WO2010085805A1 - Methods for treating acute myocardial infarctions and associated disorders - Google Patents

Methods for treating acute myocardial infarctions and associated disorders Download PDF

Info

Publication number
WO2010085805A1
WO2010085805A1 PCT/US2010/022112 US2010022112W WO2010085805A1 WO 2010085805 A1 WO2010085805 A1 WO 2010085805A1 US 2010022112 W US2010022112 W US 2010022112W WO 2010085805 A1 WO2010085805 A1 WO 2010085805A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
patient
therapeutic
ami
days
Prior art date
Application number
PCT/US2010/022112
Other languages
French (fr)
Inventor
Jeff Olgin
Karl Kossen
Original Assignee
Intermune, Inc.
The Regents Of The University Of California
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 CN2010800055907A priority Critical patent/CN102292124A/en
Priority to BRPI1006979A priority patent/BRPI1006979A2/en
Priority to AP2011005824A priority patent/AP2011005824A0/en
Priority to EP10734007A priority patent/EP2389227A4/en
Priority to JP2011548228A priority patent/JP2012515800A/en
Priority to SG2011050952A priority patent/SG172981A1/en
Application filed by Intermune, Inc., The Regents Of The University Of California filed Critical Intermune, Inc.
Priority to MX2011007854A priority patent/MX2011007854A/en
Priority to AU2010206543A priority patent/AU2010206543A1/en
Priority to CA2747251A priority patent/CA2747251A1/en
Publication of WO2010085805A1 publication Critical patent/WO2010085805A1/en
Priority to IL213526A priority patent/IL213526A0/en
Priority to TN2011000364A priority patent/TN2011000364A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system having sulfur as a ring hetero atom, e.g. ticlopidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4433Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4436Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • 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
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the invention relates to methods of treating patients who have suffered an acute myocardial infarction (AMI) and associated disorders with a therapeutic that has anti-fibrotic effects, for example, pirfenidone and analogs thereof.
  • AMI acute myocardial infarction
  • a therapeutic that has anti-fibrotic effects for example, pirfenidone and analogs thereof.
  • MI acute myocardial infarction
  • CHF congestive heart failure
  • VT ventricular tachycardia
  • Contraction of the heart is initiated by an electrical impulse generated by the sinoatrial node, a natural pacemaker, in the heart
  • the heart's electrical conduction system then conveys the impulse to the myocardium, or cardiac muscle, to stimulate contraction
  • Abnormal electrical conduction due to structural tissue remodeling after infarction may play an important role in ventricular arrhythmias, which can lead to sudden cardiac arrest and death.
  • Tissue remodeling is due in part to direct tissue damage, neurohormonal activation, cytokine release, inflammation and fibrosis.
  • a compound which inhibits fibrosis has beneficial effects on left ventricular (LV) function, infarct size, peri-mfarct fibrosis, electrophysiology of the infarct border zone and VT inducibility. It is also unexpected that such compounds offer a more targeted and effective inhibition of detrimental post-acute MI remodeling than RAA blockers.
  • novel means to prevent arrhythmias in the post-acute MI period and to improve heart contractility, improve heart function and reduce complications of acute MI such as congestive heart failure (CHF) and ventricular tachycardia (VT) and ventricular fibrillation.
  • CHF congestive heart failure
  • VT ventricular tachycardia
  • the present invention discloses a method of treating a patient who has suffered a myocardial infarction (MI), or who has not previously suffered an MI, or is within a week of suffering an MI, comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect.
  • a method of treating a patient who has suffered a myocardial infarction e.g. an acute myocardial infarction (AMI)
  • AMI acute myocardial infarction
  • the method is to limit expansion of an infarct scar due to the myocardial infarction (e.g. the AMI)
  • the invention provides a method of treating a patient who has suffered myocardial infarction (e g. an AMI) comprising administering to said patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect
  • the treatment is initiated at a time period about 1 to 42 days after suffering the myocardial infarction (e.g. the AMI), and optionally continues for up to 3 to 6 months.
  • the treatment is initiated at a time period about 3 to 14 days after suffering the myocardial infarction (e.g. the AMI), and optionally continues for up to 3 to 6 months.
  • the treatment is initiated about 5-10 days after the myocardial infarction (e.g.
  • the treatment is initiated about 2-40 days after the myocardial infarction (e.g. the AMI). In another embodiment, the treatment is initiated about 3-20 days after the myocardial infarction (e.g. the AMI). In another embodiment, the treatment is initiated about 4-15 days after the myocardial infarction (e.g. the AMI). In yet another embodiment, the treatment is initiated about 7 days after the myocardial infarction (e.g. the AMI) In some embodiments, the treatment continues for a period of at least 2 weeks. In other embodiments, the treatment after being initiated continues for a time period until about 4 weeks after the myocardial infarction (e.g. the AMI). Thus, the invention encompasses treatment of patients from about 14 days to 4 weeks after the myocardial infarction (e g the AMI)
  • the invention provides a method of reducing the incidence of congestive heart failure (CHF) in a patient who suffered a myocardial infarction (e.g., an acute myocardial infarction (AMI)), comprising administering to said patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the therapeutically effective dose reduces the incidence of congestive heart failure, and wherein optionally the treatment is initiated at a time period about 1 to 42 days after suffering the myocardial infarction (e.g. the AMI).
  • the patient is at an increased risk of congestive heart failure due to the myocardial infarction (e.g. the AMI).
  • the invention provides a method of preserving viable cardiac tissue or controlling myocardial infarct size in a patient who has suffered a myocardial infarction (e g an acute myocardial infarction (AMI)) comprising administering to said patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the administering of said therapeutic to said patient results in a relatively reduced infarct size on average compared to infarct size in a patient who has not been administered said therapeutic.
  • the treatment is initiated at a time period about 1 to 42 days after suffering the myocardial infarction (e.g. the AMI).
  • the relative reduction in infarct size is at least 5%
  • the invention provides a method of reducing the incidence of ventricular tachycardia in a patient in need thereof, comprising administering to said patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect.
  • the patient has suffered a myocardial infarction (e g. an AMI).
  • the treatment is initiated at a time period about 1 to 42 days after suffering the myocardial infarction (e.g. the AMI).
  • the administering is initiated about 7 days after suffe ⁇ ng the myocardial infarction (e.g. the AMI).
  • the invention provides a method of treating or preventing ventricular fibrillation in a patient in need thereof is provided, comprising administering to said patient a therapeutic having an anti-fibrotic effect.
  • the patient has suffered a myocardial infarction (e g an AMI)
  • the treatment is initiated at a time pe ⁇ od about 1 to 42 days after suffering the myocardial infarction (e g the AMI)
  • the administering is initiated about 7 days after the suffering of the myocardial infarction (e.g. the AMI).
  • the administering reduces the incidence of sudden cardiac death relative to the incidence of cardiac death in the absence of administration of the therapeutic.
  • the administering reduces cardiac risk of the patient relative to the cardiac risk in the absence of administration of the therapeutic.
  • cardiac risk means the risk of cardiac morbidity resulting from any one or a combination of ventricular tachycardia, sudden cardiac death, ventricular fibrillation and/or congestive heart failure.
  • the invention provides a method of controlling (e.g., reduce, reduce the incidence or severity of, or prevent the progression of) arrhythmia in a patient in need thereof is provided, comprising administering to the patient a therapeutic having an anti- fibrotic effect, wherein the administering of the therapeutic controls (e.g., reduce, reduce the incidence or severity of, or prevent the progression of) arrhythmia in the patient.
  • a method of controlling e.g., reduce, reduce the incidence or severity of, or prevent the progression of arrhythmia in a patient in need thereof is provided, comprising administering to the patient a therapeutic having an anti- fibrotic effect, wherein the administering of the therapeutic controls (e.g., reduce, reduce the incidence or severity of, or prevent the progression of) arrhythmia in the patient.
  • the administering reduces the incidence or severity of arrhythmia in the patient relative to the incidence or severity of arrhythmia in the absence of administration of the therapeutic
  • the patient has suffered a myocardial infarction (e g an AMI)
  • the administration is initiated about 1 to 42 days after the suffering of the myocardial infarction (e.g. the AMI).
  • the administration is initiated about 7 days after the suffering of the myocardial infarction (e g. the AMI).
  • the administering treats ventricular remodeling.
  • the patient is diagnosed as suffering a first myocardial infarction (e.g. a first AMI), i e. the patient has not been diagnosed as having previously suffered a myocardial infarction (e g an AMI) or the patient has not previously suffered a myocardial infarction (e g an AMI)
  • a first myocardial infarction e.g. a first AMI
  • any of the methods described herein optionally exclude treatment of patients diagnosed with chronic MI.
  • the therapeutic having an anti-fibrotic effect reduces tissue remodeling or fibrosis
  • the therapeutic having an anti-fibrotic effect reduces the activity of transforming growth factor-beta (TGF- ⁇ ), targets one or more TGF- ⁇ isoforms, inhibits TGF- ⁇ receptor kinases TGFBRl (ALK5) and/or TGFBR2, or modulates one or more post- receptor signaling pathways.
  • TGF- ⁇ transforming growth factor-beta
  • targets one or more TGF- ⁇ isoforms targets one or more TGF- ⁇ isoforms, inhibits TGF- ⁇ receptor kinases TGFBRl (ALK5) and/or TGFBR2, or modulates one or more post- receptor signaling pathways.
  • TGFBRl TGF- ⁇ receptor kinases
  • the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the TGF- ⁇ pathway and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect is an endothelin receptor antagonist, targets both endothelin receptor A and endothelin receptor B or selectively targets endothelin receptor A.
  • the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the endothelin A and/or B pathway, and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect reduces activity of connective tissue growth factor (CTGF).
  • CTGF connective tissue growth factor
  • the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the CTGF pathway and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect inhibits matrix metalloproteinase (MMP).
  • MMP matrix metalloproteinase
  • the therapeutically effective amount of such a compound may inhibit MMP and/or reduce fibrosis.
  • the therapeutically effective amount of such a compound may inhibit MMP-9 or MMP- 12.
  • the therapeutic having an anti-fibrotic effect reduces the activity of epidermal growth factor receptor (4), targets EGF receptor, or inhibits EGF receptor kinase.
  • the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the EGF pathway and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect reduces the activity of platelet derived growth factor (PDGF), targets PDGF receptor (PDGFR), inhibits PDGFR kinase activity, or inhibits post-PDGF receptor signaling pathways
  • PDGF platelet derived growth factor
  • PDGFR PDGF receptor
  • the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the PDGF pathway and/or reduce fibrosis
  • the therapeutic having an anti fibrotic effect reduces the activity of vascular endothelial growth factor (VEGF), targets one or more of VEGF, VEGF receptor 1 (VEGFRl, Fit 1), or VEGF receptor 2 (VEGFR2, KDR).
  • VEGF vascular endothelial growth factor
  • the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the VEGF pathway and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect inhibits multiple receptor kinases such as BIRB-1120 which inhibits receptor kinases for vascular endothelial growth factor, fibroblast growth factor, and platelet derived growth factor
  • the therapeutically effective amount of such a compound may inhibit one or more receptor kinases in the VEGF, FGF or PDGF pathways and/or reduce fibrosis
  • the therapeutic having an anti-fibrotic effect interferes with mtegnn function
  • the therapeutically effective amount of such a compound may inhibit integrin function and/or reduce fibrosis
  • the therapeutic having an anti-fibrotic effect may inhibit ⁇ V mtegrins.
  • the therapeutic having an anti fibrotic effect may inhibit mtegnn ⁇ V ⁇ 6 function
  • the therapeutic having an anti-fibrotic effect interferes with pro-fibrotic activities of IL-4 and IL-13, targets IL-4 receptor, IL-13 receptor.
  • the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the IL-4 and/or IL-13 pathway and/or reduce fibrosis
  • the therapeutic having an anti-fibrotic effect modulates signaling through the JAK-STAT pathway.
  • the therapeutically effective amount of such a compound may modulate signaling through the JAK-STAT pathway and/or reduce fibrosis
  • the therapeutic having an anti-fibrotic effect interferes with epithelial mesenchymal transition, or inhibits mTor
  • the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects on mesenchyma, and/or reduce fibrosis
  • the therapeutic having an anti-fibrotic effect reduces levels of copper. In such cases, the therapeutically effective amount of such a compound may reduce copper levels in circulation and/or tissue, and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect reduces oxidative stress
  • the therapeutically effective amount of such a compound may reduce oxidative stress and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect inhibits prolyl hydrolyse.
  • the therapeutically effective amount of such a compound may reduce prolyl hydrolase and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect is an agonist of proliferator- activated receptor-gamma (PPAR- ⁇ ).
  • the therapeutic having an anti-fibrotic effect inhibits phosphodiesterase 4 (PDE4) or phosphodiesterase 5 (PDE5), or modifies the arachidonic acid pathway.
  • the therapeutically effective amount of such a compound may inhibit the PDE4 and/or PDE5 pathway, or may inhibit the arachidonic acid pathway, and/or reduce fibrosis.
  • the therapeutic having an anti-fibrotic effect is combined with a pharmaceutically acceptable carrier.
  • the administration is oral.
  • the therapeutically effective amount is a total daily dose of about 50 mg to about 2400 mg of said therapeutic or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof.
  • the therapeutically effective amount is administered in divided doses three times a day or two times a day, or is administered in a single dose once a day.
  • said therapeutic is pirfenidone or compound of formula (I), (II), (III), (IV), or (V) or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof:
  • R 1 , R 2 , R 3 , R 4 , X 1 , X 2 , X 3 , X 4 , X 5 , Y 1 , Y 2 , Y 3 , and Y 4 are independently selected from the group consisting of H, deuterium, Ci-C 1 0 alkyl, Ci-Cio deuterated alkyl, substituted Q- Cio alkyl, Ci Qo alkenyl, substituted Ci Qo alkenyl, Ci Qo thioalkyl, Q Qo alkoxy, substituted Q-Qo alkoxy, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxyl, C t Ci 0 alkoxyalkyl, substituted Ci Ci 0 alkoxyalkyl, Ci-Ci
  • X 6 and X 7 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkylenylaryl, alkylenylheteroaryl, alkylenylheterocycloalkyl, alkylenylcycloalkyl, or X 6 and X 7 together form an optionally substituted 5 or 6 membered heterocyclic ring; and
  • Ar is pyndmyl or phenyl; and Z is O or S.
  • R 1 , R 2 , R 3 , R 4 , X 1 , X 2 , X 3 , X 4 , X 5 , Y 1 , Y 2 , Y 3 , and Y 4 are independently selected from the group consisting of H, deuterium, optionally substituted C 1 -C 10 alkyl, optionally substituted C 1 -C 10 deuterated alkyl, optionally substituted C 1 -C 10 alkenyl, optionally substituted C 1 -C 10 thioalkyl, optionally substituted C 1 -C 10 alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted heteroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted amido, optionally substituted sulfonyl, optionally substituted ammo, optionally substituted sulfonamide, optionally substituted sulfoxyl, cyano, nitro
  • X 6 and X 7 are independently selected from the group consisting of hydrogen, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylenylaryl, optionally substituted alkylenylheteroaryl, optionally substituted alkylenylheterocycloalkyl, optionally substituted alkylenylcycloalkyl, or X 6 and X 7 together form an optionally substituted 5 or 6 membered heterocyclic ring, and
  • Ar is optionally substituted pyndmyl or optionally substituted phenyl, and Z is O or S.
  • said therapeutic is pirfenidone or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof
  • the therapeutic administered to said patient comprises a compound of formula (II)
  • X 3 is H, OH, or C 1 -C 10 alkoxy
  • Zy is O
  • R 4 is H or hydroxyl, or a salt, ester, solvate, or prodrug thereof
  • the therapeutic administered to said patient is selected from the group consisting of , , a compound as listed in Table 1 , and pharmaceutically acceptable salts, esters, solvates, and prodrugs thereof.
  • the patient is human.
  • Figure 1 shows that the pirfenidone group (dotted line) had significantly less decline in its ejection fraction, decreasing by only 8% from week 1 to week 5.
  • the ejection fraction for controls decreased by 24% (solid line).
  • the pirfenidone group had a higher ejection fraction of 45% at 5 weeks compared to controls with a mean ejection fraction of 36%, despite the fact that the pirfemdone-treated rats had originally been randomized to a lower ejection fraction at 1 week (54% versus 60%)
  • Figure 2 depicts the conduction velocities for the normal, border, and infarct zones of both groups at various pacing cycle lengths, with pirfenidone in the circles and controls as squares.
  • Conduction velocities in the non-mfarct zones of both control and pirfenidone groups were fastest among all three zones and were similar between the two groups
  • Conduction velocities in the infarct zones of both control and pirfenidone groups were slowest among all three zones and were similar between the two groups.
  • conduction velocities in the border zones of both groups were in between those of the non-mfarct and infarct zones.
  • the conduction velocities in the border zone for the pirfemdone- treated group was significantly faster, at all pacing cycle lengths, compared to those in the border zone of control annuals.
  • Figure 3 shows a trend toward lower conduction heterogeneity for pirfemdone- treated rats (circles), compared to control rats (squares).
  • Figure 4 shows that, in terms of other electrophysiological parameters, the rise time correlates with conduction velocity.
  • An infarct is shown here to increase the time it takes to fully depolarize for both control (squares) and pirfemdone-treated (circles) rats, with the rise time being slower in the infarct zones compared to their respective normal areas.
  • the rise times in the border zones are in between the infarct and normal zones.
  • the rise time is shown to be shorter for the border zones of pirfemdone-treated rats, consistent with the faster conduction velocities in pirfenidone-treated rats.
  • Figure 5 depicts fluorescence amplitude for the three zones.
  • Figure 6 depicts the myocardial infarct size and amount of myocardial fibrosis in control versus pirfemdone-treated rats.
  • Figure 7 shows the largest measured frequency gradient over the distance that the gradient occurs for each mapped surface
  • the dark solid bars represent Control, hatched bars - congestive heart failure (CHF), and open bars - pirfenidone (PFD)
  • Figure 8 shows summary correlation coefficient (XC) data for VF activation patterns.
  • Panel A - average XC values for each mapped surface for each group. The dark solid bars represent Control, hatched bars - CHF, and open bars - PFD.
  • Panel B - average XC values for each VF activation patterns for all groups
  • Panel C coefficient of variance of the XC values for each VF activation patterns for all groups.
  • Pirfenidone is an orally active, anti-fibrotic agent. It is demonstrated herein that pirfenidone exhibits specific and potent attenuation of post-MI fibrosis, and ameliorates the arrhythmogenic potential of cardiac remodeling.
  • Pirfenidone is a small drug molecule whose chemical name is 5-mefhyl-1-phenyl-2- (1H)-pyridone. It is a non-peptide synthetic molecule with a molecular weight of 185.23 daltons. Its chemical elements are expressed as Ci 2 H 11 NO, and its structure and synthesis are known.
  • INDs pirfenidone Investigational New Drug Applications
  • Pirfenidone is being investigated for therapeutic benefits to patients suffering from fibrosis conditions such as Hermansky-Pudlak Syndrome (HPS), associated pulmonary fibrosis and idiopathic pulmonary fibrosis (IPF) Pirfenidone is also being investigated for a pharmacologic ability to prevent or remove excessive scar tissue found in fibrosis associated with injured tissues including that of lungs, skin, joints, kidneys, prostate glands, and livers.
  • HPS Hermansky-Pudlak Syndrome
  • IPF idiopathic pulmonary fibrosis
  • Pirfenidone has been reported to inhibit excessive biosynthesis or release of various cytokines such as TNF- ⁇ , TGF- ⁇ l, bFGF, PDGF, and EGF (Zhang S et al , Australian and New England J Ophthalmology 26 S74-S76 (1998); Cain et al , Int'l J Immunopharmacology 20:685-695 (1998)). Pirfenidone has also been reported to decrease collagen expression and to alter the balance of matrix metalloproteinases (MMPs) and their endogenous inhibitors (tissue inhibitor of metalloproteinases or TIMPs).
  • MMPs matrix metalloproteinases
  • TIMPs tissue inhibitor of metalloproteinases
  • AMI Acute Myocardial Infarction
  • methods are provided for treating a patient who has suffered an acute myocardial infarction (AMI) comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect.
  • AMI acute myocardial infarction
  • a method is provided for treating a condition caused by ventricular remodeling, wherein the ventricular remodeling is caused by an AMI.
  • the ventricular remodeling is fibrosis.
  • a method is provided for reducing ventricular remodeling (e g , ventricular fibrosis) in a patient who has suffered an AMI.
  • the ventricular remodeling (e.g., ventricular fibrosis) is reduced relative to an amount of ventricular remodeling (e.g., an amount of ventricular fibrosis) in the absence of administration of the therapeutic (e.g., in comparison to a patient who was not administered the therapeutic)
  • AMI Acute myocardial infarction
  • infarction damage or death
  • Coronary arterial occlusion due to thrombosis is the cause of most cases of AMI. This blockage restricts the blood supply to the muscle walls of the heart and is often accompanied by symptoms such as chest pain, heavy pressure in the chest, nausea, and shortness of breath, or shooting pain in the left arm.
  • AMI In an acute MI, severe restriction of blood flow in the coronary conduit vessels leads to reduced oxygen delivery to the myocardium and a subsequent cascade of inflammatory reactions resulting in death (infarction) of myocardial tissue Rapid restoration of blood flow to jeopardized myocardium can limit necrosis and reduce mortality.
  • AMI leads to rapid death of myocytes and vascular structures in the supplied region of the ventricle. The loss of myocytes, arterioles, and capillaries in the mfarcted area is irreversible, resulting with time in the formation of scarred tissue.
  • Patients with AMI can be diagnosed by characteristically elevated levels of troponin, creatine kinase and myoglobin Troponin levels are now considered the criterion standard in defining and diagnosing MI, according to the American College of Cardiology (ACC)/Amencan Heart Association (AHA) consensus statement on MI.
  • Cardiac troponin levels (troponin-T and troponm-I) have a greater sensitivity and specificity than myocardial muscle creatine kinase (CK MB) levels in detecting MI They have important diagnostic and prognostic roles.
  • Positive troponin levels are considered diagnostic of MI in the most recent ACC/ AHA revisions, because of their combined specificity and sensitivity in this diagnosis Serum levels typically increase within 3 12 hours from the onset of chest pain, peak at 24 48 hours, and return to baseline over 5-14 days.
  • Creatine kinase comprises 3 isoenzymes, including creatine kinase with muscle subunits (CK-MM), which is found mainly in skeletal muscle, creatine kinase with brain subunits (CK-BB), predominantly found in the brain, and myocardial muscle creatine kinase (CK-MB), which is found mainly in the heart.
  • CK-MM creatine kinase with muscle subunits
  • CK-BB creatine kinase with brain subunits
  • CK-MB myocardial muscle creatine kinase
  • Urine myoglobin levels rise within 1-4 hours from the onset of chest pain in AMI. Myoglobin levels are highly sensitive but not specific, and they may be useful within the context of other studies and in early detection of MI in the ED.
  • the electrocardiogram can be an important tool in the initial evaluation and triage of patients in whom an MI is suspected It is confirmatory of the diagnosis in approximately 80% of cases It is recommended to obtain an ECG immediately if MI is considered or suspected. In patients with infenor MI, a right-sided ECG is recorded to rule out right ventricular (RV) infarct.
  • RV right ventricular
  • Convex ST- segment elevation with upright or inverted T waves is generally indicative of MI in the appropriate clinical setting ST depression and T- wave changes may also indicate evolution of MI (non-ST-elevated MI). Progression of MI can be evaluated by performing ECGs serially, e.g. daily serial ECGs for the first 2-3 days and additionally as needed
  • Imaging studies can be helpful for diagnosis of MI, particularly if the diagnosis is questionable.
  • An echocardiogram can identify regional wall motion abnormalities indicating tissue damage or death.
  • An echocardiogram can also define the extent of the infarction and assess overall left vent ⁇ cle (LV) and right ventricle (RV) function.
  • an echocardiogram can identify complications, such as acute mitral regurgitation (MR), LV rupture, or pericardial effusion.
  • Myocardial perfusion imaging utilizes an intravenously administered radiopharmaceutical to depict the distribution of blood flow in the myocardium
  • the radiopharmaceutical distribution in the heart is imaged using a gamma camera
  • Perfusion abnormalities, or defects, are assessed and quantified as to location, extent and intensity.
  • Myocardial perfusion imaging can identify areas of reduced myocardial blood flow associated with infarct.
  • Cardiac catheterization defines the patient's coronary anatomy and the extent of the blockage(s) via cardiac angiography.
  • AMI may be distinguished from chronic myocardial infarction using any appropriate method known in the art.
  • the presence of myocardial edema involving a disruption of the energy-regulated ionic transport mechanisms across the cell membrane after the MI is indicative of AMI (Willerson et al , 1977, Am J Pathol 87:159— 188)
  • the relatively large extracellular matrix of the developed scar allows gadolmium-based contrast media to accumulate, resulting in DE.
  • T2-weighted CMR sensitively detects lnfarct- associated myocardial edema (Wisenberg et al., 1988, Am Heart J.
  • a combination of delayed enhancement (DE) and T2-weighted cardiovascular magnetic resonance (CMR) is used to differentiate acute from chronic MI (Abdel-Aty et al. , 2004, Circulation 109 2411-2416)
  • CHF Congestive Heart Failure
  • methods wherein the incidence of congestive heart failure (CHF) or complications of CHF are reduced when a therapeutic having an anti fibrotic effect is administered to said patient.
  • CHF or complications of CHF are reduced relative to the incidence of CHF or complications of CHF in the absence of administration of the therapeutic (e g , m comparison to a patient who was not administered the therapeutic)
  • the incidence of CHF may be reduced by at least 10% when a therapeutic having an anti-fibrotic effect is administered to a patient in comparison to a patient who was not administered the therapeutic
  • the incidence of CHF may be reduced by at least 15%, or at least 20%, or at least 25%, or at least 30%, or at least 35%, or at least 40%, or at least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95% or more when a therapeutic having an anti-fibrotic effect
  • CHF may be a complication of AMI and results from a decline in the pumping capacity of the heart CHF can also result from cardiac malformations, such as valve disease, or other disorders that damage cardiac tissue, e.g cardiac myopathy. Due to the activation of one or more compensatory mechanisms, the damaging changes caused by CHF can be present and ongoing even while the patient remains asymptomatic In fact, the compensatory mechanisms which maintain normal cardiovascular function du ⁇ ng the early phases of CHF may actually contribute to progression of the disease, for example by exerting deleterious effects on the heart and circulation [0068] Some of the more important pathophysiologic changes which occur in CHF are activation of the hypothalamic -pituitary- adrenal axis, systemic endothelial dysfunction and myocardial remodeling.
  • Therapies specifically directed at counteracting the activation of the hypothalamic- pituitary-adrenal axis include beta-adrenergic blocking agents ( ⁇ -blockers), angiotensin converting enzyme (ACE) inhibitors, certain calcium channel blockers, nitrates and endothelm-1 blocking agents.
  • ⁇ -blockers beta-adrenergic blocking agents
  • ACE angiotensin converting enzyme
  • calcium channel blockers calcium channel blockers
  • nitrates calcium channel blockers
  • endothelm-1 blocking agents include calcium channel blockers and nitrates, while producing clinical improvement have not been clearly shown to prolong survival whereas ⁇ -blockers and ACE inhibitors have been shown to significantly prolong life, as have aldosterone antagonists.
  • Systemic endothelial dysfunction is a well-recognized feature of CHF and is clearly present by the time signs of left ventricular dysfunction are present. Endothelial dysfunction is important with respect to the intimate relationship of the myocardial microcirculation with cardiac myocytes. The evidence suggests that microvascular dysfunction contributes significantly to myocyte dysfunction and the morphological changes which lead to progressive myocardial failure.
  • Myocardial remodeling is a complex process which accompanies the transition from asymptomatic to symptomatic heart failure, and may be described as a series of adaptive changes within the myocardium.
  • Components of myocardial remodeling may include fibrosis, alterations in myocyte biology, loss of myocytes by necrosis or apoptosis, alterations in the extracellular matrix and alterations in left ventricular chamber geometry.
  • Symptoms include dyspnea (shortness of breath) which worsens upon lying supine, fluid retention and swelling in the lungs and extremities, e.g. with pulmonary rales or edema in the legs.
  • Electrocardiogram may show anterior Q waves or left bundle branch block on the electrocardiogram.
  • the echocardiogram is the diagnostic standard for identifying congestive heart failure
  • the patient may undergo two-dimensional echocardiography with Doppler flow studies. Radionuclide angiography or contrast cineangiography may be helpful if the echocardiogram is equivocal.
