the cell is measured or detected by known procedures, including any of the methods, molecular procedures and assays known to one of skill in the art or described herein. In one embodiment of the present invention, the one or more compounds of the invention limits or prevents a decrease in the level of RyR-bound FKBP in the cells.
[0233] RyR, including RyRl; RyR2, and RyR3, has been implicated in a number of biological events in cells. For example, it has been shown that RyR2 channels play an important role in EC coupling and contractility in cardiac muscle cells. Therefore, it is clear that preventive drugs designed to limit or prevent a decrease in the level of RyR-bound FKBP in cells, particularly RyR2-bound FICPB 12.6 in cardiac muscle cells, are useful in the regulation of a number of RyR-associated biological events, including EC coupling and contractility. Thus, the one or more compounds of the invention are evaluated for effect on EC coupling and contractility in cells, particularly cardiac muscle cells, and therefore, usefulness for preventing exercise-induced sudden cardiac death.
[0234] Accordingly, the method of the present invention further comprises the steps of contacting one or more compounds of the invention with a culture of cells containing RyR; and determining if the one or more compounds has an effect on an RyR-associated biological event in the cells. As used herein, a "RyR-associated biological event" includes a biochemical or physiological process in which RyR levels or activity have been implicated. As disclosed herein, examples of RyR-associated biological events include, without limitation, EC coupling and contractility in cardiac muscle cells. According to this method of the present invention, the one or more compounds are contacted with one or more cells (such as cardiac muscle cells) in vitro. For example, a culture of the cells is incubated with a preparation containing the one or more compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m. The compounds' effect on a RyR-associated biological event then is assessed by any biological assays or methods known in the art, including immunoblotting, single-channel recordings and any others disclosed herein.
[0235] The present invention is further directed to one or more compounds of Formula I3 I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, identified by the above-described identification method, as well as a pharmaceutical composition comprising the compound and a pharmaceutically acceptable carrier and/or diluent. The compounds are useful for preventing exercise-induced sudden cardiac death in a subject, and for treating or preventing other RyR-associated conditions. As used herein, a "RyR-associated condition" is a condition, disease, or disorder in which RyR level or activity has been implicated, and includes an RyR-associated biological event. The RyR-associated condition is treated or prevented in the subject by administering to the subject an amount of the compound effective to treat or prevent the RyR-associated condition in the subject. This amount is readily determined by one skilled in the art. In one embodiment, the present invention provides a method for preventing exercise-induced sudden cardiac death in a subject, by administering the one or more compounds of the invention to the subject in an amount effective to prevent the exercise-induced sudden cardiac death in the subject.
[0236] The present invention also provides an in vivo method for assaying the effectiveness of the compounds of the invention in preventing disorders and diseases associated with the RyR recpetors. The method comprises the steps of: (a) obtaining or generating an animal containing RyR; (b) administering one or more of the compounds of the invention to the animal; (c) exposing the animal to one or more conditions known to increase phosphorylation of RyR in cells; and (d) deteπnining the extent the compound limits or prevents a decrease in the level of RyR-bound FKBP in the animal. The method further comprises the steps of: (e) administering one or more of the compounds of the invention to an animal containing RyR; and (f) determining the extent of the effect of the compound on a RyR-associated biological event in the animal. Also provided is a pharmaceutical composition comprising this compound; and a method for preventing exercise-induced sudden cardiac death in a subject, by administering this compound to the subject in an amount effective to prevent the exercise- induced sudden cardiac death in the subject.
[0237] It has been demonstrated that compounds which block PKA activation would be expected to reduce the activation of the RyR channel, resulting in less release of calcium into the cell. Compounds that bind to the RyR channel at the FKBP binding site, but do not come off the channel when the channel is phosphorylated by PKA, would also be expected to decrease the activity of the channel in response to PKA activation or other triggers that activate the RyR channel. Such compounds would also result in less calcium release into the cell.
[0238] By way of example, the diagnostic assays screen for the release of calcium into cells via the RyR channel, using calcium-sensitive fluorescent dyes (e.g., Fluo-3, Fura-2, and the like). Cells are loaded with the fluorescent dye of choice, then stimulated with RyR activators to determine the reduction of the calcium-dependent fluorescent signal (Brillantes, et al, Stabilization of calcium release channel (ryanodine receptor) function by FK506- binding protein. Cell, 77:513-23, 1994; Gillo, et al, Calcium entry during induced differentiation in murine erythroleukemia cells. Blood, 81:783-92, 1993; Jayaraman, et al, Regulation of the inositol 1 ,4,5-trisphosphate receptor by tyrosine phosphorylation. Science, 272:1492-94, 1996). Calcium-dependent fluorescent signals are monitored with a photomultiplier tube, and analyzed with appropriate software. This assay can easily be automated to screen the compounds of the invention using multiwell dishes.
[0239] To demonstrate that the compounds of inhibit the PKA-dependent activation of RyR- mediated intracellular calcium release, an assay involves the expression of recombinant RyR channels in a heterologous expression system, such as Sf9, HEK293, or CHO cells. RyR could also be co-expressed with beta-adrenergic receptors. This would permit assessment of the effect of compounds of the invention on RyR activation, in response to addition of beta- adrenergic receptor agonists.
[0240] The level of PKA phosphorylation of RyR2 which correlates with the degree of heart failure also is assayed and then used to determine the efficacy of the one or more compounds of the invention to block the PKA phosphorylation of the RyR2 channel. Such an assay is based on the use of antibodies that are specific for the RyR2 protein. For example, the RyR2- channel protein is immunoprecipitated and then back-phosphorylated with PKA and [gamma32P]-ATP. The amount of radioactive [32P] label that is transferred to the RyR2 protein then is measured using a phosphorimager (Marx, et al, PKA phosphorylation dissociates FKBP 12.6 from the calcium release channel (ryanodine receptor): defective regulation in failing hearts. Cell, 101 :365-76, 2000).
[0241] Another assay of the compounds of the invention involves use of a phosphoepitope- specifϊc antibody that detects RyRl that is PKA phosphorylated on Ser 2843 or RyR2 that is PKA phosphorylated on Ser 2809. Immunoblotting with such an antibody can be used to assess efficacy of these compounds for therapy for heart failure and cardiac arrhythmias. Additionally, RyR2 S2809A and RyR2 S2809D knock-in mice are used to assess efficacy of therapy for heart failure and cardiac arrhythmias. Such mice further provide evidence that PKA hyperphosphorylation of RyR2 is a contributing factor in heart failure and cardiac arrhythmias by showing that the RyR2 S2809A mutation inhibits heart failure and arrhythmias, and that the RyR2 S2809D mutation worsens heart failure and arrhythmias.
[0242] Therefore, in a specific embodiment, the present invention provides a method of treating heart failure, atrial fibrillation or exercise-induced cardiac arrhythmia, comprising administering to an animal in need thereof, a therapeutically effective amount of a compound selected from the compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m.
[0243] Intracellular Ca2+ leak is proposed as a principal mediator of depressed muscle performance and dystrophic muscle remodeling. Muscular dystrophies are heterogeneous hereditary diseases characterized by weakness and progressive muscle wasting. Of all forms of muscular dystrophies involving the dystrophin-associated protein complex (referred to as dystrophinopathies), Duchenne muscular dystrophy (DMD) is one of the most frequent genetic diseases (X-linked; 1 in 3,500 boys) with death usually occurring before age 30 by respiratory and/or cardiac failure in high numbers of patients. Becker muscular dystrophy (BMD) represents a milder form of the disease associated with a reduction in the amount or expression of a truncated form of the dystrophin protein whereas Duchenne patients have been characterized by complete absence or very low levels of dystrophin. Duchenne and Becker's muscular dystrophy (DMD/BMD) are caused by mutations in the gene encoding the 427-kDa cytoskeletal protein dystrophin. However, with increasing age in BMD cardiac symptoms are more common than in DMD patients and do not correlate with skeletal muscle symptoms. Since genetic screening will not eliminative DMD due to a high incidence of sporadic cases, an effective therapy is highly desirable. DMD/BMD have been consistently associated with disturbed intracellular calcium metabolism. Because alterations of intracellular Ca2+ concentrations in DMD myofϊbers are believed to represent a central pathogenic mechanism, development of a therapeutic intervention that prevents intracellular Ca + abnormalities as a cause of skeletal muscle degeneration is highly desirable.
[0244] It is well established that lack of dystrophin expression is the primary genetic defect in DMD and BMD. However, the key mechanism leading to progressive muscle damage is largely unknown. It has been suggested that elevations of intracellular Ca2+ concentrations ([Ca2+]]) under resting conditions directly contributed to toxic muscle cell (myofiber) damage and concurrent activation of Ca2+-dependent proteases. Since calpain activity is increased in necrotic muscle fibers of mdx mice and calpain dysfunction contributes to limb-girdle muscular dystrophy, preventing activation of calcium-dependent proteases by inhibiting intracellular Ca2+ elevations represents a strategy to prevent muscle wasting in DMD. Significant differences in [Ca2+]; between normal and dystrophic muscles have been reported in myotubes and animal models including the dystrophin-deficient mdx mouse. Intracellular Ca2+ elevations are prevented by administration of a pharmaceutical composition comprising a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m.
[0245] The present invention also provides a method of diagnosis of a disease or disorder in a subject, said method comprising: obtaining a cell or tissue sample from the subject; obtaining DNA from the cell or tissue; comparing the DNA from the cell or tissue with a control DNA encoding RyR to determine whether a mutation is present in the DNA from the cell or tissue, the presence of a mutation indicating a disease or disorder. In one embodiment, the mutation is a RyR2 mutation on chromosome Iq42-q43. In another embodiment, the mutation is one or more CPTV mutations. In another embodiment, the mutation may be a mutation that is present in the DNA encoding RyR2 of a SIDS subject. The diagnostic method is used to detect the presence of a disease or disorder in an adult, a child or a fetus. The disease and disorders include, but are not limited to, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome. Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure. Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof. Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence. Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss.
[0246] The present invention further provides a method of diagnosis of disorders and diseases in a subject, said method comprising: obtaining cells or tissue sample from the subject; incubating the cells or tissue sample with the compound of the invention under conditions which increase phosphorylation of RyR in cells; determining (a) whether RyR bound to calstabin (i.e. RyRl bound to calstabinl, RyR2 bound to calstabin2, or RyR3 bound to calstabinl) is increased in the cells or tissue compared to RyR bound to calstabin in control cells or tissues said control cells or tissues lacking mutant RyR calcium channels, or (b) whether a decrease in release of calcium occurs in RyR channels compared to a lack of decrease in release of calcium in the control cells; an increase in RyR-bound calstabin in (a) indicating a disorder or disease in the subject or a decrease in release of calcium in RyR channels in (b) compared to the control cells indicating a cardiac disease or disorder in the subject. The diagnostic method is used to detect the presence of a disease or disorder in an adult, a child or a fetus. The disease and disorders include, but are not limited to, cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome. Cardiac disorder and diseases include, but are not limited to, irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure. Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise-induced variants thereof. Skeletal muscular disorder and diseases include, but are not limited to, skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence. Cognitive disorders and diseases include, but are not limited to, Alzheimer's Disease, forms of memory loss, and age-dependent memory loss.
[0247] The present invention further provides a method of diagnosis of a cardiac disorder or disease in a subject, said method comprising: obtaining cardiac cells or tissue sample from the subject; incubating the cardiac cells or tissue sample with the compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, under conditions which increase phosphorylation of RyR2 in cells; determining (a) whether RyR2 bound to calstabin2 is increased in the cells or tissue compared to RyR2 bound to calstabin2 in control cells or tissues said control cells or tissues lacking mutant RyR2 calcium channels, or (b) whether a decrease in release of calcium occurs in RyR2 channels compared to a lack of decrease in release of calcium in the control cells; an increase in RyR2 -bound calstabin2 in (a) indicating a disorder or disease in the subject or a decrease in release of calcium in RyR2 channels in (b) compared to the control cells indicating a cardiac disease or disorder in the subject. The provided method is used to diagnose CPTV. The provided method also is used to diagnose sudden infant death syndrome (SIDS). The provided method additionally is used to diagnose cardiac irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure. Irregular heartbeat disorders and diseases include and exercise-induced irregular heartbeat disorders and diseases include, but are not limited to, atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPVT); and exercise- induced variants thereof.
[0248] In addition to the above-mentioned therapeutic uses, the compounds of the invention are also useful in diagnostic assays, screening assays and as research tools.
Methods of Synthesis
[0249] The present invention, provides, in a further aspect, processes for the preparation of a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, and salts, solvates, hydrates, complexes, and pro-drugs thereof, and pharmaceutically acceptable salts of such pro-drugs. More particularly, the present invention provides processes for the preparation of compounds selected from the group consisting of Sl, S2, S3, S4, S5, S6, S7, S9, SI l, S12, SB, S14, S19, S20, S22, S23, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S103, S104, S105, S107, S108, S109, SIlO, Sil l, S112, S113, S114, S115, S116, Sl 17, Sl 18, Sl 19, S120, S121, S122, and S123, and salts, solvates, hydrates, complexes, and pro-drugs thereof, and pharmaceutically acceptable salts of such pro-drugs. The various synthetic routes to the compounds are described herein.
[0250] Some of the following syntheses utilize solvents. In one embodiment, the solvent is an organic solvent. In another embodiment, the organic solvent is methylene chloride (CH2Cl2), chloroform (CCl4), formaldehyde (CH2O) or methanol (CH3OH). Some of the following syntheses also utilize a base catalyst. In one embodiment, the base catalyst is an amine compound. In another embodiment, the base catalyst is an alkylamine such as triethylamine (TEA). In still another embodiment, the base catalyst is pyridine. Some of the following syntheses also utilize basic solutions. In one embodiment, the basic solution is sodium bicarbonate or calcium carbonate. In another embodiment, the basic solution is saturated sodium bicarbonate or saturated calcium carbonate. Some of the following syntheses use acidic solutions. In one embodiment, the acidic solution is a sulfuric acid solution, a hydrochloric acid solution, or a nitric acid solution, hi one embodiment, the solution is IN HCl. One of skill in the art will appreciate still other solvents, organic solvents, base catalysts, basic solutions, and acidic solutions that are used in the embodiments, according to the description herein. The solvents, organic solvents, reactants, catalysts, wash solutions, and so forth are added at appropriate temperatures (e.g. room temperature or about 20°C-25°C, O0C, etc.).
[0251] Some of the following syntheses utilize the compound S68 as a starting material. S68 is available commercially from MicroChemistry Ltd. (Moscow, Russia). See also WO01/55118 for the preparation of S68.
[0252] Several of the following syntheses use S26 as a starting material. S26 is synthesized as an intermediate in the synthesis of S3, S4, S5, and S54, as illustrated in scheme 1 in example 4. Methods for synthesizing S26 are also described in U.S. Patent Applicatin No. 10/680,988.
[0253] Some of the following syntheses requiring purification of the reaction mixture to yield a final product. Purification of the reaction mixture involves one or more processes such as removal of any solvent, crystallization of the product, chromatographic separation of the product (including HPLC, silica gel chromatography, column chromatography, and so forth), washing with basic solution, washing with acidic solution, re-dissolving the product in another solvent, and so forth. One of skill in the art will appreciate still other processes that are used in the embodiments, according to the description herein.
[0254] The reactions are carried out as long as needed (e.g., one hour, several hours, overnight, 24 hours, etc.) to obtain the desired or optimal yields of the desired compounds. Often, the reaction mixtures are stirred. The reactions are carried out at appropriate temperatures (e.g. room temperature or about 20°C-25°C, O0C, 100 0C, etc.).
[0255] Synthon S26 is prepared according to methods described in U.S. Patent App. No. 10/680,988.
[0256] S3, S4, S5, and S54 are prepared from S26. S26 is reacted with RSO2Cl5 wherein R is CH2=CH- (S3), Me- (S4), p-Me-C6H4- (S5), or NH-2-Py (S54), to form a product. The product is purified, for example by column chromatography, to yield S3, S4, S5, or S54. In one embodiment, the reaction occurs in a solvent, such as an organic solvent like CH2Cl2, so that a reaction mixture is formed, and the solvent is removed from the reaction mixture before or during purification of the product. If necessary, a base catalyst, such as triethylamine, is used in the synthesis. Also, basic (e.g., saturated sodium bicarbonate) and acidic washes (e.g., IN HCl) are used if needed to purifying the reaction mixture and/or product, and are accompanied by drying, for example over sodium sulfate, if needed. Column chromatography, for example, is used to purifying the residue to isolate the desired product.
[0257] Sl and S2 are prepared from S3 by reaction with EDSTR1R2, where R is
Figure imgf000102_0001
(Sl) or NBu2 (S2). The product is purified, for example by column chromatography, to yield Sl or S2. In one embodiment, the reaction occurs in a solvent, such as an organic solvent like CH2Cl2, so that a reaction mixture is formed, and the solvent is removed from the reaction mixture before or during purification of the product. Column chromatography, for example, is used to purifying the residue to isolate the desired product.
[0258] S7, S9, S27 and S40 are prepared from S26 by reaction with an alcohol of formula RCOX, where X is Cl or NHS and R is ICH2- (S7), Ph- (S9), CH2=CH- (S27), or 4-N3-2-OH- C6H5 (S40). hi one embodiment, the reaction occurs in a solvent, such as an organic solvent like CH2Cl2, so that a reaction mixture is formed, and the solvent is removed from the reaction mixture before or during purification of the product. If necessary, a base catalyst, such as triethylamine, is used in the synthesis. Also, basic (e.g., saturated sodium bicarbonate) and acidic washes (e.g., IN HCl) are used if needed to purifying the reaction mixture and/or product, and are accompanied by drying, if needed. In another embodiment, S40 is formed by reaction with an alcohol of formula RCOX, where R is 4-N3^-OH-C6Hs and X is NHS. Column chromatography, for example, is used to purifying the residue to isolate the desired product.
' [0259] SI l and S12 are prepared from S26 by reaction with a compound of formula C6H4- NCX, wherein X is O (Sl 1) or S (S12). In one embodiment, the reaction occurs in a solvent, such as an organic solvent like CH2Cl2, so that a reaction mixture is formed, and the solvent is removed from the reaction mixture before or during purification of the product. If necessary, a base catalyst, such as triethylamine or pyridine, is used in the synthesis. In another embodiment, a base catalyst such as pyridine is used as the solvent in which the reaction takes place, and additional solvent, such as ethyle acetate or another appropriate organic solvent, is added after the reaction occurs. Also, basic (e.g., saturated sodium bicarbonate) and acidic washes (e.g., IN HCl) are used if needed to purifying the reaction mixture and/or product, and are accompanied by drying, if needed. Column chromatography, for example, is used to purifying the residue to isolate the desired product.
[0260] The isomers S 13 and S 14 are prepared from S26 by reaction with phenyl methoxyphosphonyl chloride (Ph(MeO)P(O)Cl). In one embodiment, the reaction occurs in a solvent, such as an organic solvent, such as methylene chloride. If necessary, abase catalyst such as triethylamine may be used, for example, by adding it to a reaction mixture formed by mixing the reactants in a solvent. Also, the reaction mixture is washed with basic solution, for example saturated sodium bicarbonate, if necessary. The isomers are separated and purified, for example, using silica gel chromatography.
[0261] S19 and 22 are prepared from S26 by reaction with a compound of formula ClOC-X-
Figure imgf000103_0001
COCl, where X is CH2-CH2 (S 19) or (S22). In one embodiment, the reaction occurs in the presence of a solvent, such as an organic solvent, such as methylene chloride. If necessary, a base catalyst such as triethylamine is added to the reaction mixture formed by mixing the reactants in a solvent. Also, base (e.g., saturated sodium bicarbonate), acid (e.g., IN HCl), and water washes are used to remove unwanted compounds from the reaction mixture, if needed.
[0262] S20 and S23 are prepared from an intermediate compound of formula
Figure imgf000103_0002
(S23). The intermediate compound is treated with H2O2. If necessary, sodium thiosulfate also is used to treat the intermediate. In one embodiment, the reaction occurs in the presence of a solvent, such as an organic solvent, such as methanol (CH3OH), forming a reaction mixture. The solvent is removed from the reaction mixture after the reaction takes place and, if desired, the residue is redissolved in another solvent, such as another organic solvent, such as ethyl acetate. The reaction mixture is washed with basic solution (e.g., saturated sodium carbonate) if desired to remove unwanted compounds from the reaction mixture. The reaction mixture is dried (e.g., using sodium sulfate) if it is washed with basic solution. The final residue is purified, for example by column chromatography, to obtain the final product. [0263] S57 is prepared from S26 and methyl chlorooxoacetate. In an embodiment, the reaction occurs in the presence of a solvent, such as an organic solvent, such as methylene chloride. A base catalyst such as pyridine is used as necessary to facilitate or hasten the reaction. The reaction mixture formed by mixing the reactants and a solvent is washed with basic solution {e.g., saturated sodium bicarbonate), acidic solution {e.g. HCl), and water. Purification such as silic gel chromatography yields S57.
[0264] S36 is prepared from S57 by reaction with sodium hydroxide. In one embodiment, the reaction takes place in a solvent, such as an organic solvent, such as methanol. The solvent is removed from the reaction mixture formed by mixing the reactants and the solvent, thereby forming a residue. The residue is dissolved in water and washed with another organic solvent, such as ether, to remove unwanted hydrophobic compounds. The aqueous phase from the basic washes is acidified and the product is extracted therefrom using an organic solvent, such as methylene chloride. Further purification is used if necessary.
[0265] S38 is prepared in a manner similar to S36, except a compound of formula
Figure imgf000104_0001
is used as the starting material in the synthesis.
[0266] S44 is prepared by treating S36 with thionyl chloride to form crude S36-C1. Excess thionyl chloride, if any, is removed from the reaction mixture. The crude S 36-Cl then is dissolved in a solvent, such as an organic solvent like methylene chloride, and reacted with mono-protected {e.g., mono-Boc protected) cystamine. A base catalyst such as pyridine is used if desired, and the reaction mixture is quenched with a basic solution {e.g., saturated sodium bicarbonate). The reaction mixture formed by mixing the cystamine and S36-C1 reactants is purified. The protecting groups {e.g., Boc) are removed using an appropriate acid or base wash {e.g., trifluoroacetic acid in an organic solvent in the case of the Boc protecting group). The final product then is purified, for example, using chromatography techniques.
[0267] S57 and S59 are prepared from S36-C1, which is reacted with methanol (S57) or ethylamine (S59).
[0268] S43 and S45 are prepared from S36-cystamine, which is prepared as disclosed herein. S-36 cystamine is reacted with an NHS activated ester of an appropriate azido compound to yield S43 and S45. The reaction takes place in a solvent, such as an organic solvent. [0269] S37 is prepared from S26 by reaction with 4-nitrophenyl chloroforniate (NO2C6HSOCOCI). The reaction takes place in a solvent and, if desired, abase catalyst such as triethylamine may be used. The reaction mixture formed by mixing the reactants and a solvent is washed with water to remove unwanted hydrophilic compounds. The solvent is removed from the reaction mixture to form a residue, which is purified (e.g., using chromatography techniques) to yield S37.
[0270] S6, S46-53, S64, S66, and S67 are prepared from S37 by reaction with an amine of formula RNH2, wherein NR is NH2 (S46), NEt2 (S48), NHCH2Ph (S49), NHOH (S51),
CH^ (S6), πJ3$ (S47), O ^ (S50), HNΥX> (S52),
(S53), (S64),
Figure imgf000105_0001
(S66), or (S67). The reaction takes place in the presence of a solvent, such as an organic solvent, such as DMF. In one embodiment, only one equivalent of amine is used in the reaction. Purification is accomplished, for example, by SiO2 column chromatography.