  • methods wherein the cardiac tissue is preserved from necrosis when a therapeutic having an anti-fibrotic effect is administered to a patient suffering an AMI, in comparison to the amount of viable cardiac tissue in the absence of administration of the therapeutic (e.g., in comparison to a patient who was not administered a therapeutic).
  • the amount of cardiac tissue preserved from necrosis can be increased at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
  • the increase in viable cardiac tissue can be determined by MRI or computerized tomography (CT) scan.
  • Control means to reduce, reduce the incidence of, or prevent the progression of a disorder.
  • methods are provided wherein the infarct size of a patient is reduced when a therapeutic is administered to said patient, in comparison to the infarct size of a patient in the absence of administration of the therapeutic (e.g., in comparison to a patient who was not administered a therapeutic).
  • the infarct size can be reduced at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
  • the reduction in infarct size can be determined by MRI and/or by voltage/conduction mapping.
  • cardiac function is preserved when a therapeutic having an anti-fibrotic effect is administered to a patient suffering an AMI, in comparison to the cardiac function of a patient suffering an AMI in the absence of administration of the therapeutic (e.g., in comparison to a patient who was not administered a therapeutic).
  • Preservation of cardiac function can be determined by measuring ejection fraction using echocardiography, wherein the ejection fraction can be improved by at least 1%, at least 3%, at least 5%, at least 7%, at least 10%, at least 12%, or at least 15%.
  • Preservation of cardiac tissue can also be determined by measuring ejection fraction using MRI, wherein the ejection fraction can be improved by at least 1%, at least 3%, at least 5%, at least 7%, at least 10%, at least 12%, or at least 15%, and/or the infarct size can be decreased by at least 1%, at least 3%, at least 5%, at least 7%, at least 10%, at least 12% or at least 15%.
  • Other methods of determining cardiac function are known in the art and include but are not limited to nuclear imaging, functional capacity, exercise capacity, New York Heart Association (NYHA) functional classification system, and myocardial oxygen consumption (MV02)
  • ventricular tachycardia in a patient is reduced when a therapeutic is administered to said patient, in compa ⁇ son to the incidence of ventricular tachycardia in a patient who was not administered the therapeutic
  • the incidence of ventricular tachycardia can be reduced at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80% at least 85%, at least 90%, or at least 95%
  • the reduction in incidence of tachycardia can be determined by electrocardiogram (ECG or EKG) or by echocardiogram
  • methods for treating or preventing vent ⁇ cular fibrillation in a patient in need thereof, comprising administering to the patient a therapeutic having an anti fibrotic effect
  • the amount or degree of ventricular fibrillation is reduced relative to the amount or degree of ventricular fibrillation in the absence of administration of the therapeutic
  • Vent ⁇ cular fibrillation is a condition in which the heart s electrical activity becomes disordered When this happens, the heart s vent ⁇ cles contract in a rapid, unsynchronized way The ventricles “quiver” rather than beat, causing the heart to pump little or no blood
  • VF is life threatening and requires prompt treatment Without medical treatment, collapse and sudden cardiac death can occur Ventricular fibrillation (VF) may occur spontaneously with unpredictable timing and requires specialized tests to acquire an accurate diagnosis.
  • VF Ventricular fibrillation
  • VF may be diagnosed using an electrocardiogram (ECG or EKG), e g a Holter Monitor --
  • ECG electrocardiogram
  • Holter monitor is a small, portable machine that records the patient's ECG and is typically worn for 24 hours This monitor may detect arrhythmias that might not show up on a resting electrocardiogram, which only records a heartbeat for a few seconds at rest
  • VF may also be diagnosed using an event monitor This is a small monitor about the size of a pager that the patient can have for up to a month. Since the arrhythmia may occur at unpredictable times, this monitor records the abnormal rhythm when the patient signals that he or she is experiencing symptoms
  • An exercise stress or treadmill test also may be used to diagnose VF, by recording the electrical activity of the patient's heart during exercise, which differs from the heart's electrical activity at rest.
  • Another method of diagnosing VF is through an electrophysiology study
  • physiciansinsert special electrode catheters long, flexible wires — into veins and guide them into the heart These catheters sense electrical impulses and also may be used to stimulate different areas of the heart Physicians can then locate the sites that are causing arrhythmias.
  • the EP study allows physicians to examine an arrhythmia under controlled conditions and acquire more accurate, detailed information than with any other diagnostic test.
  • VF can be monitored and measured by any one or more of the parameters described, for example, in Example 5 below.
  • the incidence of VF can be reduced by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%, compared to incidence of VF in a patient who was not administered the therapeutic.
  • Sudden cardiac death also called sudden arrest
  • cardiac arrest The victim may or may not have diagnosed heart disease.
  • the time and mode of death are unexpected It occurs within minutes after symptoms appear
  • AMI due to coronary heart disease
  • Other types of arrhythmia can also cause cardiac arrest.
  • cardiac arrests that lead to sudden death occur when the electrical impulses in the diseased heart become rapid (ventricular tachycardia) or chaotic (ventricular fibrillation) or both. This irregular heart rhythm (arrhythmia) causes the heart to suddenly stop beating. Some cardiac arrests are due to extreme slowing of the heart, bradycardia If a cardiac arrest was due to ventricular tachycardia or ventricular fibrillation, survivors are at higher risk for another arrest, especially if they have underlying heart disease.
  • the incidence of sudden cardiac death is reduced when a therapeutic having an anti-fibrotic effect is administered to said patient, m comparison to the incidence of cardiac death m a patient who was not administered a therapeutic.
  • the incidence of sudden cardiac death can be reduced at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
  • Methods of the invention are contemplated to control arrhythmia by administering a therapeutic having an anti-fibrotic effect
  • a method is provided to reduce the incidence or risk of arrhythmia.
  • the incidence or risk can be reduced at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
  • An arrhythmia is an abnormal heart rhythm. In an arrhythmia the heartbeats may be too slow, too rapid, too irregular, or too early. There are many types of arrhythmias, including premature atrial contractions (early extra beats that originate in the atria (upper chambers of the heart), premature ventricular contractions (PVCs) (skipped heartbeat), atrial fibrillation (an irregular heart rhythm that causes the atria, the upper chambers of the heart to contract abnormally), atrial flutter (an arrhythmia caused by one or more rapid circuits in the atrium), paroxysmal supraventricular tachycardia (PSVT) (a rapid heart rate, usually with a regular rhythm, originating from above the ventricles), accessory pathway tachycardias (a rapid heart rate due to an extra abnormal pathway or connection between the atria and the ventricles), AV nodal reentrant tachycardia (a rapid heart rate due to more than one pathway through the AV node),
  • Symptoms of arrhythmia include chest pain, fainting, fast or slow heartbeat (palpitations), light-headedness, dizziness, paleness, shortness of breath, skipping beats, changes in the pattern of the pulse,and sweating.
  • Arrythmias may be diagnosed by those of skill in the art using such methods as electrocardiogram, Holter monitor, event monitor, stress test, echocardiogram, cardiac catheterization, electrophysiology study (EPS), and head-up tilt table test
  • the amount of a therapeutic effective to control arrhythmia may be an amount effective to reduce vent ⁇ cular remodeling, e.g. in an animal model or during clinical trial.
  • Ventricular remodeling refers to the changes in size, shape, and function of the heart after injury to the left ventricle The injury is typically due to AMI.
  • the ventricular remodeling is due to ventricular fibrosis caused by an AMI.
  • the remodeling process is characterized by progressive expansion of the initial infarct area and dilation of the left ventricular lumen, with cardiomyocyte replacement by fibrous tissue deposition in the ventricular wall (Kocher et al., 2001, Nature Medicine 7(4): 430-6).
  • neoangiogenesis Another integral component of the remodeling process is the development of neoangiogenesis within the myocardial infarct scar, a process requiring activation of latent collagenase and other proteinases.
  • neoangiogenesis is insufficient to keep pace with the tissue growth required for contractile compensation and is unable to support the greater demands of the hypertrophied but viable myocardium.
  • Therapeutic agents used in the disclosed methods can be any therapeutic agent that affects fibrosis Contemplated agents include agents that reduce the activity of transforming growth factor-beta (TGF- ⁇ ) (including but not limited to GC-1008 (Genzyme/Medlmmune); lerdelimumab (CAT- 152; Trabio, Cambridge Antibody); metelimumab(CAT-192,Cambridge Antibody,); LY-2157299 (Eh Lilly); ACU-HTR-028 (Opko Health)) including antibodies that target one or more TGF- ⁇ isoforms, inhibitors of TGF- ⁇ receptor kinases TGFBRl (ALK5) and TGFB R2, and modulators of post-receptor signaling pathways; chemokme receptor signaling; endothelin receptor antagonists including inhibitors that target both endothehn receptor A and B and those that selectively target endothelin receptor A (including but not limited to ambrisentan; avosentan, bosentan,
  • prolyl hydrolase inhibitors including but not limited to 1016548, CG-0089, FG-2216, FG- 4497, FG-5615, FG-6513, fibrostatin A (Takeda), lufironil,P-1894B, and safironil
  • PPAR peroxisome proliferator- activated receptor
  • formula (I), (II), (III), (IV), or (V) defined above are examples of formula (I), (II), (III), (IV), or (V) defined above.
  • R 1 , R 2 , R 3 , R 4 , X 1 , X 2 , X 3 , X 4 , X 5 , Y 1 , Y 2 , Y 3 , and Y 4 are independently selected from the group consisting of H, deuterium, C 1 -C 10 alkyl, C 1 -C 10 deuterated alkyl, substituted Ci- Cio alkyl, C 1 -C 10 alkenyl, substituted C 1 -C 10 alkenyl, C 1 -C 10 thioalkyl, C 1 -C 10 alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxyl, Ci-Cio alkoxyalkyl, Ci-Cio carboxy, Ci
  • X 6 and X 7 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkylenylaryl, alkylenylheteroaryl, alkylenylheterocycloalkyl, alkylenylcycloalkyl, or X 6 and X 7 together form an optionally substituted 5 or 6 membered heterocyclic ring; and Ar is pyridinyl or phenyl; and Z is O or S; or a pharmaceutically acceptable salt, ester, solvate, or prodrug of pirfenidone or the compound of formula (I), (II), (III), (IV), or (V).
  • R 1 , R 2 , R 3 , R 4 , X 1 , X 2 , X 3 , X 4 , X 5 , Y 1 , Y 2 , Y 3 , and Y 4 are independently optionally substituted pyrazinyl, optionally substituted pyridazinyl, optionally substituted pyrrolyl, optionally substituted thiophenyl, optionally substituted thiazolyl, optionally substituted oxazolyl, optionally substituted imidazolyl, optionally substituted isoxazolyl, optionally substituted pyrazolyl, optionally substituted isothiazolyl, optionally substituted napthyl, optionally substituted quinolinyl, optionally substituted isoquinolinyl, optionally substituted quinoxalinyl, optionally substituted benzothiazolyl, optionally substituted benzothiophenyl, optionally substituted benzofuranyl, optional
  • Other specific therapeutic agents contemplated include relaxin, ufironil, surifonil, a TGF- ⁇ antibody, CAT-192, CAT-158; ambresentan, thelin; FG-3019, a CTGF antibody; anti- EGFR antibody;a EGFR kinase inhibitor; tarceva; gefitinib; PDGF antibody, PDGFR kinase inhibitor; gleevec; BIBF-1120, VEGF, FGF, and PDGF receptor inhibitor; anti-integ ⁇ n antibody; IL-4 antibody; tetrathiomolybdate, a copper chelating agent; interferon-gamma; NAC, a cysteine pro-drug; hepatocyte growth factor (HGF); KGF; angiotension receptor blockers, ACE inhibitors, rennin inhibitors; COX and LO inhibitors; Zileuton; monteleukast; avastin; statins; PDE5 inhibitors
  • alkyl refers to a saturated or unsaturated straight or branched chain hydrocarbon group of one to ten carbon atoms, including, but not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-hexyl, and the like.
  • alkyls of one to six carbon atoms are also contemplated
  • the term "alkyl” includes "bridged alkyl," i e , a bicyclic or polycyclic hydrocarbon group, for example, norbornyl, adamantyl, bicyclo[2.2.2]octyl, bicyclo[2.2.1]heptyl, bicyclo[3.2.1]octyl, or decahydronaphthyl.
  • Alkyl groups optionally can be substituted, for example, with hydroxy (OH), halo, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, and ammo.
  • the alkyl group consists of 1-40 carbon atoms, preferably 1-25 carbon atoms, preferably 1-15 carbon atoms, preferably 1-12 carbon atoms, preferably 1-10 carbon atoms, preferably 1-8 carbon atoms, and preferably 1-6 carbon atoms.
  • “Heteroalkyl” is defined similarly as alkyl, except the heteroalkyl contains at least one heteroatom independently selected from the group consisting of oxygen, nitrogen, and sulfur.
  • cycloalkyl refers to a cyclic hydrocarbon group, e.g., cyclopropyl, cyclobutyl, cyclohexyl, and cyclopentyl.
  • Heterocycloalkyl is defined similarly as cycloalkyl, except the ring contains one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur.
  • Nonlimiting examples of heterocycloalkyl groups include piperdine, tetrahydrofuran, tetrahydropyran, dihydrofuran, morpholine, thiophene, and the like.
  • Heterocycloalkyl groups optionally can be further N-substituted with alkyl, hydroxyalkyl, alkylenearyl, or alkyleneheteroaryl.
  • alkenyl refers to a straight or branched chain hydrocarbon group of two to ten carbon atoms containing at least one carbon double bond including, but not limited to, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl, and the like
  • alkylene used herein refers to an alkyl group having a substituent.
  • alkylene aryl refers to an alkyl group substituted with an aryl group.
  • the alkylene group is optionally substituted with one or more substituent previously listed as an optional alkyl substituent.
  • an alkylene group can be -CH2CH2-.
  • alkenylene is defined identical as “alkylene,” except the group contains at least one carbon-carbon double bond.
  • aryl refers to a monocyclic or polycyclic aromatic group, preferably a monocyclic or bicyclic aromatic group, e.g., phenyl or naphthyl. Unless otherwise indicated, an aryl group can be unsubstituted or substituted with one or more, and in particular one to four groups independently selected from, for example, halo, alkyl, alkenyl, OCF3, NO2, CN, NC, OH, alkoxy, amino, CO2H, CO2alkyl, aryl, and heteroaryl.
  • aryl groups include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl, chlorophenyl, methylphenyl, methoxyphenyl, trifluoromethylphenyl, nitrophenyl, 2,4- methoxychlorophenyl, and the like.
  • heteroaryl refers to a monocyclic or bicyclic ring system containing one or two aromatic rings and containing at least one nitrogen, oxygen, or sulfur atom in an aromatic ring. Unless otherwise indicated, a heteroaryl group can be unsubstituted or substituted with one or more, and in particular one to four, substituents selected from, for example, halo, alkyl, alkenyl, OCF 3 , NO 2 , CN, NC, OH, alkoxy, amino, CO 2 H, CO 2 alkyl, aryl, and heteroaryl.
  • heteroaryl groups include, but are not limited to, thienyl, furyl, pyridyl, oxazolyl, quinolyl, thiophenyl, isoquinolyl, indolyl, triazinyl, triazolyl, isothiazolyl, isoxazolyl, imidazolyl, benzothiazolyl, pyrazinyl, pyrimidinyl, thiazolyl, and thiadiazolyl.
  • deuterated alkyl refers to an alkyl group substituted with one or more deuterium atoms (D).
  • thioalkyl used herein refers to one or more thio groups appended to an alkyl group.
  • hydroxyalkyl refers to one or more hydroxy groups appended to an alkyl group.
  • alkoxy refers to straight or branched chain alkyl group covalently bonded to the parent molecule through an --O— linkage. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, n- butoxy, sec-butoxy, t-butoxy and the like. [0111] The term “alkoxyalkyl” used herein refers to one or more alkoxy groups appended to an alkyl group.
  • arylalkoxy refers to a group having an aryl appended to an alkoxy group.
  • a non-limiting example of an arylalkoxy group is a benzyloxy (Ph-CH 2 -O-).
  • amino refers to -NR 2 , where R is independently hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl.
  • R is independently hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl.
  • Non-limiting examples of amino groups include NH 2 and N(CH 3 ) 2 .
  • R is independently hydrogen or alkyl.
  • amido refers to -C(O)NH 2 , -C(O)NR 2 , -NRC(O)R or - NHC(O)H, where each R is independently hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl.
  • the amido group is -NHC(O)alkyl or -NHC(O)H.
  • a non-limiting example of an amido group is -NHC(O)CH 3 .
  • carboxy refers to — COOH or its deprotonated form -COO .
  • C 1-10 carboxy refers to optionally substituted alkyl or alkenyl groups having a carboxy moiety. Examples include, but are not limited to, -CH 2 COOH, - CH 2 CH(COOH)CH 3 , and -CH 2 CH 2 CH 2 COOH.
  • alkoxycarbonyl refers to — (CO) — O-alkyl, wherein the alkyl group can optionally be substituted
  • alkoxycarbonyl groups include, but are not limited to, methoxycarbonyl group, ethoxycarbonyl group, propoxycarbonyl group, and the like.
  • alkylcarbonyl refers to — (CO)-alkyl, wherein the alkyl group can optionally be substituted.
  • alkylcarbonyl groups include, but are not limited to, methylcarbonyl group, ethylcarbonyl group, propylcarbonyl group, and the like.
  • sulfonamido refers to— SO 2 NR 2 , wherein R is independently hydrogen, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl.
  • the sulfonamido group is -SO 2 NR 2 where R is independently hydrogen or an optionally substituted alkyl. Examples of a sulfonamido group include, but are not limited to, -SO 2 N(CH 3 ) 2 and -SO 2 NH 2 .
  • sulfonyl refers to -SO 2 R, where R is independently hydrogen or an optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl
  • R is independently hydrogen or an optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl
  • a sulfonyl group is -SO 2 alkyl, wherein the alkyl group can optionally be substituted
  • a sulfonyl group is methylsulfonyl (e.g., - SO 2 CH 3 )
  • sulfoxyl refers to -SOR, where each R is independently hydrogen or an optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl
  • R is independently hydrogen or an optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl
  • a sulfonyl group is methylsulfonyl (e g , SOCH 3 )
  • Carbohydrates are polyhydroxy aldehydes or ketones, or substances that yield such compounds upon hydrolysis.
  • Carbohydrates comprise the elements carbon (C), hydrogen (H) and oxygen (O) with a ratio of hydrogen twice that of carbon and oxygen.
  • carbohydrates are simple sugars or monosaccharides These simple sugars can combine with each other to form more complex carbohydrates
  • the combination of two simple sugars is a disaccharide
  • Carbohydrates consisting of two to ten simple sugars are called oligosaccharides, and those with a larger number are called polysaccharides
  • uronide refers to a monosaccharide having a carboxyl group on the carbon that is not part of the ring
  • the uronide name retains the root of the monosaccharide, but the -ose sugar suffix is changed to -uronide.
  • the structure of glucuronide corresponds to glucose.
  • a radical indicates species with a single, unpaired electron such that the species containing the radical can be covalently bonded to another species Hence, in this context, a radical is not necessarily a free radical Rather, a radical indicates a specific portion of a larger molecule.
  • the term "radical” can be used interchangeably with the term "group.”
  • substituted group is derived from the unsubstituted parent structure in which there has been an exchange of one or more hydrogen atoms for another atom or group
  • substituted group means a group selected from the following moieties •
  • the substituent group is a "size-limited substituent” or “size-limited substituent group,” which refers to a group selected from all of the substituents described above for a "substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C 1 -C 20 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C 4 -C 8 cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 4 to 8 membered heterocycloalkyl
  • the substituent group is a "lower substituent” or “lower substituent group,” which refers to a group selected from all of the substituents descnbed above for a "substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C 1 -C 8 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C 5 -C 7 cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 5 to 7 membered heterocycloalkyl
  • the substituent group(s) is (are) one or more group(s) individually and independently selected from alkyl, cycloalkyl, aryl, fused aryl, heterocyclyl, heteroaryl, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, alkoxycarbonyl, nitro, silyl, tnhalomethanesulfonyl, trifluoromethyl, and amino, including mono and di substituted amino groups, and the protected derivatives thereof.
  • Asymmetric carbon atoms can be present. All such isomers, including diastereomers and enantiomers, as well as the mixtures thereof, are intended to be included in the scope of the disclosure herein. In certain cases, compounds can exist in tautomeric forms. All tautomeric forms are intended to be included in the scope of the disclosure herein. Likewise, when compounds contain an alkenyl or alkenylene group, there exists the possibility of cis- and trans- isomeric forms of the compounds. Both cis- and trans- isomers, as well as the mixtures of cis- and trans- isomers, are contemplated.
  • One class of compounds contemplated for use in the disclosed methods is a deuterated (D) form of any of the compounds disclosed herein.
  • One specific such compound is a compound having a CD 3 moiety and/or a D to replace any or all of the methyl or hydrogens of pirfenidone. Examples include , and .
  • the synthesis of these compounds can be found in International Patent Publication No. WO 08/157786.
  • each of R , R 2 , R 3 , R 4 , and R 6 is independently selected from the group consisting of H, halo, cyano, nitro, hydroxy, optionally substituted Ci 6 alkyl, optionally substituted C 3 7 cycloalkyl, optionally substituted C 4 1 0 alkylcycloalkyl, optionally substituted C 2 6 alkenyl, optionally substituted Ci 6 alkoxy, optionally substituted C ⁇ o r 1 0 aryl > optionally substituted py ⁇ dinyl, optionally substituted py ⁇ midmyl, optionally substituted thienyl, optionally substituted furanyl, optionally substituted thiazolyl, optionally substituted oxazolyl, optionally substituted phenoxy, optionally substituted thiophenoxy, optionally substituted sulphonamido, optionally substituted urea, optionally substituted thiourea, optionally substituted amido, optionally substituted keto, optionally substituted keto
  • the salts, e.g., pharmaceutically acceptable salts, of the disclosed therapeutics may be prepared by reacting the appropriate base or acid with a stoichiometric equivalent of the therapeutic.
  • pharmaceutically acceptable derivatives e.g., esters
  • metabolites, hydrates, solvates and prodrugs of the therapeutic may be prepared by methods generally known to those skilled in the art
  • another embodiment provides compounds that are prodrugs of an active compound.
  • a prodrug is a compound which is metabolized in vivo (e.g., by a metabolic transformation such as deamination, dealkylation, de- esterification, and the like) to provide an active compound
  • a "pharmaceutically acceptable prodrug” means a compound which is, withm the scope of sound medical judgment suitable for pharmaceutical use in a patient without undue toxicity, irritation, allergic response, and the like, and effective for the intended use, including a pharmaceutically acceptable ester as well as a zwitteriomc form, where possible, of the therapeutic
  • the term “pharmaceutically acceptable ester” refers to esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkeny
  • the compounds and compositions described herein may also include metabolites
  • the term "metabolite” means a product of metabolism of a compound of the embodiments or a pharmaceutically acceptable salt, analog, or derivative thereof, that exhibits a similar activity in vitro or in vivo to a disclosed therapeutic.
  • the compounds and compositions described herein may also include hydrates and solvates.
  • the term “solvate” refers to a complex formed by a solute (herein, the therapeutic) and a solvent. Such solvents for the purpose of the embodiments preferably should not negatively interfere with the biological activity of the solute. Solvents may be, by way of example, water, ethanol, or acetic acid.
  • reference herein to a particular compound or genus of compounds will be understood to include the various forms described above, including pharmaceutically acceptable salts, esters, prodrugs, metabolites and solvates thereof.
  • therapeutically effective amount and “prophylactic ally effective amount,” as used herein, refer to an amount of a compound sufficient to treat, ameliorate, or prevent the identified disease or condition, or to exhibit a detectable therapeutic, prophylactic, or inhibitory effect The effect can be detected by, for example, an improvement in clinical condition, reduction in symptoms, or by any of the assays or clinical diagnostic tests desc ⁇ bed herein.
  • the precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration. Therapeutically and prophylactically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician.
  • the therapeutics disclosed herein can be dosed at a total amount of about 50 to about 2400 mg per day.
  • the dosage can be divided into two or three doses over the day or given in a single daily dose.
  • Specific amounts of the total daily amount of the therapeutic contemplated for the disclosed methods include about 50 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 267 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 534 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, about 1000 mg, about 1050 mg, about 1068 mg, about 1100 mg, about 1150 mg, about 1200 mg, about 1250 mg, about 1300 mg, about 1335 mg, about 1350 mg, about 1400 mg, about 1450 mg, about 1500 mg, about 1550 mg, about 1600 mg, about 1650 mg, about 1700 mg, about 1750 mg, about 1800 mg
  • Dosages of the therapeutic can alternately be administered as a dose measured in mg/kg.
  • Contemplated mg/kg doses of the disclosed therapeutics include about 1 mg/kg to about 60 mg/kg. Specific ranges of doses in mg/kg include about 1 mg/kg to about 20 mg/kg, about 5 mg/kg to about 20 mg/kg, about 10 mg/kg to about 20 mg/kg, about 25 mg/kg to about 50 mg/kg, and about 30 mg/kg to about 60 mg/kg.
  • administration of the therapeutic can be initiated at 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, 31 days, 32 days, 33 days, 34 days, 35 days, 36 days, 37 days, 38 days, 39 days, 40 days, 41 days, or 42 days after suffering the AMI.
  • initiation of the treatment about 1-40 days, about 1-30 days, about 1-25 days, about 1-20 days, about 1-14 days, about 1-10 days, about 2-40 days, about 3-40 days, about 3-38 days, about 3-30 days, about 3-25 days, about 3-20 days, about 3-15 days, about 3-14 days, about 3-10 days, about 4-36 days, about 4-30 days, about 4-25 days, about 4-20 days, about 4-14 days, about 5-40 days, about 5-34 days, about 5-30 days, about 5-25 days, about 5-20 days, about 5-14 days, about 6-40 days, about 6-32 days, about 6-30 days, about 6-25 days, about 6-20 days, about 6-14 days, about 7-40 days, about 7-30 days, about 7-25 days, about 7-20 days, about 7-14 days, about 8-28 days, about 9-26 days, about 10-24 days, about 12-22 days, about 13-20 days, or about 14-18 days after suffering the AMI.
  • Treatment e.g., continued administration of the therapeutic can continue for at least a week, at least 2 weeks, at least 3 weeks, at least a month, at least 6 weeks, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, or at least a year.
  • the treatment can be for up to 3 months, up to 4 months, up to 5 months, or up to 6 months.
  • a patient suffering an AMI continues to be administered the therapeutic for a time period up to 4 weeks after suffering the AMI, e g , the therapeutic continues to be administered on the day that is 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, and/or 28 days after suffering the AMI.
  • the compounds described herein may be formulated in pharmaceutical compositions with a pharmaceutically acceptable excipient, carrier, or diluent.
  • the compound or composition comprising the compound can be administered by any route that permits treatment of the disease or condition.
  • a preferred route of administration is oral administration.
  • the compound or composition comprising the compound may be delivered to a patient using any standard route of administration, including parenterally, such as intravenously, mtrapentoneally, mtrapulmonary, subcutaneously or intramuscularly, intrathecally, transdermally, rectally, orally, nasally or by inhalation.
  • Slow release formulations may also be prepared from the agents described herein in order to achieve a controlled release of the active agent in contact with the body fluids m the gastro intestinal tract, and to provide a substantial constant and effective level of the active agent in the blood plasma.
  • the crystal form may be embedded for this purpose m a polymer matrix of a biological degradable polymer, a water-soluble polymer or a mixture of both, and optionally suitable surfactants. Embedding can mean in this context the incorporation of micro-particles in a matrix of polymers. Controlled release formulations are also obtained through encapsulation of dispersed micro-particles or emulsified micro-droplets via known dispersion or emulsion coating technologies.
  • Administration may take the form of single dose administration, or the compound of the embodiments can be administered over a period of time, either in divided doses or in a continuous-release formulation or administration method (e g , a pump) However the compounds of the embodiments are administered to the subject, the amounts of compound administered and the route of administration chosen should be selected to permit efficacious treatment of the disease condition.
  • the pharmaceutical compositions may be formulated with pharmaceutically acceptable excipients such as carriers, solvents, stabilizers, adjuvants, diluents, etc., depending upon the particular mode of administration and dosage form.
  • the pharmaceutical compositions should generally be formulated to achieve a physiologically compatible pH, and may range from a pH of about 3 to a pH of about 11, preferably about pH 3 to about pH 7, depending on the formulation and route of administration. In alternative embodiments, it may be preferred that the pH is adjusted to a range from about pH 5 0 to about pH 8 More particularly, the pharmaceutical compositions may comprise a therapeutically or prophylactically effective amount of at least one compound as described herein, together with one or more pharmaceutically acceptable excipients.
  • the pharmaceutical compositions may comp ⁇ se a combination of the compounds described herein, or may include a second active ingredient useful in the treatment or prevention of bacterial infection (e.g., anti-bacterial or anti-microbial agents).