[0271] S6, S46-53, S64, S66, and S67 also are prepared from S26, via the S26-phosgene intermediate. The S26-phosgene intermediate is formed by reacting S26 with triphosgene. Thereafter, the S26-phosgene is reacted with an amine of formula RNH2, where NR is NH2
(S46), NEt2 (S48), NHCH2Ph (S49), NHOH (S51),
Figure imgf000105_0002
(S47), ^ (S50), 1X -O " (S52), OO ( .S.5.3..), O ~ (S64),
Figure imgf000105_0003
(S66), or (S67). The reaction takes place in the presence of a solvent, such as an organic solvent. Pn one embodiment, only one equivalent of amine is used in the reaction. Purification is accomplished, for example, by SiO2 column chromatography. [0272] S55, S56, S58, and S60-63 are prepared from S27 by reaction with HNRiR2, where
Figure imgf000106_0001
NRiR2 is (S55), ςκ£ro ( .„S5.6,.), Q (S58), -CXJy""'
(S60),
Figure imgf000106_0002
(S63). The reaction occurs in a solvent, such as an organic solvent, such as chloroform, thereby forming a reaction mixture. The solvent is removed from the reaction mixture to form a residue, which is purified, for example, by silica gel column chromatography to yield the final product.
[0273] S69-75 are prepared from S68, via the S68-phosgene intermediate. S68 is treated with triphosgene to form the intermediate, which in turn is treated with an amine RNH2,
where R is NH2 (S70), NEt2 (S75), NHOH (S74),
Figure imgf000106_0003
(S69), (S71),
Figure imgf000106_0004
(S72), and (S73). The reaction occurs in a solvent, such as an organic solvent, such as chloroform, thereby forming a reaction mixture. The solvent is removed from the reaction mixture to form a residue, which is purified, for example, by silica gel column chromatography to yield the final product.
[0274] S76 is prepared from S68 by reaction with methyl chlorooxoacetate. In an embodiment, the reaction occurs in the presence of a solvent, such as an organic solvent, such as methylene chloride. A base catalyst such as pyridine is used as necessary to facilitate or hasten the reaction. The reaction mixture formed by mixing the reactants and a solvent is washed with basic solution (e.g., saturated sodium bicarbonate), acidic solution (e.g. HCl), and water. Purification such as silic gel chromatography yields S76.
[0275] S77 is prepared from S76 by reaction with sodium hydroxide. In one embodiment, the reaction takes place in a solvent, such as an organic solvent, such as methanol. The solvent is removed from the reaction mixture formed by mixing the reactants and the solvent, thereby forming a residue. The residue is dissolved in water and washed with another organic solvent, such as ether, to remove unwanted hydrophobic compounds. The aqueous phase from the basic washes is acidified and the product is extracted therefrom using an organic solvent, such as methylene chloride. Further purification is used if necessary.
[0276] S78-S81 are prepared by treating S77 with thionyl chloride to form crude S77-C1. Excess thionyl chloride, if any, is removed from the reaction mixture. The crude S77-C1 then is dissolved in a solvent, such as an organic solvent like methylene chloride, and reacted with HX, where X is NHEt (S78), NHPh (S79), NH2 (S80), and NHCH2-pyridine (S81). The solvent is removed, and the residue is purified.
[0277] S82 is prepared from S68. S68 is reacted with CH2CHSO2Cl in a manner analogous to the production of S3. The product then is treated with HNRiR2 in a manner analogous to
Figure imgf000107_0001
the production of Sl and S2, except that NR1R2 is
[0278] S83 is prepared from S68. S68 is reacted with RCOCl, wherein R is
Figure imgf000107_0002
, in a manner analogous to the production of S 7, S9, and S40.
[0279] S84 is prepared from S68 by reaction with benzyl bromide. In an embodiment, the reaction takes place in a solvent, such as an organic solvent like methylene chloride. A base catalyst such as triethylamine is added as necessary to catalyze the reaction. The reaction mixture formed by mixing the reactants and the solvent is purified to yield S 84.
[0280] S85 is prepared from S26. S26 is reacted with di-tert-butyl dicarbonate in a solvent, for example an organic solvent like methylene chloride. A base catalyst such as triethylamine also is used, if necessary. The reaction mixture formed by mixing the reactants and the solvent is washed with saturated sodium bicarbonate solution and the aqueous layer is extracted with an organic solvent. The combined organic layers are dried and concentrated provide S85.
[0281] S86 is prepared from S85 in a solvent, for example an organic solvent. S85 is treated with BBr3 to form a reaction mixture. If necessary, a base catalyst, such as triethylamine, is used in the reaction. The reaction is quenched (e.g., in the case of triethylamine, with methanol) aid concentrated. Purification, for example by column chromatography, yields S86. [0282] S87 is prepared by reacting S86 with trifluoromethylsulfonyl anhydride. The reaction is carried out in a solvent, such as an organic solvent. A base catalyst such as triethylamine is added if necessary. In the case of triethylamine, the reaction mixture formed by mixing the reactants and the solvent is quenched with water, after which the aqueous layer is extracted with an appropriate organic solvent. If desired, the organic layers are dried (e.g., using magnesium sulfate), and the organic layers are concentrated. Purification of the concentrated organic layers yields S87.
[0283] S88 is prepared from S87 by reaction of morpholine, tris(dibenzylideneacetone)dipalladium(0), 2-(di-tert-butylphosphino)-biphenyl, and potassium phosphate. The reaction mixture is diluted with a solvent, such as methylene chloride or another appropriate organic solvent, and washed with water. The aqueous layer, formed by washing with water, is extracted with an organic solvent, such as methylene chloride. The organic layers then are dried (e.g., over magnesium sulfate) and concentrated. The residue is purified, for example by silica gel flash chromatography, to yield S88.
[0284] S89 is prepared from S87 by reaction with benzenethiol and /-Pr2NEt in a solvent, such as CH3CN or another appropriate organic solvent. After reaction, an organic solvent such as ethyl acetate is added to the reaction mixture. If necessary, the reaction mixture is washed with one or more of acidic (e.g., HCl), basic (e.g. NaOH), and water solutions. After drying (e.g. with Na2SO4), the solution is concentrated. Purification, for example by chromatography, yields S89. In an alternative, refluxing S87 with benezethiol in an appropriate solvent such as dioxane with a catalyst such as z-Pr2NEt/Pd2(dba)3/xantphos yields S89.
[0285] S90 is prepared from S87 reacted with a base, phenylboronic acid, and a catalyst. In an embodiment, the base is K2CO3 and the catalyst is (Pd(Ph3P)4). In one embodiment, the reaction occurs in a solvent, such as an organic solvent, such as dioxane. The reaction mixture formed by mixing the reactants and the solvent is diluted with a solvent (e.g. methylene chloride), and washed with water to remove unwanted hydrophilic compounds. Concentration and purification of the residue yields S90.
[0286] S92 is prepared from S87 reacted with zinc cyanide. In an embodiment, the reaction occurs in a solvent, such as an organic solvent like DMF. A catalyst such as Pd(Ph3P)4 also is used to facilitate and hasten the reaction. The reaction mixture formed by mixing the reactants and the solvent, if necessary, is diluted with water and an acidic solution and extacted with an organic solvent. The organic extracts then are washed using a salt solution, dred, filtered, and concentrated. Purification of the residue proceeds, for example, by silica gel column chromatography.
[0287] S94 is prepared for S86 by reaction with acetic anhydride. In an embodiment, the reaction takes place in a solvent, such as an organic solvent like methylene chloride. Triethylamine or another base catalyst is added as necessary. Washing with water, followed by drying (e.g., using sodium sulfate), is used as desired. Purification of the residue yields S94.
[0288] S95 is prepared from S94 by reaction with anhydrous AlCl3, in a solvent if desired. The solvent is an organic solvent like benzene. The reaction mixture is refluxed and cooled on ice. Extraction with an organic solvent, concentration, and purification of the residue yields S95.
[0289] S96 is prepared from S86 by iodination. For example, S86 is added to a solvent, such as an organic solvent like methanol, with excess NaI and Chloramine-T. The reaction mixture is quenched with Na2S2O3 solution. Concentration and purification of the residue yields S96 as a mixture of mono-iodinated and di-iodinated products.
[0290] S97 is prepared from S86 by reaction with a nitric acid. S86 is protected (e.g., using the Boc protecting groups) and added to concentrated sulfuric acid. Nitric acid is added to the reaction mixture. The reaction mixture is cooled and neutralized (e.g., using Na2CO3) to quench the reaction. Organic extraction and subsequent concentration is used to isolate the product. Purification yields S 97.
[0291] S98 is prepared by hydrogenation of S97. For example, S97 is added to a solution, such as an organic solution like methanol. H2 gas is bubbled through the solution and Pd/C catalyst or another applicable catalyst is added. Filtration to remove the catalyst and purification yields S97.
[0292] SlOO is prepared from S98. S98 is dissolved in acid solution, such as aqueous HCl. To this a solution of sodium nitrite, and then NaN3 in water, are added. The reaction mixture is extracted using an organic solvent. If needed, the extract is washed with a basic solution (e.g., saturated sodium bicarbonate) and water. Organic layers from the washing are dried using, for example, anhydrous sodium sulfate, and concentrated to form a residue. The residue is purified to yield SlOO. To prepare S99, NaN3 is substituted with NaBF4 in a similar manner. [0293] SlOl, S102, and S103 may each be prepared from S68.
[0294] SlOl may be prepared from S68 as follows. Triphosgene is reacted with S68 in the presence of a solvent (such as the organic solvent dichloromethane, CH2Cl2) to generate S68- phosgene. Optionally, a base is also present or added to scavenge acid generated during the reacton. Any suitable base may be used. For example, organic bases such as organic amines like triethylamine, di-isopropylethylamine or pyridine may be used. Inorganic bases such as as sodium bicarbonate may also be used. Then, without the need for purification, the reaction mixture containing the S68-phosgene is treated with 1 -piperonylpiperazine. If necessary, the reaction mixture is washed with one or more of acidic (e.g., HCl), basic (e.g. NaOH), and water solutions. The solvents are removed, for example under reduced pressure. The SlOl product can then be purified, for example using SiO2 column chromatography.
[0295] S 102 maybe prepared from S68 using the same scheme as for SlOl, with the exception that piperidine is used in the place of piperonylpiperazine.
[0296] S103 may be prepared from S68 using the same scheme as for SlOl, with the exception that N-Boc 1-piperazine is used in the place of piperonylpiperazine. Also, trifiuoroacetic acid (TFA) is added de-protect the Boc group.
[0297] S 104 may be prepared by reacting S36 with hydrogen peroxide (H2O2) in the presence of a solvent (such as MeOH). The solvents are removed (for example under reduced pressure), and the S 104 product can then be purified, for example by re-crystallization.
[0298] S 105 may be prepared from S68 as follows. S68 is be reacted with CH3O- C(O)C(O)Cl in the presence of a solvent (such as the organic solvent dichloromethane (CH2Cl2)) and optionally a catalyst (such as pyridine). Preferably, the CH3O-C(O)C(O)Cl should be added dropwise. If necessary, the reaction mixture is washed with one or more of acidic (for example HCl), basic (for example NaOH), and water solutions. The solvents are removed and the product may be further purified, for example by SiO2 column chromatography.
[0299] S 107 may be prepared from S26 as follows. To a solution of S26 in a solvent (such as MeOH), formaldehyde (CH2O) and sodium cyanoborohydride (NaBCNH3) are added and allowed to react. Preferably, the reaction mixture is maintained at around pH 4-5, for example by addition of a few drops of IN HCl. The solvents are then removed, for example under reduced pressure. If necessary, the residue may be dissolved in ethyl acetate and washed with with one or more of a basic solution (for example NaOH), and water. The solvents may be removed, and the product may be further purified, for example using SiO2 column chromatography.
[0300] S 108 may be prepared as follows. A mixture of N-benzyloxycarbonyl-glycine (Cbz- GIy,), Diisopropyl-carbodiimide (DIC), and N-hydroxysuccinimide (NHS), are reacted together in a solvent (such as the organic solvent dichloromethane (CH2Cl2)) for a suitable amount of time. S26 is then added to the mixture and the reaction is allowed to proceed further. If necessary, the reaction mixture is washed with one or more of acidic (for example HCl), basic (for example NaOH), and water solutions. The solvents may then be removed, for example by evaporation. The product may be further purified, for example using SiO2 column chromatography.
[0301] S109 maybe prepared from S108, as follows. S108 in a solvent (such as the organic solvent dichloromethane (CH2Cl2)) is reacted with HBrZCH3CO2H. After a suitable amount of time, the reaction mixture is evaporated, for example under reduced pressure. The residue is dissolved in a suitable solvent, such as MeOH, and is treated with propylene oxide. The solvent may then be removed, for example under reduced pressure, to provide crude S 109. The SlOO may be further purified, for example by dissolving in an acidic solution (such as HCl), washing with ethyl acetate, and evaporating.
[0302] SIlO may be prepared as follows. A mixture of S26, methyl 1 -bromoacetate and pyridine are reacted in DMF for a suitable amount of time. To this mixture, ethyl aceate is added, and if necessary, the reaction mixture is washed with a basic solution (for example NaHCO3), or water. The product S 110, as an oil, may be purified, for example by SiO2 column chromatography.
[0303] Sill may be prepared as follows. A base (such as IN NaOH) is added to Sl 10 in a solvent (such as MeOH), and the mixture is allowed to react for a suitable amount of time. The solvents are then removed, for example under reduced pressure, and the residue may then be dissolved in an aqueous solution, such as water. The aqueous phase may be washed with ethyl acetate and acidified, for example with IN HCl, to pH of around 4. The solvents may then be removed, for example under reduced pressure, to produce crude Si l l. The NaCl may be removed using an alcohol, such as ethanol, to yield pure Sil l as a solid.
[0304] Sl 12 maybe prepared as follows: To a mixture of S26 and pyridine in an solvent (such as the organic solvent dichloromethane (CH2Cl2)), SO2Cl2 is added drop-wise at around O0C and reacted for a suitable amount of time. The solvents may be removed, for example under reduced pressure. The residue can be dissolved in a suitable basic solution, such as NaOH. The aqueous solution may then be washed with ethyl acetate, and acidified (for example with IN HCl) to around pH 4. The aqueous phase may be extracted again with ethyl acetate, and the ethyl acetate phase evaporated, for example under reduced pressure, to provide Sl 12, as a powder.
[0305] S 113 may be prepared as follows. S 107 in ethyl acetate is treated with CH3I. The mixture is stirred for a suitable amount of time, and the product Sl 13, as white solid, is collected by filtration.
[0306] Sl 14 maybe prepared as follows. The compound S26, in a solvent such as the organic solvent CH2Cl2 is ideally cooled to around O0C. To this solution, triphosgene, is added. Optionally, a base is also present or added to scavenge acid generated during the reacton. Any suitable base may be used. For example, organic bases such as organic amines like triethylamine, di-isopropylethylamine or pyridine may be used. Inorganic bases such as as sodium bicarbonate may also be used. The reaction is allowed to proceed (ideally around O0C) for a suitable amount of time (for example about 1 hour). Without the need for purification, the resulting S26-phosgene in the reaction mixture may then be treated with N- Boc 1 -piperazine, again ideally at around O0C, and the reaction is allowed to proceed (ideally at around 00C) for a suitable amount of time (for example about 1 hour). If necessary, the reaction mixture is washed with one or more of acidic (for example HCl), basic (for example NaOH), and water solutions. The solvents are removed and the product may be further purified, for example by SiO2 column chromatography.
[0307] Sl 15 may be prepared as follows. A mixture of Sl 14 and Lawesson Reagent in toluene is stirred at around 9O0C for several hours. The mixture is cooled to room temperature and washed with a suitable base, such as saturated NaHCO3. The product SI l 5_may be purified, for example by SiO2 chromatography.
[0308] Sl 16 may be prepared as follows. A mixture of Sl 15 and trifluoroacetic acid (TFA) in a suitable solvent solvent (such as the organic solvent dichloromethane (CH2Cl2)) is stirred at around room temperature for a suitable amount of time (for example, about 2 hours). Evaporation of the solvents, for example under reduced pressure, produces Sl 16.
[0309] S 117 (S 117) may be prepared as follows. A solution of S057 in a suitable solvent (such as the organic solvent dichloromethane (CH2Cl2)) is cooled to about -780C. To this, IM BBr3 a suitable solvent (such as the organic solvent dichloromethane (CH2Cl2)) is added, and the mixture is stirred (still at about 780C) for a suitable amount of time (for example around 3 hours) and then wSed to room temperature. If necessary, the mixture is washed with an acid (such as IN HCl) and/or H2O. After removal of the solvents, the product Sl 17 may be purified, for example by SiO2 column chromatography.
[0310] Sl 18 maybe synthesized as follows. S26 in as suitable solvent a suitable solvent (such as the organic solvent dichloromethane (CH2Cl2)) is treated with BODIPY TMR-X, SE (Molecular Probes Inc.) for a suitable amount of time (for example, around 3 hours). If necessary, the mixture may be washed with an acid (such as 0.01 N HCl) and/or a base (such as NaHCO3). Removal of the solvents, for example under reduced pressure, will yield Sl 18.
[0311] Sl 19 may be synthesized as follows. A mixture S 107, H2O2 (for example around 50%), and an alcohol (such as MeOH), is stirred at around room temperature for a suitable amount of time (typically around 2 days). If desired, mass spectrometry may be used to monitor the disappearance of S107 and the formation of the Sl 19 product). The solvents may be removed, for example under reduced pressure, to give Sl 19.
[0312] S 120 maybe synthesized as follows. A mixture S26, benzyl bromide and Na2CO3 in a solvent (such as DMF), is reacted for a suitable amount of time, preferably overnight. Ethyl acetate is added to the reaction, and then, if necessary, the reaction is washed with a suitable solvent, for example with H2O (4x10 ml). The organic phase may be concentrated, for example under reduced pressure, and the residue may be purified, for example by column chromatography to give S 121.
[0313] S121 may be synthesized as for S120, but using 4-OH-benzyl bromide instead of benzyl bromide.
[0314] S122 may be synthesized as follows. To a cold solution of compound S26 in a solvent, such as the organic solvent in CH2Cl2, DIEA is added, and subsequently acetoxyacetyl chloride is added. The reaction is allowed to proceed for a suitable amount of time, and then diluted, (for example with 1.0 AfHCl aqueous solution) and extracted (for example using CH2Cl2). The combined organic layers may, if necessary, be washed (for example with H2O, brine), dried (for example with Na2SO4), filtered, and dried (for example by evaporation). The product maybe further purified, for example by chromatography on a silica gel column, and may be eluted with a gradient increasing in polarity from 0 to 50% of petroleum in ethyl acetate. Relevant fractions may then be combined to give the desired product.
- Ill - [0315] S 123 maybe synthesized as follows. To a solution of compound S 122 in a solvent (such as MeOH) and THF, preferably at room temperature, is added LiOH. The reaction is allowed to proceed for a suitable amount of time at a suitable temperature (ideally around room temperature, and may then be diluted (for example with with 1.0 MHCl aqueous solution) and extracted (for example with CH2Cl2). The combined organic layers may be washed (for example with H2O, brine), dried (for example with Na2SO4), filtered and dried (for example by evaporation). The crude product may be purified, for example by chromatography on a silica gel column, eluted, for example with a gradient increasing in polarity from 0 to 70% petroleum in ethyl acetate. Relevant fractions may then beO combined to give S 123.
[0316] It should be noted that the compounds used as starting materials for, or generated as intermediates in, the synthesis of the compounds of the invention, may themselves have structures encompassed by the formulae of the invention, and/or may themselves be active agents useful in the methods and compositions of the present invention. Such starting materials and intermediates may be useful for, inter alia, treating or preventing various disorders and diseases associated with RyR receptors such as muscular and cardiac disorders, treating or preventing a leak in a RyR2 receptor in a subject, or modulating the binding of RyR and FKBP in a subject. The present invention encompasses any of the starting materials or intermediates disclosed herein that have structures encompassed by Foπnula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, and/or which are useful as active agents in the methods and compositions of the present invention. For example, in one embodiment the compound S68, which is useful as a starting material for the synthesis of compounds S69 - S75, maybe used for, inter alia, treating or preventing various disorders and diseases associated with RyR receptors, treating or preventing a leak in a RyR2 receptor, or modulating the binding of RyR and FKBP in a subject.
[0317] In another embodiment, the compound S26, which is useful in the synthesis of many of the compounds described herein (including S3, S4, S5, S7, S9, Sl 1, S12, S13, S14 and other compounds) may be used for, inter alia, treating or preventing various disorders and diseases associated with RyR receptors, treating or preventing a leak in a RyR2 receptor, or modulating the binding of RyR and FKBP in a subject.
[0318] Similarly, in another embodiment, the compound S25 (see U.S. Patent Application 10/809,089) may also be used for, inter alia, treating or preventing various disorders and diseases associated with RyR receptors, treating or preventing a leak in a RyR2 receptor, or modulating the binding of RyR and FICBP in a subject.
[0319] The compounds of the present invention are prepared in different forms, such as salts, hydrates, solvates, complexes, pro-drugs or salts of pro-drugs and the invention includes all variant forms of the compounds.
[0320] The term "compound(s) of the invention" as used herein means a compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, and salts, hydrates, prodrugs and solvates thereof.
[0321] A "pharmaceutical composition" refers to a mixture of one or more of the compounds described herein, or pharmaceutically acceptable salts, hydrates or pro-drugs thereof, with other chemical components, such as physiologically acceptable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
[0322] A "pro-drug" refers to an agent which is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. They are bioavailable, for instance, by oral administration whereas the parent drug is not. The pro-drug also has improved solubility in pharmaceutical compositions over the parent drug. For example, the compound carries protective groups which are split off by hydrolysis in body fluids, e.g., in the bloodstream, thus releasing active compound or is oxidized or reduced in body fluids to release the compound.
[0323] A compound of the present invention also can be formulated as a pharmaceutically acceptable salt, e.g., acid addition salt, and complexes thereof. The preparation of such salts can facilitate the pharmacological use by altering the physical characteristics of the agent without preventing its physiological effect. Examples of useful alterations in physical properties include, but are not limited to, lowering the melting point to facilitate transmucosal administration and increasing the solubility to facilitate administering higher concentrations of the drug.
[0324] The term "pharmaceutically acceptable salt" means an acid addition salt which is suitable for or compatible with the treatment of a patient or a subject such as a human patient or an animal such as a dog.
[0325] The teπn "pharmaceutically acceptable acid addition salt" as used herein means any non-toxic organic or inorganic salt of any base compounds represented by Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or any of their intermediates. Illustrative inorganic acids which form suitable acid addition salts include hydrochloric, hydrobromic, sulfuric and phosphoric acids, as well as metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate. Illustrative organic acids that form suitable acid addition salts include mono-, di-, and tricarboxylic acids such as glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic and salicylic acids, as well as sulfonic acids such as p-toluene sulfonic and methanesulfonic acids. Either mono or di-acid salts can be formed, and such salts exist in either a hydrated, solvated or substantially anhydrous form. In general, the acid addition salts of compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m, are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms. The selection of an appropriate salt will be known to one skilled in the art. Other non-pharmaceutically acceptable salts, e.g., oxalates, are used, for example, in the isolation of compounds of the invention for laboratory use or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
[0326] The compounds of the present invention form hydrates or solvates, which are included in the scope of the claims. When the compounds of the present invention exist as regioisomers, configurational isomers, conformers or diasteroisomeric forms all such forms and various mixtures thereof are included in the scope of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m. It is possible to isolate individual isomers using known separation and purification methods, if desired. For example, when a compound of the present invention is a racemate, the racemate can be separated into the (S)-compound and (R)-compound by optical resolution. Individual optical isomers and mixtures thereof are included in the scope of Formula I, Ia, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m.
[0327] The term "solvate" as used herein means a compound of Formula I, I-a, I-b, I-c, I-d, I- e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, or a pharmaceutically acceptable salt thereof, wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered. Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a "hydrate."
[0328] The term an "effective amount," "sufficient amount" or "therapeutically effective amount" of an agent as used herein is that amount sufficient to effect beneficial or desired results, including clinical results and, as such, an "effective amount" depends upon the context in which it is being applied. The response is preventative and/or therapeutic. The term "effective amount" also includes that amount of the compound of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 or I-m, which is "therapeutically effective" and which avoids or substantially attenuates undesirable side effects.