  • a therapeutic agent that may be used alone or in combination with another therapeutic agent according to the methods of the present invention include, but are not limited to, an agent that reduces tissue remodeling or fibrosis, reduces the activity of transforming growth factor-beta (TGF- ⁇ ), targets one or more TGF- ⁇ isoforms, inhibits TGF- ⁇ receptor kinases TGFBRl (ALK5) and/or TGFBR2, or modulates one or more post- receptor signaling pathways, is an endothelin receptor antagonists, targets both endothehn receptor A and endothelin receptor B or selectively targets endothelin receptor A, reduces activity of connective tissue growth factor (CTGF), inhibits matrix metalloproteinase (MMP), particularly MMP-9 and/or M
  • CTGF connective tissue
  • Formulations e.g., for parenteral or oral administration, are most typically solids, liquid solutions, emulsions or suspensions, while inhalable formulations for pulmonary administration are generally liquids or powders, with powder formulations being generally preferred.
  • a preferred pharmaceutical composition may also be formulated as a lyophihzed solid that is reconstituted with a physiologically compatible solvent prior to administration.
  • Alternative pharmaceutical compositions may be formulated as syrups, creams, ointments, tablets, and the like
  • pharmaceutically acceptable excipient refers to an excipient for administration of a pharmaceutical agent, such as the compounds described herein.
  • the term refers to any pharmaceutical excipient that may be administered without undue toxicity.
  • compositions are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there exists a wide variety of suitable formulations of pharmaceutical compositions (see, e.g., Remington's Pharmaceutical Sciences).
  • Suitable excipients may be carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles.
  • Other exemplary excipients include antioxidants (e g., ascorbic acid), chelating agents (e.g., EDTA), carbohydrates (e.g., dextrin, hydroxyalkylcellulose, and/or hydroxyalkylmethylcellulose), stearic acid, liquids (e g , oils, water, saline, glycerol and/or ethanol) wetting or emulsifying agents, pH buffering substances, and the like.
  • Liposomes are also included withm the definition of pharmaceutically acceptable excipients.
  • compositions described herein may be formulated in any form suitable for an intended method of administration.
  • tablets, troches, lozenges, aqueous or oil suspensions, non-aqueous solutions, dispersible powders or granules (including micronized particles or nanoparticles), emulsions, hard or soft capsules, syrups or elixirs may be prepared.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
  • compositions particularly suitable for use in conjunction with tablets include, for example, inert diluents, such as celluloses, calcium or sodium carbonate, lactose, calcium or sodium phosphate; disintegrating agents, such as cross-linked povidone, maize starch, or alginic acid; binding agents, such as povidone, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc.
  • inert diluents such as celluloses, calcium or sodium carbonate, lactose, calcium or sodium phosphate
  • disintegrating agents such as cross-linked povidone, maize starch, or alginic acid
  • binding agents such as povidone, starch, gelatin or acacia
  • lubricating agents such as magnesium stearate, stearic acid or talc.
  • Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period
  • a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • Formulations for oral use may be also presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example celluloses, lactose, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with non-aqueous or oil medium, such as glycerin, propylene glycol, polyethylene glycol, peanut oil, liquid paraffin or olive oil
  • an inert solid diluent for example celluloses, lactose, calcium phosphate or kaolin
  • non-aqueous or oil medium such as glycerin, propylene glycol, polyethylene glycol, peanut oil, liquid paraffin or olive oil
  • compositions may be formulated as suspensions comprising a compound of the embodiments in admixture with at least one pharmaceutically acceptable excipient suitable for the manufacture of a suspension.
  • compositions may be formulated as dispersible powders and granules suitable for preparation of a suspension by the addition of suitable excipients
  • Excipients suitable for use in connection with suspensions include suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelhilose, sodium alginate, polyvinylpyrrolidone, gum tragacanth, gum acacia), dispersing or wetting agents (e.g., a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g.
  • suspending agents e.g., sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelhilose, sodium alginate, polyvinylpyrrolidone, gum tragacanth, gum acacia
  • dispersing or wetting agents e.g., a naturally occurring phosphatide (e.g., lecithin), a condensation product of
  • heptadecaethyleneoxycethanol a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g. , polyoxyethylene sorbitan monooleate)); and thickening agents (e.g., carbomer, beeswax, hard paraffin or cetyl alcohol).
  • the suspensions may also contain one or more preservatives (e.g., acetic acid, methyl or n- propyl p-hydroxy-benzoate); one or more coloring agents; one or more flavoring agents, and one or more sweetening agents such as sucrose or saccharin.
  • the pharmaceutical compositions may also be in the form of oil-m water emulsions.
  • the oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth; naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids; hexitol anhydrides, such as sorbitan monooleate; and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • sweetening agents such as glycerol, sorbitol or sucrose
  • Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • the pharmaceutical compositions may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous emulsion or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous emulsion or oleaginous suspension.
  • This emulsion or suspension may be formulated by a person of ordinary skill in the art using those suitable dispersing or wetting agents and suspending agents, including those mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,2-propane-diol.
  • the ste ⁇ le injectable preparation may also be prepared as a lyophilized powder.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile fixed oils may be employed as a solvent or suspending medium
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids e g., oleic acid
  • a pharmaceutically acceptable salt of a compound described herein may be dissolved in an aqueous solution of an organic or inorganic acid, such as 0.3 M solution of succinic acid, or more preferably, citric acid. If a soluble salt form is not available, the compound may be dissolved in a suitable co-solvent or combination of co-solvents Examples of suitable co- solvents include alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from about 0 to about 60% of the total volume. In one embodiment, the active compound is dissolved in DMSO and diluted with water
  • the pharmaceutical composition may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle, such as water or isotonic salme or dextrose solution.
  • an appropriate aqueous vehicle such as water or isotonic salme or dextrose solution.
  • compounds which have been modified by substitutions or additions of chemical or biochemical moieties which make them more suitable for delivery e.g., increase solubility, bioactivity, palatabihty, decrease adverse reactions, etc.
  • the compounds described herein may be formulated for oral administration in a lipid-based formulation suitable for low solubility compounds. Lipid-based formulations can generally enhance the oral bioavailability of such compounds
  • a preferred pharmaceutical composition comprises a therapeutically or prophylactically effective amount of a compound described herein, together with at least one pharmaceutically acceptable excipient selected from the group consisting of medium chain fatty acids and propylene glycol esters thereof (e g., propylene glycol esters of edible fatty acids, such as caprylic and capric fatty acids) and pharmaceutically acceptable surfactants, such as polyoxyl 40 hydrogenated castor oil.
  • a pharmaceutically acceptable excipient selected from the group consisting of medium chain fatty acids and propylene glycol esters thereof (e g., propylene glycol esters of edible fatty acids, such as caprylic and capric fatty acids) and pharmaceutically acceptable surfactants, such as polyoxyl 40 hydrogenated castor oil.
  • cyclodextrins may be added as aqueous solubility enhancers.
  • Preferred cyclodextrins include hydroxypropyl, hydroxyethyl, glucosyl, maltosyl and maltotnosyl derivatives of ⁇ -, ⁇ -, and ⁇ -cyclodextrin.
  • a particularly preferred cyclodextnn solubility enhancer is hydroxypropyl-o-cyclodext ⁇ n (BPBC), which may be added to any of the above-described compositions to further improve the aqueous solubility characteristics of the compounds of the embodiments.
  • BPBC hydroxypropyl-o-cyclodext ⁇ n
  • the composition comprises about 0 1% to about 20% hydroxypropyl-o-cyclodextrin, more preferably about 1% to about 15% hydroxypropyl-o-cyclodextrin, and even more preferably from about 2 5% to about 10% hydroxypropyl-o-cyclodextrin.
  • the amount of solubility enhancer employed will depend on the amount of the compound of the invention in the composition.
  • the methods of the embodiments also include the use of a compound or compounds as described herein together with one or more additional therapeutic agents for the treatment of disease conditions
  • the combination of active ingredients may be: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by any other combination therapy regimen known in the art
  • the methods described herein may comprise administering or delivering the active ingredients sequentially, e.g., in separate solution, emulsion, suspension, tablets, pills or capsules, or by different injections in separate syringes
  • an effective dosage of each active ingredient is administered sequentially, i.e., serially
  • simultaneous therapy effective dosages of two or more active ingredients are administered together.
  • Various sequences of intermittent combination therapy may also be used.
  • MI Experimental Myocardial Infarction
  • a protocol is described for examining the ventricular function, extent of fibrosis and VT inducibility in an ischemia reperfusion rat model after pirfenidone treatment.
  • Ventricular function was assessed via echocardiography.
  • VT inducibility was assessed by programmed stimulation and EP study The electrophysiological properties were assessed using high resolution optical mapping, and the extent of fibrosis was studied using standard histological techniques
  • the depth of entry was 2 mm, which was slightly greater than the level of the left coronary artery
  • Both suture ends were then threaded through a PE-90 polyethylene tube 6 in in length to form a "snare loop" around the artery, closed by pulling on the free ends of the suture
  • the snare loop was tested by closing and releasing after 10-seconds to demonstrate adequate ischemia and reperfusion
  • the suture was then tightened to occlude the artery for 20 minutes and then removed to allow for reperfusion.
  • the chest was then closed with 5 0 prolene suture, and the animal was allowed to recover.
  • LV left ventricular
  • ESV left ventricular
  • EDV end-systolic and end-diastolic volumes
  • LVEF LV ejection fraction
  • LVEF was calculated using the following formula: (EDV-ESV)/EDV 100 Fractional shortening (FS) was evaluated from the M mode of the parasternal long-axis view at the papillary muscle level on the basis of the percent changes of LV end-diastolic and end- systolic diameters.
  • Optical mapping is a technique to perform high-resolution electrophysiologic evaluation of the cardiac tissue. To summarize the procedure, ten thousand simultaneous optical action potentials were recorded with a 100 x 100 CMOS camera within a 19 mm x 19 mm mapping field on the epicardium of the LV anterior wall. Using a 1000-W tungsten- halogen light source, fluorescence was excited with an excitation filter of 530 nm and transmitted with an emission long-pass filter of > 630 nm. Fluorescent optical maps were acquired at 2000 Hz during programmed electrical stimulation. Optical mapping was performed 5 wks after MI.
  • Rats were injected with heparin (500 U ip) 15 min before excision of the heart, and were then anesthetized with pentobarbital sodium (50 mg/kg ip). After adequate anesthesia, the heart was rapidly excised and arrested by immersion in cold cardioplegia solution. The aorta was cannulated and retrogradely perfused, at a rate of 6 mL/min, with 37°C modified Tyrode solution containing (in mmol/L). 130 NaCl, 20 0 NaHCO 3 , 1 2 MgCl 2 , 4.0 KCl, 5.6 glucose, and 1.8 CaCl 2 , gassed with 95% 02/5% C02.
  • Extraneous tissue was carefully removed from the heart The cannulated heart was then placed in 37°C Tyrode solution in a specialized temperature-controlled optical recording chamber (maintained at 37°C) while ECG, perfusion rate, and temperature were measured continuously for the duration of the experiment.
  • Tyrode solution containing voltage- sensitive dye PGH I (10 ⁇ L of 5mM stock solution) was perfused through the preparation over a 5-mm period.
  • Ventricular epicardium bipolar pacing at a stimulus amplitude of 2X threshold, was performed on normal tissue near the infarct zone Mapping was recorded during pacing drives of 250 ms to 90 ms (decremented by 10 ms), as well as dunng S1-S2 pacing using a basic cycle length (BCL) of 200 ms and maximum S2 of 150 ms and decremented by 10 ms
  • BCL basic cycle length
  • BCL basic cycle length
  • maximum S2 150 ms and decremented by 10 ms
  • programmed stimulation with up to three extrastimuli, and burst pacing (from 90 ms to 60 ms) were used to assess arrhythmia inducibihty. Inducibility was defined as the ability to provoke sustained (> 30 s) ventricular tachycardia (VT) or ventricular fibrillation. Maps were also captured during programmed stimulation and with all episodes of arrhythmia.
  • Optical mapping data was analyzed using modified OMproCCD software (from Bum-rak Choi, Pittsburg, PA) and Matlab custom software.
  • Raw fluorescence data was viewed as a movie of normalized fluorescence intensity, which revealed activation within the field of view.
  • Quantitative data was obtained from optically derived action potentials (APs) for each of the 10,000 pixels of the CMOS camera. Activation time and action potential duration at 50% (APD50) and 80% repolarization (APD80) were measured for each paced cycle length (PCL).
  • APs optically derived action potentials
  • Activation time was calculated at the maximum rate of rise of the fluorescent AP (dF/dt) APD80 is the duration from the activation time (start of the action potential) to the time point where the action potential has recovered to 20% maximal fluorescent signal (peak of the optical AP). Isochronal maps of activation were constructed for each map. Rise time was calculated as the time between takeoff and at the peak of the action potential. The OMproCCD software was used to calculate conduction vectors representing conduction velocities and conduction direction at each pixel, as previously described. Phase differences, calculated as the average difference with neighboring activation times at each site, were measured to quantify the spatial heterogeneity of conduction, as previously described.
  • Frequency histograms were constructed for the phase differences within a recorded area These histograms were summarized as the median phase time at 50th percentile (P50), and the 5th and 95th percentiles (P5 and P95, respectively) of the distribution
  • P50 percentile
  • P5 and P95 percentiles
  • Ventricular tissue samples were fixed in 10% neutral buffered formalin. The samples were embedded in paraffin, sectioned (10- ⁇ m thick), and then stained with Masson's trichrome or Sinus red with fast green counterstain. Stained slides were examined under light microscopy, digitized using a high-resolution scanner, and analyzed using Photoshop CS software. Infarct areas on Masson's trichome corresponded tightly with areas of dense Sinus red staining with minimal to no fast green. Infarct scar area and total area of left ventricular myocardium, for all sections, were manually traced in the digital images and automatically calculated by the software. Infarct size, expressed as a percentage, was measured by dividing the sum of infarct areas from all sections by the sum of LV areas from all sections and multiplying by 100.
  • the total area of fibrosis was also assessed After excluding the infarct area (defined as dense fibrosis), fibrosis in the border and non-infarct zones was quantified from digital photomicrographs of the Sinus red-stained sections. Areas containing blood vessels and perivascular interstitial cells were also excluded from fibrosis quantification. The red pixel content of digitized images relative to the total tissue area was counted by using the Adobe Photoshop CS software
  • fibrosis leads to decoupling of muscle fibers, conduction slowing and conduction blocks, as well as "zig-zag” and chaotic conduction.
  • the distnbution of fibrosis is also important: a finger-like distribution, as opposed to a more diffuse picture, is also thought to cause more disruption of wave propagation and is therefore more arrhythmogenic [Breithardt et al. Eur Heart J (1989) 10 Suppl E: 9-18].
  • cardiac fibrosis in the infarct border zone has such a strmg- like distribution and is more likely to cause alterations of direction-directed electrical propagation with the fibrotic tissue interrupting normally tight cell-cell coupling.
  • Pirfenidone attenuated the total amount of fibrosis, as well as extra-mfarct fibrosis.
  • PFD did not affect the infarct size, compared to control infarcts. Therefore, absent the PFD intervention, ongoing remodeling changes may actually contribute to infarct expansion long after the initial ischemic insult. There is evidence that this is indeed the case, with studies indicating that cardiomyocyte death can occur in non-mfarcted myocardium, particularly within the infarct border zone, for weeks after an MI.
  • Fibrosis within the infarct border zone for PFD animals was not only decreased but its distribution appeared less heterogeneous, with less of the finger-like projections seen in control infarcts This decrease in erratic distribution, as well as in quantity of fibrosis, was associated with improved conduction velocities in PFD border zones A concurrent increase in action potential rise and faster rise time in PFD border zones further confirm these findings. These results, as well as decreased conduction heterogeneity, were likely responsible for the almost three-fold decrease in VT susceptibility in PFD animals.
  • Optical Mapping Studies A coronary perfused left ventricular preparation was used as described in Wu et al., / Cardwvasc Electrophyswl 1998; 9:1336-47. Briefly, following sedation with sodium pentothal (0.25 mg/Kg), a left lateral thoracotomy is performed and the heart was rapidly excised. It was then perfused with cardioplegic solution ((in mmol/L): NaCl 123, KCl 15, NaHCO 3 22, NaH 2 PO 4 0.65, MgCl 2 0.50, glucose 5 5, CaCl 2 2, bubbled with 95% O 2 /5% CO 2 ) retrogradely through the aorta.
  • cardioplegic solution (in mmol/L): NaCl 123, KCl 15, NaHCO 3 22, NaH 2 PO 4 0.65, MgCl 2 0.50, glucose 5 5, CaCl 2 2, bubbled with 95% O 2 /5% CO 2 ) retrogradely through the aorta.
  • ventricles were removed at approximately 1 cm below the AV ring and the left anterior descending coronary artery (LAD) was perfused.
  • LAD left anterior descending coronary artery
  • the right ventricle was removed and the left ventricle was cut to the size that was perfused by the LAD and included a papillary muscle. All ventricular branches were then ligated
  • the ventricular preparation was then transferred to a tissue chamber maintained at 37°C.
  • the perfusion line in the LAD was perfused with modified Tyrode's solution ((in mmol/L): NaCl 123, KCl 5.4, NaHCO 3 22, NaH 2 PO 4 0.65, MgCl 2 0.50, glucose 5.5, CaCl 2 2, bubbled with 95% O 2 /5% CO 2 ).
  • modified Tyrode's solution (in mmol/L): NaCl 123, KCl 5.4, NaHCO 3 22, NaH 2 PO 4 0.65, MgCl 2 0.50, glucose 5.5, CaCl 2 2, bubbled with 95% O 2 /5% CO 2 ).
  • a bolus of 30 - 40 ⁇ l of the voltage sensitive dye PGH- 1 was injected directly into the perfusate.
  • VF Two plunge electrodes were dedicated for recording a bipolar signal for monitoring the electrical activity of the preparation.
  • VF was initiated with either extra stimuli or with rapid burst pacing at a cycle length of 50 ms, a pulse width of 9.9 ms, and an output of 9.9 mA.
  • Several 4-s episodes of VF were recorded on each surface in each preparation Activation movies of the VF were then viewed, and the activation patterns were determined After termination of VF signals were obtained during pacing at 250ms and isochronal maps of activation were constructed to look at conduction Activation patterns and wave-front direction during VF were determined from raw fluorescence movies (isopotential).
  • Activation was characterized as 1) spiral (single reentrant circuit dominating the epoch), 2) focal (discrete, high frequency location of activation), 3) multiple wavefront (rapidly changing or varying wave fronts with wave-front collision), or 4) one broad wavefront (single wave-front passing through the map) VF was defined as rapid and irregular activations on the bipolar signal used for monitoring the elect ⁇ cal activity of the preparation.
  • a fast Fourier transform was calculated on the digitally filtered waveform
  • the data were detrended and multiplied by a Hamming window
  • the largest peak of the resulting magnitude spectrum was identified, and the positions of the harmonic peaks were determined on the basis of its position
  • the areas under the largest peak and three of its harmonic peaks were each calculated over a 1-Hz window This produced an area under four peaks
  • the total area of the spectrum was calculated from 2 Hz up to but not including the fifth harmonic peak.
  • the ratio of the power under the harmonic peaks to the total power in this range was calculated, and the resulting number was defined as the organization index (OI)
  • OI organization index
  • the OI was theorized to represent the organization of AF for that signal at that period in time
  • SD/mean spatial coefficient of variance
  • temporal coefficient of variance of average DFs from among AF episodes for each mapping field withm each preparation were calculated.
  • Stability was defined as persistence over at least 90% of the epoch, and if it disappeared, it would return in the same location
  • Cross Correlation Analysis Spatial correlation analysis was performed on all recorded signals between all possible paired electrogram combinations in each animal. The cross-correlation function was calculated at zero lag for each electrogram combination, and the peak value was considered the correlation coefficient, representing the degree of correlation between the two signals. All of the correlation coefficients calculated from an AF recording with optical mapping were then averaged to produce a mean correlation value for each AF episode.
  • VF Activation Patterns On examination of the optical mapping activation sequences, 4 types of activation patterns were seen - spiral wave, focal area of activation, multiple waves, and on broad wavefront sweeping through the field of view. Table 2 shows the types of activation patterns that were seen on each mapped surface for each dog.
  • Epicardial Surface For the Control group, only 2 of the 10 mapped epicardial surfaces showed evidence of focal activation. These two surfaces also corresponded to having stable, high DF areas. All others had activation patterns of either multiple wavelets or one broad wavefront dominating the field of view. The activation map, during pacing at 250 ms, shows homogeneous conduction throughout the field of view. Similar results were seen in the CHF and PFD groups. Both groups had 2/6 mapped surfaces having either focal activation or a spiral wave (1 CHF dog). These types of activation corresponded to stable, high DF areas. All other dogs had either multiple wavefronts or one broad wavefront dominating the field of view. These activation patterns had either transient DFs (multiple wavefronts) or the area was dommanted one DF (broad waveftont) The activation images show homogeneous conduction, similar to Control, but at a slower conduction velocity.
  • Endocardial Surface Mapping of the endocardial surface included the papillary muscle and only the CHF group had AF characterized by stable, high DF areas that correlated to spiral waves or focal activation patterns. Three of the five mapped endocardial surfaces in the CHF group fell into this category. Even though 2 of 7 endocardial surfaces in the Control group had activation characterized by spiral waves, no discrete, stable DFs were observed. The other 5 Controls and all of the mapped endocardial surfaces in the PFD group had either multiple or broad wavefront activation. All of the groups showed heterogeneous conduction marked by conduction slowing. This is in contrast to the homogenous conduction seen on the epicardial surface.
  • Transmural Surface The transmural surface had the highest percentage of spiral wave and focal activation when compared to the other mapped surfaces for all groups
  • CHF group the transmural surface was mapped in 5 dogs and all of them had VF activation patterns of either a spiral wave or focal activation.
  • the VF was characterized by stable, discrete, high DF areas.
  • 50% of the mapped transmural surfaces had an activation pattern of a spiral wave that correlated to stable high DF areas.
  • In the Control group 75% of the transmural surfaces had focal activation. One of these did not correlate to stable, high DF areas.
  • Each group showed heterogeneous conduction characterized by areas of conduction slowing and block.
  • Both the endocardial and transmural surfaces had VF characterized by one broad wavefront sweeping through the field of view.
  • the corresponding DF maps are characterized by a single DF.
  • the epicardial surface had VF characterized by a broad wavefront, and the corresponding DF map was dominated by a singular DF.
  • the endocardial and transmural surfaces both had VF characterized by stable, high DF areas.
  • the VF mechanisms that these DF corresponded to were a focal mechanism on the endocardial surface and a spiral wave on the transmural surface.
  • a spiral wave was seen in the transmural surface and the corresponding DF map had a stable, high DF area.
  • OI organization index
  • the Control group also showed the most temporal stability in OI levels as this group had the lowest OI temporal CoV values at all surfaces with the lowest measurements found on the endocardial surface
  • the endocardial and transmural surfaces of the CHF and PFD groups were significantly different than those of the Control group.
  • FIG. 8A shows the average correlation coefficients for each surface of each group.
  • Figure 7 shows the gradient of frequencies over distance across the endocardial surface, transmural suface and epicardial surface. Pirfendidone preserved the transmural gradient to that similar to control animals, whereas untreated animals with heart failure have a very large gradient.
  • a method of treating a patient who has suffered an acute myocardial infarction comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein optionally the treatment is initiated at a time period about 1 to 42 days after suffering the AMI, and optionally continues for up to 3 to 6 months.
  • AMI acute myocardial infarction
  • a method of reducing the incidence of congestive heart failure in a patient who suffered an acute myocardial infarction comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the therapeutically effective dose reduces the incidence of congestive heart failure.
  • a method of preserving viable cardiac tissue or controlling or reducing myocardial infarct size in a patient who has suffered an acute myocardial infarction comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic to the patient results in a relatively reduced infarct size on average compared to infarct size in a patient who has not been administered the therapeutic.
  • AMI acute myocardial infarction
  • a method of reducing the incidence of ventricular tachycardia in a patient in need thereof comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic prevents or reduces the incidence of ventricular tachycardia.
  • a method of treating or preventing ventricular fibrillation in a patient in need thereof comprising administering to the patient a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic prevents ventricular fibrillation in the patient.
  • a method of controlling arrhythmia in a patient in need thereof comprising administering to the patient a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic controls arrhythmia in the patient.
  • AMI acute myocardial infarction
  • the therapeutic having an anti-fibrotic effect is a therapeutic that reduces tissue remodeling or fibrosis, reduces the activity of transforming growth factor-beta (TGF- ⁇ ), targets one or more TGF- ⁇ isoforms, inhibits TGF- ⁇ receptor kinases TGFBRl (ALK5) and/or TGFBR2, or modulates one or more post-receptor signaling pathways; is an endothelin receptor antagonists, targets both endothelin receptor A and endothelin receptor B or selectively targets endothelin receptor A; reduces activity of connective tissue growth factor (CTGF); inhibits matrix metalloproteinase; reduces the activity of epidermal growth factor (EGF), targets the EGF receptor, or inhibits EGF receptor kinase; reduces the activity of platelet derived growth factor (PDGF), targets PDGF receptor (PDGFR), inhibits PDGFR kinase activity, or inhibits post- PDGF receptor
  • TGF- ⁇ transforming growth factor-bet
  • R 1 , R 2 , R 3 , R 4 , X 1 , X 2 , X 3 , X 4 , X 5 , Y 1 , Y 2 , Y 3 , and Y 4 are independently selected from the group consisting of H, deuterium, Ci-C io alkyl, C 1 -C 10 deuterated alkyl, substituted Ci- Cio alkyl, C 1 -C 10 alkenyl, substituted C 1 -C 10 alkenyl, C 1 -C 10 thioalkyl, C 1 -C 10 alkoxy, substituted C 1 -C 10 alkoxy, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxyl, Ci-Cio alkoxyalkyl, substituted Ci
  • X 6 and X 7 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkylenylaryl, alkylenylheteroaryl, alkylenylheterocycloalkyl, alkylenylcycloalkyl, or X 6 and X 7 together form an optionally substituted 5 or 6 membered heterocyclic ring; and
  • Ar is pyridmyl or phenyl; and Z is O or S.
  • X 3 is H, OH, or C 1 i 0 alkoxy
  • Z is O
  • R 4 is H or hydroxyl, or a salt, ester, solvate, or prodrug thereof.
  • A is N or CR 2
  • B is N or CR 4
  • E is N, N + X 4 or CX 4
  • G is N, N + X 3 or CX 3
  • J is N, N + X 2 or
  • K is N, N + X 1 or CX 1 ; a dashed line is a single or double bond,
  • R 1 , R 2 , R 3 , R 4 , X 1 , X 2 , X 3 , X 4 , X 5 , Y 1 , Y 2 , Y 3 , and Y 4 are independently selected from the group consisting of H, deuterium, optionally substituted Ci-Cio alkyl, optionally substituted C 1 -C 10 deuterated alkyl, optionally substituted C 1 -C 10 alkenyl, optionally substituted C 1 -C 10 thioalkyl, optionally substituted C 1 -C 10 alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted heteroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted amido, optionally substituted sulfonyl, optionally substituted ammo, optionally substituted sulfonamido, optionally substituted sulfoxyl, cyano,
  • X 6 and X 7 are independently selected from the group consisting of hydrogen, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylenylaryl, optionally substituted alkylenylheteroaryl, optionally substituted alkylenylheterocycloalkyl, optionally substituted alkylenylcycloalkyl, or X 6 and X 7 together form an optionally substituted 5 or 6 membered heterocyclic ring, and
  • Ar is optionally substituted pyridmyl or optionally substituted phenyl; and Z is O or S.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hospice & Palliative Care (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Quinoline Compounds (AREA)
  • Pyridine Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The invention relates to methods of treating patients who have suffered an acute myocardial infarction (AMI) with a therapeutic that has anti-fibrotic effects, for example, pirfenidone and analogs thereof The method of treating a patient who has suffered an acute myocardial infarction may include administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein optionally the treatment is initiated at a time period about 1 to 42 days after suffering the AMI, and optionally continues for up to 3 to 6 months