[0329] As used herein and as well understood in the art, "treatment" is an approach for obtaining beneficial or desired results, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized {i.e., not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment.
[0330] The terms "animal," "subject" and "patient" as used herein include all members of the animal kingdom including, but not limited to, mammals, animals {e.g., cats, dogs, horses, etc.) and humans.
[0331] The present invention further provides a composition, comprising radio labeled compounds of Formula I, I-a, I-b, I-c, I-d, I-e, I-f, I-g, I-h, I-i, I-j, I-k, 1-1 and I-m. Labeling of the compounds is accomplished using one of a variety of different radioactive labels known in the art. The radioactive label of the present invention is, for example, a radioisotope. The radioisotope is any isotope that emits detectable radiation including, without limitation, 35S, 1251, 3H, or 14C. Radioactivity emitted by the radioisotope can be detected by techniques well known in the art. For example, gamma emission from the radioisotope is detected using gamma imaging techniques, particularly scintigraphic imaging.
[0332] By way of example, radio-labeled compounds of the invention are prepared as follows. A compound of the invention may be demethylated at the phenyl ring using BBr3. The resulting phenol compound then is re-methylated with a radio-labeled methylating agent (such as 3H-dimethyl sulfate) in the presence of a base (such as NaH) to provide 3H-labeled compounds.
[0333] The present invention further provides compounds that may be classified as 1,4- benzothiazepines, including, by way of example and without limitation, Sl, S2, S3, S4, S5, S6, S7, S9, SI l, S12, S13, S14, S19, S20, S22, S23, S25, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S103, Sl 04, S105, S107, S108, S109, SIlO, Sill, S112, S113, S114, S115, S116, S117, S118, S119, S120, S121, S122, and S123.
[0334] These and any other compounds of the present invention are associated with a pharmaceutically acceptable carrier, as described above, so as to form a pharmaceutical composition.
[0335] In accordance with the method of the present invention, the decrease in the level of RyR-bound FKBP is limited or prevented in the subject by decreasing the level of phosphorylated RyR in the subject. In one embodiment, the amount of the agent effective to limit or prevent a decrease in the level of RyR2-bound FKBP12.6 in the subject is an amount of the agent effective to treat or prevent heart failure, atrial fibrillation and/or exercise- induced cardiac arrhythmia in the subject. In another embodiment, the amount of the agent effective to limit or prevent a decrease in the level of RyR2-bound FKBP 12.6 in the subject is an amount of the agent effective to prevent exercise-induced sudden cardiac death in the subject.
[0336] In view of the foregoing, the present invention further provides a method for treating or preventing exercise-induced cardiac arrhythmia in a subject, comprising administering to the subject a 1,4-benzothiazepine compound, as disclosed herein, in an amount effective to treat or prevent exercise-induced cardiac arrhythmia in the subject. Similarly, the present invention provides a method for preventing exercise-induced sudden cardiac death in a subject, comprising administering to the subject a 1,4-benzothiazepine compound, as disclosed herein, in an amount effective to prevent exercise-induced sudden cardiac death in the subject. Additionally, the present invention provides a method for treating or preventing atrial fibrillation or heart failure in a subject, comprising administering to the subject a compound, as disclosed herein, in an amount effective to treat or prevent the atrial fibrillation or heart failure in the subject. In each of these methods, the compound is selected from the group of compounds consisting of compounds of the formula:
Figure imgf000119_0001
wherein, n is 0, 1, or 2;
R is located at one or more positions on the benzene ring; each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -N3, -SO3H, acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, - SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
Ri is selected from the group consisting of H, oxo, alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl maybe substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
R2 is selected from the group consisting of H, -C=O(R5), -C=S(R6), -SO2R7, -POR8R9, - (CH2)m-Rio, alkyl, aryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
R3 is selected from the group consisting of H , CO2Y, CONY, acyl, alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N3 O, -S-, -CN3 -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl; and wherein Y is selected from the group consisting of H, alkyl, aryl, cycloalkyl, and heterocyclyl;
R4 is selected from the group consisting of H, alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, cycloalkyl, and heterocyclyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
R5 is selected from the group consisting of -NR165NHNHR16, NHOH, -OR15, CONH2NHRi6, CO2R15, CONR16, CH2X, acyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
Rn is selected from the group consisting Of-OR1S5 NHNRi6, NHOH, -NR16, CH2X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cyclό alkyl, and (hetero-)cyclyl;
R7 is selected from the group consisting Of-ORi5, -NR165NHNHR16, NHOH, CH2X, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl;
R8 and R9 independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl; R10 is selected from the group consisting of NH2, OH, -SO2Rn, -NHSO2Rn,
Figure imgf000121_0001
NHC=O(R12), -OC=O(R12), and -PORi3R14;
Rn, Ri2, R13, and Ri4 independently are selected from the group consisting of H, OH, NH2, NHNH2, NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted with one or more radicals independently selected from the group consisting of halogen, -N-, -0-, - S-, -CN, -N3, nitro, oxo, acyl, alkenyl, alkoxyl, alkyl, alkylamino, amino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, and hydroxyl;
X is selected from the group consisting of halogen, CN, CO2R15, CONRi6, -NR16, -OR1S7 - SO2R7, and -POR8R9; and
Ri5 and R16 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, -N-, -0-, -S-, -CN, -N3, nitro, oxo, acyl, alkenyl, alkoxyl, alkyl, alkylamino, amino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, and hydroxyl, and wherein each substituted acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl radical may itself be substituted with one or more radicals independently selected from the group consisting of halogen, -N-, -0-, -S-, -CN, -N3, nitro, oxo, acyl, alkenyl, alkoxyl, alkyl, alkylamino, amino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, and hydroxy; and salts, hydrates, solvates, complexes, and prodrugs thereof.
[0337] Examples of such compounds include, without limitation, Sl, S2, S3, S4, S5, S6, S7, S9, SIl, S12, S13, S14, S19, S20, S22, S23, S25, S26, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S103, S104, S105, S107, S108, S109, SI lO, Si l l, S112, S113, S114, S115, S116, S117, S118, S119, S120, S121, S122, and S123. [0338] In an embodiment of the present invention, if R2 is C=O(Rs) or SO2R7, then R is at positions 2, 3, or 5 on the benzene ring.
[0339] In another embodiment of the invention, if R2 is C=O(R5) or SO2R7, then each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -N3, -SO3H, acyl, alkyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero- )aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero- )arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl.
[0340] In another embodiment of the invention, if R2 is C=O(R5) or SO2R7, then there are at least two R groups attached to the benzene ring. Furthermore, there are at least two R groups attached to the benzene ring, and both R groups are attached at positions 2, 3, or 5 on the benzene ring. Still further, each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -N3, -SO3H, acyl, alkyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero- )arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, heterocyclyl, heterocyclylalkyl, alkenyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, -SH, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl.
[0341] In another embodiment of the invention, if R2 is C=O(Rs), then R5 is selected from the group consisting of -NR16, NHNHR16, NHOH5 -ORi5, CONH2NHRi6, CONR]65 CH2X, acyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted with one or more radicals independently selected from the group consisting of halogen, N, O, -S-, -CN, -N3, nitro, oxo, acyl, alkyl, alkoxyl, alkylamino, alkenyl, aryl, (hetero-)cycloalkyl, and (hetero-)cyclyl.
Efficacy Demonstrations
[0342] As demonstrated by Figure 1, embodiments A, B, C, and D, S36 is more potent at increasing the binding of FKBP12.6 and RyR2 than JTV-519 and does not block the L-type Ca2+ channel (Ica,L) or HERG K+ channel (Iκr)- In embodiment A, PKA phosphorylated RyR2 is generated as follows: cardiac SR membrane preparations (5 μl, 50 μg) are added to a total of 100 μl of kinase buffer (8 mM MgCl2, 10 mM EGTA, 50 rnM Tris-PIPES, pH 6.8) containing 100 μM MgATP and 40 units of PICA, and incubated at room temperature. Samples are centrifuged at 95,000 g for 10 min and the pellets are washed three times in 0.2 ml imidazole buffer. The final pellets are pooled and resuspended in imidazole buffer (final concentration ~ 10 μg /μl). To test for the FKBP12.6 rebinding efficiency of JTV-519, PICA phosphorylated cardiac SR (50 mg) is incubated for 30 minutes at room temperature with the test compounds and 250 nM FICBP12.6 in 10 mM imidizol buffer, pH 7.0. Samples then are centrifuged at 100,000 g for 10 minutes and pellets washed 3 times with imidizol buffer. After washing, proteins are size-fractionated on 15% PAGE. Immunoblots are developed using an anti-FKBP antibody (1 :3,000 dilution). The amount of rebinding is quantified using densitometry of Western blots and is compared to the amount of FKBP associated with RyR in non-phosphorylated SR. ECso's for the compounds are determined by generating FKBP binding data using concentrations of compounds ranging from 0.5-1000 nM. In embodiment B, currents through L-type Ca2+ channels in isolated mouse cardiomyocytes are recorded using whole-cell patch clamp recording conditions with Ba2+ as the charge carrier. The extracellular solution contains (in mM): N-methyl-D-glucamine, 125; BaCl2, 20; CsCl, 5; MgCl2, 1; HEPES, 10; glucose, 5; pH 7.4 (HCl). The intracellular solution contains (in mM): CsCl, 60; CaCl2, 1; EGTA, 11; MgCl2, 1; K2ATP, 5; HEPES, 10; aspartic acid, 50; pH 7.4 (CsOH). Under these conditions, it is expected that the measured current was carried by Ba2+ primarily through L-type calcium channels which is referred to as Ica,L- Drugs are applied by a local solution changer and reach the cell membrane within 1 s. The effects of nifedipine and S36 are tested with 20 ms long voltage-clamp steps to +10 or +20 mV (peak of current- voltage relation for each individual cell) from holding potentials of -80 mV or -40 mV. In embodiment C, the voltage-dependence of L-type Ca2+ current blocked by JTV-519 (1 μM) and S36 (lμM) are measured and presented.
[0343] As demonstrated by Figure 2, embodiments A, B, C, and D, S36 prevents exercise- induced sudden cardiac death at lower plasma levels compared with JTV-519. In embodiment A are shown representative ECGs of an untreated FKBP12.6+/~ mouse and JTV519-treated FICBP12.6+/" and FKBP12.6"7" mice. Mice are treated with 0.5 mg JTV- 519/per kilogram of body weight per hour for 7 days with an implanted osmotic mini-pump. JTV-519 has no effect on resting heart rate or other ECG parameters such as heart rate (HR). In embodiment B are shown sustained polymorphic ventricular tachycardia recorded by telemetry in an untreated FKBP12.6+/ mouse (upper tracing) subjected to exercise testing, immediately followed by injection with 0.5 mg epinephrine per kilogram of body weight. Representative telemetry ECG recording of a JTV-519-treated FKBP12.6+/~ mouse following the same protocol is shown in the bottom tracing, hi embodiment C are shown numbers of mice with cardiac death (left), sustained VTs (>10 beats, middle), and nonsustained VTs (3 to 10 arrhythmogenic beats, right) in experimental groups of mice subjected to exercise testing and injection with 0.5 mg/kg epinephrine. In embodiment D, the dose-dependence of pharmacological effects of JTV-519 and S36 is shown. Plasma levels of 1 μM JTV519 prevent cardiac arrhythmias and sudden cardiac death in FKBPl 2.6+/~ mice. Plasma levels of 1 μM and 0.02 μM S36 also prevent cardiac arrhythmias and sudden cardiac death in FKBP12.6+/"mice.
[0344] As demonstrated by Figure 3, S36 prevents the development of acute heart failure post-myocardial infarction. Placebo-treated or S36 (100 nM or 200 nM plasma concentrations)-treated mice are subjected to permanent ligation of the left anterior descending coronary artery resulting in myocardial infarction. S36 significantly improves fractional shortening assessed by M-mode echocardiography 2 weeks post-myocardial infarction, compared with placebo.
[0345] As demonstrated by Figure 4, S36 improves cardiac function in chronic heart failure post-myocardial infarction. Wild-type mice are subjected to permanent ligation of the left anterior descending coronary artery resulting in myocardial infarction. Seven days following myocardial infartion, mice are treated with S36 (plasma concentration 200 nM) or placebo. Heart weight to body weight (HW/BW) ratios and pressure-volume loops quantifications (dP/dt, slope of the maximum derivative of change in systolic pressure over time) show reverse remodeling and improved cardiac contractility in S36-treated mice compared with placebo.
[0346] Figure 5 is a summary graph of EC50 values of JTV-519 and compounds S1-S67 disclosed herein. The FKBP12.6 rebinding assay described above is used to determine the amount of FKBP12.6 binding to PKA-phosphorylated RyR2 at various concentrations (0.5 - 1000 nM) of the compounds shown. EC50 values are calculated using Michaelis-Menten curve fitting.
[0347] As demonstrated by Figure 6, embodiments A, B, and C, S36 normalizes CPVT- associated RyR2-P2328S channel function and structure. In embodiment A are shown representative single-channel current traces of vmphosphorylated RyR2-P2328S and PKA- phosphorylated RyR2-WT treated with S36 showing no influence of JTV-519 on baseline channel function. However, in PKA phosphorylated RyR2-P2328S, as shown in embodiment B, S36 normalizes the single channel closed state to levels approaching those seen in embodiment A, reducing the open probability from 14.4% to 0.3% following administration of 0.1 μmol/L S36. Insets in embodiments A and B show channel openings >1 pA at a higher resolution. Embodiment C shows immunoblot analysis of calstabin-2 binding of RyR2- P2328S in the presence or absence of PKA and 0.1 μmol/L S36, as indicated. RyR2-P2328S is imrnunoprecipitated and in vitro PKA was phosphorylated as described above.
[0348] As demonstrated by Figure 7, embodiments A and B, treatment with JTV-519 reduces PKA phosphorylation of RyR2 in mice with heart failure. Equivalent amounts of RyR2 are immunoprecipitated with an antibody against RyR2 (top blot). Representative immunoblots (embodiment A) and bar graphs (embodiment B) show the amount of PKA phosphorylated RyR2 at Ser-2808 bound to RyR2 in wild-type and calstabin2(FKBP12.6)"A mice. Treatment with JTV-519 (0.5 mg/kg/h) for 28 days post-myocardial infarction reduces PKA- phosphorylation of RyR2, presumably due to reverse cardiac remodeling, in wildtype but not calstabin-2 (FKBPl 2.O)^ nIiCe.
[0349] As demonstrated by Figure 8, embodiments A and B, mice in which cardiac RyR2 cannot be PKA phosphorylated (RyR2-S2808A knockin mice) have improved cardiac function following myocardial infarction. Shown in embodiment A is the quantification of M-mode echocardiograms showing improved ejection fraction in RyR2-S2808A knockin mice compared with wildtype 28 days following permanent coronary artery ligation. Shown in embodiments B and C are pressure-volume loop quantifications showing (embodiment B) improved cardiac contractility and decreased cardiac dilation (embodiment C) in RyR2- S2808A knockin mice compared with wildtype following myocardial infarction.
[0350] Figure 9, embodiments A, B, C, and D demonstrate the effect of JTV-519 on calstabin2 affinity to RyR2 in haploinsufficient calstabin2 (FKBPl 2.6)+/~ mice after exercise. The average probability that RyR2 channels will be open in calstabin2+/~ mice subjected to exercise were significantly increased compared to those of channels from exercised wild type (control; calstabin2+/+) mice, which are predominately closed under conditions that simulate diastole in the heart. As shown in embodiments C and D, treatment of exercised calstabin2+/~ mice with JTV-519 significantly reduced the channel open probability (P0) compared with that of channels from exercised mice that were not treated, consistent with increased amounts of calstabin2 in the RyR2 channel complex. Therefore, JTV-519 increases the binding affinity of calstabin2 to RyR2. In contrast, JTV-519 treatment of exercised Calstabin2~^ deficient mice did not result in channels with a low P0, indicating that the presence of calstabirώ is necessary for JTV-519 effects which are documented as binding of calstabin2 to RyR2.
[0351] Figure 10, embodiments A, B, C, D, E, and F demonstrate, respectively, normalized RyR2 channel gating and increased calstabin2 binding to RyR2 channels after treatment with JTV-519. The immunoblots in embodiments A and B show the amounts of RyR2 and calstabin2 associated with immunoprecipitated RyR2 after incubation with the indicated concentrations of JTV-519 for, respectively, wild-type RyR2 (RyR2-WT) channels and RyR2-S2809D. The binding curves in embodiment C demonstrate that JTV-519 significantly increases the affinity of calstabin2 for PKA-phosphorylated RyR2 channels. The results also show that depletion of calstabin2 from the RyR2 macromolecular complex which is associated with increased RyR2 open probability, ventricular tachycardias, and sudden cardiac death in haploinsufficient calstabin2+/~ mice is reversed by treatment with JTV-519. Therefore, the JTV-519 and related compounds prevent disorders and conditions associated with the RyR2 receptors.
[0352] As shown in Figure 11, embodiments A, B, C, D, and E RyRl channel functioning is increased and normalized in mdx (dystrophin deficient) mice treated with JTV-519. In Figure 11, channel openings are represented as upward deflections; 'c' indicates the closed state; and 4 pA current open amplitude is indicated by dash. Upper traces represent 5 sees and lower traces 500 ms; dotted lines indicate subconductance states.
[0353] Embodiment A of Figure 11 shows a single channel current trace of RyRl from soleus muscle of control (wild-type) mouse under resting conditions (cytoplasmic Ca2+ 150 nM). As seen, RyRl is predominantly closed. Embodiment C of Figure 11 shows that RyRl channel function in mdx mouse shows significantly increased open probability, increased average open, and decreased average closed dwell times, To and Tc, respectively. Increased Po in mdx mice is consistent with intracellular Ca2+ leak. The amplitude histograms in embodiments B, D, and F show multiple subconductance states consistent with calstabinl (FKBP 12) depletion in RyRl from mdx soleus muscle. Embodiment E of Figure 11 shows an mdx mouse treated with 1.0 μM JTV-519. As seen, the RyRl channels JTV-519-treated mouse demonstrates normal activity that is not significantly different from untreated wild- type trace, thereby indicating that JTV-519 can normalize RyRl channel function m mdx mice.
[0354] The data of Figure 11 are consistent with intracellular SR Ca2+ leak via RyRl channels as the cause of increased cytosolic Ca2+ leak in skeletal muscles from mdx (dystrophin deficient) mice.
[0355] Figure 12, embodiments A and B, demonstrate that mdx skeletal muscle has normal levels of RyRl PKA phosphorylation, but depleted levels of calstabml. The imrnunoblots in embodiment A show that mdx type mice have depeleted levels of calstabml compared to a control (wild-type) mouse The summary bar graphs of embodiment B show that the mdx mouse, nevertheless, has an equivalent level of PKA-phosphorylation. Therefore, it is concluded that calstabml depeletion is a defect that is consistent with the intracellular Ca2+ leak observed in skeletal muscle cells from mdx mice and myofibers from human mutation carriers. Intracellular SR Ca2+ leak is likely to contribute to myofiber death and wasting of muscle mass by toxic intracellular Ca2+ overload and activation of proteases.
[0356] Figure 13, embodiments A, B, and C, demonstrates that SR Ca2+ leak at the subcellular level m skeletal muscles of animals with heart failure is detectable. Life quality and prognosis in heart failure (HF) patients is severely decreased due to skeletal muscle dysfunction (e g , shortness of breath due to diaphragmatic weakness, and exercise intolerance due to limb skeletal muscle fatigue) in addition to depressed cardiac function. Dysregulation of intracellular SR Ca2+ release is a pathogenic mechanism underlying skeletal muscle dysfunction m HF HF in animals causes significantly accelerates intrinsic skeletal muscle fatigue.
[0357] Embodiments A and B of Figure 13 are ΔF/F fluorescence line scan images of representative examples of Ca2+ sparks in myofibers from sham and postmyocardial infarction (PMI) rats and corresponding Ca2+ spark time course. Embodiment C shows the relative distribution of the spatio-temporal properties of the Ca2+ sparks Charts indicate 25, 50, 75 percentiles, the horizontal lines indicate the range from 1-99% of the distribution. Sham, open symbols (n = 137, three animals); postmyocardial infarction (PMI), gray symbols (n = 82, two animals) *, P < 0.05. FDHM, full duration at 50% peak amplitude; FWHM, full width at 50% peak amplitude.
[0358] Figure 14, embodiments A and B, demonstrate that treatment of wild-type mice with JTV-519 improved soleus muscle fatigue times compared with placebo. Soleus muscles from JTV519-treated wild-type mice or calstabin2 mice with heart failure from myocardial infarction are more resistant to fatigue (P < 0.05) compared to placebo-treated mice. After completion of treatment, soleus muscle was dissected and mounted in a tissue bath to assess isolated skeletal muscle function. Representative 50% of maximal tetanic force fatigue time tracings are shown for wild-type and calstabin2 mice, treated with JTV519 or placebo, in embodiment A. In embodiment B are shown bar graph summarizing mean time to fatigue.
[0359] In summary, JTV-519 treatment improved skeletal muscle fatigability in heart failure animals in vivo. Interestingly, in calstabin∑1* knockout mice, fatigue times were also significantly improved in JTV519 treated mice, suggesting that the beneficial effects on isolated skeletal muscle function depend on calstabinl and not calstabin2 binding to RyRl. Indeed, calstabinl appears to be the only isoform of functional significance expressed in skeletal muscles.
[0360] Figure 15, embodiments A and B, demonstrates that in a mouse model of post- myocardial infarction heart failure, RyRl in soleus muscle is also PKA-hyperphosphorylated. Both in wild-type and calstabkώ"7" mice, JTV-519 increased binding of calstabinl to RyRl in soleus muscle, suggesting that JTV-519 improves skeletal muscle fatigability by normalizing calstabinl rebinding to the channel complex. Equivalent amounts of RyRl were immunoprecipitated with an antibody against RyRl . Embodiment A provides bar graphs show the amount of PKA phosphorylation of RyRl at Ser-2844 in mice (corresponding to human Ser-2843). The significant decrease of RyRl PKA phosphorylation in JTV519 treated wild-type animals is likely resulting from beneficial cardiac effects and secondary reduction of sympathetic nervous activity. Embodiment B are bar graphs showing the amount of calstabinl bound to RyRl from wild-type or calstabin2~'~ mice treated with JTV519 or placebo. Mice were treated with JTV519 by implantable osmotic minipumps using a dose of 0.5 mg/kg/day. In summary, JTV-519 treatment resulted in a highly significant increase of calstabinl in the RyRl complex in soleus muscles in vivo.
[0361] Figure 16, embodiments A, B, C, and D demonstrates that calstabinl rebinding to RyRl by JTV519 normalizes abnormal or leaky RyRl channel function in vivo. In embodiments A and C are shown RyRl single-channel tracings at 150 nM cytoplasmic Ca2+ representing resting conditions in skeletal muscle for wild-type mice treated with placebo and JTV-519. JTV-519 treatment of mice with heart failure and increased muscle fatigue normalized RyRl channel gating in skeletal muscle in vivo. Channel openings are upward, the dash indicates the full level of channel opening (4 pA), the dotted lines indicate subconductance levels, and 'c' indicates the closed state of the channels. For the amplitude histograms in embodiment B and D, amplitude is represented on the x axis, and events indicates the number of channel openings. Po, To and Tc values correspond to the representative traces. Treatment as indicated on top of traces. Inset shows higher resolution of open states.
[0362] In summary, the data shows that in vivo JTV519 treatment normalizes skeletal muscle function and RyRl channel dysfunction consistent with preventing intracellular SR Ca2+ leak as a cause of increased skeletal muscle fatigue.
[0363] Figure 17 demonstrate that JTV-519 also increases the binding affinity of calstabinl for RyRl in skeletal muscle in vivo. This probably explains why JTV519 treated mice with heart failure have increased levels of calstabinl bound to RyRl in soleus muscle. In embodiment A, equivalent amounts of skeletal RyRl or cardiac RyR2 were immunoprecipitated, PKA phosphorylated and incubated with calstabinl or calstabin2 at increasing concentrations of JTV519, respectively. Sediment represents only calstabin bound to RyR. Immunoblotting showed that >50 nM of JTV519 increased the binding affinity of calstabin for RyR. The graphs of embodiment B further demonstrate that PKA phosphorylation of RyRl reduced the affinity of calstabinl for RyRl (open circles), whereas treatment with JTV519 (filled circles) restored the binding affinity of calstabinl for RyRl to that of non-PKA phosphorylated RyRl (open squares).
[0364] Figure 18, embodiment A, B, C, and D demonstrates that Ser-2843 is the unique PKA phosphorylation site in skeletal RyRl channels. (A) Representative single channel traces of wild-type RyRl, (B) effect of exogenous PICA phosphorylation of RyRl (wt RyRl-P), (C) PKA does not affect RyRl-S2843A that contains a non-functional PKA phosphorylation site. Since PKA does not increase RyRl-S2843A activity, Ser-2843 appears to constitute the unique PICA phosphorylation site in RyRl channels in skeletal muscle. Accordingly, (D) constitutively phosphorylated RyRl-S2843D mimics exogenous PKA phosphorylation shown in (B) confirming that Ser-2843 is the unique PKA phosphorylation site in skeletal RyRl channels. RyRl single channel recordings in planar lipid bilayers show activity of the channels at 150 nM [Ca2+]cfa (cytosolic side) with 1 mM ATP. Recordings were at 0 mV, closed state of the channels as indicated by 'c', and channel openings are upward deflections. AU point amplitude histograms are shown on the right. Open probability (P0) and mean closed (Tc) and open (To) dwell times are indicated above each channel tracing. [0365] Figure 19, embodiments A and B, demonstrates the depletion of stabilizing calstabinl and PKA hyperphosphorylation of RyRl channels from sustained exercise. Aerobic exercise can be defined as a form of physical exercise that increases the heart rate and enhances oxygen intake to improve performance. Examples of aerobic exercise are running, cycling, and swimming. During the study of Figure 19, mice were challenged by aerobic exercise (forced swimming) for 90 mins twice daily. The animals were accustomed to swimming in preliminary training sessions: day -3 twice 30 mins, day -2 twice 45 mins, day -1 twice 60 mins, day 0 and following twice 90 mins. Mice were then exercised for 1, 7, or 21 additional, consecutive days for 90 mins twice daily. Between swimming sessions separated by a 4 hour rest period the mice are kept warm and given food and water. An adjustable-current water pool was used to exercise mice by swimming. An acrylic pool (90 cm long x 45 cm wide x 45 cm deep) filled with water to a depth of 25 cm was used. A current in the pool was generated with a pump. The current speed during the swimming session was at a constant speed of 1 1/min flow rate. The water temperature was maintained at 34°C with an electric heater. Age- and weight-matched mice were used to exclude differences in buoyancy from body fat.
[0366] Using forced swimming as an efficient protocol to increase skeletal muscle aerobic capacity in mice, the composition and phosphorylation status of the skeletal RyRl channel complex have been investigated. Unexpectedly, after 3 weeks of 90 mins swimming twice daily, C57B16 wild-type mice showed significantly increased RyRl phosphorylation by PKA while Ca2+-calmodulin kinase II (CaMKII) phosphorylation was not changed indicating specificity of the stress pathway RyRl protein expression was stable, however, RyRl channels were depleted of the stabilizing subunit calstabinl (FKBP 12). It has been shown that RyRl hyperphosphorylation and calstabinl depletion are consistent with leaky RyRl channels that cause intracellular SR Ca2+ leak.
[0367] RyRl channels are PKA hyperphosphorylated and depleted of the stabilizing calstabinl subunit after 3 weeks of 90 mins swimming twice daily. As seen in Embodiment A, the immunoprecipitated RyRl macromolecular channel complex shows increased PKA phosphorylation at Ser-2844 (corresponding to human RyRl-Ser-2843) whereas CaMKII phosphorylation at Ser-2849 (corresponding to human RyRl-Ser-2848) is unchanged. Concomitant with increased RyRl -Ser-2844 PKA hyperphosphorylation, calstabinl is depleted from the channel complex. As seen in embodiment B, normalization of phosphorylation and calstabinl content to four subunits of the tetrameric channel complex shows a significant in increase in PKA phosphorylation and depletion of the stabilizing calstabinl subunit. Control, non-exercised mice; swim, mice exercised 90 mins twice daily for 3 weeks (preliminary data). P < 0.05.
[0368] Figure 20, embodiments A and B, demonstrate that PKA phosphorylation increases for increasing durations of sustained exercise. To investigate the influence of the duration of sustained exercise on the RyRl Ca2+ release channel defect, mice were exposed to swimming for 1, 7, or 21 days followed by immediate sacrifice. Longer exposure to sustained exercise results in a significant increase of RyRl PKA hyperphosphorylation beginning at 7 days and saturating at 21 days.
[0369] In Figure 20, embodiment A, the immunoprecipitated RyRl channel complex shows significantly and above physiologic levels increased PKA phosphorylation at Ser-2844 (corresponding to human RyRl-Ser-2843) after 7 days of swimming exercise. In Figure 20, embodiment B, inormalization of RyR2-Ser-2844 phosphorylation within the tetrameric channel complex documents a significant increase in PKA phosphorylation. *, P < 0.05; **, P < 0.005.
[0370] In summary, the data of Figure 20 shows that sustained exercise results in significantly increased RyRl phosphorylation by protein kinase A (PICA) which contributes to depletion of the stabilizing calstabinl subunit from the channel complex as the cause of a gain-of-function defect.
[0371] Figure 21 provides data showing that showing that chronically increased sympathetic stimulation of skeletal muscles, results in RyRl -dependent intracellular Ca2+ leak and significantly increased muscle fatigue. What is the functional consequence of chronically increased RyRl PKA hyperphosphorylation? As shown in Figure 21 for mice and rats with heart failure from myocardial infarction, chronic RyRl PKA hyperphosphorylation results in increased muscle fatigue.
[0372] In embodiment A, it can be seen that heart failure skeletal muscle fatigues earlier than control. Rat soleus muscle (n = 5 control, n = 8 HF) was mounted in a tissue bath to assess contractile function. Representative fatigue time tracing is shown for control and HF skeletal muscles. Bar graph shows mean (± S.D.) time to 40% fatigue. *, P < 0.05. In embodiment B, it can be seen that heart failure skeletal muscle achieved maximal tetanic force more slowly than control skeletal muscles. Tetanic force was induced by high-frequency field stimulation. Bar graph shows tetanic 50% contraction time. **, P < 0.01. Embodiment C demonstrates the correlation between time to fatigue and RyRl PKA phosphorylation (r = 0.88) in rat skeletal muscle from sham and heart failure animals. Muscle function and RyRl PKA phosphorylation were assessed using contralateral soleus muscles from each animal.
[0373] In summary, Figure 21 provides data showing that sustained exercise causes RyRl PKA hyperphosphorylation and calstabinl depletion, and Figure 21 shows that the identical defect occurs in disease forms with increased sympathetic activity causing intracellular SR Ca2+ leak and significantly accelerated skeletal muscle fatigue.
[0374] An additional problem during sustained exercise and stress is skeletal muscle degeneration further contributing to decreased skeletal muscle performance. To assess structural changes during sustained exercise, histologic changes in the fast-twitch muscles of mice exposed to 3 weeks of exercise by swimming have been characterized. Results are shown in Figure 22. Cross-sections of the mouse M. extensor digitorum longus (EDL) showed histologic changes consistent with myofiber degeneration from intracellular Ca2+ overload from defective RyRl channels. Therefore sustained exercise for 90 mins twice daily triggers a dystrophic phenotype in EDL muscles of normal C57B16 mice.
[0375] Trichrome stain shows packed myofibers of similar cross-sectional dimension in non- exercised control (WT) mice (left). Three weeks swimming results in myofiber degeneration and interstitial collagen deposits with irregular fiber sizes. Hematoxylin-eosin (H&E) stain indicates nuclear changes and myofiber death. These changes are consistent with dystrophic remodeling.
[0376] The rapid delayed rectifier potassium channel (1(Kr)) is important for repolarization of the cardiac action potential. HERG is the pore-forming subunit of the I(Kr) channel. Suppression of I(Kr) function, for example as a side-effect of a drug or the result of a mutation in hERG, can lead to long-QT (LQT) syndrome, which is associated with increased risk of life-threatening arrhythmias. The compounds of the present invention exhibit a lower level of hERG blocking activity than JTV-519, as demonstrated in Figures 23- 43. Thus, the compounds of the present invention are expected to be less toxic and/or exhibit fewer side effects than JTV-519.
[0377] Figures 23 to 26 illustrate the effect of the compound ARM036 (also referred to as S36) and ARM036-Na (a sodium salt of ARM036) on hERG currents.
[0378] Figure 23 shows a typical hERG voltage-clamp current recording before (control) and after application of ARM036 at lOOμM. The voltage pulse protocol used to activate the hERG currents is illustrated below the current trace. It can be seen that, following activation by the conditioning prepulse (to +20 mV), partial repolarization (-50 mV test pulse) of the membrane evoked a large, slowly decaying outward tail current. Application of ARM036 minimally reduced the outward tail current in a concentration- and time-dependent manner.
[0379] Figure 24 shows a typical time course of the effect of ARM036 at 100 mM on the amplitude of the hERG channel current.
[0380] Figure 25 is a graph showing the concentration-dependence of the effect of ARM036 on the hERG current. Table 1 provides the numerical data that is illustrated graphically in Figure 25. Because the highest concentration of ARM036 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM036.
Table 1
Figure imgf000133_0001
[0381] Figure 26 is a graph showing the concentration-dependence of the effect of ARM036- Na on the hERG current. Table 2 provides the numerical data that is illustrated graphically in Figure 26. Because the highest concentration of ARM036-Na tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM036-NA
Table 2
[0382] Figure 27 is a graph showing the concentration-dependence of the effect of ARM047 on the hERG current. Table 3 provides the numerical data that is illustrated graphically in Figure 27. The IC50 value for ARM047 block of the hERG current was 2.496 μM.
Table 3
Figure imgf000134_0001
[0383] Figure 28 is a graph showing the concentration-dependence of the effect of ARM048 on the hERG current. Table 4 provides the numerical data that is illustrated graphically in Figure 28. Because the highest concentration of ARM048 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM048.
Table 4
Figure imgf000134_0002
[0384] Figure 29 is a graph showing the concentration-dependence of the effect of ARM050 on the hERG current. Table 5 provides the numerical data that is illustrated graphically in Figure 29. Because the highest concentration of ARM050 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM050.
Table 5
Figure imgf000135_0001
Figure 30 is a graph showing the concentration-dependence of the effect of ARM057 on the hERG current. Table 6 provides the numerical data that is illustrated graphically in Figure 30. Because the highest concentration of ARM057 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM057.
Table 6
Figure imgf000135_0002
[0385] Figure 31 is a graph showing the concentration-dependence of the effect of ARM064 on the hERG current. Table 7 provides the numerical data that is illustrated graphically in Figure 31. Because the highest concentration of ARM064 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM064.
[0386] Table 7
Figure imgf000135_0003
[0387] Figure 32 is a graph showing the concentration-dependence of the effect of ARM074 on the hERG current. Table 8 provides the numerical data that is illustrated graphically in Figure 32. Because the highest concentration of ARM050 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM074.
[0388] Table 8
Figure imgf000136_0001
[0389] Figure 33 is a graph showing the concentration-dependence of the effect of ARM075 on the hERG current. Table 9 provides the numerical data that is illustrated graphically in Figure 33. Because the highest concentration of ARM075 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM075.
Table 9
Figure imgf000136_0002
[0390] Figure 34 is a graph showing the concentration-dependence of the effect of ARM076 on the hERG current. Table 10 provides the numerical data that is illustrated graphically in Figure 34. Because the highest concentration of ARM076 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM076. Table 10
Figure imgf000137_0001
[0391] Figure 35 is a graph showing the concentration-dependence of the effect of ARM077 on the hERG current. Table 11 provides the numerical data that is illustrated graphically in Figure 35. Because the highest concentration of ARM077 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARM077.
Table 11
Figure imgf000137_0002
[0392] Figure 36 is a graph showing the concentration-dependence of the effect of ARMlOl on the hERG current. Table 12 provides the numerical data that is illustrated graphically in Figure 36. Because the highest concentration of ARMlOl tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARMlOl .
Table 12
Figure imgf000138_0001
[0393] Figure 37 is a graph showing the concentration-dependence of the effect of ARM102 on the hERG current. Table 13 provides the numerical data that is illustrated graphically in Figure 37. Because the highest concentration of ARM102 tested resulted in less than 50% current inhibition, it was not possible to determine an ICs0 value for ARMl 02.
Table 13
Figure imgf000138_0002
[0394] Figure 38 is a graph showing the concentration-dependence of the effect of ARM103 on the hERG current. Table 14 provides the numerical data that is illustrated graphically in Figure 38. Because the highest concentration of ARM103 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARMl 03.
Table 14
Figure imgf000139_0001
[0395] Figure 39 is a graph showing the concentration-dependence of the effect of ARM 104 on the hERG current. Table 15 provides the numerical data that is illustrated graphically in Figure 39. Because the highest concentration of ARM104 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARMl 04.
Table 15
Figure imgf000139_0002
[0396] Figure 40 is a graph showing the concentration-dependence of the effect of ARM 106 on the hERG current. Table 16 provides the numerical data that is illustrated graphically in Figure 40. Because the highest concentration of ARM106 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARMl 06.
Table 16
Figure imgf000140_0001
[0397] Figure 41 is a graph showing the concentration-dependence of the effect of ARM 107 on the hERG current. Table 17 provides the numerical data that is illustrated graphically in Figure 41. Because the highest concentration of ARM 107 tested resulted in less than 50% current inhibition, it was not possible to determine an IC50 value for ARMl 07.
Table 17
Figure imgf000140_0002
[0398] Figure 42 is a graph showing the concentration-dependence of the effect of S26 on the hERG current. Table 18 provides the numerical data that is illustrated graphically in Figure 42. The IC50 value for S26 was 7.029 μM.
Table 18
Figure imgf000141_0001
[0399] Figure 43 is a graph showing the concentration-dependence of the effect of JTV-519 (referred to in the figure as "ARMOXX") on the hERG current. Tablel9 provides the numerical data that is illustrated graphically in Figure 43. The IC50 value for JTV-519 was 0.463 μM.
Table 19
Figure imgf000141_0002
[0400] The antiarrhythmic drug E-4031, a known blocker of hERG currents, was used as a positive control. E-4031 blocked the hERG current with an ICs0 of 0.5 μM (n = 6).
[0401] In summary, the compounds of the present invention exhibit reduced hERG blocking activity as compared to JTV-519. Thus, the compounds of the invention are expected to be less toxic and/or exhibit fewer side effects than JTV-519.
[0402] Table 20 below provides EC50 values for compounds Sl -S 107. These EC50 data were obtained using thee FKBP12.6 rebinding assay described above to determine the amount of FKBP 12.6 binding to PKA-phosphorylated RyR2 at various concentrations (0.5 - 1000 nM) of the compounds shown in Table 20. The EC50 values are calculated using Michaelis- Menten curve fitting. Table 20
Figure imgf000142_0001
High-throughput screening method
[0403] In addition to the compounds disclosed herein, other compounds can be discovered that are capable of modulating calcium ion channel activity, in particular those channels related to the RyR series of caclium ion channels. Provided herein is a highly-efficient assay for high-throughput screening of other compounds that are capable of modulating calcium ion channel activity.
[0404] By way of example, and as shown in Example 5 below, a highly-efficient assay for high-throughput screening for small molecules is developed by immobilizing FKBP, either FKBP 12 or FKBP12.6 {e.g., wild-type FKBP12.6 or a fusion protein, such as GST- FKBP 12.6) onto a 96-well plate coated with glutathione, using standard procedures. PKA- phosphorylated ryanodine receptor (RyR), specifically RyRl or RyR3 in the case of FKBP 12 and RyR2 in the case of FKBP12.6, is loaded onto the FKBP-coated plate, and incubated with compounds at various concentrations (10-100 nM) for 30 min. Thereafter, the plate is washed to remove the unbound RyR, and then incubated with anti-RyR antibody (e.g., for 30 min). The plate is washed again to remove unbound anti-RyR antibody, and then treated with florescent-labeled secondary antibody. The plate is read by an automatic fluorescent plate reader for binding activity.
[0405] Alternatively, RyR is PKA-phosphorylated in the presence Of32P-ATP. Radioactive PKA-phosphorylated RyR is loaded onto an FKBP-coated, 96-well plate, in the presence of JTV-519 analogues and other compounds at various concentrations (10-100 nM) for 30 min. The plate is washed to remove the unbound radiolabeled RyR, and then read by an automatic plate reader. PKA-phosphorylated RyR also is coated to the plate, and incubated with 32S- labeled FKBP in the presence of the compounds.
[0406] The present invention is described in the following Examples, which are set forth to aid in the understanding of the invention and should not be construed to limit in any way the scope of the invention as defined in the claims which follow thereafter.
EXAMPLES
EXAMPLE 1 - RvR2 PKA PHOSPHORYLATION AND FKBP 12.6 BINDING
[0407] Cardiac SR membranes are prepared, as previously described (Marx, et al, PKA phosphorylation dissociates FKBP 12.6 from the calcium release channel (ryanodine receptor): defective regulation in failing hearts. Cell, 101:365-76, 2000; Kaftan, et al, Effects of rapamycin on ryanodine receptor/Ca^2+^-release channels from cardiac muscle. Circ. Res., 78:990-97, 1996). 35S-labelled FKBP12.6 was generated using the TNT™ Quick Coupled Transcription/Translation system from Promega (Madison, WI). [3H] ryanodine binding is used to quantify RyR2 levels. 100 μg of microsomes are diluted in 100 μl of 10-mM imidazole buffer (pH 6.8), incubated with 250-nM (final concentration) [35S] -FKBP 12.6 at 37°C for 60 min, then quenched with 500 μl of ice-cold imidazole buffer. Samples are centrifuged at 100,000 g for 10 min and washed three times in imidazole buffer. The amount of bound [35S]-FKBP 12.6 is determined by liquid scintillation counting of the pellet.
EXAMPLE 2 - IMMUNOBLOTS [0408] Immunoblotting of microsomes (50 μg) is performed as described, with anti- FKBP12/12.6 (1:1,000), anti-RyR-5029 (1:3,000) (Jayaraman, et al, FK506 binding protein associated with the calcium release channel (ryanodine receptor). /. Biol. Chem., 267:9474- 77, 1992), or anti-phosphoRyR2-P2809 (1:5,000) for 1 h at room temperature (Reiken, et al., Beta-blockers restore calcium release channel function and improve cardiac muscle performance in human heart failure. Circulation, 107:2459-66, 2003). The P2809- phosphoepitope-specific anti-RyR2 antibody is an affinity-purified polyclonal rabbit antibody, custom-made by Zymed Laboratories (San Francisco, CA) using the peptide, CRTRRI-(pS)-QTSQ, which corresponds to RyR2 PKA-phosphorylated at Ser2809. After incubation with HRP -labeled anti-rabbit IgG (1 :5,000 dilution; Transduction Laboratories, Lexington, KY), the blots are developed using ECL (Amersham Pharmacia, Piscataway, NJ).
EXAMPLE 3 - SINGLE-CHANNEL RECORDINGS
[0409] Single-channel recordings of native RyR2 from mouse hearts, or recombinant RyR2, are acquired under voltage-clamp conditions at 0 mV, as previously described (Marx, et al., PKA phosphorylation dissociates FKBP12.6 from the calcium release channel (ryanodine receptor): defective regulation in failing hearts. Cell, 101:365-76, 2000). Symmetric solutions used for channel recordings are: trans compartment - HEPES, 250 mmol/L; Ba(OH)2, 53 mmol/L (in some experiments, Ba(OH)2 is replaced by Ca(OH)2); pH 7.35; and cis compartment - HEPES, 250 mmol/L; Tris-base, 125 mmol/L; EGTA, 1.0 mmol/L; and CaCl2, 0.5 mmol/L; pH 7.35. Unless otherwise indicated, single-channels recordings are made in the presence of 150-nM [Ca2+] and 1.0-mM [Mg2+] in the cis compartment. Ryanodine (5 mM) is applied to the cis compartment to confirm identity of all channels. Data is analyzed from digitized current recordings using Fetchan software (Axon Instruments, Union City, CA). All data is expressed as mean ± SE. The unpaired Student's t- testis used for statistical comparison of mean values between experiments. A value of p<Q.O5 is considered statistically significant.
EXAMPLE 4 - COMPOUNDS AND METHODS FOR THEIR SYNTHESIS Scheme 1 : Synthesis of S3, S4, S5 and S54
[0410] Synthon S26 was prepared according to methods described in U.S. Patent App. No. 10/680,988.
[0411] Synthesis of S3 (Scheme 1): To a stirred solution of vinylsulfonic acid (22 mg, 0.2 mmol) in anhydrous CH2Cl2 (5 ml) is added thionyl chloride (2M in CH2Cl2, 0.1 ml, 0.2 mmol). The reaction mixture is stirred at room temperature overnight and evaporated under vacuum. The residue is dissolved in CH2Cl2 (5 ml). To this solution, a solution of S26 (20 mg, 0.1 mmol) in CH2Cl2 (3 ml) is added drop-wise at O0C. The reaction mixture is stirred at 00C for one hour and at room temperature for another hour and washed with saturated sodium bicarbonate and IN HCl. After removal of the solvent, the product S3 is purified by SiO2 column chromatography as a colorless oil (18 mg, 65%).
[0412] Synthesis of S4 (Scheme 1): To a stirred solution of S26 (20 mg, 0.1 mmol) in CH2Cl2 (5 ml) is added methylsulfonyl chloride (26 mg, 0.2 mmol) and triethylamine (30 mg, 0.3 mmol) at 0°C. The resulting mixture is stirred at O0C for one hour and at room temperature overnight. The organic phase is washed with aqueous saturated sodium bicarbonate and dried over sodium sulfate. After filtration and evaporation of the organic solvents, the product S4 is purified by SiO2 column chromatography (25 mg oil, yield: 90%). Similarly, S5 and S54 are synthesized in 95% and 91% yields respectively.
Scheme 2: Synthesis of S1 and S2
Figure imgf000145_0002
[0413] Synthesis of Sl and S2 (Scheme 2): To a solution of S3 (28 mg, 0.1 mmol) in chloroform (5 ml) is added 4-benzylpiperidine (18 mg, 0.1 mmol). The resulting mixture is stirred at room temperature for 1 day. After removal of organic solvent, the residue is purified on silica gel column. Product Sl is obtained as a colorless oil (34 mg, yield 75%). S2 is synthesized similarly from S3 and dibutylamine in 78% yield.
Scheme 3: Synthesis of S7, S9 and S40
H5-
Figure imgf000146_0001
[0414] Synthesis of S7, S9, S27 and S40 (Scheme 3): To a stirred solution of iodoacetic acid (37 mg, 0.2 mmol) in CH2Cl2 (10 ml) is added thionyl chloride (2 M solution in CH2Cl2, 0.1 ml, 0.2 mmol). The resulting mixture is stirred at 00C for one hour and at room temperature overnight. After removal of solvent, the crude acid chloride is added to a stirred solution of S26 (20 mg, 0.1 mmol) and triethylamine (30 mg, 0.3 mmol) in CH2Cl2 (10 ml) at O0C. The mixture is stirred at 00C for one hour and at room temperature overnight. The organic phase is washed with saturated sodium bicarbonate and IN HCl. The crude product is purified by column chromatography to give S7 as a colorless oil (34 mg, yield, 95%). Similarly, S9 is synthesized in 95% yield; synthon S27 is synthesized in 96% yield; and S40 is synthesized in 91% yield using N-hydroxysuccinimidyl 4-azidosalicylic acid (NHS-ASA).
Scheme 4: Synthesis of S11 and S12
Figure imgf000146_0002
[0415] Synthesis of SIl and S12 (Scheme 4): To a solution of S26 (20 mg, 0.1 mmol) in pyridine (1 ml) is added phenyl isocyanate (18 mg, 0.15 mmol). The resulting mixture is stirred at room temperature for 24 hours. Then ethyl acetate (10 ml) is added and the organic phase is washed with IN HCl and saturated sodium bicarbonate. The product SIl is purified by SiO2 column chromatography as a white solid (27 mg, yield: 86%). Similarly, S12 is synthesized from S26 and phenyl isothiocyanate in 85% yield.
Scheme 5: Synthesis of S13 and S14
Figure imgf000146_0003
S26 S13/S14 [0416] Synthesis of S13 and S14 (Scheme 5): To S26 (20 mg, 0.1 mmol) in CH2Cl2 (5 ml) is added triethylamine (30 mg, 0.3 mmol) and phenyl methoxyphosphonyl chloride (38 mg, 0.2 mmol) at 00C. After stirring for 2 hours at room temperature, the reaction mixture is washed with saturated sodium bicarbonate. Isomers are separated and purified by silica gel column to yield S13 (14 mg, yield: 40%) and S14 (16 mg, yield: 45%).
Scheme 6: Synthesis of S19, S22
Figure imgf000147_0001
S19: X=CH2-CH2 S22: X=]
[0417] Synthesis of S19 (Scheme 6): To a stirred solution of S26 (20 mg, 0.1 mmol) and triethylamine (30 mg, 0.3 mmol) in CH2Cl2 (5 ml) is added 1,4-butyldiacid chloride (8 mg, 0.05 mmol) at 00C. The resulting mixture is stirred at 00C for one hour and at room temperature overnight. The organic phase is washed with saturated sodium bicarbonate and IN HCl and water. After removal of solvent, product S19 is purified by column chromatography (oil, 19 mg, 80% yield). Similarly S22 is prepared from 2,6 pyridyl dicarboxylic acid dichloride.
Scheme 7: Synthesis of S20 and S23
Figure imgf000147_0002
[0418] Synthesis of S20 and S23 (Scheme 7): S27 (25 mg, 0.1 mmol) in MeOH (5 ml) is treated with H2O2 (30%, 0.5 ml) at room temperature for 1 day. After treatment with sodium thiosulfate solution, methanol is removed by evaporation. The resulting residue is dissolved in ethyl acetate (10 ml) and washed with saturated sodium carbonate. After drying over sodium sulfate, solvent is evaporated to provide a crude product which is purified by silica gel column chromatography to yield S20 as colorless oil (16 mg, 60% yield). Similarly, S23 is synthesized from SlO. Scheme 8: Synthesis of S36, S43, S44, S45, S57, S59
X X==ιOCH3
Figure imgf000148_0001
Figure imgf000148_0002
[0419] Synthesis of S36 and S57 (Scheme 8): To a stirred solution of S26 (0.85g, 4.4mmol) and pyridine (0.7Og, 8.8mmol) in CH2Cl2 (50 ml) at O0C is added drop-wise methyl chlorooxoacetate (0.8 Ig, 6.6mmol). The reaction mixture is stirred at 00C for 2 hours then washed with saturated sodium bicarbonate, IN HCl, and water. Silica gel column chromatography provides S57 as a white solid (l.lg, 90% yield). S57 (1.Ig, 3.9mmol) is dissolved in methanol (10ml) and then a solution of sodium hydroxide (0.3g, 7.5mmol) in water (10ml) is added. The reaction mixture is stirred at room temperature for one hour. After solvent is removed, the residue is dissolved in water (10 ml) and washed with ether (2xl0ml). The aqueous phase is acidified with IN HCl to pH=2. The product is extracted with CH2Cl2 (2xl0ml). Removal of solvent yields product S36 as a white solid (1.Og, yield 100%). The product can be further purified by recrystalization. S38 is similarly synthesized (see Structure List).
[0420] Synthesis of S43, S44, S45 and S59 (Scheme 8): S36 (150mg, 0.56mmol) is treated with thionyl chloride (5ml) at room temperature overnight. After removal of the excess thionyl chloride, the crude product S36-C1 is dissolved in CH2Cl2 (10ml) and, to this solution, mono-Boc protected cystamine and pyridine (0.2ml, 196mg, 2.48mmol) are added at O0C. The reaction mixture is stirred at 00C for one hour and at room temperature overnight and quenched with saturated sodium bicarbonate. The organic phase is separated and the solvent is removed to give intermediate S36-cystamine, which is purified by SiO2 column chromatography in 80% yield. Deprotection of the Boc-group is achieved with trifluoroacetic acid in CH2Cl2, and the deprotected S36-cystamine is used for the synthesis of S43 and S45 by reaction with NHS-activated ester of azido compounds. Yield is 75% for S43 and 80% for S45. [0421] S44 is synthesized as a by-product of the following reaction: S36 (50mg, 0.19mmol) is treated with thionyl chloride (2ml) at room temperature overnight. After removal of the excess of thionyl chloride, the crude product is dissolved in CH2Cl2 (5ml). To this solution, cystamine (134mg, O.88mmol) and pyridine (98mg, 1.23mmol) in CH2Cl2 (10ml) is added and the reaction mixture is stirred at room temperature overnight. S44 is purified by column as a white solid (20mg, 16%). Similarly, S57 and S59 are synthesized by reaction of S36-C1 with methanol or ethylamine (Scheme 8).
Scheme 9: Synthesis of urea-based analogs S6,S46-S53,S64,S66, S67
Figure imgf000149_0001
S26-phosgene
S6: S47: NR=