Description

METHODS FOR TREATING ACUTE MYOCARDIAL INFARCTIONS AND
ASSOCIATED DISORDERS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U S Provisional Application No 61/147,340, filed January 26, 2009, the disclosure of which is incorporated herein by reference in its entirety
FIELD OF THE INVENTION
[0002] The invention relates to methods of treating patients who have suffered an acute myocardial infarction (AMI) and associated disorders with a therapeutic that has anti-fibrotic effects, for example, pirfenidone and analogs thereof.
BACKGROUND
[0003] There are approximately 1.5 million cases of acute myocardial infarction (AMI) m the United States each year, resulting in more than 500,000 deaths. Many of the deaths resulting from AMI occur before the patient can reach the hospital. Despite medical and interventional advances in the treatment of acute coronary syndromes over the last two decades, patients continue to face significant morbidity and mortality following a myocardial infarction Post-myocardial infarction (MI) complications include congestive heart failure (CHF) and ventricular tachycardia (VT).
[0004] Contraction of the heart is initiated by an electrical impulse generated by the sinoatrial node, a natural pacemaker, in the heart The heart's electrical conduction system then conveys the impulse to the myocardium, or cardiac muscle, to stimulate contraction Abnormal electrical conduction due to structural tissue remodeling after infarction may play an important role in ventricular arrhythmias, which can lead to sudden cardiac arrest and death. Tissue remodeling is due in part to direct tissue damage, neurohormonal activation, cytokine release, inflammation and fibrosis.
[0005] Medical therapeutics, including drug therapy aimed at suppressing and preventing ventricular arrhythmias have thus far been disappointing. Earlier agents, including class IC anti-arrhythmics, were unexpectedly pro- arrhythmic in the setting of coronary artery disease and raised a cautionary note. Current post-MI pharmacotherapies include renin-angiotensin- aldosterone (RAA) blockers, which improve cardiac remodeling but do not specifically target fibrosis. It is an object of the present invention to provide novel therapies and therapeutic regimens for treating acute myocardial infarction. SUMMARY
[0006] It has been unexpectedly found that a compound which inhibits fibrosis has beneficial effects on left ventricular (LV) function, infarct size, peri-mfarct fibrosis, electrophysiology of the infarct border zone and VT inducibility. It is also unexpected that such compounds offer a more targeted and effective inhibition of detrimental post-acute MI remodeling than RAA blockers. Provided herein are novel means to prevent arrhythmias in the post-acute MI period, and to improve heart contractility, improve heart function and reduce complications of acute MI such as congestive heart failure (CHF) and ventricular tachycardia (VT) and ventricular fibrillation.
[0007] Without being bound by a theory of the invention, early fibrosis in response to cardiac injury is believed to be important in forming a healing scar and serves as a compensatory function in preventing infarct expansion, aneurysm formation, and cardiac perforation. However, late-onset and excessive fibrosis beyond the infarct, and into the infarct border zone and other viable tissues, can contribute to adverse cardiac remodeling. Cardiac fibrosis can cause altered propagation, leading to non-uniform anisotropic conduction that eventually causes the formation of re-entry circuits and potentially wave breaks that predispose to arrhythmogenesis. The results described herein indicate that inhibiting late-onset fibrosis can provide measurable beneficial effects in the post-acute MI setting
[0008] In the broadest feature, the present invention discloses a method of treating a patient who has suffered a myocardial infarction (MI), or who has not previously suffered an MI, or is within a week of suffering an MI, comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect. In another aspect, the present invention discloses a method of treating a patient who has suffered a myocardial infarction (e.g. an acute myocardial infarction (AMI)) comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein optionally the treatment is initiated immediately after suffering the myocardial infarction (e.g. the AMI), and optionally continues for up to 3 to 6 months, hi some aspects, the method is to limit expansion of an infarct scar due to the myocardial infarction (e.g. the AMI)
[0009] In another aspect, the invention provides a method of treating a patient who has suffered myocardial infarction (e g. an AMI) comprising administering to said patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect In some embodiments, the treatment is initiated at a time period about 1 to 42 days after suffering the myocardial infarction (e.g. the AMI), and optionally continues for up to 3 to 6 months. In other embodiments, the treatment is initiated at a time period about 3 to 14 days after suffering the myocardial infarction (e.g. the AMI), and optionally continues for up to 3 to 6 months. In another embodiment, the treatment is initiated about 5-10 days after the myocardial infarction (e.g. the AMI) In another embodiment, the treatment is initiated about 2-40 days after the myocardial infarction (e.g. the AMI). In another embodiment, the treatment is initiated about 3-20 days after the myocardial infarction (e.g. the AMI). In another embodiment, the treatment is initiated about 4-15 days after the myocardial infarction (e.g. the AMI). In yet another embodiment, the treatment is initiated about 7 days after the myocardial infarction (e.g. the AMI) In some embodiments, the treatment continues for a period of at least 2 weeks. In other embodiments, the treatment after being initiated continues for a time period until about 4 weeks after the myocardial infarction (e.g. the AMI). Thus, the invention encompasses treatment of patients from about 14 days to 4 weeks after the myocardial infarction (e g the AMI)
[0010] In an embodiment, the invention provides a method of reducing the incidence of congestive heart failure (CHF) in a patient who suffered a myocardial infarction (e.g., an acute myocardial infarction (AMI)), comprising administering to said patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the therapeutically effective dose reduces the incidence of congestive heart failure, and wherein optionally the treatment is initiated at a time period about 1 to 42 days after suffering the myocardial infarction (e.g. the AMI). In some aspects, the patient is at an increased risk of congestive heart failure due to the myocardial infarction (e.g. the AMI).
[0011] In an embodiment, the invention provides a method of preserving viable cardiac tissue or controlling myocardial infarct size in a patient who has suffered a myocardial infarction (e g an acute myocardial infarction (AMI)) comprising administering to said patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the administering of said therapeutic to said patient results in a relatively reduced infarct size on average compared to infarct size in a patient who has not been administered said therapeutic. In some embodiments, the treatment is initiated at a time period about 1 to 42 days after suffering the myocardial infarction (e.g. the AMI). In further embodiments, the relative reduction in infarct size is at least 5%
[0012] In an embodiment, the invention provides a method of reducing the incidence of ventricular tachycardia in a patient in need thereof, comprising administering to said patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect. In some embodiments, the patient has suffered a myocardial infarction (e g. an AMI). In further embodiments, the treatment is initiated at a time period about 1 to 42 days after suffering the myocardial infarction (e.g. the AMI). In another embodiment, the administering is initiated about 7 days after suffeπng the myocardial infarction (e.g. the AMI).
[0013] In an embodiment, the invention provides a method of treating or preventing ventricular fibrillation in a patient in need thereof is provided, comprising administering to said patient a therapeutic having an anti-fibrotic effect. In some embodiments, the patient has suffered a myocardial infarction (e g an AMI) In further embodiments, the treatment is initiated at a time peπod about 1 to 42 days after suffering the myocardial infarction (e g the AMI) In another embodiment, the administering is initiated about 7 days after the suffering of the myocardial infarction (e.g. the AMI). In another embodiment, the administering reduces the incidence of sudden cardiac death relative to the incidence of cardiac death in the absence of administration of the therapeutic. In still another embodiment, the administering reduces cardiac risk of the patient relative to the cardiac risk in the absence of administration of the therapeutic. As used herein, the term "cardiac risk" means the risk of cardiac morbidity resulting from any one or a combination of ventricular tachycardia, sudden cardiac death, ventricular fibrillation and/or congestive heart failure.
[0014] In some embodiments, the invention provides a method of controlling (e.g., reduce, reduce the incidence or severity of, or prevent the progression of) arrhythmia in a patient in need thereof is provided, comprising administering to the patient a therapeutic having an anti- fibrotic effect, wherein the administering of the therapeutic controls (e.g., reduce, reduce the incidence or severity of, or prevent the progression of) arrhythmia in the patient. In some embodiments, the administering reduces the incidence or severity of arrhythmia in the patient relative to the incidence or severity of arrhythmia in the absence of administration of the therapeutic In some embodiments the patient has suffered a myocardial infarction (e g an AMI) In further embodiments the administration is initiated about 1 to 42 days after the suffering of the myocardial infarction (e.g. the AMI). In still further embodiments the administration is initiated about 7 days after the suffering of the myocardial infarction (e g. the AMI). In other embodiments, the administering treats ventricular remodeling.
[0015] In some embodiments of any of the preceding methods, the patient is diagnosed as suffering a first myocardial infarction (e.g. a first AMI), i e. the patient has not been diagnosed as having previously suffered a myocardial infarction (e g an AMI) or the patient has not previously suffered a myocardial infarction (e g an AMI) In some embodiments, any of the methods described herein optionally exclude treatment of patients diagnosed with chronic MI.
[0016] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect reduces tissue remodeling or fibrosis, hi some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect reduces the activity of transforming growth factor-beta (TGF-β), targets one or more TGF-β isoforms, inhibits TGF- β receptor kinases TGFBRl (ALK5) and/or TGFBR2, or modulates one or more post- receptor signaling pathways. In such cases, the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the TGF-β pathway and/or reduce fibrosis.
[0017] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect is an endothelin receptor antagonist, targets both endothelin receptor A and endothelin receptor B or selectively targets endothelin receptor A. In such cases, the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the endothelin A and/or B pathway, and/or reduce fibrosis.
[0018] In other embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect reduces activity of connective tissue growth factor (CTGF). In such cases, the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the CTGF pathway and/or reduce fibrosis.
[0019] In further embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect inhibits matrix metalloproteinase (MMP). In such cases, the therapeutically effective amount of such a compound may inhibit MMP and/or reduce fibrosis. In certain embodiments, the therapeutically effective amount of such a compound may inhibit MMP-9 or MMP- 12.
[0020] In still other embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect reduces the activity of epidermal growth factor receptor (4), targets EGF receptor, or inhibits EGF receptor kinase. In such cases, the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the EGF pathway and/or reduce fibrosis.
[0021] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect reduces the activity of platelet derived growth factor (PDGF), targets PDGF receptor (PDGFR), inhibits PDGFR kinase activity, or inhibits post-PDGF receptor signaling pathways In such cases, the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the PDGF pathway and/or reduce fibrosis
[0022] In some embodiments of any of the preceding methods, the therapeutic having an anti fibrotic effect reduces the activity of vascular endothelial growth factor (VEGF), targets one or more of VEGF, VEGF receptor 1 (VEGFRl, Fit 1), or VEGF receptor 2 (VEGFR2, KDR). In such cases, the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the VEGF pathway and/or reduce fibrosis.
[0023] In other embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect inhibits multiple receptor kinases such as BIRB-1120 which inhibits receptor kinases for vascular endothelial growth factor, fibroblast growth factor, and platelet derived growth factor In such cases, the therapeutically effective amount of such a compound may inhibit one or more receptor kinases in the VEGF, FGF or PDGF pathways and/or reduce fibrosis
[0024] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect interferes with mtegnn function In such cases, the therapeutically effective amount of such a compound may inhibit integrin function and/or reduce fibrosis In further embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect may inhibit αV mtegrins. In other embodiments, the therapeutic having an anti fibrotic effect may inhibit mtegnn αVβ6 function
[0025] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect interferes with pro-fibrotic activities of IL-4 and IL-13, targets IL-4 receptor, IL-13 receptor. In such cases, the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects in the IL-4 and/or IL-13 pathway and/or reduce fibrosis
[0026] In further embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect modulates signaling through the JAK-STAT pathway. In such cases, the therapeutically effective amount of such a compound may modulate signaling through the JAK-STAT pathway and/or reduce fibrosis
[0027] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect interferes with epithelial mesenchymal transition, or inhibits mTor In such cases, the therapeutically effective amount of such a compound may exhibit one or more of the foregoing effects on mesenchyma, and/or reduce fibrosis [0028] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect reduces levels of copper. In such cases, the therapeutically effective amount of such a compound may reduce copper levels in circulation and/or tissue, and/or reduce fibrosis.
[0029] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect reduces oxidative stress, hi such cases, the therapeutically effective amount of such a compound may reduce oxidative stress and/or reduce fibrosis.
[0030] In still further embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect inhibits prolyl hydrolyse. In such cases, the therapeutically effective amount of such a compound may reduce prolyl hydrolase and/or reduce fibrosis.
[0031] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect is an agonist of proliferator- activated receptor-gamma (PPAR-γ).
[0032] In some embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect inhibits phosphodiesterase 4 (PDE4) or phosphodiesterase 5 (PDE5), or modifies the arachidonic acid pathway. In such cases, the therapeutically effective amount of such a compound may inhibit the PDE4 and/or PDE5 pathway, or may inhibit the arachidonic acid pathway, and/or reduce fibrosis.
[0033] In various embodiments of any of the preceding methods, the therapeutic having an anti-fibrotic effect is combined with a pharmaceutically acceptable carrier. In other embodiments of any of the preceding methods, the administration is oral.
[0034] In some embodiments of any of the preceding methods, the therapeutically effective amount is a total daily dose of about 50 mg to about 2400 mg of said therapeutic or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof.
[0035] In some embodiments of any of the preceding methods, the therapeutically effective amount is administered in divided doses three times a day or two times a day, or is administered in a single dose once a day.
[0036] In various embodiments of any of the preceding methods, said therapeutic is pirfenidone or compound of formula (I), (II), (III), (IV), or (V) or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof:
Figure imgf000010_0001
wherein
A is N or CR2; B is N or CR4; E is Nor CX4; G is N or CX3; J is N or CX2; K is N or CX1; a dashed line is a single or double bond,
R1, R2, R3, R4, X1, X2, X3, X4, X5, Y1, Y2, Y3, and Y4 are independently selected from the group consisting of H, deuterium, Ci-C 10 alkyl, Ci-Cio deuterated alkyl, substituted Q- Cio alkyl, Ci Qo alkenyl, substituted Ci Qo alkenyl, Ci Qo thioalkyl, Q Qo alkoxy, substituted Q-Qo alkoxy, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxyl, Ct Ci0 alkoxyalkyl, substituted Ci Ci0 alkoxyalkyl, Ci-Cio carboxy, substituted Q -C 10 carboxy, Q-Qo alkoxycarbonyl, substituted C1-C10 alkoxycarbonyl, CO-uronide, CO-monosacchande, CO-ohgosaccharide, and CO- polysacchande;
X6 and X7 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkylenylaryl, alkylenylheteroaryl, alkylenylheterocycloalkyl, alkylenylcycloalkyl, or X6 and X7 together form an optionally substituted 5 or 6 membered heterocyclic ring; and
Ar is pyndmyl or phenyl; and Z is O or S.
[0037] In some embodiments, A is N or CR2; B is N or CR4, E is N, N+X4 or CX4; G is N, N+X3 or CX3, J is N, N+X2 or CX2; K is N, N+X1 or CX1; a dashed line is a single or double bond,
R1, R2, R3, R4, X1, X2, X3, X4, X5, Y1, Y2, Y3, and Y4 are independently selected from the group consisting of H, deuterium, optionally substituted C1-C10 alkyl, optionally substituted C1-C10 deuterated alkyl, optionally substituted C1-C10 alkenyl, optionally substituted C1-C10 thioalkyl, optionally substituted C1-C10 alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted heteroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted amido, optionally substituted sulfonyl, optionally substituted ammo, optionally substituted sulfonamide, optionally substituted sulfoxyl, cyano, nitro, halogen, hydroxyl, SO2H2, optionally substituted C1-C10 alkoxyalkyl, optionally substituted C1-C10 carboxy, optionally substituted C1-C10 alkoxycarbonyl, CO-uronide, CO-monosaccharide, CO-oligosacchaπde, and CO-polysacchaπde
X6 and X7 are independently selected from the group consisting of hydrogen, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylenylaryl, optionally substituted alkylenylheteroaryl, optionally substituted alkylenylheterocycloalkyl, optionally substituted alkylenylcycloalkyl, or X6 and X7 together form an optionally substituted 5 or 6 membered heterocyclic ring, and
Ar is optionally substituted pyndmyl or optionally substituted phenyl, and Z is O or S.
[0038] In some embodiments of any of the preceding methods, said therapeutic is pirfenidone or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof
[0039] In various embodiments of any of the preceding methods, the therapeutic administered to said patient comprises a compound of formula (II)
Figure imgf000011_0001
wherein
X3 is H, OH, or C1-C10 alkoxy, Zy is O, and R2 is methyl, C(=0)H, C(=O)CH3, C(=O)O- glucosyl, fluoromethyl, difluoromethyl, trifluoromethyl, methylmethoxyl, methylhydroxyl, or phenyl, and R4 is H or hydroxyl, or a salt, ester, solvate, or prodrug thereof
[0040] In still further embodiments of any of the preceding methods, the therapeutic administered to said patient is selected from the group consisting of
Figure imgf000012_0001
Figure imgf000013_0001
, , a compound as listed in Table 1 , and pharmaceutically acceptable salts, esters, solvates, and prodrugs thereof.
[0041] In some embodiments of any of the preceding methods, the patient is human. BRIEF DESCRIPTION OF THE DRAWINGS
[0042] Figure 1 shows that the pirfenidone group (dotted line) had significantly less decline in its ejection fraction, decreasing by only 8% from week 1 to week 5. The ejection fraction for controls decreased by 24% (solid line). The pirfenidone group had a higher ejection fraction of 45% at 5 weeks compared to controls with a mean ejection fraction of 36%, despite the fact that the pirfemdone-treated rats had originally been randomized to a lower ejection fraction at 1 week (54% versus 60%)
[0043] Figure 2 depicts the conduction velocities for the normal, border, and infarct zones of both groups at various pacing cycle lengths, with pirfenidone in the circles and controls as squares. Conduction velocities in the non-mfarct zones of both control and pirfenidone groups were fastest among all three zones and were similar between the two groups Conduction velocities in the infarct zones of both control and pirfenidone groups were slowest among all three zones and were similar between the two groups. Finally, conduction velocities in the border zones of both groups were in between those of the non-mfarct and infarct zones. However, the conduction velocities in the border zone for the pirfemdone- treated group was significantly faster, at all pacing cycle lengths, compared to those in the border zone of control annuals.
[0044] Figure 3 shows a trend toward lower conduction heterogeneity for pirfemdone- treated rats (circles), compared to control rats (squares).
[0045] Figure 4 shows that, in terms of other electrophysiological parameters, the rise time correlates with conduction velocity. An infarct is shown here to increase the time it takes to fully depolarize for both control (squares) and pirfemdone-treated (circles) rats, with the rise time being slower in the infarct zones compared to their respective normal areas. The rise times in the border zones are in between the infarct and normal zones. The rise time is shown to be shorter for the border zones of pirfemdone-treated rats, consistent with the faster conduction velocities in pirfenidone-treated rats. [0046] Figure 5 depicts fluorescence amplitude for the three zones. Normal areas had the highest amplitude, infarct areas the least, and border areas in the middle There was a trend toward higher amplitudes of fluorescence in the border zones of pirfenidone -treated rats, as compared to those of the controls.
[0047] Figure 6 depicts the myocardial infarct size and amount of myocardial fibrosis in control versus pirfemdone-treated rats.
[0048] Figure 7 shows the largest measured frequency gradient over the distance that the gradient occurs for each mapped surface The dark solid bars represent Control, hatched bars - congestive heart failure (CHF), and open bars - pirfenidone (PFD)
[0049] Figure 8 shows summary correlation coefficient (XC) data for VF activation patterns. Panel A - average XC values for each mapped surface for each group. The dark solid bars represent Control, hatched bars - CHF, and open bars - PFD. Panel B - average XC values for each VF activation patterns for all groups Panel C - coefficient of variance of the XC values for each VF activation patterns for all groups.
DETAILED DESCRIPTION OF THE INVENTION
[0050] Pirfenidone (PFD) is an orally active, anti-fibrotic agent. It is demonstrated herein that pirfenidone exhibits specific and potent attenuation of post-MI fibrosis, and ameliorates the arrhythmogenic potential of cardiac remodeling.
[0051] Pirfenidone is a small drug molecule whose chemical name is 5-mefhyl-1-phenyl-2- (1H)-pyridone. It is a non-peptide synthetic molecule with a molecular weight of 185.23 daltons. Its chemical elements are expressed as Ci2H11NO, and its structure and synthesis are known. Several pirfenidone Investigational New Drug Applications (INDs) are currently on file with the U.S. Food and Drug Administration Human investigations are ongoing or have recently been completed for pulmonary fibrosis, renal glomerulosclerosis, and liver cirrhosis. There have been other Phase II studies that used pirfenidone to attempt to treat benign prostate hypertrophy, hypertrophic scarring (keloids), and rheumatoid arthritis.
[0052] Pirfenidone is being investigated for therapeutic benefits to patients suffering from fibrosis conditions such as Hermansky-Pudlak Syndrome (HPS), associated pulmonary fibrosis and idiopathic pulmonary fibrosis (IPF) Pirfenidone is also being investigated for a pharmacologic ability to prevent or remove excessive scar tissue found in fibrosis associated with injured tissues including that of lungs, skin, joints, kidneys, prostate glands, and livers. [0053] Pirfenidone has been reported to inhibit excessive biosynthesis or release of various cytokines such as TNF- α, TGF-βl, bFGF, PDGF, and EGF (Zhang S et al , Australian and New England J Ophthalmology 26 S74-S76 (1998); Cain et al , Int'l J Immunopharmacology 20:685-695 (1998)). Pirfenidone has also been reported to decrease collagen expression and to alter the balance of matrix metalloproteinases (MMPs) and their endogenous inhibitors (tissue inhibitor of metalloproteinases or TIMPs).
Acute Myocardial Infarction (AMI)
[0054] In some embodiments, methods are provided for treating a patient who has suffered an acute myocardial infarction (AMI) comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect. In some embodiments, a method is provided for treating a condition caused by ventricular remodeling, wherein the ventricular remodeling is caused by an AMI. In some embodiments, the ventricular remodeling is fibrosis. Thus, in some embodiments a method is provided for reducing ventricular remodeling (e g , ventricular fibrosis) in a patient who has suffered an AMI. The ventricular remodeling (e.g., ventricular fibrosis) is reduced relative to an amount of ventricular remodeling (e.g., an amount of ventricular fibrosis) in the absence of administration of the therapeutic (e.g., in comparison to a patient who was not administered the therapeutic)
[0055] Acute myocardial infarction (AMI) refers to infarction (damage or death) of heart tissue due to an acute, immediate blockage of one or more of the coronary arteries. Coronary arterial occlusion (blockage) due to thrombosis is the cause of most cases of AMI. This blockage restricts the blood supply to the muscle walls of the heart and is often accompanied by symptoms such as chest pain, heavy pressure in the chest, nausea, and shortness of breath, or shooting pain in the left arm. In an acute MI, severe restriction of blood flow in the coronary conduit vessels leads to reduced oxygen delivery to the myocardium and a subsequent cascade of inflammatory reactions resulting in death (infarction) of myocardial tissue Rapid restoration of blood flow to jeopardized myocardium can limit necrosis and reduce mortality. AMI leads to rapid death of myocytes and vascular structures in the supplied region of the ventricle. The loss of myocytes, arterioles, and capillaries in the mfarcted area is irreversible, resulting with time in the formation of scarred tissue.
[0056] After the initial cell death due to lack of oxygen, there is a later phase of myocardial cell injury that likely results from an ensuing acute inflammatory reaction (Entman M. L. et al , 1991, FASEB J 5- 2529). Initially, the importance of an inflammatory reaction in mediating myocardial cell injury during AMI was recognized in animal studies which showed that corticosteroids could reduce infarction size by 20 to 35% (Libby P et al , 1973, J Clin Invest 52 599, Maclean D et al 1978, J Clin Invest 61 541) However, clinical application of methyl-predmsolone in AMI to minimize myocardial necrosis, was not successful mamly because this treatment interfered with scar formation and healing, leading in some patients to the development of aneurysm and rupture of the ventricle wall (Roberts R et al., 1976, Circulation 53 Suppl. I: 204) A similar effect has been observed in long-term experiments in rats (Maclean D. et al., 1978, J Clin Invest 61: 541). These disappointing results discouraged further clinical studies that aimed at reducing infarction size by attenuating the inflammatory reaction following AMI
[0057] Patients with AMI can be diagnosed by characteristically elevated levels of troponin, creatine kinase and myoglobin Troponin levels are now considered the criterion standard in defining and diagnosing MI, according to the American College of Cardiology (ACC)/Amencan Heart Association (AHA) consensus statement on MI. Cardiac troponin levels (troponin-T and troponm-I) have a greater sensitivity and specificity than myocardial muscle creatine kinase (CK MB) levels in detecting MI They have important diagnostic and prognostic roles. Positive troponin levels are considered diagnostic of MI in the most recent ACC/ AHA revisions, because of their combined specificity and sensitivity in this diagnosis Serum levels typically increase within 3 12 hours from the onset of chest pain, peak at 24 48 hours, and return to baseline over 5-14 days.
[0058] Creatine kinase comprises 3 isoenzymes, including creatine kinase with muscle subunits (CK-MM), which is found mainly in skeletal muscle, creatine kinase with brain subunits (CK-BB), predominantly found in the brain, and myocardial muscle creatine kinase (CK-MB), which is found mainly in the heart. Serial measurements of CK-MB isoenzyme levels were previously the standard criterion for diagnosis of MI CK-MB levels typically increase within 3-12 hours of onset of chest pain reach peak values within 24 hours, and return to baseline after 48-72 hours Levels peak earlier (wash out) if reperfusion occurs Sensitivity is approximately 95%, with high specificity However, sensitivity and specificity are not as high as for troponin levels
[0059] Urine myoglobin levels rise within 1-4 hours from the onset of chest pain in AMI. Myoglobin levels are highly sensitive but not specific, and they may be useful within the context of other studies and in early detection of MI in the ED [0060] The electrocardiogram (ECG) can be an important tool in the initial evaluation and triage of patients in whom an MI is suspected It is confirmatory of the diagnosis in approximately 80% of cases It is recommended to obtain an ECG immediately if MI is considered or suspected. In patients with infenor MI, a right-sided ECG is recorded to rule out right ventricular (RV) infarct. Convex ST- segment elevation with upright or inverted T waves is generally indicative of MI in the appropriate clinical setting ST depression and T- wave changes may also indicate evolution of MI (non-ST-elevated MI). Progression of MI can be evaluated by performing ECGs serially, e.g. daily serial ECGs for the first 2-3 days and additionally as needed
[0061] Imaging studies can be helpful for diagnosis of MI, particularly if the diagnosis is questionable. An echocardiogram can identify regional wall motion abnormalities indicating tissue damage or death. An echocardiogram can also define the extent of the infarction and assess overall left ventπcle (LV) and right ventricle (RV) function. In addition, an echocardiogram can identify complications, such as acute mitral regurgitation (MR), LV rupture, or pericardial effusion.
[0062] Myocardial perfusion imaging (MPI) utilizes an intravenously administered radiopharmaceutical to depict the distribution of blood flow in the myocardium The radiopharmaceutical distribution in the heart is imaged using a gamma camera Perfusion abnormalities, or defects, are assessed and quantified as to location, extent and intensity. Myocardial perfusion imaging can identify areas of reduced myocardial blood flow associated with infarct.
[0063] Cardiac catheterization defines the patient's coronary anatomy and the extent of the blockage(s) via cardiac angiography.
[0064] AMI may be distinguished from chronic myocardial infarction using any appropriate method known in the art. In some embodiments, the presence of myocardial edema involving a disruption of the energy-regulated ionic transport mechanisms across the cell membrane after the MI is indicative of AMI (Willerson et al , 1977, Am J Pathol 87:159— 188) The relatively large extracellular matrix of the developed scar allows gadolmium-based contrast media to accumulate, resulting in DE. T2-weighted CMR sensitively detects lnfarct- associated myocardial edema (Wisenberg et al., 1988, Am Heart J. 115.510-518, Higgjns et al., 1983, Am J Cardiol 52 184-188; Garcia-Dorado et al., 1993, Cardiovasc Res 27:1462- 1469) and may be used to differentiate acute from chronic MI. In certain embodiments, a combination of delayed enhancement (DE) and T2-weighted cardiovascular magnetic resonance (CMR) is used to differentiate acute from chronic MI (Abdel-Aty et al. , 2004, Circulation 109 2411-2416)
Congestive Heart Failure (CHF)
[0065] In some embodiments, methods are provided wherein the incidence of congestive heart failure (CHF) or complications of CHF are reduced when a therapeutic having an anti fibrotic effect is administered to said patient. The incidence of CHF or complications of CHF are reduced relative to the incidence of CHF or complications of CHF in the absence of administration of the therapeutic (e g , m comparison to a patient who was not administered the therapeutic) The incidence of CHF may be reduced by at least 10% when a therapeutic having an anti-fibrotic effect is administered to a patient in comparison to a patient who was not administered the therapeutic In further embodiments, the incidence of CHF may be reduced by at least 15%, or at least 20%, or at least 25%, or at least 30%, or at least 35%, or at least 40%, or at least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95% or more when a therapeutic having an anti-fibrotic effect is administered to a patient in comparison to a patient who was not administered the therapeutic
[0066] The prevalence of congestive heart failure has been growing as the population ages and as cardiologists are more successful at reducing mortality from ischemic heart disease, the most common cause of congestive heart failure Roughly 4.6 million people in the United States have heart failure with an incidence approaching 10 per 1000 after age 65 years Hospital discharges for congestive heart failure rose from 377,000 in 1979 to 957,000 in 1997 making congestive heart failure the most common discharge diagnosis in people age 65 and over The five year mortality from congestive heart failure approaches 50%