Figure imgf000149_0003
Figure imgf000149_0002
S48: NR=NEt2 , S49: NR=NHCH2Ph
NR= N
Figure imgf000149_0004
[0422] Synthesis of urea-based analogs S6, S46-S53, S64, S66, S67 (Scheme 9). S26 (195 mg, 1.0 mmol) in CH2Cl2 (20 ml) is added 4-nitrophenyl chloroformate (220 mg, 1.1 mmol) and triethylamine (120 mg, 1.2 mmol) at O0C. The reaction mixture is stirred for 2 hours at room temperature and washed with water. Removal of the solvents, followed by purification using column chromatography provides compound S37 (330 mg, 91%). Reaction of S37 (36 mg, 0.1 mmole) with one equivalent of amine in DMF (3 ml) overnight provides urea-based compounds in >60% yield after purification by SiO2 column chromatography. Alternatively, the urea-based compounds can be synthesized through a versatile and more reactive intermediate S26-phosgene shown in Scheme 9. Scheme 10: Synthesis of S55, S56, S58, S60-S63
Figure imgf000150_0001
[0423] Synthesis of S55, S56, S58, S60-S63 (Scheme 10): The reaction mixture of S27 (25mg, O.lmmol) and 4-(4-aminobenzyl)piperidine (19 mg, O.lmmol) in chloroform (5 ml) is stirred at room temperature for 2 days. After removal of solvent, the product S60 is purified by silica gel column chromatography as a white solid (36mg, yield 90%). S55, S56, S58, and S61-S63 similarly are synthesized according to method described above.
Experimental for new compounds
Scheme 11 : Synthesis of urea-based analogs S69-S75
triphosgene
Figure imgf000150_0003
S68
Figure imgf000150_0002
S68-phosgene
Figure imgf000150_0004
[0424] Analogs S69-S75, having no methoxyl groups on the benzene πng (R=H in Formula I), are synthesized as shown in Scheme 11 in a similar manner to that employed in the synthesis of S46-S53 (see Scheme 9). The synthesis starts with commercially available S68 and involves a versatile intermediate, S68-phosgene, to provide S69-S75 in 60-95% yield. Scheme 12: Synthesis of S76-S81
Figure imgf000151_0001
Scheme 13: Synthesis of S82-S84
Figure imgf000151_0002
[0425] By analogy to the syntheses of S36, S43-S45, S57, and S59 (Scheme 8), S76-S81 are synthesized from commercially available S68 as shown in Scheme 12 in 70-95% yield. Using S68 as starting material, the compounds S82, S83, and S84, as shown in Scheme 13, also are synthesized similarly to the compounds which have a methoxy group on the benzene ring (R=4-OCH3 in Formula I).
Scheme 14: Synthesis of S88-S93
Figure imgf000151_0003
S90: R= ^Jh- S91 : R= -^~ S92: R=CN S93: R=PhCH2NH [0426] Synthesis of S85-S93 is accomplished as shown in Scheme 14. The following are examples of the synthesis.
[0427] Synthesis of S85: A solution of S26 (10 mmol), di-tert-bxxtyl dicarbonate (11 mmol), and triethylamine (12 mmol) in dichloromethane (100 ml) is stirred at room temperature for 5 hours. The reaction mixture is washed with saturated sodium bicarbonate solution (10 ml) and the aqueous layer is extracted with dichloromethane (2 x 15 ml). The combined organic layers are dried over magnesium sulfate and concentrated under vacuum to provide S85 as colorless oil (2.9Og, 98% yield).
[0428] Synthesis of S86: To a solution of S85 (2.36 g, 8 mmol) in dichloromethane (100 ml) at -78°C is added BBr3 (1.0 M solution in dichloromethane) (18 ml, 18 mmol) drop-wise. The solution is warmed to room temperature and the reaction mixture is quenched with methanol (100 ml) and concentrated under vacuum. The product S86 is purified by column chromatography.
[0429] Synthesis of S87: To a solution of S86 (6 mmol) in dichloromethane (40 ml) at 00C is added triethylamine (7 mmol) followed by trifluoromethylsulfonyl anhydride (7 mmol). The solution is stirred at room temperature for 30 minutes, and the reaction mixture is quenched with water (10 ml). The aqueous layer is extracted with dichloromethane (2 x 15 ml), and the combined organic layers are dried over magnesium sulfate and concentrated under vacuum. The crude product is purified by silica gel flash chromatography to provide S87 in 75% yield.
[0430] Synthesis of S88: A mixture of S87 (1 mmol), morpholine (8 ml), tris(dibenzylideneacetone)dipalladium(0) (5 mol %), 2-(di-ført-butylphosphino)-biphenyl (20 mol %), and potassium phosphate (1.2 mmol) is heated at 80°C in a sealed tube for 12 hours. The reaction mixture is cooled to room temperature, diluted with dichloromethane (50 ml), and washed with water (10 ml). The aqueous layer is extracted with dichloromethane (2 x 15 ml), and the combined organic layers are dried over magnesium sulfate and concentrated under vacuum. The crude product is purified by silica gel flash chromatography to give S88 in 81% yield.
[0431] Synthesis of S89: A solution of S87 (1 mmol), benzenethiol (2 mmol) and Z-Pr2NEt (2 mmol) in CH3CN (20 ml) is heated at 8O0C for 18 hours. After cooling, ethyl acetate (30 ml) is added and then washed with IN HCl, water, and then IN NaOH. After drying with Na2SO4, the solution was concentrated. The product S89 was purified by chromatography in 59% yield. Alternatively, S89 is synthesized by refluxing of S87 with benzenethiol in dioxane for 10 hours using z"-Pr2NEt/Pd2(dba)3/xantphos as catalyst.
[0432] Synthesis of S90 : To a solution of S87 (1.0 mmol) in dioxane (10 mL) are added K2CO3 (2 mmol), phenylboronic acid (1 mmol), and (Pd(Ph3P)4 (0.11 mmol), and the mixture is stirred at 90 0C for 16 hours. The reaction mixture is cooled to 25 0C, diluted with CH2Cl2 (30 mL), washed with water (10 mL), and the organic phase is evaporated to dryness under vacuum. Purification by column chromatography gives S90 in 40% yield.
[0433] Synthesis of S92: To a solution of S87 (1.0 mmol) in DMF (5 mL) are added zinc cyanide (1 mmol) and Pd(Ph3P)4 (0.11 mmol). The reaction mixture is stirred and heated at 1000C for 1 hour, followed by cooling, dilution with water (50 mL) and 2 M sulfuric acid (5 mL), and extraction with EtOAc (3χ). The combined organic extracts are washed with brine (2x), dried over magnesium sulfate, filtered, and evaporated under vacuum. The product S92 is purified by silica gel column chromatography in 80% yield.
Scheme 15: Synthesis of S94-S100
Figure imgf000153_0001
[0434] Synthesis of S94: To a solution of S86 (lmmol) in CH2Cl2 (10 ml) is added at O0C acetic anhydride (1.2 mmol) and triethylamine (1.3 mmol). The reaction mixture is stirred at room temperature overnight, then washed with H2O. After drying with Na2SO4, the solvent is evaporated and the product S94 (98% yield by NMR) is used for the next reaction without further purification. [0435] Synthesis of S95: To a stirred solution of S84 (0.5 mmol) in benzene (20 ml) is added anhydrous AlCl3 (0.6 mmol) drop-wise. The reaction mixture is refluxed for 5 hours and poured on to crushed ice (10 g). After extraction and concentration, the product S95 is purified by silica gel column chromatography in 83% yield.
[0436] Synthesis of S96: To a solution of S86 (0.1 mmol) in methanol (5 ml) is added NaI (10 mg, excess) and Chloramine-T (0.3 mmol). The reaction mixture is stirred for 30 minutes and quenched with Na2S2O3 solution. The solvent is evaporated. The product is purified by silica gel column chromatography as a mixture of mono-iodinated or di-iodinated products in a combined yield of 60%.
[0437] Synthesis of S97: S86 (3 mmol) is added to concentrated H2SO4 (2 ml). To the stirred mixture is added, slowly, concentrated HNO3 (2 ml) drop- wise. After 10 minutes, the reaction mixture is poured on to crushed ice (5 g) and neutralized with Na2CO3 to pH= 7. The Boc-deprotected nitro intermediate is collected by extraction with EtOAc and converted back to S97 by reaction with BoC2O. Purification by silica gel column chromatography provides S97 in 78% yield.
[0438] Synthesis of S98: A mixture of S97 (2 mmol) and 10% Pd/C (0.1 g) in methanol (20ml) is bubbled through with H2 gas for 2 hours. After filtration and concentration, the amine product is used for the next reactions without further purification.
[0439] Synthesis of S99 and SlOO: S98 (1 mmol) is dissolved in aqueous HCl (2 mmol HCl, 10ml H2O). To this solution is added at 00C slowly a solution of sodium nitrite (1 mmol) in water (5 ml). The reaction mixture is stirred at 0°C for 1 hour, then NaN3 (2 mmol) in water (2ml) is added drop-wise at O0C. The resulting mixture is stirred at O0C for 1 hour and at room temperature overnight. The product is extracted with ethyl acetate and washed with saturated sodium bicarbonate and water. The organic layer is dried over anhydrous sodium sulfate and concentrated to give crude product S98. Column purification on silica gel provide the product in 71 % yield. Similarly, S99 is synthesized in 60% yield.
[0440] Synthesis of SlOl, S102, and S103 (also referred to as ARMlOl, ARM102, and
ARM103, respectively) may be accomplished as shown in Scheme 16. The following are examples of the synthesis.
Scheme 16: Synthesis of ARMlOl, ARM102, and ARM103
Figure imgf000155_0001
a) N-Bocl-piperaziπe b) TFA
Figure imgf000155_0002
[0441] Synthesis of SlOl: A solution of S68 (165 mg. lmmol) in CH2Cl2 (50 ml) was cooled to O0C. To this solution, triphosgene (150 mg, 0.5 mmol) and pyridine ( 0.5 ml. excess) were added and stirred at O0C for lhour. Without purification, the resulting S68-phosgene in the reaction mixture was treated with 1-piperonylpiperazine (233 mg, 1.1 mmol) at O0C. After stirring at 00C for 1 hour, the reaction mixture was washed with H2O (2 x 10 ml), IN HCl (2 x 10 ml) and satuarted NaHCO3 (2 x 10 ml), and the solvents were removed under reduced pressure. Purification hy SiO2 column chromatography provided ARMlOl having a yield of 80%. The structure of the product was confirmed by nuclear magnetic resonance (NMR), mass spectroscopy (MS) and/or by elemental analysis.
[0442] Synthesis of S102: S102 was synthesized from S68 using the same method used to synthesize SlOl, with the exception that piperidine was used in place of 1- piperonylpiperazine. The structure of the product was confirmed by nuclear magnetic resonance (NMR), mass spectroscopy (MS) and/or by elemental analysis.
[0443] Synthesis of S103: S103 was synthesized from S68 using the same method used to synthesize SlOl, with the exception that N-Boc 1-piperazine was used in place of 1- piperonylpiperazine, and in a subsequent step the Boc group was deprotected using trifluoroacetic acid (TFA). The structure of the product was confirmed by nuclear magnetic resonance (NMR), mass spectroscopy (MS) and/or by elemental analysis. Scheme 17: Synthesis of ARM104
Figure imgf000156_0001
[0444] Synthesis of S104 (ARM104) maybe accomplished as shown in Scheme 17. The following is an example of the synthesis. A mixture ARM036 (S36) (27 mg, 0.1 mmol), 50% H2O2 (1 ml), and MeOH (3 ml) was stirred at room temperature for 2 days to generate the ARMl 04 product. Mass spectroscopy (MS) was used to monitor the disappearance of ARM036 and the appearance of the product ARM104. The solvents were removed under reduced pressure, and the product was purified by re-crystallization. The final yield was 26 mg of ARM104 at 85% purity. The structure of the final product was deteπnined by nuclear magnetic resonance (NMR) and/or MS.
Scheme 18: Synthesis of ARM105
Figure imgf000156_0002
[0445] Synthesis of S105 (ARM105) maybe accomplished as shown in Scheme 18. The following is an example of the synthesis: To a stirred solution of S68 (80 mg, 0.48 mmol) and pyridine (0.1 ml, excess) in CH2Cl2 (50 ml) at O0C, CH3O-C(O)C(O)Cl (70 mg, 0.58 mmol) was added dropwise. The reaction mixture was stirred at O0C for 2 hours and washed with IN HCl, saturated sodium bicarbonate, and water. Removal of the solvents and purification by SiO2 column chromatography were performed to produce the ARM105 product as a white solid (yield: 95 mg, 94%).
Scheme 19: Synthesis of ARM107
Figure imgf000157_0001
[0446] Synthesis of S107 (ARM107) maybe accomplished as shown in Scheme 19. The following is an example of the synthesis: To S26 (180 mg, 0.92 mmol) in MeOH (20 ml) was added 30% CH2O solution (1.5 ml, excess) and sodium cyanoborohydride (NaBCNH3) (0.4g excess). The reaction mixture was stirred at room temperature, and the pH of the solution was maintained at around pH 4-5 by addition of a few drop of IN HCl. After 3 hours, the solvents were removed under reduced pressure. The residue was dissolved in 20 ml ethyl acetate and washed with H2O and saturated NaHCO3 (2 x 10 ml). The solvents were removed and the ARM107 was purified by SiO2 column chromatography to give a yield: 170 mg, 93%.
Scheme 20: Synthesis of ARMl 08
Figure imgf000157_0002
[0447] Synthesis of S108 (ARM108) maybe accomplished as shown in Scheme 20. The following is an example of the synthesis: A mixture of N-benzyloxycarbonyl-glycine (Cbz- Gly, 129 mg, 0.61 mmol), Diisopropyl-carbodiimide (DIC, 90 mg, 0.71 mmol), N- hydroxysuccinimide (NHS, 70.4 mg, 0.71 nmol) in CH2Cl2 (50 ml) was stirred for 0.5 h at room temperature. To this mixture was added S26 (100 mg, 0.51 mmol) and the mixture was stirred at room temperature for overnight. After washing with INCl (2x1 OmI) and saturated NaHCO3 solution (2x1 OmI), the solvents were removed by evaporation. The product ARM108 was purified by SiO2 column chromatography, to give a yield of 120 mg, 61%.
Scheme 21: Synthesis of ARMl 09
Figure imgf000158_0001
[0448] Synthesis of S 109 (ARM 109) maybe accomplished as shown in Scheme 21. The following is an example of the synthesis: ARM108 (40 mg, 0.1 mmol) in CH2Cl2 (5 ml) was treated with 1 ml of 30% HBrZCH3CO2H. After stirring at room temperature overnight, the reaction mixture was evaporated under reduced pressure. The residue was dissolved in MeOH (3 ml) and treated with propylene oxide (1 ml). The solvents were removed under reduced pressure to provide crude ARM109 which was further purified by dissolving in 0.15 N HCl H2O solution (3.5 ml), followed by washing with ethyl acetate ( 3 ml) and evaporation. The yield of ARM109 was 28.3 mg, 95% (white powder, HCl salt).
Scheme 22: Synthesis of ARMl 10
Figure imgf000158_0002
[0449] Synthesis of SIlO (ARMIlO) maybe accomplished as shown in Scheme 22. The following is an example of the synthesis: A mixture of S26 (100 mg, 0.51 mmol) and methyl 1 -bromoacetate (100 mg, 1 ,2 eq.) and pyridine (50mg) in DMF (what is DMF?) (5 ml) was stirred at room temperature overnight. To this mixture, ethyl aceate (50 ml) was added and washed with saturated NaHCO3 solution (2x1 OmI) and H2O (2xl0ml). The product ARMIlO as an oil was purified by SiO2 column chromatography, to give a yield of 32 mg, 23%.
Scheme 23 : Synthesis of ARMl 11
Figure imgf000158_0003
[0450] Synthesis of Sill (ARMlIl) maybe accomplished as shown in Scheme 23. The following is an example of the synthesis: To a mixture of ARMIlO (16 mg, 0.06 mmol) in MeOH (2 ml) was added IN NaOH (0.1 ml) and the mixture was stirred at room temperature overnight. The solvents were removed under reduced pressure and the residue was dissolved in H2O (10 ml). The aqueous phase was washed with ethyl acetate (2x5 ml) and acidified with IN HCl to pH=4. Removal of the solvents under reduced pressure provided crude ARMlIl. The NaCl was removed using ethanol to yield pure ARMlIl as solid, having a yield of 13mg, 87%.
Scheme 24: Synthesis of Arml 12
Figure imgf000159_0001
[0451] Synthesis of S112 (ARM112) maybe accomplished as shown in Scheme 24. The following is an example of the synthesis: To a mixture of S26 (100 mg, 0.51 mmol) and pyridine (lOOmg) in CH2Cl2 (20 ml), SO2Cl2 (89 mg, 1.2 eq.) was added drop-wise at O0C and stirred at room temperature overnight. The solvents were removed under reduced pressure and the residue was dissolved in 5.5 ml NaOH solution (5 ml H2O +0.5 ml IN NaOH). The water solution was washed with ethyl acetate (2x5 ml), and acidified with IN HCl to pH 4. The aqueous phase was extracted with ethyl acetate (3x5ml) and the ethyl acetate phase was evaporated under reduced pressure to provide ARM 112, as powder, with a yield of 9 mg.
Scheme 25: Synthesis of ARMl 13
Figure imgf000159_0002
[0452] Synthesis of S113 (ARM113) maybe accomplished as shown in Scheme 25. The following is an example of the synthesis: ARM107 (45 mg, 0.21 mmol) in ethyl acetate (2 ml) was treated with CH3I ( 200 mg, excess). The mixture was stirred at room temperature overnight and the product ARM113, as white solid, was collected by filtration to give a yield of 69 mg, 97%.
Scheme 25 A: Synthesis of Arml 14
Figure imgf000160_0001
[0453] Synthesis of S114 (ARM114) maybe accomplished as shown in Scheme 25A. The following is an example of the synthesis. S26 (195 mg, lmmol) in CH2Cl2 (50 ml) was cooled to O0C. To this solution, triphosgene (150 mg, 0.5 mmol) and pyridine ( 0.5 ml. excess) was added and stirred at O0C for 1 hour. Without purification, the resulting S26- phosgene in the reaction mixture was treated withN-Boc 1-piperazine (200 mg, 1.1 mmol) at O0C. After stirring at O0C for 1 hour, the reaction mixture was washed with H2O (2X10 ml), IN HCl (2X10 ml), and saturated NaHCO3 (2X10 ml), and the solvents were removed under reduced pressure. Purification by SiO2 column chromatography provided ARMl 14 with a yield of 80%.
Scheme 26: Synthesis of Arml 15
Figure imgf000160_0002
[0454] Synthesis of S115 (ARM115) may be accomplished as shown in Scheme 26. The following is an example of the synthesis: A mixture of ARMl 14 (200mg, 0.49 mmol) and Lawesson Reagent (400 mg) in toluene (50 ml) was stirred at 9O0C for 5 hours. The mixture was cooled to room temperature and washed with saturated NaHCO3 (2x20ml). The product ARM115_was purified by SiO2 column chromatography to give a yield of 160 mg, 75%.
Scheme 27: Synthesis of Arml 16
Figure imgf000161_0001
[0455] Synthesis of S116 (ARM116) maybe accomplished as shown in Scheme 27. The following is an example of the synthesis: A mixture of ARM115 (lOmg, 0.02 mmol) and trifiuoroacetic acid (TFA, 0.5 ml) in CH2Cl2 (10 ml) was stirred at room temperature for 2 hours. Evaporation of the solvents under reduced pressure produced ARMl 16 with yield of 6 mg, 92%.
Scheme 28: Synthesis of Arml 17
Figure imgf000161_0002
[0456] Synthesis of S117 (ARM117) may be accomplished as shown in Scheme 28. The following is an example of the synthesis: A solution of ARM057 (200mg, 0.71 mmol) in CH2Cl2 (20 ml) was cooled to -780C. To this, IM BBr3 in CH2Cl2 (1 ml) was added, and the mixture was stirred at -780C for 3 hours and then warmed to room temperature overnight. The mixture was washed with IN HCl (2x10 ml) and H2O (lxlOml). After removal of the solvents, the product ARM117 was purified by SiO2 column chromatography to give a yield of δO mg, 33%.
Scheme 29 Synthesis of Arml 18
Figure imgf000162_0001
[0457] Synthesis of S118 (ARM118) may be accomplished as shown in Scheme 29. The following is an example of the synthesis: S26 (3.6 mg, 0.018 mmol) in CH2Cl2 (3 ml) was treated with BODIPY TMR-X, SE (Molecular Probes Inc.) (4 mg, 0.006 mmol) for 3 hours. The mixture was washed with 0.01 N HCl (2x1 ml) and saturated NaHCO3 (2xlml). Removal of the solvents under reduced pressure yielded pure ARM118 (98%).
Scheme 30: Synthesis of ARMl 19
Figure imgf000162_0002
[0458] Synthesis of S119 (ARM119) maybe accomplished as shown in Scheme 30. The following is an example of the synthesis: A mixture ARM107 (50 mg, 0.24 mmol), 50% H2O2 (1 ml), MeOH (3 ml) was stirred at room temperature for 2 days (mass spectrometry was used to monitor the disappearance of ARM107 and the formation of the product). The solvents were removed under reduced pressure to give ARMIlO, having a yield of 26 mg, 45%.
Scheme 31: Synthesis of ARM120 and ARM 121
Figure imgf000163_0001
S28 R=OH1 Arm 120 R=H, Arm121
[0459] Synthesis of S120 (ARM120) and S121 (ARM121) may be accomplished as shown in Scheme 31. The following is an example of the synthesis: A mixture S26 (195 mg, 1 mmol), benzyl bromide (1.1 mmol) and Na2CO3 (10 mmol) in DMF (10 ml) was stirred overnight. Ethyl acetate (30 ml) was added to the reaction, and then the reaction was washed with H2O (4x10 ml). The organic phase was concentrated under reduced pressure and the residue was purified by column chromatography to give S121 as a white powder, with a yield of 280 mg, at 98%. S120 was similarly synthesized, but using 4-OH-benzyl bromide instead of benzyl bromide.
[0460] Synthesis of S122 (ARM122) (LB21300-30). The following is an example of the synthesis: To a cold solution of compound S26 (250 mg, 1.28 mmol, 1 equivalent) in CH2Cl2 (5OmL) at O0C was added DIEA (0.67 mL, 3.8 mmol, 3.0 equivalent), followed by acetoxyacetyl chloride (0.17 mL, 1.58 mmol, 1.24 equivalent). Then, the reaction mixture was stirred at O0C for 20 min, diluted with 1.0 MHCl aqueous solution (100 mL) and extracted by CH2Cl2 (3x50 mL). The combined organic layers were washed (H2O, brine), dried (Na2SO4), filtered, and evaporated to dry. The crude product was purified by chromatography on a silica gel column, eluting with a gradient increasing in polarity from 0 to 50% petroleum in ethyl acetate. Relevant fractions were combined to give the desired compound (350 mg, 93%).
[0461] Synthesis of S123 (ARM123) (LB21300-34). The following is an example of the synthesis: To a solution of compound S122 (287 mg, 0.97 mmol, 1 equivalent) in MeOH (5mL) and THF (8 mL) at 230C was added LiOH (140 mg, 3.33 mmol, 3.44 equivale in H2O 5 mL). The reaction mixture was stirred at 230C for 20 minutes, diluted with 1.0 M HCl aqueous solution (100 mL) and extracted by CH2Cl2 (3x50 mL). The combined organic layers were washed (H2O, brine), dried (Na2SO4), filtered and evaporated to dry. The crude product was purified by chromatography on a silica gel column, eluting with a gradient increasing in polarity from 0 to 70% petroleum in ethyl acetate. Relevant fractions were combined to give S123 (244 mg, 100%).
EXAMPLE 5 - HIGH-THROUGHPUT SCREENING METHOD
[0462] Assays for screening biologically-active small molecules have been developed. These assays are based on rebinding of FKBP 12 protein to RyR.
[0463] A highly-efficient assay for high-throughput screening for small molecules is developed by immobilization of FKBP 12.6 (GST-fusion protein) onto a 96-well plate coated with glutathione. PKA-phosphorylated ryanodine receptor type 2 (RyR2) is loaded onto the FKBP12.6-coated plate, and incubated with JTV-519 analogues at various concentrations (10-100 nM) for 30 min. Thereafter, the plate is washed to remove the unbound RyR2, and then incubated with anti-RyR2 antibody for 30 min. The plate is again washed to remove unbound anti-RyR2 antibody, and then treated with florescent-labeled secondary antibody. The plate is read by an automatic fluorescent plate reader for binding activity.
[0464] In an alternative assay, RyR2 is PKA-phosphorylated in the presence of 32P-ATP. Radioactive PKA-phosphorylated RyR2 is loaded onto an FKBP12.6-coated, 96-well plate, in the presence of JTV-519 analogues at various concentrations (10-100 nM) for 30 min. The plate is washed to remove the unbound radiolabeled RyR2, and then read by an automatic plate reader.
EXAMPLE 6 - EFFECT OF ARM036 COMPOUNDS ON hERG CURRENTS
[0465] The effects of the compounds of the invention on hERG currents were studied using cultured human embryonic kidney 293 (HEK 293) cells which had been stably tranfected with hERG cDNA. HEK 293 cells do not express endogenous hERG. HEK293 cells were transfected with a plasmid containing the hERG cDNA and a neomycin resistance gene. Stable transfectants were selected by culturing the cells in the presence of G418. The selection pressure was maintained by continued culture in the presence of G418. Cells were cultures in Dulbecco's Modified Eagle Medium/Nutreint Mizture F-12 (D-MEM/F-12) supplemented with 10% fetal bovin serum, 199U/ml penicillin G sodium, lOμg/mL streptomycin sulfate and 500 μg/mL G418. Cells for use in electrophysiology were cultured in 35 mm dishes.
[0466] Electrophysiological recordings (using the whole-cell patch clamp method) were performed at room temperature (18°C-24°C). Each cell acted as its own control. The effect of ARM0036 was evaluated at two concentrations: 10 and lOOμM. Each concentration was tested in at least three cells (n > 3). 90 nM Cisapride (commercially available from TOCRIS Bioscience) was used as a positive control for hERG blockade. For recording, cells were transferred to the recording chamber and superfused with vehicle control solution. The patch pipette solution for whole cell recording contained 130 mM potassium aspartate, 5 mM MgC12, 5 mM EGTA, 4 mM ATP and 10 mM HEPES. The pH was adjusted to 7.2 with KOH. The pipette solution was prepared in batches, aliquoted, and stored frozen. A fresh aliquot was thawed and used each day. Patch pipettes were made from glass capillary tubing using a P-97 micropipette puller (Sutter Instruments, Novato, CA). A commercial patch clamp amplifier was used for whole cell recordings. Before digitization, current records were low-pass filtered at one-fifth of the sampling frequency.
[0467] Onset and steady stae block of hERG current was measured using a pulse pattern with fixed amplitudes (conditioning prepulse: +20 mV for 2 seconds; test pulse: _50 mV for 2 seconds) repeated at 10 second intervals, from a holding potential of -80 mV. Peak tail current was measured during the 2 second step to _50 mV. A steady state was maintained for at least 30 seconds before applying the test compound or the positive control. Peak tail current was monitored until a new steady state was achieved. Test compound concentrations were applied cumulatively in ascending order without washout between applications.
[0468] Data acquisition and analysis was performed using the suite of pCLAMP (Vre. 8.2) programs (Axon Instruments, Union City, CA). Steady state was defined by the limiting constant rate of change with time (linear time dependence). The steady state before and after application of the test or control compounds was used to calculate the percentage of current inhibited at each concentration. Concentratioon-response data were fit to an equation of the form:
%Block = {l-l/[Test]/IC50)N]}xl00
[0469] where [Test] is the concentration of the test compound, IC50 (inhibitory concentration 50) is the concentration of the test compound producing half-maximal inhibition, N is the Hill coefficient, and % Block is the percentage of hERG current inhibited at each concentration of the test compound. Nonlinear squares fits were solved with the Solver add-in for Excel 2000 (Microsoft, Redmond, WA). For some compounds it was not possible to determine the IC50 because the highest concentration of the test compound used did not block the hERG channel by 50% or more. EXAMPLE 7 - EFFECT OF VARIOUS COMPOUNDS ON hERG CURRENTS
[0470] Multiple compounds of the invention were tested for their effects on hERG currents. The compounds tested were: ARM036-Na ARM047, ARM048, ARM050, ARM057, ARM064, ARM074, ARM075, ARM076, ARM077, ARMlOl, ARM102, ARM103, ARM104, ARM106, ARM107 and ARM26. By way of comparison, the effect of JTV-519 (referred to in the figures as ARMOXX) on hERG currents was also tested. Electrophysiological recordings were made using the PatchXpress 7000A (Molecular Devices) automatic parallel patch clamp system. Each compound was tested at 0.01, 0.1, 1 and 1OmM, with each concentration tested in 2 cells (n > 2). The duration of exposure to each test concentration was 5 minutes. Other aspects of the experimental protocols were essentially similar to those described in Example 6. For some compounds it was not possible to determine the IC50 because the highest concentration of the test compound used did not block the hERG channel by 50% or more.
[0471] AU publications, references, patents and patent applications cited herein are incorporated by reference in their entirety to the same extent as if each individual application, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.
[0472] While the foregoing invention has been described in some detail for purposes of clarity and understanding, it will be appreciated by one skilled in the art, from a reading of the disclosure, that various changes in form and detail can be made without departing from the true scope of the invention in the appended claims.

Claims

What is claimed is: What is claimed is:
1. A compound of formula:
Figure imgf000167_0001
wherein, n is 0, 1, or 2; q is O, 1, 2, 3, or 4; each R is independently selected from the group consisting of halogen, -OH, -NH2, - NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, -O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted or unsubstituted;
Ri is selected from the group consisting of H, oxo, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted;
R2 is selected from the group consisting of H, -C(=O)R5, -C(=S)R6, -SO2R7, - P(=0)RgR9, -(CH2)m-R)o, alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl may be substituted or unsubstituted; R3 is selected from the group consisting of H , -CO2Y, -C(=O)NHY, acyl, -O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted; and wherein Y is selected from the group consisting of H, alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl, and wherein each alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl maybe substituted or unsubstituted;
R4 is selected from the group consisting of H, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted;
R5 is selected from the group consisting of-NRi5Ri6, -(CH2)qNR15Ri6, -NHNRiSR16, - NHOH, -OR]5,
Figure imgf000168_0001
-CH2X3 acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted, and wherein q is 1, 2, 3, 4, 5, or 6;
Rδ is selected from the group consisting Of-ORj5, -NHNR15R16, -NHOH, -NR15R16, - CH2X, acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R7 is selected from the group consisting Of-OR15, -NRisRi6, -NHNR15R16, -NHOH, - CH2X, alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
Rs and R9 independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; R1O is selected from the group consisting of -NR15R1O, OH, -SO2Rn, -NHSO2R11, CC=O)(R12), NHC=O(R12), -OC=O(R12), and -P(K))R13Ri4;
R11, R12, R13, and R14 independently are selected from the group consisting of H, OH, NH2, -NHNH2, -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
X is selected from the group consisting of halogen, -CN, -CO2Ri5, -C(O)NRi5Ri6, - NR15Ri6, -OR15, -SO2R7, and -P(O)R8R9; and
Ri5 and Ri6 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; and optionally R15 and Riβ together with the N to which they are bonded may form a heterocycle which may be substituted; the nitrogen in the benzothiazepine ring may optionally be a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and prodrugs thereof; provided that when q is O and n is O, then R2 is not H, Et, -C(=O)NH2, (=O)NHPh, - C(=S)NH-nButyl, -C(=O)NHC(=O)CH2C1, -C(=O)H, -C(O)Me, -C(O)Et, - C(O)CH=CH2, -S(O)2Me, or -S(O)2Et; further provided that when q is O and n is 1 or 2, then R2 is not -C(O)Me, -C(O)Et, -S(O)2Me5 Or -S(O)2Et; further provided that when q is 1, and R is Me, Cl, or F at the 6 position of the benzothiazepene ring, then R2 is not H, Me, -C(O)H, -C(O)Me, -C(O)Et, - C(O)Ph, -S(O)2Me, or -S(O)2Et; and further provided that when q is 1, n is 0, and R is OCT3, OH, Ci-C3 alkoxyl at the 7 position of the benzothiazepene ring, then R2 is not H, -C(=O)CH=CH2, or
Figure imgf000170_0001
2. The compound of claim 1 , wherein the compound is selected from the group consisting of Sl, S2, S3, S4, S5, S6, S7, S9, SI l, S12, S13, S14, S19, S20, S22, S23, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S103, S 104, S107, S108, S109, SIlO, Sill, S112, S113, Sl 14, S115, S116, S117, S118, S119, S120, S121, S122, and S123.
3. The compound of claim 1 , wherein the compound is selected from the group consisting of S47, S50, S64, S74, S75, S77, S85, SlOl, S102, S103, S104, and S107.
4. The compound of claim 1, wherein the compound is S47.
5. The compound of claim 1, wherein the compound is S50.
6. The compound of claim 1, wherein the compound is S64.
7. The compound of claim 1, wherein the compound is S74.
8. The compound of claim 1, wherein the compound is S75.
9. The compound of claim 1, wherein the compound is S77.
10. The compound of claim 1 , wherein the compound is S85.
11. The compound of claim 1 , wherein the compound is S 101.
12. The compound of claim 1 , wherein the compound is S 102.
13. The compound of claim 1 , wherein the compound is S 103.
14. The compound of claim 1, wherein the compound is S 104.
15. The compound of claim 1 , wherein the compound is S 107.
6. A pharmaceutical composition comprising a compound of formula:
Figure imgf000171_0001
wherein, n is O, I, or 2; q is 0, 1, 2, 3, or 4; each R is independently selected from the group consisting of halogen, -OH, -NH2, - NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(^O)2CF3, acyl, ■ O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, -O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted or unsubstituted;
Ri is selected from the group consisting of H, oxo, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted;
R2 is selected from the group consisting of H, -C(=O)R5, -C(^S)R6, -SO2R7, - P(=0)RsR9, -(CH2)In-Ri0, alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl may be substituted or unsubstituted;
R3 is selected from the group consisting of H , -CO2Y, -C(=0)NHY, acyl, -O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted; and wherein Y is selected from the group consisting of H, alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl, and wherein each alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl maybe substituted or unsubstituted;
R4 is selected from the group consisting of H, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted;
R5 is selected from the group consisting of -NRi5Ri6, -(CH2)qNRi5R16, -NHNR1SRi6, - NHOH, -OR15, -C(O)NHNR15R16, -CO2R15, -C(K))NRi5Ri6, -CH2X, acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted, and wherein q is 1, 2, 3, 4, 5, or 6;
R6 is selected from the group consisting Of-OR15, -NHNR15Ri6, -NHOH, -NRi5Ri6, - CH2X, acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R7 is selected from the group consisting Of-OR15, -NRi5Ri6, -NHNRi5Ri6, -NHOH3 - CH2X, alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
Rs and R9 independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
Rio is selected from the group consisting of -NRi5RiS, OH, -SO2Rn, -NHSO2Rn, C(K))(Ri2), NHC=O(Ri2), -OC=O(Ri2), and -P(K))Ri3Ri4;
Rn, R12, R13, and Ri4 independently are selected from the group consisting of H, OH, NH2, -NHNH2, -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted;
X is selected from the group consisting of halogen, -CN, -CO2R15, -C(=O)NRi5Ri6, - NR15R16, -OR15, -SO2R7, and -PC=O)R8R9; and
Ri5 and Ri6 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; and optionally Ri5 and Ri6 together with the N to which they are bonded may form a heterocycle which may be substituted; the nitrogen in the benzothiazepine ring may optionally be a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and prodrugs thereof; provided that when q is O and n is O, then R2 is not H, Et, -C(=O)NH2, (-O)NHPh, - C(=S)NH-nButyl, -C(=O)NHC(=O)CH2C1, -C(=O)H, -C(=0)Me, -C(K))Et, - C(=O)CH=CH2, -S(O)2Me, or -S(O)2Et; further provided that when q is O and n is 1 or 2, then R2 is not -C(=0)Me, -C(=O)Et, -S(O)2Me, or -S(O)2Et; further provided that when q is 1, and R is Me, Cl, or F at the 6 position of the benzothiazepene ring, then R2 is not H, Me, -C(O)H, -C(O)Me, -C(O)Et, - C(O)Ph, -S(O)2Me, or -S(O)2Et; and further provided that when q is 1, n is O, and R is OCT3, OH, C1-C3 alkoxyl at the 7 position of the benzothiazepene ring, then R2 is not H, -C(O)CH=CH2, or
Figure imgf000173_0001
17. The pharmaceutical composition of claim 16, wherein the compound is selected from the group consisting of Sl, S2, S3, S4, S5, Sβ, S7, S9, SIl, S12, S13, S14, S19, S20, S22, S23, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, 866, S67, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, SSl, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S103, S 104, S107, S108, S109, SIlO, Sil l, S112, S113, S114, S115, S116, S117, S118, S119, S120, S121, S122, and S123.
18. The pharmaceutical composition of claim 16, further comprising at least one additive selected from the group consisting of analgesic agents, antioxidants, aromatics, buffers, binders, colorants, disintegrants, diluents, emulsifϊers, excipients, extenders, flavor-improving agents, gellants, glidants, preservatives, skin-penetration enhancers, solubilizers, stabilizers, suspending agents, sweeteners, tonicity agents, vehicles and viscosity-increasing agents.
19. The pharmaceutical composition of claim 16, wherein the composition is presented in a form selected from the group consisting of capsules, granules, powders, solutions, suspensions, and tablets.
20. The pharmaceutical composition of claim 16, wherein the composition is administered by a method selected from the group consisting of oral, sublingual, buccal, parenteral, intravenous, transdermal, inhalation, intranasal, vaginal, intramuscular, and rectal modes of administration.
21. The pharmaceutical composition of claim 16, wherein the composition is effective to treat or prevent disorders and diseases associated with the RyR receptors that regulate calcium channel functioning in cells.
22. A method of treating or preventing disorders and diseases in a subject, comprising administering to the subject a therapeutically effective amount of a compound having the formula:
Figure imgf000174_0001
wherein, n is 0, 1, or 2; q is O, 1, 2, 3, or 4; each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(O)2CF3, acyl, -O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, -O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino may be substituted or unsubstituted;
Ri is selected from the group consisting of H, oxo, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted;
R2 is selected from the group consisting of H, -C(=O)R5, -C(=S)Rδ, -SO2R7, - P(=O)R8R9, -(CH2)m-R10, alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl may be substituted or unsubstituted;
R3 is selected from the group consisting of H , -CO2Y, -C(=0)NHY, acyl, -O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl maybe substituted or unsubstituted; and wherein Y is selected from the group consisting of H, alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl, and wherein each alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted;
R4 is selected from the group consisting of H, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl; wherein each alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, and heterocyclyl may be substituted or unsubstituted;
R5 is selected from the group consisting Of-NR15Ri6, -(CH2)qNRisRi6, -NHNR15Ri6, - NHOH, -OR]5, -C(=O)NHNRi5R16, -CO2R15, -C(=O)NRi5Ri6, -CH2X, acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted, and wherein q is I3 2, 3, 4, 5, or 6;
R.6 is selected from the group consisting Of-ORi5, -NHNR15Ri6, -NHOH, -NRi5R1O, - CH2X, acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted;
R7 is selected from the group consisting of — OR15, -NR15R16, -NHNRi5Ri6, -NHOH, - CH2X, alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R8 and R9 independently are selected from the group consisting of OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted;
Rio is selected from the group consisting of -NR15Ri6, OH, -SO2Rn, -NHSO2Rn, CeO)(R12), NHC=O(R12), -OC=O(R12), and
Figure imgf000176_0001
Rn, Ri2, R]3, and R14 independently are selected from the group consisting of H, OH, NH2, -NHNH2, -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
X is selected from the group consisting of halogen, -CN, -CO2Ri5, -C(=O)NRi5Ri6, - NRi5Ri6, -ORi5, -SO2R7, and -Pt=O)R8R9; and
Ri5 and Ri6 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; and optionally R1S and R^ together with the N to which they are bonded may form a heterocycle which may be substituted; the nitrogen in the benzothiazepine ring may optionally be a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and prodrugs thereof.
23. The method of claim 22, wherein the compound is selected from the group consisting of Sl, S2, S3, S4, S5, S6, S7, S9, SIl, S12, S13, S14, S19, S20, S22, S23, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, SlOO, SlOl, S102, S103, S104, S107, S108, S109, SIlO, Si ll, S112, S113, S114, S115, S116, S117, S118, S119, S120, S121, S122, and S123.
24. The method of claim 22, wherein the disorders and diseases are associated with the RyR receptors that regulate calcium channel functioning in cells.
25. The method of claim 24, wherein the disorders and diseases are selected from the group consisting of cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome.
26. The method of claim 25, wherein the cardiac disorder and diseases are selected from the group consisting of irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
27. The method of claim 26, wherein the irregular heartbeat disorders and diseases and exercise-induced irregular heartbeat disorders and diseases are selected from the group consisting of atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPTV); and exercise-induced variants thereof.
28. The method of claim 25, wherein the skeletal muscular disorder and diseases are selected from the group consisting of skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence.
29. The method of claim 25, wherein the cognitive disorders and diseases are selected from the group consisting of Alzheimer's Disease, forms of memory loss, and age- dependent memory loss.
30. The method of claim 22, wherein the subject is an in vitro or in vivo system.
31. The method of claim 22, wherein the subject is a human.
32. A high-throughput screening method to detect compounds that are active towards the RyR receptor, comprising: immobilizing FKBP protein on a structure; loading PKA-phosphorylated ryanodine receptor (RyR) onto the structure; incubating the structure with the RyR and FKBP compounds; washing the structure to substantially remove any unbound RyR; incubating the structure with an anti-RyR antibody; washing the structure to substantially remove any unbound anti-RyR antibody ; treating the structure with fluorescent-labeled secondary antibody; and reading the structure with a fluorescent reader for binding activity.
33. The method of claim 32, wherein the structure is a multi-well plate.
34. A method of diagnosis of a disease or disorder in a subject, said method comprising: obtaining a cell or tissue sample from the subject; obtaining DNA from the cell or tissue; comparing the DNA from the cell or tissue with a control nucleic acid encoding RyR to determine whether a mutation is present in the DNA from the cell or tissue, the presence of a mutation indicating a disease or disorder.
35. The method of claim 34, wherein the subject is an adult, a child or a fetus.
36. The method of claim 34, wherein the mutation is present in a SIDS subject.
37. The method of claim 34, wherein the mutation is a mutation which is present in the DNA encoding RyRl .
38. The method of claim 34, wherein the mutation is a mutation which is present in the DNA encoding RyR2.
39. The method of claim 38, wherein the RyR2 mutation is on chromosome 1 q42-q43.
40. The method of claim 38, wherein the mutation is a CPTV mutation.
41. The method of claim 34, wherein the disorders and diseases are associated with the RyR receptors that regulate calcium channel functioning in cells.
42. The method of claim 41 , wherein the disorders and diseases are selected from the group consisting of cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome.
43. The method of claim 42, wherein the cardiac disorder and diseases are selected from the group consisting of irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
44. The method of claim 43, wherein the irregular heartbeat disorders and diseases and exercise-induced irregular heartbeat disorders and diseases are selected from the group consisting of atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPTV); and exercise-induced variants thereof.
45. The method of claim 42, wherein the skeletal muscular disorder and diseases are selected from the group consisting of skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence.
46. The method of claim 42, wherein the cognitive disorders and diseases are selected from the group consisting of Alzheimer's Disease, forms of memory loss, and age- dependent memory loss.
47. A method of diagnosis of a disorder or disease in a subject, said method comprising: obtaining cells or tissue sample from the subject; incubating the cells or tissue sample with the compound of Formula 1 under conditions which increase phosphorylation of RyR in the cells; determining
(a) whether RyR bound to calstabin is increased in the cells or tissue compared to RyR bound to calstabin in control cells or tissues, said control cells or tissues lacking mutant RyR calcium channels, or
(b) whether a decrease in release of calcium occurs in RyR channels compared to a lack of decrease in release of calcium in the control cells; an increase in RyR-bound calstabin in (a) indicating a disorder or disease in the subject or a decrease in release of calcium in RyR channels in (b) compared to the control cells indicating a disease or disorder in the subject.
48. The method of claim 47, wherein the subject is an adult, a child or a fetus.
49. The method of claim 47, wherein the disorders and diseases are associated with the RyR receptors that regulate calcium channel functioning in cells.
50. The method of claim 49, wherein the disorders and diseases are selected from the group consisting of cardiac disorders and diseases, skeletal muscular disorders and diseases, cognitive disorders and diseases, malignant hyperthermia, diabetes, and sudden infant death syndrome.
51. The method of claim 50, wherein the cardiac disorder and diseases are selected from the group consisting of irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
52. The method of claim 51 , wherein the irregular heartbeat disorders and diseases and exercise-induced irregular heartbeat disorders and diseases are selected from the group consisting of atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPTV); and exercise-induced variants thereof.
53. The method of claim 50, wherein the skeletal muscular disorder and diseases are selected from the group consisting of skeletal muscle fatigue, exercise-induced skeletal muscle fatigue, muscular dystrophy, bladder disorders, and incontinence.
54. The method of claim 50, wherein the cognitive disorders and diseases are selected from the group consisting of Alzheimer's Disease, forms of memory loss, and age- dependent memory loss.
55. A method of diagnosis of a cardiac disorder or disease in a subject, said method comprising: obtaining cardiac cells or tissue sample from the subject; incubating the cardiac cells or tissue sample with the compound of claim 1 under conditions which increase phosphorylation of RyR2 in cells; determining
(a) whether RyR2 bound to calstabin is increased in the cells or tissue compared to RyR2 bound to calstabin in control cells or tissues said control cells or tissues lacking mutant RyR2 calcium channels, or
(b) whether a decrease in release of calcium occurs in RyR2 channels compared to a lack of decrease in release of calcium in the control cells; an increase in RyR2-bound calstabin in (a) indicating a disorder or disease in the subject or a decrease in release of calcium in RyR2 channels in (b) compared to the control cells indicating a cardiac disease or disorder in the subject .
56. The method of claim 55, wherein the subject is an adult, a child or a fetus.
57. The method of claim 55, wherein the cardiac disorder and diseases are selected from the group consisting of irregular heartbeat disorders and diseases; exercise-induced irregular heartbeat disorders and diseases; sudden cardiac death; exercise-induced sudden cardiac death; congestive heart failure; chronic obstructive pulmonary disease; and high blood pressure.
58. The method of claim 55, wherein the irregular heartbeat disorders and diseases and exercise-induced irregular heartbeat disorders and diseases are selected from the group consisting of atrial and ventricular arrhythmia; atrial and ventricular fibrillation; atrial and ventricular tachyarrhythmia; atrial and ventricular tachycardia; catecholaminergic polymorphic ventricular tachycardia (CPTV); and exercise-induced variants thereof.
59. A compound of formula I-a:
Figure imgf000182_0001
wherein: n is 0, 1, or 2; q is O, 1, 2, 3, or 4; each R is independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, -S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; wherein each acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero- )arylamino may be substituted or unsubstituted;
R2 is selected from the group consisting of H, -C=O(Rs), -C=S(R6), -SO2R7, - P(=0)RsR9, -(CH2)m-Rio, alkyl, aryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl; wherein each alkyl, aryl, heteroaryl, cycloalkyl, cycloalkylalkyl, and heterocyclyl maybe substituted or unsubstituted;
R5 is selected from the group consisting Of-NRi5R16, -NHNR1SRi6, -NHOH, -OR15, - C(=O)NHNR15Ri6, -CO2R15,
Figure imgf000182_0002
-CH2X, acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R6 is selected from the group consisting Of-ORi5, -NHNRi5Ri6, -NHOH, -NRi5Ri6, - CH2X, acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted;
R7 is selected from the group consisting of H, -OR15, -NRiSR16, -NHNRi5Ri6, - NHOH, -CH2X, alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, akenyl, alkynyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
R8 and R9 independently are selected from the group consisting of -OH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted;
Rio is selected from the group consisting of -NR15R16, OH, -SO2Rn, -NHSO2R11, - C(=O)R12, NH(C=O)R12, -0(C=O)R12, and -P(O)Ri3R14; m is 0, 1, 2, 3, or 4;
Ru, Ri2, R13, and R14 independently are selected from the group consisting of H, OH, NH2, -NHNH2, -NHOH, acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
X is selected from the group consisting of halogen, -CN, -CO2Ri5, -C(=0)NRisRi6, - NRi5Ri6, -ORi5, -SO2R7, and -P(=O)R8R9; and
Ri5 and Ri6 independently are selected from the group consisting of H, acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; and optionally Ri5 and Rig together with the N to which they are bonded may form a heterocycle which may be substituted or unsubstituted; the nitrogen in the benzothiazepine ring may be optionally a quaternary nitrogen; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes, and prodrugs thereof; provided that when q is 0 and n is 0, then R2 is not H, Et, -CC=O)NH2, C=O)NHPh, - C(=S)NH-nButyl, -C(=O)NHC(=O)CH2C1, -C(O)H, -C(O)Me, -C(O)Et, - C(=O)CH=CH2, -S(=O)2Me, or -S(=O)2Et; further provided that when q is O and n is 1 or 2, then R2 is not -C(O)Me, -C(O)Et, -SC=O)2Me, or -S(O)2Et; further provided that when q is 1, and R is Me, Cl, or F at the 6 position of the benzothiazepene ring, then R2 is not H, Me, -C(O)H, -C(O)Me, -C(O)Et, - C(^O)Ph, -SC=O)2Me, or -S(O)2Et; and further provided that when q is 1, n is O, and R is OCT3, OH, C1-C3 alkoxyl at the 7 position of the benzothiazepene ring, then R2 is not H, -C(=O)CH=CH2, or
Figure imgf000184_0001
60. The compound of claim 59, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(O)2Ci- C4alkyl, -SC=O)C1 -C4alkyl, -S-Ci-C4alkyl, -OSC=O)2CF3, Ph, -NHCH2Ph, -C(O)Me, - OC(=O)Me, morpholinyl and propenyl; and n is O.
61. The compound of claim 60, wherein R2 is selected from the group consisting of - C=O(R5), -C=S(R6), -SO2R7, -PC=O)R8R9, and -(CH2V-Ri0.
62. The compound of claim 59, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, - S(O)2Ci-C4alkyl, -S(=O)Ci-C4alkyl, -S-Q-Qalkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, - C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is 1 or 2.
63. The compound of claim 62, wherein R2 is selected from the group consisting of — C=O(R5), -C=S(R6), -SO2R7, -PC=O)R8R9, and -(CH2V-Ri0.
64. The compound of claim 59 having the formula of I-b:
Figure imgf000185_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, - S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R2 and n are as defined in claim 59; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
65. The compound of claim 64, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -St=O)2C1- Qalkyl, -S(=O)C,-C4alkyl, -S-C1-C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, - OC(=O)Me, morpholinyl and propenyl; and n is O.
66. The compound of claim 65, wherein R2 is selected from the group consisting of- C=O(R5), -C=S(R6), -SO2R7, -P(=O)R8R9, and -(CH2)m-R10.
67. The compound of claim 66, wherein R is OMe, and R" is H.
68. The compound of claim 66, wherein R' is H, and R" is H.
69. The compound of claim 64, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci- C4alkyl, -S(=O)Ci-C4alkyl, -S-d-C4alkyl, -OS(O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, - OC(=O)Me, morpholinyl and propenyl; and n is 1 or 2.
70. The compound of claim 66, wherein R2 is selected from the group consisting of- C=O(R5), -C=S(R6), -SO2R7, -P(=O)R8R9, and -(CH2)m-Ri0.
71. The compound of claim 67, wherein R' is OMe, and R" is H.
72. The compound of claim 67, wherein R' is H, and R" is H.
73. The compound of claim 59 having the formula of I-e:
Figure imgf000186_0001
wherein each R, R5, q and n is as defined in claim 59; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and prodrugs thereof.
74. The compound of claim 73, wherein each R is independently selected from the group consisting of H, halogen, -OH3 OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SC=O)2Ci- C4alkyl, -S(=O)C1-C4alkyl, -S-C1-C4alkyl, -OSO=O)2CF3, Ph, -NHCH2Ph, -CC=O)Me, - OC(=O)Me, morpholinyl and propenyl; and n is O.
75. The compound of claim 74, wherein R5 is selected from the group consisting of — NRi5R16, -NHOH, -OR15, -CH2X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted.
76. The compound of claim 73, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SC=O)2C1- Qalkyl, -S(=O)C!-C4alkyl, -S-CrC4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, - 0C(=0)Me, morpholinyl and propenyl; and n is 1 or 2.
77. The compound of claim 76, wherein R5 is selected from the group consisting of- NRi5R16, -NHOH, -OR15, -CH2X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted.
78. The compound of claim 77, wherein R5 is an alkyl substituted by at least one labeling group.
79. The compound of claim 78, wherein the labeling group is selected from the group consisting of a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group.
80. The compound of claim 78, wherein the labeling group is a fluorescent labeling group selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4',6-Diamidino-2- phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.
81. The compound of claim 78, wherein the labeling group is Bodipy.
82. The compound of claim 59 having the formula of I-f:
Figure imgf000187_0001
wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, - S(=O)alkyl, -OS(=O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio may be substituted or unsubstituted;
R5 and n are as defined in claim 59; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
83. The compound of claim 82, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Cr C4alkyl, -S(=O)d-C4alkyl, -S-C1-C4alkyl, -OSC=O)2CF3, Ph, -NHCH2Ph, -C(=0)Me, - OC(=O)Me, morpholinyl and propenyl; and n is O.
84. The compound of claim 82, wherein R5 is selected from the group consisting of- NR15R16, -NHOH, -OR15, -CH2X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted or unsubstituted.
85. The compound of claim 84, wherein R' is OMe, and R is H.
86. The compound of claim 84, wherein R' is H, and R is H.
87. The compound of claim 82, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SC=O)2C1- Qalkyl, -S(=O)C1-C4alkyl, -S-C1-C4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph3 -C(=0)Me, - 0C(=0)Me, morpholinyl and propenyl; and n is 1 or 2.
88. The compound of claim 87, wherein R5 is selected from the group consisting of- NR15R16, -NHOH, -OR15, -CH2X, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each acyl, alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted.
89. The compound of claim 88, wherein R' is OMe, and R" is H.
90. The compound of claim 88, wherein R' is H, and R" is H.
91. The compound of claim 59 having the formula of I-g:
Figure imgf000188_0001
wherein W is S or O; each R, Ri5, R16, q, and n is as defined in claim 59; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
92. The compound of claim 91, wherein each R is independently selected from the group consisting of H3 halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SC=O)2C1- Qalkyl, -S(=O)d-C4alkyl, -S-Q-Qalkyl, -OS(=O)2CF3? Ph, -NHCH2Ph, -C(=0)Me, - OC(=O)Me, morpholinyl and propenyl; and n is O.
93. The compound of claim 92, wherein W is S.
94. The compound of claim 92, wherein Ri5 and Ri6 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted; and optionally R15 and R16 together with the N to which they are bonded may form a heterocycle which may be substituted.
95. The compound of claim 92, wherein W is O.
96. The compound of claim 95, wherein Ri5 and Ri6 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted; and optionally Ri5 and Ri6 together with the N to which they are bonded may form a heterocycle which may be substituted.
97. The compound of claim 91, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -S(=O)2Ci- C4alkyl, -S(=O)Ci-C4alkyl, -S-Ci-C4alkyl, -OS(O)2CF3, Ph, -NHCH2Ph, -C(=O)Me, - OC(=O)Me, morpholinyl and propenyl; and n is 1 or 2.
98. The compound of claim 97, wherein W is S.
99. The compound of claim 98, wherein Ri5 and Ri6 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted; and optionally R.15 and R1S together with the N to which they are bonded may form a heterocycle which may be substituted.
100. The compound of claim 97, wherein W is O.
101. The compound of claim 100, wherein R15 and R16 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted; and optionally Ri5 and R^ together with the N to which they are bonded may form a heterocycle which may be substituted.
102. The compound of claim 59 having the formula of I-h:
Figure imgf000190_0001
wherein W is S or O; wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -SO3H, -S(=O)2alkyl, - S(=O)alkyl, -OS(^O)2CF3, acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio, and (hetero-)arylamino; and wherein each acyl, alkyl, alkoxyl, alkylamino, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, (hetero-)aryl, (hetero-)arylthio maybe substituted or unsubstituted;
Ris, Ri6 and n are as defined in claim 59; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
103. The compound of claim 102, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, - SC=O)2C1 -C4alkyl, -8(=O)C]-C4alkyl, -S-CrC4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, - C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is 0.
104. The compound of claim 103, wherein W is S.
105. The compound of claim 103, wherein R15 and R16 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted; and optionally Ris and R16 together with the N to which they are bonded may form a heterocycle which may be substituted.
106. The compound of claim 105, wherein R' is OMe, and R is H.
107. The compound of claim 105, wherein R' is H, and R is H.
108. The compound of claim 103 , wherein W is O.
109. The compound of claim 108, wherein R15 and Ri6 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterpcyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted; and optionally Ri5 and Ri6 together with the N to which they are bonded may form a heterocycle which may be substituted.
110. The compound of claim 109, wherein R' is OMe, and R is H.
111. The compound of claim 109, wherein R' is H, and R is H.
112. The compound of claim 102, wherein R' and R" are independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, - S(=O)2CrC4alkyl, -S(=O)Ci-C4alkyl, -S-CrC4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, - C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is 1 or 2.
113. The compound of claim 112, wherein W is S .
114. The compound of claim 113, wherein R15 and R16 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted; and optionally R15 and Ri6 together with the N to which they are bonded may form a heterocycle which may be substituted.
115. The compound of claim 114, wherein R' is OMe, and R is H.
116. The compound of claim 114, wherein R' is H, and R is H.
117. The compound of claim 112, wherein W is O.
118. The compound of claim 117, wherein R15 and R16 independently are selected from the group consisting of H, OH, NH2, alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl maybe substituted; and optionally Ri5 and R16 together with the N to which they are bonded may form a heterocycle which may be substituted.
119. The compound of claim 117, wherein R' is OMe, and R is H.
120. The compound of claim 117, wherein R' is OMe, and R" is H.
121. The compound of claim 59 having the formula of I-i:
Figure imgf000192_0001
wherein Rj7 is selected from the group consisting Of-NRi5R16, -NHNRigRi6, -NHOH, -ORi5, -CH2X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted; each R, q, and n is as defined in claim 59; and enantiomers, diastereomers, tautomers, pharmaceutically acceptable salts, hydrates, solvates, complexes and pro-drugs thereof.
122. The compound of claim 121, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, - S(=O)2C1-C4alkyl, -S(=O)C1-C4alkyl, -S-CrC4alkyl, -OS(=O)2CF3, Ph, -NHCH2Ph, - C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is O.
123. The compound of claim 122, wherein R17 is
Figure imgf000193_0001
and -OR15.
124. The compound of claim 122, wherein R17 is -OH, -OMe, -NEt, -NHEt, -NHPh, - NH2, or -NHCH2pyridyl.
125. The compound of claim 122, wherein each R is independently selected from the group consisting of H, halogen, -OH, OMe, -NH2, -NO2, -CN, -CF3, -OCF3, -N3, -
Figure imgf000193_0002
-S(=O)C1-C4alkyl, -S-CrC4alkyl, -OS(O)2CF3, Ph, -NHCH2Ph, - C(=O)Me, -OC(=O)Me, morpholinyl and propenyl; and n is 1 or 2.
126. The compound of claim 125, wherein R17 is -NR^R16, and -OR1S.
127. The compound of claim 125, wherein R17 is -OH, -OMe, -NEt, -NHEt, -NHPh, - NH2, -NHCH2pyridyl.
128. A method for the synthesis of compounds of the formula Al :
Figure imgf000193_0003
wherein R' is OMe or H; n is O, 1 , or 2; and NR15Ri6 is selected from the group
Figure imgf000193_0004
consisting of NH2, NEt2, NHCH2Ph, NHOH,
Figure imgf000193_0005
comprising: (i) reacting a compound of formula A2:
Figure imgf000194_0001
with triphosgene to form a compound of formula A3:
Figure imgf000194_0002
(ii) reacting the compound of formula A3 with an amine of formula HNR^Riό, wherein NR^R^ is defined as above, to give the compound of formula Al .
129. A method for the synthesis of compounds of formula Al:
Figure imgf000194_0003
wherein R' is OMe or H; n is 0, 1, or 2; and NR15R16 is selected from the group
Figure imgf000195_0001
consisting of NH2, NEt2, NHCH2Ph, NHOH,
Figure imgf000195_0002
compπsmg:
(i) reacting an amine of formula HNRi5R16 with triphosgene to form a compound of formula A4:
Cl3CO(C=O)NR15Ri6 (A4); and
(ii) reacting the compound of formula A4 with a compound of formula A2:
Figure imgf000195_0003
wherein R' and n are as defined above, to give the compound of formula Al.
130. A method for the synthesis of compounds of formula A5 :
Figure imgf000195_0004
wherein R' is OMe or H; n is 0, 1, or 2; and Raa is C1-C4 alkyl or aryl; comprising: reacting a compound of formula A2:
Figure imgf000196_0001
wherein R' and n are as defined above, with an acid chloride of formula ClC(=O)C(=O)ORaa, wherein Raa is defined as above, in the presence of a base to give the compound of formula A5.
131. The method of claim 130, wherein Raa is Me or Et.
132. A method for the synthesis of compounds of formula A6 :
Figure imgf000196_0002
wherein R' is OMe or H; n is 0, 1, or 2; comprising: reacting a compound of formula A5:
Figure imgf000196_0003
wherein R' and n are defined as above, and Raa is C1-C4 alkyl or aryl, with an acid or a base.
133. The method of claim 132, wherein the acid is trifluoroacetic acid (TFA).
134. The method of claim 132, wherein the base is selected from the group consisting of NaOH, KOH, and LiOH.
135. The method of claim 132, wherein Raa is Me or Et.
PCT/US2006/032405 2004-01-22 2006-08-17 Agents for preventing and treating disorders involving modulation of the ryr receptors WO2007024717A2 (en)

Priority Applications (28)