[0067] CHF may be a complication of AMI and results from a decline in the pumping capacity of the heart CHF can also result from cardiac malformations, such as valve disease, or other disorders that damage cardiac tissue, e.g cardiac myopathy. Due to the activation of one or more compensatory mechanisms, the damaging changes caused by CHF can be present and ongoing even while the patient remains asymptomatic In fact, the compensatory mechanisms which maintain normal cardiovascular function duπng the early phases of CHF may actually contribute to progression of the disease, for example by exerting deleterious effects on the heart and circulation [0068] Some of the more important pathophysiologic changes which occur in CHF are activation of the hypothalamic -pituitary- adrenal axis, systemic endothelial dysfunction and myocardial remodeling.
[0069] Therapies specifically directed at counteracting the activation of the hypothalamic- pituitary-adrenal axis include beta-adrenergic blocking agents (β-blockers), angiotensin converting enzyme (ACE) inhibitors, certain calcium channel blockers, nitrates and endothelm-1 blocking agents. Calcium channel blockers and nitrates, while producing clinical improvement have not been clearly shown to prolong survival whereas β-blockers and ACE inhibitors have been shown to significantly prolong life, as have aldosterone antagonists.
[0070] Systemic endothelial dysfunction is a well-recognized feature of CHF and is clearly present by the time signs of left ventricular dysfunction are present. Endothelial dysfunction is important with respect to the intimate relationship of the myocardial microcirculation with cardiac myocytes. The evidence suggests that microvascular dysfunction contributes significantly to myocyte dysfunction and the morphological changes which lead to progressive myocardial failure.
[0071] Myocardial remodeling is a complex process which accompanies the transition from asymptomatic to symptomatic heart failure, and may be described as a series of adaptive changes within the myocardium. Components of myocardial remodeling may include fibrosis, alterations in myocyte biology, loss of myocytes by necrosis or apoptosis, alterations in the extracellular matrix and alterations in left ventricular chamber geometry.
[0072] The diagnosis of congestive heart failure is most often a clinical one that is based on knowledge of the patient's pertinent medical history, a careful physical examination, and selected laboratory tests. Symptoms include dyspnea (shortness of breath) which worsens upon lying supine, fluid retention and swelling in the lungs and extremities, e.g. with pulmonary rales or edema in the legs.
[0073] Congestive heart failure is strongly suggested by the presence of cardiomegaly (enlarged heart) or pulmonary vascular congestion on chest X-ray. Electrocardiogram (ECG) may show anterior Q waves or left bundle branch block on the electrocardiogram. The echocardiogram is the diagnostic standard for identifying congestive heart failure The patient may undergo two-dimensional echocardiography with Doppler flow studies. Radionuclide angiography or contrast cineangiography may be helpful if the echocardiogram is equivocal. Preservation of Viable Cardiac Tissue and Reduction of Infarct Size
[0074] In some embodiments, methods are provided wherein the cardiac tissue is preserved from necrosis when a therapeutic having an anti-fibrotic effect is administered to a patient suffering an AMI, in comparison to the amount of viable cardiac tissue in the absence of administration of the therapeutic (e.g., in comparison to a patient who was not administered a therapeutic). The amount of cardiac tissue preserved from necrosis can be increased at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%. The increase in viable cardiac tissue can be determined by MRI or computerized tomography (CT) scan.
[0075] Methods are also provided herein to control or reduce myocardial infarct size. "Control" or "controlling" as used herein means to reduce, reduce the incidence of, or prevent the progression of a disorder. In some cases, methods are provided wherein the infarct size of a patient is reduced when a therapeutic is administered to said patient, in comparison to the infarct size of a patient in the absence of administration of the therapeutic (e.g., in comparison to a patient who was not administered a therapeutic). The infarct size can be reduced at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%. The reduction in infarct size can be determined by MRI and/or by voltage/conduction mapping.
[0076] In some embodiments, methods are provided wherein the cardiac function is preserved when a therapeutic having an anti-fibrotic effect is administered to a patient suffering an AMI, in comparison to the cardiac function of a patient suffering an AMI in the absence of administration of the therapeutic (e.g., in comparison to a patient who was not administered a therapeutic). Preservation of cardiac function can be determined by measuring ejection fraction using echocardiography, wherein the ejection fraction can be improved by at least 1%, at least 3%, at least 5%, at least 7%, at least 10%, at least 12%, or at least 15%. Preservation of cardiac tissue can also be determined by measuring ejection fraction using MRI, wherein the ejection fraction can be improved by at least 1%, at least 3%, at least 5%, at least 7%, at least 10%, at least 12%, or at least 15%, and/or the infarct size can be decreased by at least 1%, at least 3%, at least 5%, at least 7%, at least 10%, at least 12% or at least 15%. Other methods of determining cardiac function are known in the art and include but are not limited to nuclear imaging, functional capacity, exercise capacity, New York Heart Association (NYHA) functional classification system, and myocardial oxygen consumption (MV02)
Reduction in the Incidence of Ventricular Tachycardia
[0077] In other cases, methods are provided wherein the incidence of ventricular tachycardia in a patient is reduced when a therapeutic is administered to said patient, in compaπson to the incidence of ventricular tachycardia in a patient who was not administered the therapeutic The incidence of ventricular tachycardia can be reduced at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80% at least 85%, at least 90%, or at least 95% The reduction in incidence of tachycardia can be determined by electrocardiogram (ECG or EKG) or by echocardiogram
Ventricular Fibrillation
[0078] In some embodiments, methods are provided for treating or preventing ventπcular fibrillation in a patient in need thereof, comprising administering to the patient a therapeutic having an anti fibrotic effect In some embodiments, the amount or degree of ventricular fibrillation is reduced relative to the amount or degree of ventricular fibrillation in the absence of administration of the therapeutic
[0079] Ventπcular fibrillation (VF) is a condition in which the heart s electrical activity becomes disordered When this happens, the heart s ventπcles contract in a rapid, unsynchronized way The ventricles "quiver" rather than beat, causing the heart to pump little or no blood
[0080] VF is life threatening and requires prompt treatment Without medical treatment, collapse and sudden cardiac death can occur Ventricular fibrillation (VF) may occur spontaneously with unpredictable timing and requires specialized tests to acquire an accurate diagnosis.
[0081] VF may be diagnosed using an electrocardiogram (ECG or EKG), e g a Holter Monitor -- A Holter monitor is a small, portable machine that records the patient's ECG and is typically worn for 24 hours This monitor may detect arrhythmias that might not show up on a resting electrocardiogram, which only records a heartbeat for a few seconds at rest
[0082] VF may also be diagnosed using an event monitor This is a small monitor about the size of a pager that the patient can have for up to a month. Since the arrhythmia may occur at unpredictable times, this monitor records the abnormal rhythm when the patient signals that he or she is experiencing symptoms
[0083] An exercise stress or treadmill test also may be used to diagnose VF, by recording the electrical activity of the patient's heart during exercise, which differs from the heart's electrical activity at rest.
[0084] Another method of diagnosing VF is through an electrophysiology study In an electrophysiology (EP) study, physiciansinsert special electrode catheters — long, flexible wires — into veins and guide them into the heart These catheters sense electrical impulses and also may be used to stimulate different areas of the heart Physicians can then locate the sites that are causing arrhythmias. The EP study allows physicians to examine an arrhythmia under controlled conditions and acquire more accurate, detailed information than with any other diagnostic test.
[0085] VF can be monitored and measured by any one or more of the parameters described, for example, in Example 5 below. In some embodiments, the incidence of VF can be reduced by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%, compared to incidence of VF in a patient who was not administered the therapeutic.
Sudden Cardiac Death
[0086] Sudden cardiac death (also called sudden arrest) is death resulting from an abrupt loss of heart function (cardiac arrest). The victim may or may not have diagnosed heart disease. The time and mode of death are unexpected It occurs within minutes after symptoms appear The most common underlying reason for patients to die suddenly from cardiac arrest is AMI due to coronary heart disease Other types of arrhythmia can also cause cardiac arrest.
[0087] Most of the cardiac arrests that lead to sudden death occur when the electrical impulses in the diseased heart become rapid (ventricular tachycardia) or chaotic (ventricular fibrillation) or both. This irregular heart rhythm (arrhythmia) causes the heart to suddenly stop beating. Some cardiac arrests are due to extreme slowing of the heart, bradycardia If a cardiac arrest was due to ventricular tachycardia or ventricular fibrillation, survivors are at higher risk for another arrest, especially if they have underlying heart disease.
[0088] Therefore, in some cases, methods are provided wherein the incidence of sudden cardiac death is reduced when a therapeutic having an anti-fibrotic effect is administered to said patient, m comparison to the incidence of cardiac death m a patient who was not administered a therapeutic. The incidence of sudden cardiac death can be reduced at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
Arrhythmia
[0089] Methods of the invention are contemplated to control arrhythmia by administering a therapeutic having an anti-fibrotic effect In some embodiments, a method is provided to reduce the incidence or risk of arrhythmia. The incidence or risk can be reduced at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
[0090] An arrhythmia is an abnormal heart rhythm. In an arrhythmia the heartbeats may be too slow, too rapid, too irregular, or too early. There are many types of arrhythmias, including premature atrial contractions (early extra beats that originate in the atria (upper chambers of the heart), premature ventricular contractions (PVCs) (skipped heartbeat), atrial fibrillation (an irregular heart rhythm that causes the atria, the upper chambers of the heart to contract abnormally), atrial flutter (an arrhythmia caused by one or more rapid circuits in the atrium), paroxysmal supraventricular tachycardia (PSVT) (a rapid heart rate, usually with a regular rhythm, originating from above the ventricles), accessory pathway tachycardias (a rapid heart rate due to an extra abnormal pathway or connection between the atria and the ventricles), AV nodal reentrant tachycardia (a rapid heart rate due to more than one pathway through the AV node), ventricular tachycardia (VT) (a rapid heart rhythm originating from the lower chambers (or ventricles) of the heart), ventricular fibrillation (an erratic, disorganized firing of impulses from the ventricles), bradyarrhythmias (slow heart rhythms, which may arise from disease in the heart's electrical conduction system), and/or long QT syndrome (the QT interval is the area on the electrocardiogram (ECG) that represents the time it takes for the heart muscle to contract and then recover, or for the electrical impulse to fire impulses and then recharge). When the QT interval is longer than normal, it increases the risk for "torsade de pomtes," a life-threatening form of ventricular tachycardia.
[0091] Symptoms of arrhythmia include chest pain, fainting, fast or slow heartbeat (palpitations), light-headedness, dizziness, paleness, shortness of breath, skipping beats, changes in the pattern of the pulse,and sweating. Arrythmias may be diagnosed by those of skill in the art using such methods as electrocardiogram, Holter monitor, event monitor, stress test, echocardiogram, cardiac catheterization, electrophysiology study (EPS), and head-up tilt table test
[0092] The amount of a therapeutic effective to control arrhythmia may be an amount effective to reduce ventπcular remodeling, e.g. in an animal model or during clinical trial. Ventricular remodeling refers to the changes in size, shape, and function of the heart after injury to the left ventricle The injury is typically due to AMI. In some embodiments, the ventricular remodeling is due to ventricular fibrosis caused by an AMI. The remodeling process is characterized by progressive expansion of the initial infarct area and dilation of the left ventricular lumen, with cardiomyocyte replacement by fibrous tissue deposition in the ventricular wall (Kocher et al., 2001, Nature Medicine 7(4): 430-6). Another integral component of the remodeling process is the development of neoangiogenesis within the myocardial infarct scar, a process requiring activation of latent collagenase and other proteinases. Under normal circumstances, the contribution of neoangiogenesis to the lnfarct- bed capillary network is insufficient to keep pace with the tissue growth required for contractile compensation and is unable to support the greater demands of the hypertrophied but viable myocardium. The relative lack of oxygen and nutrients to the hypertrophied myocytes might be an important etiological factor in the death of otherwise viable myocardium, resulting in progressive infarct extension and fibrous replacement Late reperfusion of the infarct vascular bed in both humans and animal models is known to significantly benefit ventricular remodeling and survival (Kocher et al., 2001, Nature Medicine 7(4): 430-6).
Therapeutic Agents
[0093] Therapeutic agents used in the disclosed methods can be any therapeutic agent that affects fibrosis Contemplated agents include agents that reduce the activity of transforming growth factor-beta (TGF-β) (including but not limited to GC-1008 (Genzyme/Medlmmune); lerdelimumab (CAT- 152; Trabio, Cambridge Antibody); metelimumab(CAT-192,Cambridge Antibody,); LY-2157299 (Eh Lilly); ACU-HTR-028 (Opko Health)) including antibodies that target one or more TGF-β isoforms, inhibitors of TGF-β receptor kinases TGFBRl (ALK5) and TGFB R2, and modulators of post-receptor signaling pathways; chemokme receptor signaling; endothelin receptor antagonists including inhibitors that target both endothehn receptor A and B and those that selectively target endothelin receptor A (including but not limited to ambrisentan; avosentan, bosentan, clazosentan, darusentan; BQ- 153; FR- 139317, L-744453, macitentan, PD-145065; PD-156252, PD163610,PS-433540; S-0139; sitaxentan sodium; TBC-3711; zibotentan); agents that reduce the activity of connective tissue growth factor (CTGF) (including but not limited to FG-3019, FibroGen), and also including other CTGF-neutralizing antibodies; matrix metalloproteinase (MMP) inhibitors (including but not limited to MMPI- 12, PUP-I and tigapotide tnflutate); agents that reduce the activity of epidermal growth factor receptor (EGFR) including but not limed to erlotinib, gefitmib, BMS-690514, cetuximab , antibodies targeting EGF receptor, inhibitors of EGF receptor kinase, and modulators of post-receptor signaling pathways; agents that reduce the activity of platelet derived growth factor (PDGF) (including but not limited to Imatinib mesylate (Novartis)) and also including PDGF neutralizing antibodies, antibodies targeting PDGF receptor (PDGFR), inhibitors of PDGFR kinase activity, and post-receptor signaling pathways; agents that reduce the activity of vascular endothelial growth factor (VEGF) (including but not limited to axitmib, bevacizumab, BIBF-1120, CDP-791, CT-322, DVIC- 18Fl, PTC- 299, and ramucirumab) and also including VEGF-neutralizmg antibodies, antibodies targeting the VEGF receptor 1 (VEGFRl, FIt-I) and VEGF receptor 2 (VEGFR2, KDR), the soluble form of VEGFRl (sFlt) and derivatives thereof which neutralize VEGF, and inhibitors of VEGF receptor kinase activity, inhibitors of multiple receptor kinases such as BIBF-1120 which inhibits receptor kinases for vascular endothelial growth factor, fibroblast growth factor, and platelet derived growth factor; agents that interfere with integrin function (including but not limited to STX-100 and IMGN-388) and also including mtegrin targeted antibodies; agents that interfere with the pro-fibrotic activities of IL-4 (including but not limited to AER-001, AMG-317, APG-201, and sIL-4Rα) and IL-13 (including but not limited to AER-001, AMG-317, anrukinzumab, CAT-354, cmtredekin besudotox, MK-6105, QAX-576, SB-313, SL-102, and TNX-650) and also including neutralizing anti-bodies to either cytokine, antibodies that target IL-4 receptor or IL-13 receptor, the soluble form of IL- 4 receptor or derivatives thereof that is reported to bind and neutralize both IL-4 and IL-13, chimeric proteins including all or part of IL-13 and a toxin particularly pseudomonas endotoxin, signaling though the JAK-STAT kinase pathway; agents that interfere with epithelial mesenchymal transition including inhibitors of mTor (including but not limited to AP-23573); agents that reduce levels of copper such as tetrathiomolybdate; agents that reduce oxidative stress including N-acetyl cysteine and tetrathiomolybdate; and interferon gamma Also contemplated are agents that are inhibitors of phosphodiesterase 4 (PDE4) (including but not limited to Roflumilast); inhibitors of phosphodiesterase 5 (PDE5) (including but not limited to mrrodenafil, PF-4480682, sildenafil citrate, SLx-2101, tadalafil, udenafil, UK- 369003, vardenafil, and zapπnast); or modifiers of the arachidonic acid pathway including cyclooxygenase and 5-lipoxegenase inhibitors (including but not limited to Zileuton). Further contemplated are compounds that reduce tissue remodeling or fibrosis including prolyl hydrolase inhibitors (including but not limited to 1016548, CG-0089, FG-2216, FG- 4497, FG-5615, FG-6513, fibrostatin A (Takeda), lufironil,P-1894B, and safironil) and peroxisome proliferator- activated receptor (PPAR)-gamma agonists, (including but not limited to pioglitazone and rosiglitazone,)
[0094] In some embodiments, formula (I), (II), (III), (IV), or (V) defined above are
Figure imgf000026_0001
wherein
R1, R2, R3, R4, X1, X2, X3, X4, X5, Y1, Y2, Y3, and Y4 are independently selected from the group consisting of H, deuterium, C1-C10 alkyl, C1-C10 deuterated alkyl, substituted Ci- Cio alkyl, C1-C10 alkenyl, substituted C1-C10 alkenyl, C1-C10 thioalkyl, C1-C10 alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxyl, Ci-Cio alkoxyalkyl, Ci-Cio carboxy, Ci-Cio alkoxycarbonyl, CO-uronide, CO-monosaccharide, CO-oligosaccharide, and CO- polysacchande;
X6 and X7 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkylenylaryl, alkylenylheteroaryl, alkylenylheterocycloalkyl, alkylenylcycloalkyl, or X6 and X7 together form an optionally substituted 5 or 6 membered heterocyclic ring; and Ar is pyridinyl or phenyl; and Z is O or S; or a pharmaceutically acceptable salt, ester, solvate, or prodrug of pirfenidone or the compound of formula (I), (II), (III), (IV), or (V).
[0095] In some embodiments, R1, R2, R3, R4, X1, X2, X3, X4, X5, Y1, Y2, Y3, and Y4 are independently optionally substituted pyrazinyl, optionally substituted pyridazinyl, optionally substituted pyrrolyl, optionally substituted thiophenyl, optionally substituted thiazolyl, optionally substituted oxazolyl, optionally substituted imidazolyl, optionally substituted isoxazolyl, optionally substituted pyrazolyl, optionally substituted isothiazolyl, optionally substituted napthyl, optionally substituted quinolinyl, optionally substituted isoquinolinyl, optionally substituted quinoxalinyl, optionally substituted benzothiazolyl, optionally substituted benzothiophenyl, optionally substituted benzofuranyl, optionally substituted indolyl, or optionally substituted benzimidazolyl,
[0096] In some cases, the therapeutic is a compound of formula (II), wherein X is H, OH, or C1-i0alkoxy, Z is O, and R2 is methyl, C(=O)H, C(=O)CH3, C(=O)O-glucosyl, fluoromethyl, difluoromethyl, trifluoromethyl, methylmethoxyl, methylhydroxyl, or phenyl; and R4 is H or hydroxy!
[0097] Some specific contemplated compounds of formula (II) include
Figure imgf000027_0001
Figure imgf000027_0002
Figure imgf000028_0001
, a compound listed in Table 1, below, and pharmaceutically acceptable salts, esters, solvates, and prodrugs thereof.
[0098] Other specific therapeutic agents contemplated include relaxin, ufironil, surifonil, a TGF-β antibody, CAT-192, CAT-158; ambresentan, thelin; FG-3019, a CTGF antibody; anti- EGFR antibody;a EGFR kinase inhibitor; tarceva; gefitinib; PDGF antibody, PDGFR kinase inhibitor; gleevec; BIBF-1120, VEGF, FGF, and PDGF receptor inhibitor; anti-integπn antibody; IL-4 antibody; tetrathiomolybdate, a copper chelating agent; interferon-gamma; NAC, a cysteine pro-drug; hepatocyte growth factor (HGF); KGF; angiotension receptor blockers, ACE inhibitors, rennin inhibitors; COX and LO inhibitors; Zileuton; monteleukast; avastin; statins; PDE5 inhibitors, such as sildenafil, udenafil, tadalafil, vardenafil, or zaprinast; rofumilast; etanercept (Enbrel); procoagulant; prostaglandins, such as PGE2, PRX- 08066, a 5HT2B receptor antagonist; cintredekin besudotox, a chimeric human ILl 3 conjugated to a genetically engineered Pseudomonas exotoxin; roflumilast, a PDE4 inhibitor; FG-3019, an anti-connective tissue growth factor human monoclonal antibody; GC-1008, a TGF-β human monoclonal antibody; treprostinil, a prostacyclin analog; interferon-α; QAX- 576, a IL13 modulator, WEB 2086, a PAF-receptor antagonist, imatinib mesylate, FG-1019; Suramin; Bosentan; IFN — Ib; anti-IL-4; anti-IL-13; taurine, niacin, NF-κB antisense oligonucleotides; and nitric oxide synthase inhibitors Also contemplated are peroxisome proliferator-activated receptor (PPAR)-gamma agonists, including but not limited to pioglitazone and rosightazone
[0099] The term "alkyl" used herein refers to a saturated or unsaturated straight or branched chain hydrocarbon group of one to ten carbon atoms, including, but not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-hexyl, and the like. Alkyls of one to six carbon atoms are also contemplated The term "alkyl" includes "bridged alkyl," i e , a bicyclic or polycyclic hydrocarbon group, for example, norbornyl, adamantyl, bicyclo[2.2.2]octyl, bicyclo[2.2.1]heptyl, bicyclo[3.2.1]octyl, or decahydronaphthyl. Alkyl groups optionally can be substituted, for example, with hydroxy (OH), halo, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, and ammo. It is specifically contemplated that in the analogs described herein the alkyl group consists of 1-40 carbon atoms, preferably 1-25 carbon atoms, preferably 1-15 carbon atoms, preferably 1-12 carbon atoms, preferably 1-10 carbon atoms, preferably 1-8 carbon atoms, and preferably 1-6 carbon atoms. "Heteroalkyl" is defined similarly as alkyl, except the heteroalkyl contains at least one heteroatom independently selected from the group consisting of oxygen, nitrogen, and sulfur.
[0100] As used herein, the term "cycloalkyl" refers to a cyclic hydrocarbon group, e.g., cyclopropyl, cyclobutyl, cyclohexyl, and cyclopentyl. "Heterocycloalkyl" is defined similarly as cycloalkyl, except the ring contains one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur. Nonlimiting examples of heterocycloalkyl groups include piperdine, tetrahydrofuran, tetrahydropyran, dihydrofuran, morpholine, thiophene, and the like. Cycloalkyl and heterocycloalkyl groups can be saturated or partially unsaturated ring systems optionally substituted with, for example, one to three groups, independently selected from the group consisting of alkyl, alkyleneOH, C(O)NH2, NH2, oxo (=O), aryl, haloalkyl, halo, and OH. Heterocycloalkyl groups optionally can be further N-substituted with alkyl, hydroxyalkyl, alkylenearyl, or alkyleneheteroaryl.
[0101] The term "alkenyl" used herein refers to a straight or branched chain hydrocarbon group of two to ten carbon atoms containing at least one carbon double bond including, but not limited to, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl, and the like
[0102] The term "halo" used herein refers to fluoro, chloro, bromo, or iodo. [0103] The term "alkylene" used herein refers to an alkyl group having a substituent. For example, the term "alkylene aryl" refers to an alkyl group substituted with an aryl group. The alkylene group is optionally substituted with one or more substituent previously listed as an optional alkyl substituent. For example, an alkylene group can be -CH2CH2-.
[0104] As used herein, the term "alkenylene" is defined identical as "alkylene," except the group contains at least one carbon-carbon double bond.
[0105] As used herein, the term "aryl" refers to a monocyclic or polycyclic aromatic group, preferably a monocyclic or bicyclic aromatic group, e.g., phenyl or naphthyl. Unless otherwise indicated, an aryl group can be unsubstituted or substituted with one or more, and in particular one to four groups independently selected from, for example, halo, alkyl, alkenyl, OCF3, NO2, CN, NC, OH, alkoxy, amino, CO2H, CO2alkyl, aryl, and heteroaryl. Exemplary aryl groups include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl, chlorophenyl, methylphenyl, methoxyphenyl, trifluoromethylphenyl, nitrophenyl, 2,4- methoxychlorophenyl, and the like.
[0106] As used herein, the term "heteroaryl" refers to a monocyclic or bicyclic ring system containing one or two aromatic rings and containing at least one nitrogen, oxygen, or sulfur atom in an aromatic ring. Unless otherwise indicated, a heteroaryl group can be unsubstituted or substituted with one or more, and in particular one to four, substituents selected from, for example, halo, alkyl, alkenyl, OCF3, NO2, CN, NC, OH, alkoxy, amino, CO2H, CO2alkyl, aryl, and heteroaryl. Examples of heteroaryl groups include, but are not limited to, thienyl, furyl, pyridyl, oxazolyl, quinolyl, thiophenyl, isoquinolyl, indolyl, triazinyl, triazolyl, isothiazolyl, isoxazolyl, imidazolyl, benzothiazolyl, pyrazinyl, pyrimidinyl, thiazolyl, and thiadiazolyl.
[0107] The term "deuterated alkyl" used herein refers to an alkyl group substituted with one or more deuterium atoms (D).
[0108] The term "thioalkyl" used herein refers to one or more thio groups appended to an alkyl group.
[0109] The term "hydroxyalkyl" used herein refers to one or more hydroxy groups appended to an alkyl group.
[0110] The term "alkoxy" used herein refers to straight or branched chain alkyl group covalently bonded to the parent molecule through an --O— linkage. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, n- butoxy, sec-butoxy, t-butoxy and the like. [0111] The term "alkoxyalkyl" used herein refers to one or more alkoxy groups appended to an alkyl group.
[0112] The term "arylalkoxy" used herein refers to a group having an aryl appended to an alkoxy group. A non-limiting example of an arylalkoxy group is a benzyloxy (Ph-CH2-O-).
[0113] The term "amino" as used herein refers to -NR2, where R is independently hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl. Non-limiting examples of amino groups include NH2 and N(CH3)2. In some cases, R is independently hydrogen or alkyl.
[0114] The term "amido" as used herein refers to -C(O)NH2, -C(O)NR2, -NRC(O)R or - NHC(O)H, where each R is independently hydrogen, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl. In some cases, the amido group is -NHC(O)alkyl or -NHC(O)H. A non-limiting example of an amido group is -NHC(O)CH3.
[0115] The term "carboxy" or "carboxyl" used herein refers to — COOH or its deprotonated form -COO . C1-10carboxy refers to optionally substituted alkyl or alkenyl groups having a carboxy moiety. Examples include, but are not limited to, -CH2COOH, - CH2CH(COOH)CH3, and -CH2CH2CH2COOH.
[0116] The term "alkoxycarbonyl" refers to — (CO) — O-alkyl, wherein the alkyl group can optionally be substituted Examples of alkoxycarbonyl groups include, but are not limited to, methoxycarbonyl group, ethoxycarbonyl group, propoxycarbonyl group, and the like.
[0117] The term "alkylcarbonyl" refers to — (CO)-alkyl, wherein the alkyl group can optionally be substituted. Examples of alkylcarbonyl groups include, but are not limited to, methylcarbonyl group, ethylcarbonyl group, propylcarbonyl group, and the like.
[0118] The term "sulfonamido" refers to— SO2NR2, wherein R is independently hydrogen, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl. In some cases, the sulfonamido group is -SO2NR2 where R is independently hydrogen or an optionally substituted alkyl. Examples of a sulfonamido group include, but are not limited to, -SO2N(CH3)2 and -SO2NH2. [0119] The term "sulfonyl" refers to -SO2R, where R is independently hydrogen or an optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl In some cases, a sulfonyl group is -SO2alkyl, wherein the alkyl group can optionally be substituted One example of a sulfonyl group is methylsulfonyl (e.g., - SO2CH3)
[0120] The term "sulfoxyl" refers to -SOR, where each R is independently hydrogen or an optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl One example of a sulfonyl group is methylsulfonyl (e g , SOCH3)
[0121] Carbohydrates are polyhydroxy aldehydes or ketones, or substances that yield such compounds upon hydrolysis. Carbohydrates comprise the elements carbon (C), hydrogen (H) and oxygen (O) with a ratio of hydrogen twice that of carbon and oxygen. In their basic form, carbohydrates are simple sugars or monosaccharides These simple sugars can combine with each other to form more complex carbohydrates The combination of two simple sugars is a disaccharide Carbohydrates consisting of two to ten simple sugars are called oligosaccharides, and those with a larger number are called polysaccharides
[0122] The term "uronide" refers to a monosaccharide having a carboxyl group on the carbon that is not part of the ring The uronide name retains the root of the monosaccharide, but the -ose sugar suffix is changed to -uronide. For example, the structure of glucuronide corresponds to glucose.
[0123] As used herein, a radical indicates species with a single, unpaired electron such that the species containing the radical can be covalently bonded to another species Hence, in this context, a radical is not necessarily a free radical Rather, a radical indicates a specific portion of a larger molecule. The term "radical" can be used interchangeably with the term "group."
[0124] As used herein, a substituted group is derived from the unsubstituted parent structure in which there has been an exchange of one or more hydrogen atoms for another atom or group A "substituent group," as used herein, means a group selected from the following moieties
(A) OH, NH2, SH, CN, CF3, NO2, oxo, halogen, unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, unsubstituted alkoxy, unsubstituted aryloxy, tπhalomethanesulfonyl, tnfluoromethyl, and
(B) alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, ammo, amido, carbonyl, thiocarbonyl, alkoxycarbonyl, silyl, sulfonyl, sulfoxyl, alkoxy, aryloxy, and heteroaryl, substituted with at least one substituent selected from:
(i) -OH, -NH2, -SH, -CN, -CF3, -NO2, oxo, halogen, unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, unsubstituted alkoxy, unsubstituted aryloxy, trihalomethanesulfonyl, tnfluoromethyl, and
(11) alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, ammo, amido, carbonyl, thiocarbonyl, alkoxycarbonyl, silyl, sulfonyl, sulfoxyl, alkoxy, aryloxy, and heteroaryl, substituted with at least one substituent selected from/
(a) -OH, -NH2, -SH, -CN, -CF3, -NO2, oxo, halogen, unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, unsubstituted alkoxy, unsubstituted aryloxy, tπhalomethanesulfonyl, tnfluoromethyl, and
(b) alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, ammo, amido, carbonyl, thiocarbonyl, alkoxycarbonyl, silyl, sulfonyl, sulfoxyl, alkoxy, aryloxy, and heteroaryl, substituted with at least one substituent selected from -OH, -NH2, -SH, -CN, -CF3, -NO2, oxo, halogen, unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, unsubstituted alkoxy, unsubstituted aryloxy, trihalomethanesulfonyl, tnfluoromethyl
[0125] In some embodiments, the substituent group is a "size-limited substituent" or "size- limited substituent group," which refers to a group selected from all of the substituents described above for a "substituent group," wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C1-C20 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C4-C8 cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 4 to 8 membered heterocycloalkyl
[0126] In some embodiments, the substituent group is a "lower substituent" or "lower substituent group," which refers to a group selected from all of the substituents descnbed above for a "substituent group," wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C1-C8 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C5-C7 cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 5 to 7 membered heterocycloalkyl
[0127] In some cases, the substituent group(s) is (are) one or more group(s) individually and independently selected from alkyl, cycloalkyl, aryl, fused aryl, heterocyclyl, heteroaryl, hydroxy, alkoxy, aryloxy, mercapto, alkylthio, arylthio, cyano, halo, carbonyl, thiocarbonyl, alkoxycarbonyl, nitro, silyl, tnhalomethanesulfonyl, trifluoromethyl, and amino, including mono and di substituted amino groups, and the protected derivatives thereof.