Application Number Priority Date Filing Date Title
NZ566822A NZ566822A (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the RyR receptor
SI200631612T SI1928850T1 (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the ryr receptors
BRPI0615097-7A BRPI0615097B1 (en) 2005-08-25 2006-08-17 COMPOUND, PHARMACEUTICAL COMPOSITION AND USE OF THE SAME
PL06801887T PL1928850T3 (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the ryr receptors
KR1020127003439A KR101456342B1 (en) 2005-08-25 2006-08-17 AGENTS FOR PREVENTING AND TREATING DISORDERS INVOLVING MODULATION OF RyR RECEPTORS
ES06801887T ES2421159T3 (en) 2005-08-25 2006-08-17 Agents to avoid and treat disorders that involve modulation of RyR receptors
CA2620183A CA2620183C (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the ryr receptors
RS20130294A RS52852B (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the ryr receptors
CN2006800395739A CN101500576B (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the RYR receptors
EA200800665A EA014941B1 (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the ryr receptors
JP2008528020A JP5342877B2 (en) 2005-08-25 2006-08-17 Drug for preventing and treating disorders involved in the regulation of RYR receptor
EP06801887.8A EP1928850B1 (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the ryr receptors
DK06801887.8T DK1928850T3 (en) 2005-08-25 2006-08-17 Agents for the prevention and treatment of disorders involving modulation of the RyR receptors
AU2006283534A AU2006283534C1 (en) 2005-08-25 2006-08-17 Agents for preventing and treating disorders involving modulation of the RyR receptors
PCT/US2007/009715 WO2007127145A2 (en) 2006-04-25 2007-04-23 Methods and compositions for treatment of cardiac arrhythmia in non-human animals
TW096119821A TW200815381A (en) 2006-06-02 2007-06-01 Methods for treating or reducing muscle fatigue
PCT/US2007/012936 WO2008060332A2 (en) 2006-06-02 2007-06-01 Methods for treating or reducing muscle fatigue
ARP070102392A AR061174A1 (en) 2006-06-02 2007-06-01 METHOD FOR DIAGNOSING CONDITIONS AND DISEASES ASSOCIATED WITH RIANODINE RECEPTORS AND METHODS FOR IDENTIFYING COMPOUNDS THAT AFFECT RIANODINE RECEPTORS
PCT/US2007/012969 WO2007143112A2 (en) 2006-06-02 2007-06-01 Methods for diagnosing disorders and diseases associated with ryanodine receptors and methods for identifying compounds that affect ryanodine receptors
ARP070102391A AR061173A1 (en) 2006-06-02 2007-06-01 METHODS TO TREAT OR REDUCE MUSCLE FATIGUE
PCT/US2007/018138 WO2008021432A2 (en) 2006-08-17 2007-08-15 Compositions and methods for treatment of conditions affecting the nervous system
PCT/US2007/018147 WO2008021439A2 (en) 2006-08-17 2007-08-15 Compositions and methods for treatment of cardiac hypertrophy
IL189675A IL189675A (en) 2005-08-25 2008-02-21 Compounds, salts thereof, pharmaceutical compositions comprising the same, uses thereof in the preparation of medicaments for treating or preventing disorders associated with an ryr that regulates calcium channel functioning in cells and methods of synthesizing the same
TNP2008000078A TNSN08078A1 (en) 2005-08-25 2008-02-21 Agents for preventing and treating disorders involving modulation of the ryr receptors
NO20081421A NO20081421L (en) 2005-08-25 2008-03-19 Means for the prevention and treatment of disorders involving the modulation of RYR receptors
HK08113342.0A HK1119705A1 (en) 2005-08-25 2008-12-08 Agents for preventing and treating disorders involving modulation of the ryr receptors
US12/938,098 US8710045B2 (en) 2004-01-22 2010-11-02 Agents for preventing and treating disorders involving modulation of the ryanodine receptors
HRP20130669TT HRP20130669T1 (en) 2005-08-25 2013-07-15 Agents for preventing and treating disorders involving modulation of the ryr receptors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11/212,413 2005-08-25
US11/212,413 US7704990B2 (en) 2005-08-25 2005-08-25 Agents for preventing and treating disorders involving modulation of the RyR receptors

Publications (2)

Publication Number Publication Date
WO2007024717A2 true WO2007024717A2 (en) 2007-03-01
WO2007024717A3 WO2007024717A3 (en) 2009-01-08

Family

ID=37772221

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/032405 WO2007024717A2 (en) 2004-01-22 2006-08-17 Agents for preventing and treating disorders involving modulation of the ryr receptors

Country Status (35)

Country Link
US (1) US7704990B2 (en)
EP (4) EP2764867A1 (en)
JP (1) JP5342877B2 (en)
KR (2) KR20080037741A (en)
CN (2) CN102558093B (en)
AP (1) AP3091A (en)
AR (1) AR057776A1 (en)
AU (1) AU2006283534C1 (en)
BR (1) BRPI0615097B1 (en)
CA (1) CA2620183C (en)
CR (1) CR9812A (en)
CY (3) CY1113974T1 (en)
DK (3) DK1928850T3 (en)
EA (1) EA014941B1 (en)
EC (1) ECSP088306A (en)
ES (3) ES2421159T3 (en)
GE (1) GEP20105134B (en)
HK (3) HK1119705A1 (en)
HN (1) HN2008000299A (en)
HR (3) HRP20130353T1 (en)
IL (1) IL189675A (en)
MA (1) MA29785B1 (en)
MY (1) MY144622A (en)
NO (1) NO20081421L (en)
NZ (1) NZ566822A (en)
PL (3) PL2177224T3 (en)
PT (3) PT2177224E (en)
RS (3) RS52780B (en)
SI (3) SI2311464T1 (en)
SV (1) SV2008002828A (en)
TN (1) TNSN08078A1 (en)
TW (1) TWI486338B (en)
UA (1) UA93388C2 (en)
WO (1) WO2007024717A2 (en)
ZA (1) ZA200802338B (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008144483A2 (en) * 2007-05-18 2008-11-27 Armgo Pharma, Inc. Agents for treating disorders involving modulation of ryanodine receptors
EP2127654A1 (en) * 2004-03-25 2009-12-02 The Trustees of Columbia University in the City of New York Benzothiazepine derivatives as ryanodine receptor (RYR2) inhibitors and their use in the treatment of cardiac diseases
EP2402333A1 (en) * 2009-02-25 2012-01-04 Noboru Kaneko 1,4-benzothiazepine-1-oxide derivative and pharmaceutical composition utilizing the same
WO2012019076A1 (en) 2010-08-06 2012-02-09 The Trustees Of Columbia University In The City Of New York Compositions and methods for preventing and treating cardiac ischemia/reperfusion injury
EP2418950A1 (en) * 2009-04-15 2012-02-22 State Of Oregon By&Through The State Board Of Higher Education On Behalf Of Portland State Unv. Compounds and methods for modulating activity of calcium release channels
EP2653466A1 (en) 2012-04-18 2013-10-23 Les Laboratoires Servier Agents for treating disorders involving modulation of ryanodine receptors
US8710045B2 (en) 2004-01-22 2014-04-29 The Trustees Of Columbia University In The City Of New York Agents for preventing and treating disorders involving modulation of the ryanodine receptors
WO2021015221A1 (en) 2019-07-22 2021-01-28 株式会社アエタスファルマ Method for producing optically active 1, 4-benzothiazepine-1-oxide derivative
US11504383B2 (en) 2021-01-08 2022-11-22 Armgo Pharma, Inc. Crystalline forms of a Ryanodine receptor modulator and uses thereof
US11717526B2 (en) 2021-05-20 2023-08-08 Armgo Pharma, Inc. Pharmaceutical compositions comprising a ryanodine receptor modulator and uses thereof

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8022058B2 (en) * 2000-05-10 2011-09-20 The Trustees Of Columbia University In The City Of New York Agents for preventing and treating disorders involving modulation of the RyR receptors
US7393652B2 (en) * 2000-05-10 2008-07-01 The Trustees Of Columbia University In The City Of New York Methods for identifying a chemical compound that directly enhances binding of FKBP12.6 to PKA-phosphorylated type 2 ryanodine receptor (RyR2)
US20040229781A1 (en) * 2000-05-10 2004-11-18 Marks Andrew Robert Compounds and methods for treating and preventing exercise-induced cardiac arrhythmias
US7879840B2 (en) * 2005-08-25 2011-02-01 The Trustees Of Columbia University In The City Of New York Agents for preventing and treating disorders involving modulation of the RyR receptors
US20060293266A1 (en) * 2000-05-10 2006-12-28 The Trustees Of Columbia Phosphodiesterase 4D in the ryanodine receptor complex protects against heart failure
US6489125B1 (en) * 2000-05-10 2002-12-03 The Trustees Of Columbia University In The City Of New York Methods for identifying chemical compounds that inhibit dissociation of FKBP12.6 binding protein from type 2 ryanodine receptor
US20040048780A1 (en) * 2000-05-10 2004-03-11 The Trustees Of Columbia University In The City Of New York Method for treating and preventing cardiac arrhythmia
US7544678B2 (en) * 2002-11-05 2009-06-09 The Trustees Of Columbia University In The City Of New York Anti-arrythmic and heart failure drugs that target the leak in the ryanodine receptor (RyR2)
AU2004220548A1 (en) * 2003-03-07 2004-09-23 The Trustees Of Columbia University, In The City Of New York Type 1 ryanodine receptor-based methods
US7704990B2 (en) 2005-08-25 2010-04-27 The Trustees Of Columbia University In The City Of New York Agents for preventing and treating disorders involving modulation of the RyR receptors
WO2009111463A1 (en) * 2008-03-03 2009-09-11 Armgo Pharma, Inc. Process for preparing benzothiazepines from gamma-aminoalkylbenzenes
AU2008243144A1 (en) * 2008-11-06 2010-05-20 Quark Technologies Australia Pty Ltd Improvements in Radiopharmaceutical Purification
CN101812523B (en) * 2010-04-09 2012-08-22 广州益善生物技术有限公司 SNP (Single Nucleotide 0olymorphism) detection specific primer, liquid-phase chip and detection method of RYR1 (Ryanodine Receptors 1) gene
US9464322B2 (en) 2011-09-09 2016-10-11 University Of Kentucky Research Foundation Methods for diagnosing and treating alzheimer's disease (AD) using the molecules that stabilize intracellular calcium (Ca2+) release
US9572528B1 (en) 2012-08-06 2017-02-21 Los Angeles Biomedical Research Insitute at Harbor-UCLA Medical Center Monitor for SIDS research and prevention
US10214574B2 (en) 2015-04-15 2019-02-26 Ohio State Innovation Foundation Engineered calmodulin for treatment of ryanopathies
ES2643856B1 (en) 2016-05-24 2018-08-03 Universidad Del Pais Vasco / Euskal Herriko Unibertsitatea Triazoles for the regulation of intracellular calcium homeostasis
US11758928B2 (en) 2016-12-15 2023-09-19 Société des Produits Nestlé S.A. Compositions and methods that modulate phosphorus or enzymes in a companion animal
AU2018297171A1 (en) * 2017-07-06 2020-01-30 Children's Medical Center Corporation Compositions and methods for treating or preventing catecholaminergic polymorphic ventricular tachycardia
CN108588085B (en) * 2018-05-30 2021-11-09 上海市第十人民医院 Malignant arrhythmia screening kit, animal model establishing method and application
US11129871B1 (en) 2018-06-19 2021-09-28 Washington University Compositions and methods for treating and preventing endoplasmic reticulum (ER) stress-mediated kidney diseases