[0128] The protecting groups that can form the protective derivatives of the above substituents are known to those of skill in the art and can be found in references such as Greene and Wuts, Protective Groups in Organic Synthesis; 3rd Edition, John Wiley and Sons: New York, 2006. Wherever a substituent is described as "optionally substituted" that substituent can be substituted with the above-described substituents.
[0129] Asymmetric carbon atoms can be present. All such isomers, including diastereomers and enantiomers, as well as the mixtures thereof, are intended to be included in the scope of the disclosure herein. In certain cases, compounds can exist in tautomeric forms. All tautomeric forms are intended to be included in the scope of the disclosure herein. Likewise, when compounds contain an alkenyl or alkenylene group, there exists the possibility of cis- and trans- isomeric forms of the compounds. Both cis- and trans- isomers, as well as the mixtures of cis- and trans- isomers, are contemplated.
[0130] Compounds that can be used in the disclosed methods include those described in U.S. Patent Publication No. 2007/0049624 (US national stage of WO 05/0047256), International Publication No. WO 03/068230, WO 08/003141, WO 08/157786, or in U.S. Patent Nos. 5,962,478; 6,300,349; 6,090,822; 6,114,353; Re. 40,155; 6,956,044; or 5,310,562. Synthesis of the compounds used in the disclosed methods can be by any means known in the art, including those described in the patents and patent publications listed herein. Other synthetic means can be used and are within the knowledge of the skilled artisan
[0131] One class of compounds contemplated for use in the disclosed methods is a deuterated (D) form of any of the compounds disclosed herein. One specific such compound is a compound having a CD3 moiety and/or a D to replace any or all of the methyl or hydrogens of pirfenidone. Examples include
Figure imgf000035_0001
, and . The synthesis of these compounds can be found in International Patent Publication No. WO 08/157786.
[0132] Some specific compounds of formula (I), (II), (III), or (IV) are listed in Table 1. Description of the synthesis of these compounds can be found in U.S. Provisional Application Nos. 61/058,436, filed June 3, 2008 and 61/074,446, filed June 20, 2008, the disclosures of which are each incorporated by reference herein.
Table 1
Cmpd Structure Cmpd Structure Cmpd Structure
No. No. No.
Figure imgf000035_0002
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000039_0002
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
373 375 Intentionally blank
Figure imgf000057_0001
Figure imgf000057_0002
Figure imgf000058_0001
Figure imgf000058_0002
Figure imgf000058_0003
Figure imgf000059_0001
Figure imgf000059_0002
Figure imgf000060_0001
Figure imgf000060_0002
Figure imgf000060_0003
Figure imgf000061_0001
Figure imgf000061_0002
Figure imgf000061_0003
Figure imgf000062_0001
Figure imgf000062_0002
Figure imgf000062_0003
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000064_0002
Figure imgf000064_0003
Figure imgf000065_0001
[0133] Other specific compounds of formula (I), (II), (III), or (IV) also include the following compounds.
Figure imgf000065_0002
Figure imgf000066_0001
Figure imgf000066_0002
Figure imgf000066_0004
Figure imgf000066_0003
Figure imgf000067_0001
Figure imgf000068_0001
[0134] Other compounds contemplated for use m the disclosed methods include compounds of Genus F, II', III', and IV, below. Synthesis of compounds of Genus F, IF, III', and IV are described in detail in International Patent Publication No. WO 07/062167, incorporated by reference in its entirety herein.
Figure imgf000068_0002
wherein each of R , R2 , R3 , R4 , and R6 is independently selected from the group consisting of H, halo, cyano, nitro, hydroxy, optionally substituted Ci 6 alkyl, optionally substituted C3 7 cycloalkyl, optionally substituted C4 10 alkylcycloalkyl, optionally substituted C26 alkenyl, optionally substituted Ci 6 alkoxy, optionally substituted Cόor 10 aryl> optionally substituted pyπdinyl, optionally substituted pyπmidmyl, optionally substituted thienyl, optionally substituted furanyl, optionally substituted thiazolyl, optionally substituted oxazolyl, optionally substituted phenoxy, optionally substituted thiophenoxy, optionally substituted sulphonamido, optionally substituted urea, optionally substituted thiourea, optionally substituted amido, optionally substituted keto, optionally substituted carboxyl, optionally substituted carbamyl, optionally substituted sulphide, optionally substituted sulphoxide, optionally substituted sulphone, optionally substituted amino, optionally substituted alkoxyammo, optionally substituted alkyoxyheterocyclyl, optionally substituted alkylammo, optionally substituted alkylcarboxy, optionally substituted carbonyl, optionally substituted spirocyclic cycloalkyl, optionally substituted pyrazinyl, optionally substituted pyridazmyl, optionally substituted pyrrolyl, optionally substituted thiophenyl, optionally substituted thiazolyl, optionally substituted oxazolyl, optionally substituted lmidazolyl, optionally substituted isoxazolyl, optionally substituted pyrazolyl, optionally substituted isothiazolyl, optionally substituted napthyl, optionally substituted quinolmyl, optionally substituted lsoqumolinyl, optionally substituted quinoxalmyl, optionally substituted benzothiazolyl, optionally substituted benzothiophenyl, optionally substituted benzofuranyl, optionally substituted indolyl, and optionally substituted benzimidazolyl, or a pharmaceutically acceptable salt, ester, solvate or prodrug thereof.
[0135] The salts, e.g., pharmaceutically acceptable salts, of the disclosed therapeutics may be prepared by reacting the appropriate base or acid with a stoichiometric equivalent of the therapeutic. Similarly, pharmaceutically acceptable derivatives (e.g., esters), metabolites, hydrates, solvates and prodrugs of the therapeutic may be prepared by methods generally known to those skilled in the art Thus, another embodiment provides compounds that are prodrugs of an active compound. In general, a prodrug is a compound which is metabolized in vivo (e.g., by a metabolic transformation such as deamination, dealkylation, de- esterification, and the like) to provide an active compound A "pharmaceutically acceptable prodrug" means a compound which is, withm the scope of sound medical judgment suitable for pharmaceutical use in a patient without undue toxicity, irritation, allergic response, and the like, and effective for the intended use, including a pharmaceutically acceptable ester as well as a zwitteriomc form, where possible, of the therapeutic As used herein, the term "pharmaceutically acceptable ester" refers to esters that hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms Representative examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates. Examples of pharmaceutically-acceptable prodrug types are described m Higuchi and Stella, Pro-drugs as Novel Delivery Systems, VoI 14 of the A.C S Symposium Series, and m Roche, ed , Bwreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference
[0136] The compounds and compositions described herein may also include metabolites As used herein, the term "metabolite" means a product of metabolism of a compound of the embodiments or a pharmaceutically acceptable salt, analog, or derivative thereof, that exhibits a similar activity in vitro or in vivo to a disclosed therapeutic. The compounds and compositions described herein may also include hydrates and solvates. As used herein, the term "solvate" refers to a complex formed by a solute (herein, the therapeutic) and a solvent. Such solvents for the purpose of the embodiments preferably should not negatively interfere with the biological activity of the solute. Solvents may be, by way of example, water, ethanol, or acetic acid. In view of the foregoing, reference herein to a particular compound or genus of compounds will be understood to include the various forms described above, including pharmaceutically acceptable salts, esters, prodrugs, metabolites and solvates thereof.
Dosing and Pharmaceutical Formulations
[0137] The terms "therapeutically effective amount" and "prophylactic ally effective amount," as used herein, refer to an amount of a compound sufficient to treat, ameliorate, or prevent the identified disease or condition, or to exhibit a detectable therapeutic, prophylactic, or inhibitory effect The effect can be detected by, for example, an improvement in clinical condition, reduction in symptoms, or by any of the assays or clinical diagnostic tests descπbed herein. The precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration. Therapeutically and prophylactically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician.
[0138] The therapeutics disclosed herein can be dosed at a total amount of about 50 to about 2400 mg per day. The dosage can be divided into two or three doses over the day or given in a single daily dose. Specific amounts of the total daily amount of the therapeutic contemplated for the disclosed methods include about 50 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 267 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 534 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, about 1000 mg, about 1050 mg, about 1068 mg, about 1100 mg, about 1150 mg, about 1200 mg, about 1250 mg, about 1300 mg, about 1335 mg, about 1350 mg, about 1400 mg, about 1450 mg, about 1500 mg, about 1550 mg, about 1600 mg, about 1650 mg, about 1700 mg, about 1750 mg, about 1800 mg, about 1850 mg, about 1869 mg, about 1900 mg, about 1950 mg, about 2000 mg, about 2050 mg, about 2100 mg, about 2136 mg, about 2150 mg, about 2200 mg, about 2250 mg, about 2300 mg, about 2350 mg, and about 2400 mg.
[0139] Dosages of the therapeutic can alternately be administered as a dose measured in mg/kg. Contemplated mg/kg doses of the disclosed therapeutics include about 1 mg/kg to about 60 mg/kg. Specific ranges of doses in mg/kg include about 1 mg/kg to about 20 mg/kg, about 5 mg/kg to about 20 mg/kg, about 10 mg/kg to about 20 mg/kg, about 25 mg/kg to about 50 mg/kg, and about 30 mg/kg to about 60 mg/kg.
[0140] In methods where the patient has suffered an AMI, administration of the therapeutic can be initiated at 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, 31 days, 32 days, 33 days, 34 days, 35 days, 36 days, 37 days, 38 days, 39 days, 40 days, 41 days, or 42 days after suffering the AMI. Also contemplated is initiation of the treatment about 1-40 days, about 1-30 days, about 1-25 days, about 1-20 days, about 1-14 days, about 1-10 days, about 2-40 days, about 3-40 days, about 3-38 days, about 3-30 days, about 3-25 days, about 3-20 days, about 3-15 days, about 3-14 days, about 3-10 days, about 4-36 days, about 4-30 days, about 4-25 days, about 4-20 days, about 4-14 days, about 5-40 days, about 5-34 days, about 5-30 days, about 5-25 days, about 5-20 days, about 5-14 days, about 6-40 days, about 6-32 days, about 6-30 days, about 6-25 days, about 6-20 days, about 6-14 days, about 7-40 days, about 7-30 days, about 7-25 days, about 7-20 days, about 7-14 days, about 8-28 days, about 9-26 days, about 10-24 days, about 12-22 days, about 13-20 days, or about 14-18 days after suffering the AMI. Treatment, e.g., continued administration of the therapeutic can continue for at least a week, at least 2 weeks, at least 3 weeks, at least a month, at least 6 weeks, at least 2 months, at least 3 months, at least 4 months, at least 5 months, at least 6 months, or at least a year. For example, the treatment can be for up to 3 months, up to 4 months, up to 5 months, or up to 6 months. In some embodiments, a patient suffering an AMI continues to be administered the therapeutic for a time period up to 4 weeks after suffering the AMI, e g , the therapeutic continues to be administered on the day that is 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, and/or 28 days after suffering the AMI.
[0141] As described elsewhere herein, the compounds described herein may be formulated in pharmaceutical compositions with a pharmaceutically acceptable excipient, carrier, or diluent. The compound or composition comprising the compound can be administered by any route that permits treatment of the disease or condition. A preferred route of administration is oral administration. Additionally, the compound or composition comprising the compound may be delivered to a patient using any standard route of administration, including parenterally, such as intravenously, mtrapentoneally, mtrapulmonary, subcutaneously or intramuscularly, intrathecally, transdermally, rectally, orally, nasally or by inhalation. Slow release formulations may also be prepared from the agents described herein in order to achieve a controlled release of the active agent in contact with the body fluids m the gastro intestinal tract, and to provide a substantial constant and effective level of the active agent in the blood plasma. The crystal form may be embedded for this purpose m a polymer matrix of a biological degradable polymer, a water-soluble polymer or a mixture of both, and optionally suitable surfactants. Embedding can mean in this context the incorporation of micro-particles in a matrix of polymers. Controlled release formulations are also obtained through encapsulation of dispersed micro-particles or emulsified micro-droplets via known dispersion or emulsion coating technologies.
[0142] Administration may take the form of single dose administration, or the compound of the embodiments can be administered over a period of time, either in divided doses or in a continuous-release formulation or administration method (e g , a pump) However the compounds of the embodiments are administered to the subject, the amounts of compound administered and the route of administration chosen should be selected to permit efficacious treatment of the disease condition.
[0143] In an embodiment, the pharmaceutical compositions may be formulated with pharmaceutically acceptable excipients such as carriers, solvents, stabilizers, adjuvants, diluents, etc., depending upon the particular mode of administration and dosage form. The pharmaceutical compositions should generally be formulated to achieve a physiologically compatible pH, and may range from a pH of about 3 to a pH of about 11, preferably about pH 3 to about pH 7, depending on the formulation and route of administration. In alternative embodiments, it may be preferred that the pH is adjusted to a range from about pH 5 0 to about pH 8 More particularly, the pharmaceutical compositions may comprise a therapeutically or prophylactically effective amount of at least one compound as described herein, together with one or more pharmaceutically acceptable excipients. Optionally, the pharmaceutical compositions may compπse a combination of the compounds described herein, or may include a second active ingredient useful in the treatment or prevention of bacterial infection (e.g., anti-bacterial or anti-microbial agents). In various embodiments, examples of a therapeutic agent that may be used alone or in combination with another therapeutic agent according to the methods of the present invention include, but are not limited to, an agent that reduces tissue remodeling or fibrosis, reduces the activity of transforming growth factor-beta (TGF-β), targets one or more TGF-β isoforms, inhibits TGF- β receptor kinases TGFBRl (ALK5) and/or TGFBR2, or modulates one or more post- receptor signaling pathways, is an endothelin receptor antagonists, targets both endothehn receptor A and endothelin receptor B or selectively targets endothelin receptor A, reduces activity of connective tissue growth factor (CTGF), inhibits matrix metalloproteinase (MMP), particularly MMP-9 and/or MMP-12, reduces the activity of epidermal growth factor receptor (EGFR), targets the EGF receptor, or inhibits EGF receptor kinase, reduces the activity of platelet derived growth factor (PDGF), targets PDGF receptor (PDGFR), inhibits PDGFR kinase activity, or inhibits post- PDGF receptor signaling pathways, reduces the activity of vascular endothelial growth factor (VEGF), targets one or more of VEGF receptor 1 (VEGFRl, FIt-I), VEGF receptor 2 (VEGFR2, KDR), inhibits multiple receptor kinases as in the case of BIRB-1120 which inhibits receptor kinases for vascular endothelial growth factor, fibroblast growth factor, and platelet derived growth factor, interferes with integrin function, particularly integrin αVβ6, interferes with pro-fibrotic activities of IL-4 and IL-13, targets IL-4 receptor, IL-13 receptor, modulates signaling though the JAK-STAT kinase pathway, interferes with epithelial mesenchymal transition, inhibits mTor, reduces levels of copper, reduces oxidative stress, inhibits prolyl hydrolase, inhibits phosphodiesterase 4 (PDE4) or phosphodiesterase 5 (PDE5), modifies the arachidomc acid pathway, or acts as an agonist of PPAR-γ.
[0144] Formulations, e.g., for parenteral or oral administration, are most typically solids, liquid solutions, emulsions or suspensions, while inhalable formulations for pulmonary administration are generally liquids or powders, with powder formulations being generally preferred. A preferred pharmaceutical composition may also be formulated as a lyophihzed solid that is reconstituted with a physiologically compatible solvent prior to administration. Alternative pharmaceutical compositions may be formulated as syrups, creams, ointments, tablets, and the like
[0145] The term "pharmaceutically acceptable excipient" refers to an excipient for administration of a pharmaceutical agent, such as the compounds described herein. The term refers to any pharmaceutical excipient that may be administered without undue toxicity.
[0146] Pharmaceutically acceptable excipients are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there exists a wide variety of suitable formulations of pharmaceutical compositions (see, e.g., Remington's Pharmaceutical Sciences).
[0147] Suitable excipients may be carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Other exemplary excipients include antioxidants (e g., ascorbic acid), chelating agents (e.g., EDTA), carbohydrates (e.g., dextrin, hydroxyalkylcellulose, and/or hydroxyalkylmethylcellulose), stearic acid, liquids (e g , oils, water, saline, glycerol and/or ethanol) wetting or emulsifying agents, pH buffering substances, and the like. Liposomes are also included withm the definition of pharmaceutically acceptable excipients.
[0148] The pharmaceutical compositions described herein may be formulated in any form suitable for an intended method of administration. When intended for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, non-aqueous solutions, dispersible powders or granules (including micronized particles or nanoparticles), emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
[0149] Pharmaceutically acceptable excipients particularly suitable for use in conjunction with tablets include, for example, inert diluents, such as celluloses, calcium or sodium carbonate, lactose, calcium or sodium phosphate; disintegrating agents, such as cross-linked povidone, maize starch, or alginic acid; binding agents, such as povidone, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. [0150] Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
[0151] Formulations for oral use may be also presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example celluloses, lactose, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with non-aqueous or oil medium, such as glycerin, propylene glycol, polyethylene glycol, peanut oil, liquid paraffin or olive oil
[0152] In another embodiment, pharmaceutical compositions may be formulated as suspensions comprising a compound of the embodiments in admixture with at least one pharmaceutically acceptable excipient suitable for the manufacture of a suspension.
[0153] In yet another embodiment, pharmaceutical compositions may be formulated as dispersible powders and granules suitable for preparation of a suspension by the addition of suitable excipients
[0154] Excipients suitable for use in connection with suspensions include suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelhilose, sodium alginate, polyvinylpyrrolidone, gum tragacanth, gum acacia), dispersing or wetting agents (e.g., a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g. , heptadecaethyleneoxycethanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g. , polyoxyethylene sorbitan monooleate)); and thickening agents (e.g., carbomer, beeswax, hard paraffin or cetyl alcohol). The suspensions may also contain one or more preservatives (e.g., acetic acid, methyl or n- propyl p-hydroxy-benzoate); one or more coloring agents; one or more flavoring agents, and one or more sweetening agents such as sucrose or saccharin.
[0155] The pharmaceutical compositions may also be in the form of oil-m water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth; naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids; hexitol anhydrides, such as sorbitan monooleate; and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
[0156] Additionally, the pharmaceutical compositions may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous emulsion or oleaginous suspension. This emulsion or suspension may be formulated by a person of ordinary skill in the art using those suitable dispersing or wetting agents and suspending agents, including those mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,2-propane-diol.
[0157] The steπle injectable preparation may also be prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile fixed oils may be employed as a solvent or suspending medium For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids (e g., oleic acid) may likewise be used in the preparation of injectables
[0158] To obtain a stable water-soluble dose form of a pharmaceutical composition, a pharmaceutically acceptable salt of a compound described herein may be dissolved in an aqueous solution of an organic or inorganic acid, such as 0.3 M solution of succinic acid, or more preferably, citric acid. If a soluble salt form is not available, the compound may be dissolved in a suitable co-solvent or combination of co-solvents Examples of suitable co- solvents include alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from about 0 to about 60% of the total volume. In one embodiment, the active compound is dissolved in DMSO and diluted with water
[0159] The pharmaceutical composition may also be in the form of a solution of a salt form of the active ingredient in an appropriate aqueous vehicle, such as water or isotonic salme or dextrose solution. Also contemplated are compounds which have been modified by substitutions or additions of chemical or biochemical moieties which make them more suitable for delivery (e.g., increase solubility, bioactivity, palatabihty, decrease adverse reactions, etc.), for example by esterification, glycosylation, PEGylation, etc [0160] In a preferred embodiment, the compounds described herein may be formulated for oral administration in a lipid-based formulation suitable for low solubility compounds. Lipid-based formulations can generally enhance the oral bioavailability of such compounds
[0161] As such, a preferred pharmaceutical composition comprises a therapeutically or prophylactically effective amount of a compound described herein, together with at least one pharmaceutically acceptable excipient selected from the group consisting of medium chain fatty acids and propylene glycol esters thereof (e g., propylene glycol esters of edible fatty acids, such as caprylic and capric fatty acids) and pharmaceutically acceptable surfactants, such as polyoxyl 40 hydrogenated castor oil.
[0162] In an alternative preferred embodiment, cyclodextrins may be added as aqueous solubility enhancers. Preferred cyclodextrins include hydroxypropyl, hydroxyethyl, glucosyl, maltosyl and maltotnosyl derivatives of α-, β-, and γ-cyclodextrin. A particularly preferred cyclodextnn solubility enhancer is hydroxypropyl-o-cyclodextπn (BPBC), which may be added to any of the above-described compositions to further improve the aqueous solubility characteristics of the compounds of the embodiments. In one embodiment, the composition comprises about 0 1% to about 20% hydroxypropyl-o-cyclodextrin, more preferably about 1% to about 15% hydroxypropyl-o-cyclodextrin, and even more preferably from about 2 5% to about 10% hydroxypropyl-o-cyclodextrin. The amount of solubility enhancer employed will depend on the amount of the compound of the invention in the composition.
[0163] The methods of the embodiments also include the use of a compound or compounds as described herein together with one or more additional therapeutic agents for the treatment of disease conditions Thus, for example, the combination of active ingredients may be: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by any other combination therapy regimen known in the art When delivered in alternation therapy, the methods described herein may comprise administering or delivering the active ingredients sequentially, e.g., in separate solution, emulsion, suspension, tablets, pills or capsules, or by different injections in separate syringes In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in simultaneous therapy, effective dosages of two or more active ingredients are administered together. Various sequences of intermittent combination therapy may also be used.
[0164] The invention will be more fully understood by reference to the following examples which detail exemplary embodiments of the invention. They should not, however, be construed as limiting the scope of the invention. All citations throughout the disclosure are hereby expressly incorporated by reference
EXAMPLES EXAMPLE 1
Experimental Myocardial Infarction (MI) Protocol
[0165] In this example, a protocol is described for examining the ventricular function, extent of fibrosis and VT inducibility in an ischemia reperfusion rat model after pirfenidone treatment. Ventricular function was assessed via echocardiography. VT inducibility was assessed by programmed stimulation and EP study The electrophysiological properties were assessed using high resolution optical mapping, and the extent of fibrosis was studied using standard histological techniques
[0166] After baseline echocardiography, thirty male Sprague-Dawley rats, ages 6-10 weeks, underwent myocardial infarction using an ischemia-reperfusion model Briefly, rats were anesthetized using inhaled isoflurane (5% induction, 2 5% maintenance, 02 output 1 L/min) and positioned supine on an electrically warmed animal surgery platform Rats were intubated using a 16 gauge i.v. catheter and then ventilated using a Harvard rodent respirator After a left thoracotomy and pericardiotomy were performed, a 7-0 Ticron suture was introduced into the myocardium, using the left atrial appendage and right outflow tract as landmarks. The depth of entry was 2 mm, which was slightly greater than the level of the left coronary artery Both suture ends were then threaded through a PE-90 polyethylene tube 6 in in length to form a "snare loop" around the artery, closed by pulling on the free ends of the suture The snare loop was tested by closing and releasing after 10-seconds to demonstrate adequate ischemia and reperfusion The suture was then tightened to occlude the artery for 20 minutes and then removed to allow for reperfusion. The chest was then closed with 5 0 prolene suture, and the animal was allowed to recover. After one week and repeat echocardiography, rats were randomized to placebo rodent feed (control group, n=15) or rodent feed mixed with 1.2% pirfenidone (PFD) (treatment group, n=15) for four weeks. All experiments and data analyses were performed with the operator blinded to treatment group
Statistical Analysis
[0167] Statistical comparisons for the studies described herein were made between groups by using the paired or unpaired t tests, unless noted otherwise. Fisher's Exact test was used to compare VT inducibility between control and PFD treatment groups. All values are reported as means ± SEM. P < 0.05 was considered significant
EXAMPLE 2
Echocardiographic Analysis of Experimental Models
[0168] At baseline, and at 1 wk and 5 wk after infarction, a commercially available high- resolution echocardiographic system (Vevo 660, VisualSonics, Toronto, ON, Canada) equipped with a 25-MHz mechanical transducer was used for echocardiography. Rats were placed supine on a warming platform, and ECG limb electrodes were attached. To minimize ultrasound attenuation, the chests were shaved and cleaned with a chemical hair remover (Nair). Aquasonic 100 gel (Parker Laboratories, Fairfield, NJ) was applied to the thoracic surface to optimize visibility of the cardiac chambers. Parasternal long-axis and parasternal short-axis two-dimensional views were acquired.
[0169] Using the long-axis view, left ventricular (LV) end-systolic and end-diastolic volumes (ESV and EDV), as well as LV ejection fraction (LVEF), were calculated by using frames with the maximal and minimal cross-sectional area and width. The system software utilizes a formula based on a cylindrical-hemiellipsoid model (volume = 8 area2 ÷ 3 ÷ length). LVEF was calculated using the following formula: (EDV-ESV)/EDV 100 Fractional shortening (FS) was evaluated from the M mode of the parasternal long-axis view at the papillary muscle level on the basis of the percent changes of LV end-diastolic and end- systolic diameters. LV mass was estimated using the following equation at end diastole. LV mass = 1.05 (epicardial volume - endocardial volume), where volume is based on the cylindrical-hemiellipsoid model. These evaluations of LV function in the rodent are well validated. Echocardiographic acquisition and analysis were obtained while blinded to the treatment group.
[0170] Serial echocardiography at baseline, 1 week post-MI, and 5 weeks post-MI, showed evidence of progressive LV remodeling for rats in both groups, including LV dilatation, increases in EDV and ESV, and decreases in ejection fraction. However, the pirfenidone- treated group had significantly less decline in its ejection fraction (from 68±6% to 45±14% in the control group and from 66±5% to 36+15% in the PFD treated group) (Figure 1). During the treatment period (week 1 to week 5) there was a significantly (p=0.005) lower percent decrease in EF in the pirfenidone-treated rats (8 6%) compared to controls (24.3%). EXAMPLE 3
Electrophysiologic Analysis and Evaluation of Arrhythmias in Experimental Models
[0171] Optical mapping is a technique to perform high-resolution electrophysiologic evaluation of the cardiac tissue. To summarize the procedure, ten thousand simultaneous optical action potentials were recorded with a 100 x 100 CMOS camera within a 19 mm x 19 mm mapping field on the epicardium of the LV anterior wall. Using a 1000-W tungsten- halogen light source, fluorescence was excited with an excitation filter of 530 nm and transmitted with an emission long-pass filter of > 630 nm. Fluorescent optical maps were acquired at 2000 Hz during programmed electrical stimulation. Optical mapping was performed 5 wks after MI. Rats were injected with heparin (500 U ip) 15 min before excision of the heart, and were then anesthetized with pentobarbital sodium (50 mg/kg ip). After adequate anesthesia, the heart was rapidly excised and arrested by immersion in cold cardioplegia solution. The aorta was cannulated and retrogradely perfused, at a rate of 6 mL/min, with 37°C modified Tyrode solution containing (in mmol/L). 130 NaCl, 20 0 NaHCO3, 1 2 MgCl2, 4.0 KCl, 5.6 glucose, and 1.8 CaCl2, gassed with 95% 02/5% C02. Extraneous tissue was carefully removed from the heart The cannulated heart was then placed in 37°C Tyrode solution in a specialized temperature-controlled optical recording chamber (maintained at 37°C) while ECG, perfusion rate, and temperature were measured continuously for the duration of the experiment. Before optical recordings, Tyrode solution containing voltage- sensitive dye PGH I (10 μL of 5mM stock solution) was perfused through the preparation over a 5-mm period.
[0172] Once a cannulated heart was perfused with PGH I, it was placed in the optical chamber with its LV anterior wall pressed against the imaging window In order to include areas of normal, border zone, and infarct tissues within the mapping field, comparable mapping positions were used for all the hearts. During optical recordings, contractility was blocked with 15 mM butadione monoxime (BDM). Ventricular epicardium bipolar pacing, at a stimulus amplitude of 2X threshold, was performed on normal tissue near the infarct zone Mapping was recorded during pacing drives of 250 ms to 90 ms (decremented by 10 ms), as well as dunng S1-S2 pacing using a basic cycle length (BCL) of 200 ms and maximum S2 of 150 ms and decremented by 10 ms Programmed stimulation, with up to three extrastimuli, and burst pacing (from 90 ms to 60 ms) were used to assess arrhythmia inducibihty. Inducibility was defined as the ability to provoke sustained (> 30 s) ventricular tachycardia (VT) or ventricular fibrillation. Maps were also captured during programmed stimulation and with all episodes of arrhythmia.
[0173] Optical mapping data was analyzed using modified OMproCCD software (from Bum-rak Choi, Pittsburg, PA) and Matlab custom software. Raw fluorescence data was viewed as a movie of normalized fluorescence intensity, which revealed activation within the field of view. Quantitative data was obtained from optically derived action potentials (APs) for each of the 10,000 pixels of the CMOS camera. Activation time and action potential duration at 50% (APD50) and 80% repolarization (APD80) were measured for each paced cycle length (PCL). Activation time was calculated at the maximum rate of rise of the fluorescent AP (dF/dt) APD80 is the duration from the activation time (start of the action potential) to the time point where the action potential has recovered to 20% maximal fluorescent signal (peak of the optical AP). Isochronal maps of activation were constructed for each map. Rise time was calculated as the time between takeoff and at the peak of the action potential. The OMproCCD software was used to calculate conduction vectors representing conduction velocities and conduction direction at each pixel, as previously described. Phase differences, calculated as the average difference with neighboring activation times at each site, were measured to quantify the spatial heterogeneity of conduction, as previously described. Frequency histograms were constructed for the phase differences within a recorded area These histograms were summarized as the median phase time at 50th percentile (P50), and the 5th and 95th percentiles (P5 and P95, respectively) of the distribution The absolute degree of heterogeneity, or heterogeneity range, was quantified as the width of the distribution, P95-P5, while heterogeneity index was defined as the heterogeneity range divided by the median phase (P95-P5)/P50. All parameters were determined for both control and PFD groups and their respective non-infarct, border, and infarct zones. These zones were identified using the amplitude map of fluorescence, as previously described and validated. Transitions from areas of high amplitude (non-mfarct) to lowest amplitude (infarct) were considered border zones Further evidence from triphenyltetrazohum chloride (TTC) staining, imaging of the heart under normal light conditions, and from fluorescence images, were also used to corroborate amplitude maps.
VT lnducibility and Electrophysiologic Characterization
[0174] The rate of VT induction was 73 3% in control MI rats, which is consistent with what has been shown in the art The rate of VT induction for PFD animals, however, was significantly decreased, at 28.6% (p=0.027) [0175] Optical mapping was used to analyze conduction action potential properties Figure 2 shows the conduction velocities measured in the 3 areas of the LV in all animals. Conduction velocities at all paced cycle lengths in the remote non-mfarct zones of both control and PFD groups were similar between the two groups (Figure T). Conduction velocities in the infarct zones of both control and PFD groups were significantly slower than normal (and border zone areas) and were similar between the two groups (Figure 2) Conduction velocities in the border zones (the area that predisposes to post-MI ventricular tachycardia) of both groups were intermediate to that of the remote non-mfarct and infarct zones However, the conduction velocities in the border zones for the PFD group were significantly faster, at all PCLs, compared to those in the border zones of control animals (p <005, Figure T)
[0176] Figure 3 shows the conduction heterogeneity (which has been shown to be related to an increased propensity for arrhythmias) measured in both groups across all tested cycle lengths. There was a trend toward higher conduction heterogeneity indices in control animals compared to those of PFD animals (p=0.146). The difference in conduction through infarcts of similar size, for control and PFD animals were visualized in representative activation movies and showed more slowing and increased heterogeneity of conduction for the control animal All of these parameters have previously been demonstrated to be related to enhanced substrate for ventricular arrhythmias.
[0177] The maximal rate of AP rise (dF/dt) and πse times (duration from AP takeoff to peak of fluorescent AP) for control and PFD non-mfarct zones were similar respectively, as were the rise and rise times for control and PFD infarct zones respectively. However, there was a trend, at all PCLs, for the rise of PFD border zones to be faster than the rise of control border zones. Conversely, there was a trend, at all PCLs, for the rise times of PFD border zones to be lower than those of control border zones (Figure 4). This comparison is statistically significant at the lowest PCL tested (Figure 4)
[0178] The amount of fluorescence amplitude for the three zones was also quantified, shown in Figure 5. Normal areas had the highest amplitude, infarct areas the least, and border areas in the middle. A trend toward higher amplitudes of fluorescence in the border zones of pirfemdone-treated rats was noted, as compared to those of the controls (Figure 5). This suggested that pirfenidone may have had an impact on infarct expansion in the border zone (decreased scar expansion), since the pirfenidone border zones likely had more viable cardiomyocytes to emit the additional fluorescence. This was validated histologically by examining infarct sizes for these hearts (see below). EXAMPLE 4
Histological Analysis of Infarct Size and Fibrosis
[0179] Ventricular tissue samples were fixed in 10% neutral buffered formalin. The samples were embedded in paraffin, sectioned (10-μm thick), and then stained with Masson's trichrome or Sinus red with fast green counterstain. Stained slides were examined under light microscopy, digitized using a high-resolution scanner, and analyzed using Photoshop CS software. Infarct areas on Masson's trichome corresponded tightly with areas of dense Sinus red staining with minimal to no fast green. Infarct scar area and total area of left ventricular myocardium, for all sections, were manually traced in the digital images and automatically calculated by the software. Infarct size, expressed as a percentage, was measured by dividing the sum of infarct areas from all sections by the sum of LV areas from all sections and multiplying by 100.
[0180] The total area of fibrosis was also assessed After excluding the infarct area (defined as dense fibrosis), fibrosis in the border and non-infarct zones was quantified from digital photomicrographs of the Sinus red-stained sections. Areas containing blood vessels and perivascular interstitial cells were also excluded from fibrosis quantification. The red pixel content of digitized images relative to the total tissue area was counted by using the Adobe Photoshop CS software
Implications of Examples Described Above
[0181] The amount of infarct fibrosis was quantified as percent of total myocardium. Controls had almost twice as large an infarct (18%±2.7%) as the PFD group (10+1 9%, p=0.022) (Figure 6) The amount of fibrosis (including border zones and non-MI areas and infarct scar) was also less in the PFD group (13+3%), compared to controls (23+2%; p=0.01) (Figure 6).
[0182] Previous research [Breithardt et al. Eur Heart J (1989) 10 Suppl E: 9-18 ; Spach. Circ Res (2007) 101(8): 743-5; Spach et al J Cardiovasc Electrophysiol (1994) 5(2): 182- 209, Jacobson et al Heart Rhythm (2006) 3(2). 189-97; Marchlinski et al Circulation (2004) 110(16): 2293-8; Verheule et al Circ Res (2004) 94(11): 1458-65] has shown that fibrosis is strongly correlated with atrial and ventricular arrhythmias. Increased fibrosis leads to decoupling of muscle fibers, conduction slowing and conduction blocks, as well as "zig-zag" and chaotic conduction. The distnbution of fibrosis is also important: a finger-like distribution, as opposed to a more diffuse picture, is also thought to cause more disruption of wave propagation and is therefore more arrhythmogenic [Breithardt et al. Eur Heart J (1989) 10 Suppl E: 9-18]. After an MI, cardiac fibrosis in the infarct border zone has such a strmg- like distribution and is more likely to cause alterations of direction-directed electrical propagation with the fibrotic tissue interrupting normally tight cell-cell coupling. This is believed to contribute to slowing and heterogeneous conduction velocities, eventually setting up the formation of re-entrant circuits that predispose to ventricular arrhythmias. In the rodent ischemia-reperfusion model described herein, significant remodeling occurred over the course of 5 weeks post-MI Control animals had progressive LV dilation with decreased LVEF. Fibrosis occurred not only withm the infarct scar but also in the areas bordering the infarct (infarct border zone) and in normal myocardium distant to the infarct. Nonmfarct fibrosis is a well-described phenomenon after an MI and is believed to contribute to deleterious remodeling (both mechanically and electrophysiologically)
[0183] The observed fibrosis, particularly in the infarct border zone, correlated with slower conduction velocities in the border zone of control animals and suggests that the fibrosis had led to electrical uncoupling. Furthermore, compared to normal myocardium, the action potential rise was lower, and its rise time was longer in the border zone of control infarcts; these findings are all consistent with slower conduction velocities and increased conduction heterogeneity The altered and heterogeneous conduction velocities led to more inducible VT. These results are very similar to previously reported optical mapping studies for myocardial infarction in rodents, larger animals and humans.
[0184] The results highlight the role of fibrosis attenuation in the post-MI setting and its impact on LV function and VT mducibility. PFD, an antifibrotic drug, was shown to be able to decrease the amount of fibrosis in an ischemia-reperfusion rat model. This decrease in fibrosis correlated with a decrease m infarct expansion as well as with improved left ventricular function by echocardiography Further, it was shown that decreased fibrosis was associated with decreased VT susceptibility This was related to an improvement in conduction velocity and conduction heterogeneity, which are important contributors to the substrate for VT in the post-MI setting.
[0185] The animals undergoing ischemia-reperfusion myocardial infarction were not randomized to PFD treatment until after 1 week post-MI. Because clinical studies with antiinflammatory agents, particularly corticosteroids, have shown adverse outcomes in the post- MI setting, one concern was that treatment so early in the post infarct period would have impaired wound healing, thus causing a weaker scar and possibly increasing mortality due to CHF or cardiac rupture. Several studies have shown that 1 week after a myocardial infarction in rodents is a safe and efficacious time frame. No increased mortality, CHF, or arrhythmias in animals treated with PFD were noted On the contrary, and surprisingly, animals treated with PFD appeared to have less infarct expansion, improved LV function, and decreased VT susceptibility.
[0186] Pirfenidone attenuated the total amount of fibrosis, as well as extra-mfarct fibrosis. Despite delaying treatment until 1 week after the MI, PFD appeared to have an effect on decreasing the infarct size, compared to control infarcts. Therefore, absent the PFD intervention, ongoing remodeling changes may actually contribute to infarct expansion long after the initial ischemic insult. There is evidence that this is indeed the case, with studies indicating that cardiomyocyte death can occur in non-mfarcted myocardium, particularly within the infarct border zone, for weeks after an MI. Underlying mechanisms associated with this pathology include wall restructuring, side-to-side slippage of cells, and cardiac dilatation (Cheng, Kajstura et al. 1996; Olivetti, Capasso et al. 1990). Thus, by decreasing fibrosis, PFD improved cardiac remodeling, as evidenced by the improvement in LV function, and this likely contributed to the decrease in infarct size.
[0187] Fibrosis within the infarct border zone for PFD animals was not only decreased but its distribution appeared less heterogeneous, with less of the finger-like projections seen in control infarcts This decrease in erratic distribution, as well as in quantity of fibrosis, was associated with improved conduction velocities in PFD border zones A concurrent increase in action potential rise and faster rise time in PFD border zones further confirm these findings. These results, as well as decreased conduction heterogeneity, were likely responsible for the almost three-fold decrease in VT susceptibility in PFD animals.
EXAMPLE 5
Ventricular Fibrillation Mapping
[0188] Animal Models: Twenty-four dogs weighing 25-30 Kg were divided into three groups control (n=ll), congestive heart failure (n=7), and congestive heart failure with the antifibrotic drug pirfenidone (n = 6). Heart Failure (CHF) was induced m 7 dogs via four weeks of rapid ventricular pacing via a lead placed in the RV and pulse generator set to pace at 240 bpm followed by ablation of the AV node to create complete heart block, as described m Li, et al., Circulation 1999; 100:87-95. Ventricular function was monitored weekly with transthoracic echocardiography for 4 weeks. At 4 weeks, the optical mapping study was performed. Four weeks was chosen based on previous data demonstrating significant ventricular dilatation and remodeling, and decreased contractility in that time.
[0189] Heart Failure with Pirfenidone (PFD): Heart failure was induced in 6 dogs as described above and PFD was administered as described in Lee et al., Circulation 2006; 114; 1703-12. Oral PFD (800 mg 3 times per day; InterMune, Brisbane, CA) was started 2 days before the initiation of pacing and was discontinued >6 half- lives (24 hours) before the optical mapping study.
[0190] Optical Mapping Studies: A coronary perfused left ventricular preparation was used as described in Wu et al., / Cardwvasc Electrophyswl 1998; 9:1336-47. Briefly, following sedation with sodium pentothal (0.25 mg/Kg), a left lateral thoracotomy is performed and the heart was rapidly excised. It was then perfused with cardioplegic solution ((in mmol/L): NaCl 123, KCl 15, NaHCO3 22, NaH2PO4 0.65, MgCl20.50, glucose 5 5, CaCl2 2, bubbled with 95% O2/5% CO2) retrogradely through the aorta. The ventricles were removed at approximately 1 cm below the AV ring and the left anterior descending coronary artery (LAD) was perfused. The right ventricle was removed and the left ventricle was cut to the size that was perfused by the LAD and included a papillary muscle. All ventricular branches were then ligated
[0191] The ventricular preparation was then transferred to a tissue chamber maintained at 37°C. The perfusion line in the LAD was perfused with modified Tyrode's solution ((in mmol/L): NaCl 123, KCl 5.4, NaHCO3 22, NaH2PO40.65, MgCl2 0.50, glucose 5.5, CaCl2 2, bubbled with 95% O2/5% CO2). Prior to optical recordings, a bolus of 30 - 40 μl of the voltage sensitive dye PGH- 1 was injected directly into the perfusate.
[0192] With an optical mapping system described in Wu et al., J Cardwvasc Electrophysiol 1998; 9:1336-47, optical recordings were then made from 4-cm2 area on 3 surfaces of the preparation (epicardial, endocardial (including the papillary muscle, and transmural) by a 16 x 16 photodiode array (C4657 Hamamatsu, Bπdgewater, NJ) that recorded 256 simultaneous optical action potentials. During optical recordings from a preparation, contractility was blocked with 15 mM 2,3-butadione monoxime (BDM; Sigma- Aldnch) 11. Plunge electrodes were placed on the recording surface around the field of view for both pacing and monitoring. Two plunge electrodes were dedicated for recording a bipolar signal for monitoring the electrical activity of the preparation. VF was initiated with either extra stimuli or with rapid burst pacing at a cycle length of 50 ms, a pulse width of 9.9 ms, and an output of 9.9 mA. Several 4-s episodes of VF were recorded on each surface in each preparation Activation movies of the VF were then viewed, and the activation patterns were determined After termination of VF signals were obtained during pacing at 250ms and isochronal maps of activation were constructed to look at conduction Activation patterns and wave-front direction during VF were determined from raw fluorescence movies (isopotential). Activation was characterized as 1) spiral (single reentrant circuit dominating the epoch), 2) focal (discrete, high frequency location of activation), 3) multiple wavefront (rapidly changing or varying wave fronts with wave-front collision), or 4) one broad wavefront (single wave-front passing through the map) VF was defined as rapid and irregular activations on the bipolar signal used for monitoring the electπcal activity of the preparation.
[0193] Signal Processing and Frequency Domain Analysis: The signals obtained from the optical mapping recordings were sampled at 2,000 Hz, and for each signal the dominant frequency (DF) was determined and the organization was calculated as described Everett, et al., IEEE Trans Biomed Eng 2001,48:969 78. Briefly, a fast Fourier transform (FFT) was calculated on the digitally filtered waveform The data were detrended and multiplied by a Hamming window The largest peak of the resulting magnitude spectrum was identified, and the positions of the harmonic peaks were determined on the basis of its position The areas under the largest peak and three of its harmonic peaks were each calculated over a 1-Hz window This produced an area under four peaks The total area of the spectrum was calculated from 2 Hz up to but not including the fifth harmonic peak. The ratio of the power under the harmonic peaks to the total power in this range was calculated, and the resulting number was defined as the organization index (OI) The OI was theorized to represent the organization of AF for that signal at that period in time To calculate the variance of the DFs, spatial coefficient of variance (SD/mean) of the DFs during a single episode of AF among all recording sites and temporal coefficient of variance of average DFs from among AF episodes for each mapping field withm each preparation were calculated. Discrete, stable, high frequency areas were noted Stability was defined as persistence over at least 90% of the epoch, and if it disappeared, it would return in the same location
[0194] Cross Correlation Analysis: Spatial correlation analysis was performed on all recorded signals between all possible paired electrogram combinations in each animal. The cross-correlation function was calculated at zero lag for each electrogram combination, and the peak value was considered the correlation coefficient, representing the degree of correlation between the two signals. All of the correlation coefficients calculated from an AF recording with optical mapping were then averaged to produce a mean correlation value for each AF episode.
[0195] Statistical Analysis: Data were expressed as the mean + DF. For comparisons among all mapping analysis variables, a range of mixed effects models was used. The models employed dog-specific (independently and identically distributed) random effects to account for the repeated measures made on a dog both within and across recording locations. Various contrasts (sub-models of the overall model) were explored to determine the importance of the study groups, recording site, and the group by recording site interaction effects. These contrasts were tested with a Chi-squared likelihood ratio test in a nested model fashion. Statistical significance was defined as p<0.05.
[0196] VF Activation Patterns: On examination of the optical mapping activation sequences, 4 types of activation patterns were seen - spiral wave, focal area of activation, multiple waves, and on broad wavefront sweeping through the field of view. Table 2 shows the types of activation patterns that were seen on each mapped surface for each dog.
Figure imgf000088_0001
[0197] Epicardial Surface: For the Control group, only 2 of the 10 mapped epicardial surfaces showed evidence of focal activation. These two surfaces also corresponded to having stable, high DF areas. All others had activation patterns of either multiple wavelets or one broad wavefront dominating the field of view. The activation map, during pacing at 250 ms, shows homogeneous conduction throughout the field of view. Similar results were seen in the CHF and PFD groups. Both groups had 2/6 mapped surfaces having either focal activation or a spiral wave (1 CHF dog). These types of activation corresponded to stable, high DF areas. All other dogs had either multiple wavefronts or one broad wavefront dominating the field of view. These activation patterns had either transient DFs (multiple wavefronts) or the area was dommanted one DF (broad waveftont) The activation images show homogeneous conduction, similar to Control, but at a slower conduction velocity.
[0198] Endocardial Surface: Mapping of the endocardial surface included the papillary muscle and only the CHF group had AF characterized by stable, high DF areas that correlated to spiral waves or focal activation patterns. Three of the five mapped endocardial surfaces in the CHF group fell into this category. Even though 2 of 7 endocardial surfaces in the Control group had activation characterized by spiral waves, no discrete, stable DFs were observed. The other 5 Controls and all of the mapped endocardial surfaces in the PFD group had either multiple or broad wavefront activation. All of the groups showed heterogeneous conduction marked by conduction slowing. This is in contrast to the homogenous conduction seen on the epicardial surface.
[0199] Transmural Surface: The transmural surface had the highest percentage of spiral wave and focal activation when compared to the other mapped surfaces for all groups In the CHF group, the transmural surface was mapped in 5 dogs and all of them had VF activation patterns of either a spiral wave or focal activation. The VF was characterized by stable, discrete, high DF areas. In the PFD group, 50% of the mapped transmural surfaces had an activation pattern of a spiral wave that correlated to stable high DF areas. In the Control group, 75% of the transmural surfaces had focal activation. One of these did not correlate to stable, high DF areas. Each group showed heterogeneous conduction characterized by areas of conduction slowing and block.
[0200] Dominant Frequencies: Frequency domain analysis was used as a method to quantify the activation patterns that were recorded during VF. Table 2 shows where the stable, discrete high DF areas were seen. Six of 7 CHF dogs had at least one surface with a stable, high DF area. In this group, all of the transmural surfaces that were mapped had VF characterized by a discrete, stable high DF area. Only 3 of the 11 Controls and 3 of 6 PFD dogs had at least one surface with VF that was characterized by high DF areas. The epicardial surface of the control group had a VF mechanism of multiple wavefronts. High DF areas were noted in some examples, but these were not stable. Both the endocardial and transmural surfaces had VF characterized by one broad wavefront sweeping through the field of view. The corresponding DF maps are characterized by a single DF. For the CHF group, the epicardial surface had VF characterized by a broad wavefront, and the corresponding DF map was dominated by a singular DF. The endocardial and transmural surfaces both had VF characterized by stable, high DF areas. The VF mechanisms that these DF corresponded to were a focal mechanism on the endocardial surface and a spiral wave on the transmural surface. For the PFD group, a spiral wave was seen in the transmural surface and the corresponding DF map had a stable, high DF area. A focal mechanism was seen in the epicardial surface which resulted in a high DF area. The endocardial surface had VF characterized by multiple wavefronts and only transient DF areas were seen. Summary DF data is listed in Table 3. From the statistical analysis, only the coefficient of variance for temporal and spatial DFs had significant group and surface effects.
Figure imgf000091_0001
[0201] Organization and Cross Correlation Analysis: To further analyze the spatiotemporal organization of the VF recorded on each of the surfaces of each of the models, the organization index (OI) was used to measure the organization of the recordings by quantifying the differences in the resulting FFTs. Summary data from OI maps are shown in Table 4. As the table shows, the Control group had higher mean and maximum OI values than either the CHF or PFD groups. These differences reached significance in the endocardial surface. Within groups, the endocardial surface of the control group had higher OI levels than either the epicardial or transmural surfaces. In the PFD group, the endocardial surface had the lowest OI levels and this reached significance when compared to the transmural surface. The Control group also showed the most temporal stability in OI levels as this group had the lowest OI temporal CoV values at all surfaces with the lowest measurements found on the endocardial surface The endocardial and transmural surfaces of the CHF and PFD groups were significantly different than those of the Control group.
[0202] F or each VF episode, all possible pairs of signals were cross-correlated, and the average correlation coefficients for each surface of each group is shown in Figure 8A. Figure 7 shows the gradient of frequencies over distance across the endocardial surface, transmural suface and epicardial surface. Pirfendidone preserved the transmural gradient to that similar to control animals, whereas untreated animals with heart failure have a very large gradient.
Figure imgf000093_0001
Examples of embodiments of the invention include
1 A method of treating a patient who has suffered an acute myocardial infarction (AMI) comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein optionally the treatment is initiated at a time period about 1 to 42 days after suffering the AMI, and optionally continues for up to 3 to 6 months.
2 The method of paragraph 1 , wherein the method is to limit expansion of an infarct scar due to the AMI.
3 The method of paragraph 1, wherein the treatment is initiated about 5-10 days after the AMI.
4 The method of paragraph 3, wherein the treatment is initiated about 7 days after the AMI.
5 The method of any one of paragraphs 1-4, wherein the treatment is for at least 2 weeks.
6 A method of reducing the incidence of congestive heart failure in a patient who suffered an acute myocardial infarction (AMI), comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the therapeutically effective dose reduces the incidence of congestive heart failure.
7 The method of paragraph 6, wherein the patient is at an increased risk of congestive heart failure due to the AMI.
8 The method of paragraph 6 or 7, wherein the treatment is initiated about 1 to 42 days after the suffering of the AMI
9 A method of preserving viable cardiac tissue or controlling or reducing myocardial infarct size in a patient who has suffered an acute myocardial infarction (AMI) comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic to the patient results in a relatively reduced infarct size on average compared to infarct size in a patient who has not been administered the therapeutic.
10. The method of paragraph 9, wherein the administering is initiated 1-42 days after suffering the AMI. 11. The method of paragraph 9 or 10, wherein the relative reduction in infarct size is at least 5%.
12. A method of reducing the incidence of ventricular tachycardia in a patient in need thereof, comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic prevents or reduces the incidence of ventricular tachycardia.
13. The method of paragraph 12, wherein the patient has suffered an acute myocardial infarction (AMI).
14. The method of paragraph 13, wherein the administering is initiated about 1 to 42 days after the suffering of the AMI.
15. The method of paragraph 14, wherein the administering is initiated about 7 days after the suffering of the AMI.
16. A method of treating or preventing ventricular fibrillation in a patient in need thereof, comprising administering to the patient a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic prevents ventricular fibrillation in the patient.
17. The method of paragraph 16, wherein the patient has suffered an acute myocardial infarction (AMI)
18. The method of paragraph 17, wherein the administration is initiated about 1 to 42 days after the suffering of the AMI.
19. The method of paragraph 18, wherein the administration is initiated about 7 days after the suffering of the AMI.
20. The method of any one of paragraphs 16-19, wherein the administering reduces the incidence of sudden cardiac death.
21. The method of any one of paragraphs 16-20, wherein the administering reduces cardiac risk of the patient.
22. A method of controlling arrhythmia in a patient in need thereof, comprising administering to the patient a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic controls arrhythmia in the patient.
23. The method of paragraph 22, wherein the patient has suffered an acute myocardial infarction (AMI). 24. The method of paragraph 23, wherein the administration is initiated about 1 to 42 days after the suffering of the AMI.
25. The method of paragraph 24, wherein the administration is initiated about 7 days after the suffering of the AMI.
26. The method of any one of paragraphs 22-25, wherein the administering treats ventricular remodeling.
27. The method of any one of paragraphs 1-26, wherein the patient had not previously suffered an AMI.
28. The method of any one of paragraphs 1-27, wherein the therapeutic having an anti-fibrotic effect is a therapeutic that reduces tissue remodeling or fibrosis, reduces the activity of transforming growth factor-beta (TGF- β), targets one or more TGF-β isoforms, inhibits TGF-β receptor kinases TGFBRl (ALK5) and/or TGFBR2, or modulates one or more post-receptor signaling pathways; is an endothelin receptor antagonists, targets both endothelin receptor A and endothelin receptor B or selectively targets endothelin receptor A; reduces activity of connective tissue growth factor (CTGF); inhibits matrix metalloproteinase; reduces the activity of epidermal growth factor (EGF), targets the EGF receptor, or inhibits EGF receptor kinase; reduces the activity of platelet derived growth factor (PDGF), targets PDGF receptor (PDGFR), inhibits PDGFR kinase activity, or inhibits post- PDGF receptor signaling pathways; reduces the activity of vascular endothelial growth factor (VEGF), targets one or more of VEGF receptor 1 (VEGFRl, FIt-I), VEGF receptor 2 (VEGFR2, KDR), the soluble form of VEGFRl (sFlt) and derivatives thereof which neutralize VEGF, inhibits VEGF receptor kinase activity; inhibits multiple receptor kinases such as BIRB-1120 which inhibits receptor kinases for vascular endothelial growth factor, fibroblast growth factor, and platelet derived growth factor; interferes with integrin function; interferes with pro-fibrotic activities of IL-4 and IL-13, targets IL-4 receptor, IL-13 receptor, the soluble form of IL-4 receptor or derivatives thereof; modulates signaling though the JAK-STAT kinase pathway; interferes with epithelial mesenchymal transition, inhibits mTor; reduces levels of copper, reduces oxidative stress; inhibits prolyl hydrolase; inhibits phosphodiesterase 4 (PDE4) or phosphodiesterase 5 (PDE5), or modifies the arachidonic acid pathway.
29. The method of any one of paragraphs 1-28, wherein the therapeutic is pirfenidone or compound of formula (I), (II), (III), (IV), or (V) or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof
Figure imgf000097_0001
wherein
A is N or CR2; B is N or CR4; E is N or CX4; G is N or CX3; J is N or CX2; K is N or CX1; a dashed line is a single or double bond,
R1, R2, R3, R4, X1, X2, X3, X4, X5, Y1, Y2, Y3, and Y4 are independently selected from the group consisting of H, deuterium, Ci-C io alkyl, C1-C10 deuterated alkyl, substituted Ci- Cio alkyl, C1-C10 alkenyl, substituted C1-C10 alkenyl, C1-C10 thioalkyl, C1-C10 alkoxy, substituted C1-C10 alkoxy, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxyl, Ci-Cio alkoxyalkyl, substituted Ci-Cio alkoxyalkyl, C1-C10 carboxy, substituted Ci -C io carboxy, C1-C10 alkoxycarbonyl, substituted C1-C10 alkoxycarbonyl, CO-uronide, CO-monosacchaπde, CO-oligosacchaπde, and CO- polysacchande;
X6 and X7 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkylenylaryl, alkylenylheteroaryl, alkylenylheterocycloalkyl, alkylenylcycloalkyl, or X6 and X7 together form an optionally substituted 5 or 6 membered heterocyclic ring; and
Ar is pyridmyl or phenyl; and Z is O or S.
30. The method of any one of paragraphs 1-29, wherein a therapeutically effective amount of pirfenidone or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof is administered to the patient
31. The method of any one of paragraphs 1-29, wherein the therapeutic administered to the patient comprises a compound of formula (II)
Figure imgf000098_0001
wherein
X3 is H, OH, or C1 i0alkoxy, Z is O, R2 is methyl, C(=0)H, Q=O)CH3, C(=O)O- glucosyl, fluoromethyl, difluoromethyl, trifluoromethyl, methylmethoxyl, methylhydroxyl, or phenyl; and R4 is H or hydroxyl, or a salt, ester, solvate, or prodrug thereof.
32 The method of paragraph any one of paragraphs 1-29, wherein the therapeutic administered to the patient is selected from the group consisting of
Figure imgf000098_0002
Figure imgf000099_0001
, , a compound as listed in Table 1 , and pharmaceutically acceptable salts, esters, solvates, and prodrugs thereof.
33. The method of any one of paragraphs 1-28, wherein the therapeutic is a compound of formula (I), (II), (III), (IV), or (V) or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof
Figure imgf000099_0002
Figure imgf000100_0001
wherein
A is N or CR2, B is N or CR4, E is N, N+X4 or CX4, G is N, N+X3 or CX3, J is N, N+X2 or
CX2; K is N, N+X1 or CX1; a dashed line is a single or double bond,
R1, R2, R3, R4, X1, X2, X3, X4, X5, Y1, Y2, Y3, and Y4 are independently selected from the group consisting of H, deuterium, optionally substituted Ci-Cio alkyl, optionally substituted C1-C10 deuterated alkyl, optionally substituted C1-C10 alkenyl, optionally substituted C1-C10 thioalkyl, optionally substituted C1-C10 alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted heteroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted amido, optionally substituted sulfonyl, optionally substituted ammo, optionally substituted sulfonamido, optionally substituted sulfoxyl, cyano, nitro, halogen, hydroxyl, SO2H2, optionally substituted C1-C 10 alkoxyalkyl, optionally substituted C1-C10 carboxy, optionally substituted Ci Cio alkoxycarbonyl, CO uronide, CO monosaccharide, CO oligosaccharide, and CO-polysaccharide,
X6 and X7 are independently selected from the group consisting of hydrogen, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylenylaryl, optionally substituted alkylenylheteroaryl, optionally substituted alkylenylheterocycloalkyl, optionally substituted alkylenylcycloalkyl, or X6 and X7 together form an optionally substituted 5 or 6 membered heterocyclic ring, and
Ar is optionally substituted pyridmyl or optionally substituted phenyl; and Z is O or S.
34 The method of any one of paragraphs 1-33, wherein the therapeutic is combined with a pharmaceutically acceptable carrier
35. The method of any one of paragraphs 1 34, wherein the administering is oral 36. The method of any one of paragraphs 1-35, wherein the therapeutically effective amount is a total daily dose of about 50 mg to about 2400 mg of the therapeutic or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof.
37. The method of paragraph 36, wherein the therapeutically effective amount is administered in divided doses three times a day or two times a day, or is administered in a single dose once a day.
38. The method of any one of paragraphs 1-37, wherein the patient is human.