Family Cites Families (204)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1060786A (en) * 1963-10-09 1967-03-08 Wander Ag Dr A Process for the preparation of lactams
US3519647A (en) 1966-10-12 1970-07-07 Squibb & Sons Inc 2,3,4,5-tetrahydro-1,5-benzothiazepines
DE3561317D1 (en) * 1984-01-27 1988-02-11 Ajinomoto Kk Manufacture of heptanoic acid derivatives
EP0238700B1 (en) 1986-03-27 1991-01-23 Topy Industries, Limited Carbon monoxide oxidizing catalyst
US4567254A (en) * 1984-09-19 1986-01-28 Kikkoman Corporation Method for preparing N6,8-disubstituted 3',5'-cyclic adenosine monophosphate and salt thereof
JPS6260A (en) 1985-02-26 1987-01-06 Ajinomoto Co Inc Production of cyclopentenylheptanoic acid
US4841055A (en) * 1985-03-25 1989-06-20 Japan Tobacco Inc. Desmosine derivatives and reagent for preparing artificial antigens
US4723012A (en) * 1985-03-25 1988-02-02 Japan Tobacco Inc. Desmosine derivatives having a disulfide bond and preparation of artificial antigen using the same
US6956032B1 (en) * 1986-04-18 2005-10-18 Carnegie Mellon University Cyanine dyes as labeling reagents for detection of biological and other materials by luminescence methods
JPS63227599A (en) 1987-03-14 1988-09-21 Kirin Brewery Co Ltd Anthracycline compound and use thereof
US5210266A (en) * 1987-12-03 1993-05-11 Dainippon Pharmaceutical Co., Ltd. N-substituted mercaptopropanamide derivatives
US5179125A (en) * 1987-12-03 1993-01-12 Dainippon Pharmaceutical Co., Ltd. N-substituted mercaptopropanamide derivatives
DE58901634D1 (en) * 1988-11-05 1992-07-16 Bayer Ag METHOD FOR THE NUCLEAR CHLORINATION OF AROMATIC HYDROCARBONS.
JPH0662567B2 (en) 1988-12-27 1994-08-17 麒麟麦酒株式会社 Pyridinecarboximidamide derivative, production intermediate thereof, production method and use
US5272164A (en) 1988-12-27 1993-12-21 Kirin Beer Kabushiki Kaisha Carboximidamide derivatives
US5223508A (en) * 1988-12-27 1993-06-29 Kirin Beer Kabushiki Kaisha Pyridyl carboximidamide compounds useful in treating blood pressure
US5142647A (en) 1989-04-18 1992-08-25 Japan Tobacco, Inc. Magnus measuring apparatus
CA1341094C (en) * 1989-09-25 2000-09-05 Ronald G. Worton Diagnosis for malignant hyperthermia
ES2084706T3 (en) * 1989-09-30 1996-05-16 Kirin Brewery PLANTS PRODUCTION METHOD.
US5153184A (en) 1989-10-10 1992-10-06 Dowelanco ((N-heterocyclyl)carbonyl)phosphoramidothioate ester insecticides
US5075293A (en) 1989-10-10 1991-12-24 The Dow Chemical Company ((N-heterocyclyl)carbonyl)phosphoramidothioate ester insecticides
KR940000166B1 (en) * 1989-11-09 1994-01-08 니혼다바고 상교오 가부시기가이샤 Novel glucosamine derivative and liposome containing the same as membrane component
US4963671A (en) 1989-11-20 1990-10-16 E. R. Squibb & Sons, Inc. Process for resolving chiral intermediates used in making calcium channel blockers
US5214056A (en) * 1989-12-27 1993-05-25 Japan Tobacco Inc. 1,3,2-dioxathiolane oxide derivative
US5204462A (en) * 1990-03-30 1993-04-20 Japan Tobacco, Inc. 4h-3,1-benzoxazin-4-one derivative
JP2651043B2 (en) * 1990-07-10 1997-09-10 麒麟麦酒株式会社 Diphenylmethylpiperazine derivative
US5082847A (en) 1990-07-18 1992-01-21 Syntex (U.S.A.) Inc. Carbostyril compounds connected via an oxyalkyl group with a piperidine ring and having pharmaceutical utility
US5064810A (en) 1990-09-14 1991-11-12 Jeffrey Askanazi Use of branched chain amino acids to effect diaphragm contractility and fatigue
JP2703408B2 (en) 1990-12-28 1998-01-26 麒麟麦酒株式会社 1,4-benzothiazepine derivatives
DE4102103A1 (en) * 1991-01-25 1992-08-20 Bayer Ag SUBSTITUTED BENZOXAZEPINE AND BENZTHIAZEPINE, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE IN MEDICINAL PRODUCTS
CA2084997A1 (en) 1991-04-10 1992-10-11 Koji Kobayashi Oxazinone derivative
US5182272A (en) * 1991-05-03 1993-01-26 G. D. Searle & Co. 8-substituted-dibenz[b,f][1,4]oxazepine-10(11)-carboxylic acid, substituted hydrazides, pharmaceutical compositions, and methods for treating pain
US5180720A (en) * 1991-05-03 1993-01-19 G. D. Searle & Co. 2- and 3-alkoxy or hydroxy-8-substituted-dibenz[b,f]-[1,4]oxazepine-10(11H)-carboxylic acid, substituted hydrazides and methods for treating pain
KR0152100B1 (en) 1991-07-04 1998-10-01 시게루 미즈노 Preparation process of a t-cell differentiated and matured in respect of the phenotype of differentiation antigen and its funtion
KR100255493B1 (en) * 1991-10-11 2000-08-01 가마쿠라 아키오 Remedy for osteoporosis and diazepine compounds
WO1993013082A1 (en) * 1991-12-20 1993-07-08 G.D. Searle & Co. Substituted dibenzoxazepines or dibenzothiazepines and their use as prostaglandin e2 antagonists
MX9300433A (en) * 1992-01-28 1994-07-29 Kirin Brewery PIRIDINCARBOXIMIDAMINE COMPOUNDS AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
GB9203347D0 (en) * 1992-02-17 1992-04-01 Wellcome Found Hypolipidaemic compounds
JP2667351B2 (en) 1992-03-24 1997-10-27 麒麟麦酒株式会社 Dietary lipid digestion and absorption inhibitors and foods and beverages
US5387684A (en) * 1992-03-25 1995-02-07 The Green Cross Corporation Isoindazole compound
JP3093419B2 (en) 1992-03-30 2000-10-03 麒麟麦酒株式会社 1,4-benzothiazepine derivatives
US5304644A (en) * 1992-04-15 1994-04-19 G. D. Searle & Co. 1-,2-,3-,4-,5-,6-,7-,8- and/or 9 substituted dibenzoxazepine compounds, pharmaceutical compositions and methods for treating pain
US5478832A (en) 1992-05-08 1995-12-26 The Green Cross Corporation Quinoline compounds
CA2139362A1 (en) 1992-07-02 1994-01-20 Kooji Kagara Novel intermediate for synthesis and production of amino acid derivative
WO1994004523A1 (en) * 1992-08-21 1994-03-03 Japan Tobacco Inc. Dioxacycloalkane compound with renin-inhibiting activity
US5260286A (en) 1992-10-16 1993-11-09 Japan Tobacco, Inc. 2-piperidinecarboxylic acid derivatives useful as NMDA receptor antagonists
UA41342C2 (en) 1992-11-09 2001-09-17 Дзе Бутс Компані Плс 2,3,4,5-TETRAHYDRO -1,4-BENZOTHIAZEPINES, stereisomers thereof or pharmaceutically acceptable salts thereof, a method for the preparation thereof and a pharmaceutical composition based thereon
US5354758A (en) 1992-12-16 1994-10-11 Japan Tobacco Inc. Benzomorphans useful as NMDA receptor antagonists
ZA94284B (en) * 1993-01-27 1994-08-17 Shionogi & Co Process for preparing benzothiazepine derivatives
IL108634A0 (en) 1993-02-15 1994-05-30 Wellcome Found Hypolipidaemic heterocyclic compounds, their prepatation and pharmaceutical compositions containing them
IL108633A (en) * 1993-02-15 1998-07-15 Wellcome Found Hypolipidaemic benzothiazepine derivatives their preparation and pharmaceutical compositions containing them
JP4020958B2 (en) 1993-04-15 2007-12-12 キリンファーマ株式会社 Novel glycosphingolipids and uses thereof
US5461047A (en) 1993-06-16 1995-10-24 G. D. Searle & Co. 2-,3-,4-,5-,6-,7-,8-,9- and/or 10-substituted dibenzoxazepine and dibenzthiazepine compounds, pharmaceutical compositions and methods of use
US5354747A (en) 1993-06-16 1994-10-11 G. D. Searle & Co. 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9- and/or 10-substituted dibenzoxazepine and dibenzthiazepine compounds, pharmaceutical compositions and methods of use
CA2147355A1 (en) 1993-08-19 1995-02-23 Toru Hiyoshi Dna encoding atp-dependent fructose 6-phosphate 1-phosphotransferase originating from plant, recombinant vector containing the same and method for changing sugar content in plant cells under low temperature
FR2709753B1 (en) 1993-09-09 1995-12-01 Hoechst Lab Organic nitrates, process for their preparation and their use as medicaments.
KR0170534B1 (en) * 1993-11-10 1999-02-18 미즈노 시게루 Chroman derivative and medical use thereof
JPH08127594A (en) * 1993-11-10 1996-05-21 Mochida Pharmaceut Co Ltd New protein binding to fas antigen and dna coding the same
US6897295B1 (en) * 1993-11-10 2005-05-24 Mochida Pharmaceutical Co., Ltd. Antibodies and fragments thereof to Fas ligand and Fas ligand derived polypeptides
JP3914272B2 (en) 1993-12-28 2007-05-16 中外製薬株式会社 Gene encoding adseverin
JP2706755B2 (en) * 1994-02-10 1998-01-28 日本たばこ産業株式会社 Novel benzylaminoethoxybenzene derivative
US5457182A (en) 1994-02-15 1995-10-10 Merck & Co., Inc. FK-506 cytosolic binding protein, FKBP12.6
US5449675A (en) 1994-06-09 1995-09-12 G. D. Searle & Co. Substituted dibenzoxazepine and dibenzthiazepine urea compounds, pharmaceutical compositions and methods of use
US20040175814A1 (en) 1994-06-15 2004-09-09 Kirin Beer Kubushiki Kaisha Novel transferase and amylase, process for producing the enzymes, use thereof, and gene coding for the same
DE69535865D1 (en) * 1994-06-15 2008-11-27 Kirin Brewery Transferase and amylase, methods of making these enzymes, their use and coding genes
ZA956647B (en) 1994-08-10 1997-02-10 Wellcome Found Hypolipidaemic compounds.
WO1996015152A1 (en) 1994-11-11 1996-05-23 International Reagents Corporation Antiannexin-v monoclonal antibody, process for producing the same, and use therof
WO1996016062A1 (en) 1994-11-18 1996-05-30 Japan Tobacco Inc. Osteoporosis remedy and triazepine compound
US6632976B1 (en) * 1995-08-29 2003-10-14 Kirin Beer Kabushiki Kaisha Chimeric mice that are produced by microcell mediated chromosome transfer and that retain a human antibody gene
JP3193301B2 (en) * 1995-09-14 2001-07-30 麒麟麦酒株式会社 Bioactive protein p160
US6660837B1 (en) 1995-09-14 2003-12-09 Kirin Beer Kabushiki Kaisha Modified protein derived from protein kinase N
US5906819A (en) * 1995-11-20 1999-05-25 Kirin Beer Kabushiki Kaisha Rho target protein Rho-kinase
US5866341A (en) * 1996-04-03 1999-02-02 Chugai Pharmaceutical Co., Ltd. Compositions and methods for screening drug libraries
DE69740158D1 (en) 1996-07-08 2011-05-12 Kyowa Hakko Kirin Co Ltd COMPOUNDS INFLUENCING THE ACTIVITY OF CALCIUM RECEPTORS
JP3861187B2 (en) * 1996-08-02 2006-12-20 住友精化株式会社 Method for producing thiazepine derivatives
US6111072A (en) 1996-08-26 2000-08-29 Kirin Beer Kabushiki Kaisha Rho target protein human mDia and gene encoding same
US6338955B2 (en) * 1996-12-12 2002-01-15 Kirin Beer Kabushiki Kaisha β1-4 N-acetylglucosaminyltransferase and gene encoding
TW555562B (en) * 1996-12-27 2003-10-01 Kirin Brewery Method for activation of human antigen-presenting cells, activated human antigen-presenting cells and use thereof
JP2894445B2 (en) * 1997-02-12 1999-05-24 日本たばこ産業株式会社 Compounds effective as CETP activity inhibitors
JP3521382B2 (en) 1997-02-27 2004-04-19 日本たばこ産業株式会社 Cell surface molecules that mediate cell-cell adhesion and signal transduction
US7112655B1 (en) 1997-02-27 2006-09-26 Japan Tobacco, Inc. JTT-1 protein and methods of inhibiting lymphocyte activation
JP3885177B2 (en) 1997-03-26 2007-02-21 大塚製薬株式会社 Human gene
US6011036A (en) 1997-04-15 2000-01-04 Dr. Reddy's Research Foundation Heterocyclic compounds having antidiabetic hypolipidemic antihypertensive properties process for their preparation and pharmaceutical compositions containing them
DE19722317C1 (en) 1997-05-28 1998-10-08 Boehringer Ingelheim Int New human ryanodin receptor type 3 polypeptide
AUPO941497A0 (en) 1997-09-24 1997-10-16 Fujisawa Pharmaceutical Co., Ltd. Novel compounds
US20030064406A1 (en) * 1997-10-08 2003-04-03 Noboru Kaneko Process for analyzing annexin-V in urine, and application thereof
GB9724813D0 (en) 1997-11-25 1998-01-21 Univ Nottingham Reducing muscle fatigue
JP2959765B2 (en) * 1997-12-12 1999-10-06 日本たばこ産業株式会社 3-piperidyl-4-oxoquinazoline derivative and pharmaceutical composition containing the same
AU1682699A (en) * 1997-12-25 1999-07-19 Japan Tobacco Inc. Monoclonal antibody against connective tissue growth factor and medicinal uses thereof
US6583157B2 (en) 1998-01-29 2003-06-24 Tularik Inc. Quinolinyl and benzothiazolyl modulators
US6051714A (en) 1998-03-12 2000-04-18 Reilly Industries, Inc. Processes for dechlorinating pyridines
US6410561B1 (en) * 1998-03-26 2002-06-25 Japan Tobacco Inc. Amide derivatives and nociceptin antagonists
WO1999052895A1 (en) * 1998-04-10 1999-10-21 Japan Tobacco Inc. Amidine compounds
EP1089749A4 (en) * 1998-06-08 2001-04-11 Advanced Medicine Inc Novel therapeutic agents for membrane transporters
EP1122311A4 (en) * 1998-07-31 2002-04-17 Kirin Brewery Remedies for neuropathy containing as the active ingredient galectin-1 or its derivatives
KR20010075107A (en) 1998-09-17 2001-08-09 다께다 가즈히꼬 Remedies for photochemotherapy
CA2357038C (en) 1998-12-28 2008-10-21 Japan Tobacco Inc. Medical compositions comprising 1,4-benzothiazepine derivatives for the treatment of atrial fibrillation
AU754808B2 (en) * 1999-03-30 2002-11-28 Amgen Fremont Inc. Method for preparing Monoclonal Antibody
CA2373484C (en) 1999-05-19 2009-01-20 Noboru Kaneko Use of 1,4-benzothiazepine derivatives as drugs for overcoming resistance to anticancer drugs
GB9914745D0 (en) * 1999-06-24 1999-08-25 Knoll Ag Therapeutic agents
JP3871503B2 (en) 1999-08-30 2007-01-24 日本たばこ産業株式会社 Immune disease treatment
WO2001016321A1 (en) * 1999-09-01 2001-03-08 Otsuka Pharmaceutical Co., Ltd. Platelet membrane glycoprotein vi (gpvi) dna and protein sequences, and uses thereof
AU776745B2 (en) * 1999-09-30 2004-09-23 Noboru Kaneko Anticancer agents
US7135466B2 (en) 1999-12-24 2006-11-14 Kirin Beer Kabushiki Kaisha Quinoline and quinazoline derivatives and drugs containing the same
JP2003518494A (en) * 1999-12-29 2003-06-10 グラクソ グループ リミテッド Methods and compositions relating to modulators of annexin and cartilage homeostasis
US6808873B2 (en) 2000-01-14 2004-10-26 Mitokor, Inc. Screening assays using intramitochondrial calcium
IL150651A0 (en) 2000-01-20 2003-02-12 Eisai Co Ltd Nitrogen-containing cyclic compound and pharmaceutical compositions containing the same
EP1254898A4 (en) * 2000-01-26 2003-03-12 Ono Pharmaceutical Co Benzene-fused heterocycle derivatives and drugs containing the same as the active ingredient
US6824973B2 (en) 2000-02-03 2004-11-30 Kirin Beer Kabushiki Kaisha Method of promoting stem cell proliferation or survival by contacting a cell with a stem cell factor-like polypeptide
DK1258484T3 (en) 2000-02-18 2009-04-20 Kyowa Hakko Kirin Co Ltd New isoxazole and thiazole compounds and their use as drugs
CA2402436C (en) 2000-03-24 2010-02-16 Pharmacia Corporation Amidino compound and salts thereof useful as nitric oxide synthase inhibitors
US6545170B2 (en) * 2000-04-13 2003-04-08 Pharmacia Corporation 2-amino-5, 6 heptenoic acid derivatives useful as nitric oxide synthase inhibitors
US6787668B2 (en) 2000-04-13 2004-09-07 Pharmacia Corporation 2-amino-4,5 heptenoic acid derivatives useful as nitric oxide synthase inhibitors
AR034120A1 (en) 2000-04-13 2004-02-04 Pharmacia Corp HALOGENATED DERIVATIVE COMPOUND OF HEPTENOIC ACID 2-AMINO-4,5, PHARMACEUTICAL COMPOSITION THAT INCLUDES IT AND THE USE OF SUCH COMPOUND AND SUCH COMPOSITION IN THE MANUFACTURE OF A MEDICINAL PRODUCT TO INHIBIT OR MODULATE NITRIC ACID SYNTHESIS
AR032318A1 (en) 2000-04-13 2003-11-05 Pharmacia Corp HALOGENATED DERIVATIVE COMPOUND OF HEPTENOIC ACID 2-AMINO-5,6; PHARMACEUTICAL COMPOSITION THAT INCLUDES IT AND ITS USE IN THE MANUFACTURE OF A USEFUL MEDICINAL PRODUCT AS AN INHIBITOR OF NITRICAL SYNTHEASE OXIDE
US20060293266A1 (en) 2000-05-10 2006-12-28 The Trustees Of Columbia Phosphodiesterase 4D in the ryanodine receptor complex protects against heart failure
US20040048780A1 (en) * 2000-05-10 2004-03-11 The Trustees Of Columbia University In The City Of New York Method for treating and preventing cardiac arrhythmia
US20040229781A1 (en) * 2000-05-10 2004-11-18 Marks Andrew Robert Compounds and methods for treating and preventing exercise-induced cardiac arrhythmias
US7718644B2 (en) 2004-01-22 2010-05-18 The Trustees Of Columbia University In The City Of New York Anti-arrhythmic and heart failure drugs that target the leak in the ryanodine receptor (RyR2) and uses thereof
US7393652B2 (en) 2000-05-10 2008-07-01 The Trustees Of Columbia University In The City Of New York Methods for identifying a chemical compound that directly enhances binding of FKBP12.6 to PKA-phosphorylated type 2 ryanodine receptor (RyR2)
US8022058B2 (en) 2000-05-10 2011-09-20 The Trustees Of Columbia University In The City Of New York Agents for preventing and treating disorders involving modulation of the RyR receptors
US7879840B2 (en) 2005-08-25 2011-02-01 The Trustees Of Columbia University In The City Of New York Agents for preventing and treating disorders involving modulation of the RyR receptors
US6489125B1 (en) 2000-05-10 2002-12-03 The Trustees Of Columbia University In The City Of New York Methods for identifying chemical compounds that inhibit dissociation of FKBP12.6 binding protein from type 2 ryanodine receptor
EP1280533A2 (en) 2000-05-11 2003-02-05 Pharmacia Corporation Aldosterone antagonist composition for release during aldosterone acrophase
US20030220312A1 (en) 2000-05-11 2003-11-27 G.D. Searle & Co. Epoxy-steroidal aldosterone antagonist and calcium channel blocker combination therapy for treatment of cardiovascular disorders
JP3597140B2 (en) 2000-05-18 2004-12-02 日本たばこ産業株式会社 Human monoclonal antibody against costimulatory molecule AILIM and pharmaceutical use thereof
CN101498731A (en) 2000-05-18 2009-08-05 日本烟草产业株式会社 Human monoclonal antibody against a costimulatory signal transduction molecule ATLIM and pharmaceutical use thereof
EP1316548A4 (en) 2000-07-05 2004-05-19 Ajinomoto Kk Acylsulfonamide derivatives
WO2002002101A1 (en) 2000-07-05 2002-01-10 Ajinomoto Co., Inc. Hypoglycemics
WO2002009761A2 (en) * 2000-07-27 2002-02-07 Pharmacia Corporation Epoxy-steroidal aldosterone antagonist and calcium channel blocker combination therapy for treatment of congestive heart failure
WO2002010139A1 (en) 2000-08-01 2002-02-07 Pharmacia Corporation Hexahydro-7-1h-azepin-2-yl-haxanoic acid derivatives as inhibitors of inducible nitric oxide synthase
AR030741A1 (en) * 2000-09-15 2003-09-03 Pharmacia Corp DERIVATIVES OF ACIDS 2-AMINO-2-ALQUIL-5-HEPTENOICO AND - HEPTINOICO USEFUL AS INHIBITORS OF NITRICAL SYNTHEASE OXIDE
AR031129A1 (en) * 2000-09-15 2003-09-10 Pharmacia Corp DERIVATIVES OF ACIDS 2-AMINO-2-ALQUIL-4-HEXENOICO AND -HEXINOICO USEFUL AS INHIBITORS OF NITRICO OXIDE SYNTHEASE
AR031608A1 (en) 2000-09-15 2003-09-24 Pharmacia Corp DERIVATIVES OF ACIDS 2-AMINO-2-ALQUIL-3-HEXENOICO AND -HEXINOICO USEFUL AS INHIBITORS OF NITRICO OXIDE SYNTHEASE
US6936426B2 (en) * 2000-10-06 2005-08-30 Harbor-Ucla Research And Education Institute Detection of antibody mediated inflammatory auto-immune disorders
IL155977A0 (en) * 2000-11-30 2003-12-23 Medarex Inc Transgenic transchromosomal rodents for making human antibodies
US6673904B2 (en) * 2000-12-23 2004-01-06 Kirin Beer Kabushiki Kaisha Stem cell growth factor-like polypeptides
US6962926B2 (en) 2001-01-31 2005-11-08 Telik, Inc. Antagonist of MCP-1 function, and compositions and methods of use thereof
WO2002061076A1 (en) * 2001-02-01 2002-08-08 Mochida Pharmaceutical Co., Ltd. Adiponectin-associated protein
JP4817514B2 (en) * 2001-03-09 2011-11-16 協和発酵キリン株式会社 Novel animal cell vectors and uses thereof
JP4175113B2 (en) * 2001-03-14 2008-11-05 小野薬品工業株式会社 A therapeutic agent for depression containing an EP1 antagonist as an active ingredient
US20030054531A1 (en) * 2001-03-19 2003-03-20 Decode Genetics Ehf, Human stroke gene
WO2002078625A2 (en) 2001-03-28 2002-10-10 Pharmacia Corporation Therapeutic combinations for cardiovascular and inflammatory indications
EP1382604B1 (en) * 2001-04-27 2005-12-28 Kirin Beer Kabushiki Kaisha Quinoline derivative having azolyl group and quinazoline derivative
JP4230681B2 (en) 2001-07-06 2009-02-25 株式会社東芝 High voltage semiconductor device
US20030220310A1 (en) 2001-07-27 2003-11-27 Schuh Joseph R. Epoxy-steroidal aldosterone antagonist and calcium channel blocker combination therapy for treatment of congestive heart failure
GB0121621D0 (en) * 2001-09-07 2001-10-31 Astrazeneca Ab Chemical compounds
EP1435946B8 (en) * 2001-09-14 2013-12-18 Amgen Inc. Linked biaryl compounds
JP2003095977A (en) * 2001-09-21 2003-04-03 Masafumi Yano Therapeutic agent or prophylactic against disease associated with functional disorder of muscular contraction and relaxation
JP4087338B2 (en) 2001-11-15 2008-05-21 キリンファーマ株式会社 Chimeric non-human animal
JP2003145746A (en) * 2001-11-16 2003-05-21 Seiko Epson Corp Method of inkjet recording and inkjet recorder
AU2002354056A1 (en) * 2001-11-19 2003-06-10 Ono Pharmaceutical Co., Ltd. Remedies for urinary frequency
EP1452530A4 (en) * 2001-12-03 2005-11-30 Japan Tobacco Inc Azole compound and medicinal use thereof
US20040171613A1 (en) 2001-12-27 2004-09-02 Japan Tobacco Inc. Therapeutic agent for non-immediate type allergic diseases
WO2003061699A1 (en) 2001-12-27 2003-07-31 Japan Tobacco, Inc. Remedies for allergic diseases
JP2005521653A (en) * 2002-01-17 2005-07-21 ファルマシア コーポレイション Novel alkyl / arylhydroxy or ketothiepine compounds as inhibitors of advanced sodium co-dependent bile acid transport (ASBT) and taurocholate uptake
EP1489077A4 (en) 2002-02-01 2006-06-21 Dainippon Sumitomo Pharma Co 2-furancarboxylic acid hydrazides and pharmaceutical compositions containing the same
CN1870983B (en) 2002-02-14 2010-12-01 麒麟麦酒株式会社 Compositions and foods for improving lipid metabolism
WO2003091280A1 (en) 2002-04-26 2003-11-06 Kirin Beer Kabushiki Kaisha Polypeptide having an activity to support proliferation or survival of hematopoietic stem cell or hematopoietic progenitor cell, and dna coding for the same
JPWO2003090570A1 (en) * 2002-04-26 2005-08-25 日本たばこ産業株式会社 Rod-shaped article forming device
JP4113042B2 (en) * 2002-05-24 2008-07-02 シチズンホールディングス株式会社 Display device and color display method
DE10237723A1 (en) 2002-08-17 2004-07-08 Aventis Pharma Deutschland Gmbh Use of IKappaB kinase inhibitors in pain therapy
DE60311821T2 (en) * 2002-08-30 2007-10-31 Japan Tobacco Inc. DIBENZYLAMINE COMPOUND AND THEIR MEDICAL USE
JP2004103256A (en) 2002-09-04 2004-04-02 Nippon Chemicon Corp Emergency light
EP1550660A1 (en) 2002-09-12 2005-07-06 Kirin Beer Kabushiki Kaisha Isoquinoline derivatives having kinasae inhibitory activity and drugs containing the same
KR20050062551A (en) 2002-09-20 2005-06-23 기린 비루 가부시키가이샤 HEPATITIS C VIRUS INHIBITOR COMPRISING α-GLYCOSYLCERAMIDE AS THE ACTIVE INGREDIENT
KR20050048666A (en) 2002-10-04 2005-05-24 기린 비루 가부시키가이샤 Human artificial chromosome(hac) vector
US7544678B2 (en) 2002-11-05 2009-06-09 The Trustees Of Columbia University In The City Of New York Anti-arrythmic and heart failure drugs that target the leak in the ryanodine receptor (RyR2)
WO2004042389A2 (en) 2002-11-08 2004-05-21 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with human phosphodiesterase 4d (pde4d)
JP3988830B2 (en) 2002-11-22 2007-10-10 日本たばこ産業株式会社 Fused bicyclic nitrogen-containing heterocycle
EP1439221B1 (en) 2002-12-17 2007-01-24 F. Hoffmann-La Roche Ag PDE core construct
ATE517887T1 (en) 2003-02-13 2011-08-15 Msd Kk NEW 2-PYRIDINECARBONIC ACID AMIDE DERIVATIVES
AU2004220548A1 (en) 2003-03-07 2004-09-23 The Trustees Of Columbia University, In The City Of New York Type 1 ryanodine receptor-based methods
TWI220798B (en) 2003-03-07 2004-09-01 Hitachi Cable Light-emitting diode array
MXPA05009976A (en) 2003-03-17 2005-11-04 Japan Tobacco Inc Method for increasing the oral bioavailability of s-`2- (``1-(2- ethylbutyl) cyclohexyl! carbonyl! amino) phenyl!-2- methylpropanethioate.
CA2519432C (en) 2003-03-17 2009-06-09 Japan Tobacco Inc. Pharmaceutical compositions comprising cetp inhibitors and a water-insoluble concentration-enhancing additive for the treatment of cardiovascular disease
US20040235162A1 (en) 2003-03-18 2004-11-25 Kirin Beer Kabushiki Kaisha Method of preparing immunoregulatory dendritic cells and the use thereof
CL2004000544A1 (en) 2003-03-18 2005-01-28 Pharmacia Corp Sa Organizada B USE OF A PHARMACEUTICAL COMBINATION, OF AN ALGOSTERONE RECEIVER ANTAGONIST AND A NEUTRAL ENDOPEPTIDASE INHIBITOR, USEFUL FOR THE TREATMENT AND PREVENTION OF A PATHOLOGICAL CONDITION RELATED TO HYPERTENSION, RENAL DYSFUNCTION, INSULINOPATI
US20050032210A1 (en) * 2003-03-18 2005-02-10 Kirin Beer Kabushiki Kaisha Method of preparing immuno-regulatory dendritic cells and the use thereof
JP4926473B2 (en) 2003-03-27 2012-05-09 協和発酵キリン株式会社 COMPOUND INHIBITING IN VIVO PHOSPHORUS TRANSPORT AND PHARMACEUTICAL COMPRISING THE SAME
MXPA05010945A (en) 2003-04-09 2005-11-25 Japan Tobacco Inc Heteroaromatic pentacyclic compound and medicinal use thereof.
US20060135506A1 (en) 2003-04-22 2006-06-22 Pharmacia Corporation Compositions of a cyclooxygenase-2 selective inhibitor and a calcium modulating agent for the treatment of pain, inflammation or inflammation mediated disorders
WO2004093816A2 (en) 2003-04-22 2004-11-04 Pharmacia Corporation Compositions comprising a selective cox-2 inhibitor and a calcium modulating agent
WO2004093896A1 (en) * 2003-04-22 2004-11-04 Pharmacia Corporation Compositions of a cyclooxygenase-2 selective inhibitor and a potassium ion channel modulator for the treatment of central nervous system damage
US20040229803A1 (en) 2003-04-22 2004-11-18 Pharmacia Corporation Compositions of a cyclooxygenase-2 selective inhibitor and a potassium ion channel modulator for the treatment of pain, inflammation or inflammation mediated disorders
TWI393560B (en) * 2003-05-02 2013-04-21 Japan Tobacco Inc Combination comprising s-[2-([[1-(2-ethylbutyl)cyclohexyl]carbonyl]amino)phenyl]2-methylpropanethioate and an hmg coa reductase inhibitor
US20050070543A1 (en) * 2003-07-11 2005-03-31 Pharmacia Corporation Compositions of a chromene or phenyl acetic acid cyclooxygenase-2 selective inhibitor and an ACE inhibitor for the treatment of central nervous system damage
JP2007501801A (en) * 2003-08-07 2007-02-01 日本たばこ産業株式会社 Pyrrolo [1,2-b] pyridazine derivatives
CA2536173A1 (en) * 2003-08-20 2005-03-03 Nitromed, Inc. Nitrosated and nitrosylated cardiovascular compounds, compositions and methods of use
JP2007504136A (en) * 2003-08-28 2007-03-01 ニトロメッド インコーポレーティッド Nitrosated and nitrosylated diuretic compounds, compositions and methods of use
US20050187221A1 (en) 2003-09-08 2005-08-25 Japan Tobacco Inc. Method of treating ischemia reperfusion injury
AU2004308932A1 (en) 2003-12-22 2005-07-14 Amgen Inc Aryl sulfonamide compounds and uses related thereto
US7165879B2 (en) 2004-03-25 2007-01-23 Red Devil Equipment Company Clamp lock apparatus and method for a paint mixer
JP2007533328A (en) * 2004-04-22 2007-11-22 キリンホールディングス株式会社 Transgenic animals and uses thereof
AU2005238389B2 (en) 2004-04-28 2009-08-06 Aetas Pharma Co., Ltd. Muscle relaxation accelerator and therapeutic agent for muscular tissue diseases such as muscle relaxation failure
US7378423B2 (en) * 2004-06-11 2008-05-27 Japan Tobacco Inc. Pyrimidine compound and medical use thereof
JP3968358B2 (en) * 2004-06-30 2007-08-29 日本電信電話株式会社 Thin flat twist pair clearance cable and clearance navigator unit
JP4018739B2 (en) * 2004-07-15 2007-12-05 日本たばこ産業株式会社 Condensed benzamide compound and vanilloid receptor type 1 (VR1) activity inhibitor
JP2006094849A (en) 2004-08-30 2006-04-13 Kirin Brewery Co Ltd Pluripotent cell with improved homologous recombination efficiency and use thereof
AR051780A1 (en) * 2004-11-29 2007-02-07 Japan Tobacco Inc FUSIONED RING COMPOUNDS CONTAINING NITROGEN AND USING THEMSELVES
EP1855688A4 (en) 2005-02-24 2011-09-14 Nicox Sa Nitric oxide enhancing diuretic compounds, compositions and methods of use
AU2006219231B2 (en) 2005-02-28 2010-01-14 Japan Tobacco Inc. Novel aminopyridine compound with Syk inhibitory activity
WO2006098394A1 (en) 2005-03-14 2006-09-21 Japan Tobacco Inc. Method for inhibiting lipid absorption and lipid absorption inhibitor
US7704990B2 (en) 2005-08-25 2010-04-27 The Trustees Of Columbia University In The City Of New York Agents for preventing and treating disorders involving modulation of the RyR receptors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8710045B2 (en) 2004-01-22 2014-04-29 The Trustees Of Columbia University In The City Of New York Agents for preventing and treating disorders involving modulation of the ryanodine receptors
EP2127654A1 (en) * 2004-03-25 2009-12-02 The Trustees of Columbia University in the City of New York Benzothiazepine derivatives as ryanodine receptor (RYR2) inhibitors and their use in the treatment of cardiac diseases
WO2008144483A2 (en) * 2007-05-18 2008-11-27 Armgo Pharma, Inc. Agents for treating disorders involving modulation of ryanodine receptors
WO2008144483A3 (en) * 2007-05-18 2012-08-16 Armgo Pharma, Inc. Agents for treating disorders involving modulation of ryanodine receptors
KR101337244B1 (en) 2009-02-25 2013-12-06 노보루 카네코 1,4-benzothiazepine-1-oxide derivative and pharmaceutical composition utilizing the same
EP2402333A1 (en) * 2009-02-25 2012-01-04 Noboru Kaneko 1,4-benzothiazepine-1-oxide derivative and pharmaceutical composition utilizing the same
CN102333774A (en) * 2009-02-25 2012-01-25 金子升 1, 4-benzothiaza-1-oxide derivatives and pharmaceutical compositions using the derivatives
EP2402333A4 (en) * 2009-02-25 2012-08-01 Noboru Kaneko 1,4-benzothiazepine-1-oxide derivative and pharmaceutical composition utilizing the same
US8933129B2 (en) 2009-04-15 2015-01-13 State Of Oregon By And Through The State Board Of Higher Education On Behalf Of Portland State University Compounds and methods for modulating activity of calcium release channels
EP2418950A1 (en) * 2009-04-15 2012-02-22 State Of Oregon By&Through The State Board Of Higher Education On Behalf Of Portland State Unv. Compounds and methods for modulating activity of calcium release channels
EP2418950A4 (en) * 2009-04-15 2012-10-24 State Of Oregon By & Through The State Board Of Higher Education On Behalf Of Portland State Unv Compounds and methods for modulating activity of calcium release channels
WO2012019076A1 (en) 2010-08-06 2012-02-09 The Trustees Of Columbia University In The City Of New York Compositions and methods for preventing and treating cardiac ischemia/reperfusion injury
EA027922B1 (en) * 2012-04-18 2017-09-29 Ле Лаборатуар Сервье Agents for treating disorders involving modulation of ryanodine receptors
US8853198B2 (en) 2012-04-18 2014-10-07 Les Laboratoires Servier Agents for treating disorders involving modulation of ryanodine receptors
WO2013156505A1 (en) 2012-04-18 2013-10-24 Les Laboratoires Servier Agents for treating disorders involving modulation of ryanodine receptors
AU2013248313B2 (en) * 2012-04-18 2016-11-10 Armgo Pharma Inc. Agents for treating disorders involving modulation of ryanodine receptors
KR101731459B1 (en) 2012-04-18 2017-04-28 르 라보레또레 쎄르비에르 Agents for treating disorders involving modulation of ryanodine receptors
EP2653466A1 (en) 2012-04-18 2013-10-23 Les Laboratoires Servier Agents for treating disorders involving modulation of ryanodine receptors
MD4489C1 (en) * 2012-04-18 2018-01-31 Les Laboratoires Servier 1,4-Benzothiazepine derivatives, method for the synthesis thereof, pharmaceutical compositions comprising the same, and use thereof for treating and preventing disorders involving modulation of ryanodine receptors
EP2708535A1 (en) 2012-05-11 2014-03-19 Les Laboratoires Servier Agents for treating disorders involving modulation of ryanodine receptors
WO2021015221A1 (en) 2019-07-22 2021-01-28 株式会社アエタスファルマ Method for producing optically active 1, 4-benzothiazepine-1-oxide derivative
US11504383B2 (en) 2021-01-08 2022-11-22 Armgo Pharma, Inc. Crystalline forms of a Ryanodine receptor modulator and uses thereof
US11717526B2 (en) 2021-05-20 2023-08-08 Armgo Pharma, Inc. Pharmaceutical compositions comprising a ryanodine receptor modulator and uses thereof

Also Published As

Publication number Publication date
TW200800930A (en) 2008-01-01
IL189675A (en) 2013-12-31
GEP20105134B (en) 2010-12-27
KR20120025626A (en) 2012-03-15
HRP20130353T1 (en) 2013-06-30
IL189675A0 (en) 2008-06-05
CY1114091T1 (en) 2016-07-27
EP2177224B1 (en) 2013-02-27
EP2177224A3 (en) 2010-06-30
ECSP088306A (en) 2008-07-30
EA200800665A1 (en) 2008-08-29
HK1137150A1 (en) 2010-07-23
PL2311464T3 (en) 2013-07-31
CY1113974T1 (en) 2016-07-27
RS52780B (en) 2013-10-31
PT2311464E (en) 2013-05-15
NZ566822A (en) 2011-04-29
DK2177224T3 (en) 2013-05-13
CR9812A (en) 2009-09-14
EP2311464B1 (en) 2013-02-13
SI1928850T1 (en) 2013-09-30
PL2177224T3 (en) 2013-07-31
HK1153664A1 (en) 2012-04-05
HRP20130669T1 (en) 2013-08-31
DK2311464T3 (en) 2013-05-13
NO20081421L (en) 2008-05-23
CN102558093A (en) 2012-07-11
JP2009506034A (en) 2009-02-12
PT1928850E (en) 2013-08-23
SI2177224T1 (en) 2013-06-28
PL1928850T3 (en) 2013-11-29
EP2764867A1 (en) 2014-08-13
US20070049572A1 (en) 2007-03-01
EP1928850A4 (en) 2009-06-17
AU2006283534C1 (en) 2012-08-02
ES2405780T3 (en) 2013-06-03
HRP20130360T1 (en) 2013-06-30
SV2008002828A (en) 2010-07-20
EP2177224A2 (en) 2010-04-21
AU2006283534B2 (en) 2010-11-04
TNSN08078A1 (en) 2009-07-14
RS52781B (en) 2013-10-31
WO2007024717A3 (en) 2009-01-08
EP1928850B1 (en) 2013-06-19
DK1928850T3 (en) 2013-09-23
BRPI0615097A2 (en) 2009-07-14
EA014941B1 (en) 2011-04-29
CA2620183A1 (en) 2007-03-01
ES2421159T3 (en) 2013-08-29
CN101500576A (en) 2009-08-05
CN101500576B (en) 2013-08-14
MA29785B1 (en) 2008-09-01
CA2620183C (en) 2014-04-15
PT2177224E (en) 2013-05-15
JP5342877B2 (en) 2013-11-13
AP3091A (en) 2015-01-31
KR20080037741A (en) 2008-04-30
KR101456342B1 (en) 2014-11-03
CN102558093B (en) 2014-10-15
MY144622A (en) 2011-10-14
AR057776A1 (en) 2007-12-19
US7704990B2 (en) 2010-04-27
HN2008000299A (en) 2011-07-11
ZA200802338B (en) 2009-01-28
EP1928850A2 (en) 2008-06-11
AU2006283534A1 (en) 2007-03-01
UA93388C2 (en) 2011-02-10
CY1113975T1 (en) 2016-07-27
ES2405765T3 (en) 2013-06-03
RS52852B (en) 2013-12-31
EP2311464A1 (en) 2011-04-20
HK1119705A1 (en) 2009-03-13
SI2311464T1 (en) 2013-06-28
TWI486338B (en) 2015-06-01
BRPI0615097B1 (en) 2021-06-01

Similar Documents

Publication Publication Date Title
CA2620183C (en) Agents for preventing and treating disorders involving modulation of the ryr receptors
US7879840B2 (en) Agents for preventing and treating disorders involving modulation of the RyR receptors
US8022058B2 (en) Agents for preventing and treating disorders involving modulation of the RyR receptors
WO2008144483A2 (en) Agents for treating disorders involving modulation of ryanodine receptors
AU2005329398A1 (en) Novel anti-arrythmic and heart failure drugs that target the leak in the ryanodine receptor (RyR2)
US20140187536A1 (en) Agents for preventing and treating disorders involving modulation of the ryanodine receptors
AU2005329399A1 (en) Novel anti-arrythmic and heart failure drugs that target the leak in the ryanodine receptor (RyR2)
AU2004281672A1 (en) Compounds and methods for treating and preventing exercise-induced cardiac arrhythmias
MX2008002687A (en) Agents for preventing and treating disorders involving modulation of the ryr receptors

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680039573.9

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 189675

Country of ref document: IL

Ref document number: 2008020304

Country of ref document: EG

ENP Entry into the national phase

Ref document number: 2620183

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: AP/P/2008/004363

Country of ref document: AP

Ref document number: 12008500467

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/002687

Country of ref document: MX

Ref document number: 2008528020

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006801887

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006283534

Country of ref document: AU

Ref document number: DZP2008000157

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: 08027353

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 566822

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 10576

Country of ref document: GE

Ref document number: 2453/DELNP/2008

Country of ref document: IN

Ref document number: 200800665

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 1020087007255

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2006283534

Country of ref document: AU

Date of ref document: 20060817

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: PI0615097

Country of ref document: BR

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 1020127003439

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: P-2013/0294

Country of ref document: RS