Claims

What is Claimed is:
1 A method of treating a patient who has suffered an acute myocardial infarction (AMI) comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein optionally the treatment is initiated at a time period about 1 to 42 days after suffering the AMI, and optionally continues for up to 3 to 6 months.
2 The method of claim 1 , wherein the method is to limit expansion of an infarct scar due to the AMI.
3 The method of claim 1, wherein the treatment is initiated about 5-10 days after the AMI.
4 The method of claim 3, wherein the treatment is initiated about 7 days after the AMI.
5 The method of any one of claims 1-4, wherein the treatment is for at least 2 weeks.
6 A method of reducing the incidence of congestive heart failure in a patient who has suffered an acute myocardial infarction (AMI), comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the therapeutically effective dose reduces the incidence of congestive heart failure.
7 The method of claim 6, wherein the patient is at an increased risk of congestive heart failure due to the AMI.
8 The method of claim 6 or 7, wherein the treatment is initiated about 1 to 42 days after the suffering of the AMI.
9 A method of preserving viable cardiac tissue or controlling or reducing myocardial infarct size in a patient who has suffered an acute myocardial infarction (AMI) comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic to the patient results in a relatively reduced infarct size on average compared to infarct size in a patient who has not been administered the therapeutic.
10. The method of claim 9, wherein the administering is initiated 1-42 days after suffering the AMI
11. The method of claim 9 or 10, wherein the relative reduction in infarct size is at least 5%.
12. A method of reducing the incidence of ventricular tachycardia in a patient in need thereof, comprising administering to the patient a therapeutically effective dose of a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic prevents or reduces the incidence of ventncular tachycardia.
13. The method of claim 12, wherein the patient has suffered an acute myocardial infarction (AMI).
14. The method of claim 13, wherein the administering is initiated about 1 to 42 days after the suffering of the AMI.
15. The method of claim 14, wherein the administering is initiated about 7 days after the suffering of the AMI
16. A method of treating or preventing ventricular fibrillation in a patient in need thereof, comprising administering to the patient a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic prevents ventricular fibrillation in the patient.
17. The method of claim 16, wherein the patient has suffered an acute myocardial infarction (AMI).
18. The method of claim 17, wherein the administration is initiated about 1 to 42 days after the suffering of the AMI.
19. The method of claim 18, wherein the administration is initiated about 7 days after the suffering of the AMI.
20. The method of any one of claims 16-19, wherein the administering reduces the incidence of sudden cardiac death.
21. The method of any one of claims 16-20, wherein the administering reduces cardiac risk of the patient.
22. A method of controlling arrhythmia in a patient in need thereof, comprising administering to the patient a therapeutic having an anti-fibrotic effect, wherein the administering of the therapeutic controls arrhythmia in the patient.
23. The method of claim 22, wherein the patient has suffered an acute myocardial infarction (AMI).
24. The method of claim 23, wherein the administration is initiated about 1 to 42 days after the suffering of the AMI.
25. The method of claim 24, wherein the administration is initiated about 7 days after the suffering of the AMI.
26. The method of any one of claims 22-25, wherein the administering treats ventricular remodeling.
27. The method of any one of claims 1-26, wherein the patient had not previously suffered an AMI.
28. The method of any one of claims 1-27, wherein the therapeutic having an anti- fibrotic effect is a therapeutic that reduces tissue remodeling or fibrosis, reduces the activity of transforming growth factor-beta (TGF- β), targets one or more TGF-β isoforms, inhibits TGF-β receptor kinases TGFBRl (ALK5) and/or TGFBR2, or modulates one or more post-receptor signaling pathways; is an endothelin receptor antagonists, targets both endothelin receptor A and endothelin receptor B or selectively targets endothelin receptor A; reduces activity of connective tissue growth factor (CTGF); inhibits matrix metalloproteinase; reduces the activity of epidermal growth factor (EGF), targets the EGF receptor, or inhibits EGF receptor kinase; reduces the activity of platelet derived growth factor (PDGF), targets PDGF receptor (PDGFR), inhibits PDGFR kinase activity, or inhibits post- PDGF receptor signaling pathways; reduces the activity of vascular endothelial growth factor (VEGF), targets one or more of VEGF receptor 1 (VEGFRl, FIt-I), VEGF receptor 2 (VEGFR2, KDR), the soluble form of VEGFRl (sFlt) and derivatives thereof which neutralize VEGF, inhibits VEGF receptor kinase activity; inhibits multiple receptor kinases such as BIRB-1120 which inhibits receptor kinases for vascular endothelial growth factor, fibroblast growth factor, and platelet derived growth factor; interferes with integrin function; interferes with pro-fibrotic activities of IL-4 and IL-13, targets IL-4 receptor, IL-13 receptor, the soluble form of IL-4 receptor or derivatives thereof; modulates signaling though the JAK-STAT kinase pathway; interferes with epithelial mesenchymal transition, inhibits mTor; reduces levels of copper, reduces oxidative stress; inhibits prolyl hydrolase; inhibits phosphodiesterase 4 (PDE4) or phosphodiesterase 5 (PDE5), or modifies the arachidonic acid pathway.
29. The method of any one of claims 1-28, wherein the therapeutic is pirfenidone or compound of formula (I), (II), (III), (IV), or (V) or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof
Figure imgf000105_0001
wherein
A is N or CR2; B is N or CR4; E is N or CX4; G is N or CX3; J is N or CX2; K is N or CX1; a dashed line is a single or double bond,
R1, R2, R3, R4, X1, X2, X3, X4, X5, Y1, Y2, Y3, and Y4 are independently selected from the group consisting of H, deuterium, Ci-C io alkyl, C1-C10 deuterated alkyl, substituted Ci- Cio alkyl, C1-C10 alkenyl, substituted C1-C10 alkenyl, C1-C10 thioalkyl, C1-C10 alkoxy, substituted C1-C10 alkoxy, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, heteroalkyl, substituted heteroalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxyl, Ci-Cio alkoxyalkyl, substituted Ci-Cio alkoxyalkyl, C1-C10 carboxy, substituted Ci -C io carboxy, C1-C10 alkoxycarbonyl, substituted C1-C10 alkoxycarbonyl, CO-uronide, CO-monosacchaπde, CO-oligosacchaπde, and CO- polysacchande;
X6 and X7 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkylenylaryl, alkylenylheteroaryl, alkylenylheterocycloalkyl, alkylenylcycloalkyl, or X6 and X7 together form an optionally substituted 5 or 6 membered heterocyclic ring; and
Ar is pyridmyl or phenyl; and Z is O or S.
30. The method of any one of claims 1-29, wherein a therapeutically effective amount of pirfenidone or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof is administered to the patient
31. The method of any one of claims 1-29, wherein the therapeutic administered to the patient comprises a compound of formula (II)
Figure imgf000106_0001
wherein
X3 is H, OH, or C1 i0alkoxy, Z is O, R2 is methyl, C(=0)H, Q=O)CH3, C(=O)O- glucosyl, fluoromethyl, difluoromethyl, trifluoromethyl, methylmethoxyl, methylhydroxyl, or phenyl; and R4 is H or hydroxyl, or a salt, ester, solvate, or prodrug thereof.
32 The method of claim any one of claims 1-29, wherein the therapeutic administered to the patient is selected from the group consisting of
Figure imgf000106_0002
Figure imgf000107_0001
, , a compound as listed in Table 1 , and pharmaceutically acceptable salts, esters, solvates, and prodrugs thereof.
33. The method of any one of claims 1-28, wherein the therapeutic is a compound of formula (I), (II), (III), (IV), or (V) or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof:
Figure imgf000107_0002
Figure imgf000108_0001
wherein
A is N or CR2, B is N or CR4, E is N, N+X4 or CX4, G is N, N+X3 or CX3, J is N, N+X2 or
CX2; K is N, N+X1 or CX1; a dashed line is a single or double bond,
R1, R2, R3, R4, X1, X2, X3, X4, X5, Y1, Y2, Y3, and Y4 are independently selected from the group consisting of H, deuterium, optionally substituted Ci-Cio alkyl, optionally substituted C1-C10 deuterated alkyl, optionally substituted C1-C10 alkenyl, optionally substituted C1-C10 thioalkyl, optionally substituted C1-C10 alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted heteroalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted amido, optionally substituted sulfonyl, optionally substituted ammo, optionally substituted sulfonamido, optionally substituted sulfoxyl, cyano, nitro, halogen, hydroxyl, SO2H2, optionally substituted C1-C 10 alkoxyalkyl, optionally substituted C1-C10 carboxy, optionally substituted Ci Cio alkoxycarbonyl, CO uronide, CO monosaccharide, CO oligosaccharide, and CO-polysaccharide,
X6 and X7 are independently selected from the group consisting of hydrogen, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkylenylaryl, optionally substituted alkylenylheteroaryl, optionally substituted alkylenylheterocycloalkyl, optionally substituted alkylenylcycloalkyl, or X6 and X7 together form an optionally substituted 5 or 6 membered heterocyclic ring, and
Ar is optionally substituted pyridmyl or optionally substituted phenyl; and Z is O or S.
34 The method of any one of claims 1-33, wherein the therapeutic is combined with a pharmaceutically acceptable carrier
35. The method of any one of claims 1 34, wherein the administering is oral
36. The method of any one of claims 1-35, wherein the therapeutically effective amount is a total daily dose of about 50 mg to about 2400 mg of the therapeutic or a pharmaceutically acceptable salt, ester, solvate, or prodrug thereof.
37. The method of claim 36, wherein the therapeutically effective amount is administered in divided doses three times a day or two times a day, or is administered in a single dose once a day.
38. The method of any one of claims 1-37, wherein the patient is human.
PCT/US2010/022112 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders WO2010085805A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
BRPI1006979A BRPI1006979A2 (en) 2009-01-26 2010-01-26 methods for treating acute myocardial infarction and associated disorders
AP2011005824A AP2011005824A0 (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders.
EP10734007A EP2389227A4 (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders
JP2011548228A JP2012515800A (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarction and related disorders
SG2011050952A SG172981A1 (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders
CN2010800055907A CN102292124A (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders
MX2011007854A MX2011007854A (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders.
AU2010206543A AU2010206543A1 (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders
CA2747251A CA2747251A1 (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders
IL213526A IL213526A0 (en) 2009-01-26 2011-06-13 Methods for treating acute myocardial infarctions and associated disorders
TN2011000364A TN2011000364A1 (en) 2009-01-26 2011-07-22 Methods for treating acute myocardial infarctions and associated disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US14734009P 2009-01-26 2009-01-26
US61/147,340 2009-01-26

Publications (1)

Publication Number Publication Date
WO2010085805A1 true WO2010085805A1 (en) 2010-07-29

Family

ID=42354646

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/022112 WO2010085805A1 (en) 2009-01-26 2010-01-26 Methods for treating acute myocardial infarctions and associated disorders

Country Status (16)

Country Link
US (2) US20100190731A1 (en)
EP (1) EP2389227A4 (en)
JP (1) JP2012515800A (en)
KR (1) KR20110114684A (en)
CN (1) CN102292124A (en)
AP (1) AP2011005824A0 (en)
AU (1) AU2010206543A1 (en)
BR (1) BRPI1006979A2 (en)
CA (1) CA2747251A1 (en)
CL (1) CL2011001811A1 (en)
IL (1) IL213526A0 (en)
MX (1) MX2011007854A (en)
NI (1) NI201100147A (en)
SG (1) SG172981A1 (en)
TN (1) TN2011000364A1 (en)
WO (1) WO2010085805A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8377932B2 (en) 2009-05-25 2013-02-19 Central South University Preparation of 1-(substituted benzyl)-5-trifluoromethyl-2(1H)pyridone compounds and salts thereof and their applications
US8383823B2 (en) 2007-06-20 2013-02-26 Auspex Pharmaceuticals Substituted N-aryl pyridinones
US8426407B2 (en) 2009-05-25 2013-04-23 Central South University Preparation of 1-(substituted aryl)-5-trifluoromethyl-2-(1H)pyridone compounds and salts thereof and their applications
WO2013078283A1 (en) 2011-11-22 2013-05-30 Intermune, Inc. Methods of diagnosing and treating idiopathic pulmonary fibrosis
US20140037545A1 (en) * 2012-05-08 2014-02-06 Northwestern University Inhibition of fibrosis and af by tgf-beta inhibition in the posterior left atrium (pla)
WO2014055548A1 (en) * 2012-10-02 2014-04-10 Intermune, Inc. Anti-fibrotic pyridinones
JP2014518880A (en) * 2011-05-25 2014-08-07 インターミューン, インコーポレイテッド Pirfenidone and antifibrotic therapy in selected patients
US8969347B2 (en) 2008-06-03 2015-03-03 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
US9018232B2 (en) 2011-03-08 2015-04-28 Auspex Pharmaceuticals, Inc. Substituted N-aryl pyridinones
US9770443B2 (en) 2014-01-10 2017-09-26 Genoa Pharmaceuticals, Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
US10092552B2 (en) 2011-01-31 2018-10-09 Avalyn Pharma Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
US10105356B2 (en) 2011-01-31 2018-10-23 Avalyn Pharma Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
US10233195B2 (en) 2014-04-02 2019-03-19 Intermune, Inc. Anti-fibrotic pyridinones
US11890275B2 (en) 2017-04-18 2024-02-06 Celgene Quanticel Research, Inc. Therapeutic compounds

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TR201903101T4 (en) * 2010-09-01 2019-03-21 Genzyme Corp Treatment of myocardial infarction using TGF-beta antagonists.
FI20115234A0 (en) 2011-03-08 2011-03-08 Biotie Therapies Corp New pyridazinone and pyridone compounds
WO2013169914A1 (en) * 2012-05-08 2013-11-14 Northwestern University Using intracardiac electrograms to predict location of fibrosis and autonomic nerves in the heart
RU2701156C9 (en) 2012-07-18 2019-12-18 Саншайн Лейк Фарма Ко., Лтд. Nitrogen-containing heterocyclic derivatives and use thereof in pharmaceutical preparations
EP2968292A4 (en) * 2013-03-15 2016-11-23 Intermune Inc Methods of improving microvascular integrity
WO2015090232A1 (en) 2013-12-19 2015-06-25 Sunshine Lake Pharma Co., Ltd. Nitrogenous heterocyclic derivatives and their application in drugs
AU2016279897A1 (en) 2015-06-15 2017-12-14 The Board Of Trustees Of The Leland Stanford Junior University Methods for diagnosing and treating affective disorders
CN105085383B (en) * 2015-08-19 2017-09-01 四川大学 5 methyl 2 (1H) Pyridione derivatives and its production and use
CN105669562A (en) * 2016-04-05 2016-06-15 叶芳 Fluoro-substited pyrimidine compound and preparation method thereof
CA3020779A1 (en) * 2016-04-14 2017-10-19 Shijiazhuang Sagacity New Drug Development Co., Ltd. Pyridone derivative and its use for treating fibrosis and inflammatory diseases
CN107556234A (en) * 2016-06-30 2018-01-09 陕西合成药业股份有限公司 A kind of new pyridine ketone compound and preparation method thereof and application medically
WO2018124236A1 (en) * 2016-12-27 2018-07-05 国立大学法人大阪大学 Medicinal composition for treating intractable heart disease
CN107641098A (en) * 2017-09-06 2018-01-30 南阳师范学院 A kind of method for realizing pyridone C6 positions alkenyl
KR20200118098A (en) * 2018-02-02 2020-10-14 제넨테크, 인크. Pharmaceutical compounds, salts thereof, formulations thereof, and methods of preparation and use thereof
EP3810652A1 (en) * 2018-06-22 2021-04-28 Shire Human Genetic Therapies, Inc. Anti-flt-1 antibodies in treating bronchopulmonary dysplasia

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050101581A1 (en) * 2002-08-28 2005-05-12 Reading Christopher L. Therapeutic treatment methods 2
WO2007075388A2 (en) * 2005-12-15 2007-07-05 X-Cell Medical Incorporated Methods of locally treating and preventing cardiac disorders
WO2008073395A2 (en) * 2006-12-11 2008-06-19 3D Matrix, Inc. Compositions and methods for cardiac tissue protection and regeneration

Family Cites Families (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1049411A (en) * 1972-12-18 1979-02-27 Affiliated Medical Research N-substituted pyridone and general method for preparing pyridones
US5310562A (en) * 1989-11-22 1994-05-10 Margolin Solomon B Composition and method for reparation and prevention of fibrotic lesions
US5716632A (en) * 1989-11-22 1998-02-10 Margolin; Solomon B. Compositions and methods for reparation and prevention of fibrotic lesions
US5518729A (en) * 1989-11-22 1996-05-21 Margolin; Solomon B. Compositions and methods for reparation and prevention of fibrotic lesions
US5591766A (en) * 1993-12-03 1997-01-07 Cheil Foods & Chemicals, Inc. Solid oral formulations of pyridone carboxylic acids
US6090822A (en) * 1995-03-03 2000-07-18 Margolin; Solomon B. Treatment of cytokine growth factor caused disorders
US6114353A (en) * 1995-03-03 2000-09-05 Margolin; Solomon B. Compositions and method for treatment of lymphomas, leukemias, and leiomyomas
US5962478A (en) * 1995-09-19 1999-10-05 Margolin; Solomon B. Inhibition of tumor necrosis factor α
US6294350B1 (en) * 1997-06-05 2001-09-25 Dalhousie University Methods for treating fibroproliferative diseases
JP2002526447A (en) * 1998-09-18 2002-08-20 メファ・アクチェンゲゼルシャフト Topical formulations of alkylphenylpyridones
US6956044B1 (en) * 2000-02-21 2005-10-18 Margolin Solomon B Compositions and methods for treatment of epilepsy
EP1455813B1 (en) * 2001-12-18 2015-07-15 mondoBIOTECH AG Interferon gamma in combination with a diagnostic array for use in the improved treatment of idiopathic pulmonary fibrosis
CA2496577A1 (en) * 2002-08-28 2004-03-11 Intermune, Inc. Combination therapy for treatment of fibrotic disorders
JP4542743B2 (en) * 2002-12-26 2010-09-15 Kdl株式会社 Solution pharmaceutical composition of pyridone derivatives
DE10307650A1 (en) * 2003-02-21 2004-09-02 Endress + Hauser Gmbh + Co. Kg Process for transferring data via a fieldbus in process automation technology
US20070072181A1 (en) * 2003-02-28 2007-03-29 Blatt Lawrence M Combination therapy for treating alphavirus infection and liver fibrosis
US20070032457A1 (en) * 2003-05-16 2007-02-08 Blatt Lawrence M Combination therapy for cancer treatment
WO2005016241A2 (en) * 2003-05-16 2005-02-24 Intermune, Inc. Synthetic chemokine receptor ligands and methods of use thereof
WO2004103296A2 (en) * 2003-05-16 2004-12-02 Intermune, Inc. Methods of treating idiopathic pulmonary fibrosis
US20080025986A1 (en) * 2003-06-06 2008-01-31 Ozes Osman N Methods of Treating Tnf-Mediated Disorders
US7407973B2 (en) * 2003-10-24 2008-08-05 Intermune, Inc. Use of pirfenidone in therapeutic regimens
EP1683788B1 (en) * 2003-11-14 2012-03-21 Shanghai Genomics, Inc. Derivatives of pyridone and use thereof
WO2005067454A2 (en) * 2003-12-23 2005-07-28 Valeant Pharmaceuticals North America Combination therapy for treating hepatitis c virus infection
US7235530B2 (en) * 2004-09-27 2007-06-26 Dyax Corporation Kallikrein inhibitors and anti-thrombolytic agents and uses thereof
WO2006057951A2 (en) * 2004-11-22 2006-06-01 Beth Israel Deaconess Medical Center Methods and compositions for the treatment of graft failure
WO2006099445A2 (en) * 2005-03-14 2006-09-21 Massachusetts Institute Of Technology Nanocells for diagnosis and treatment of diseases and disorders
EA015252B1 (en) * 2005-05-10 2011-06-30 Интермьюн, Инк. Method of modulating stress-activated protein kinase system
EP2305696A3 (en) * 2005-07-25 2011-10-12 Intermune, Inc. Macrocyclic inhibitors of Hepatitis C virus replication
NZ591443A (en) * 2005-09-22 2013-04-26 Intermune Inc Granule formation of pirfenidone and pharmaceutically acceptable excipients
US20090170804A1 (en) * 2005-10-31 2009-07-02 Roderick Phillips Pirfenidone/toll-like receptor (tlr) agonist compositions and methods for using them to stimulate production of granulocyte colonizing stimulating factor (g-csf)
WO2007053610A2 (en) * 2005-11-01 2007-05-10 The Regents Of The University Of California Methods of treating atrial fibrillation wtih pirfenidone
US20070203202A1 (en) * 2005-12-02 2007-08-30 Robinson Cynthia Y Methods of reducing adverse events associated with pirfenidone therapy
RU2435585C2 (en) * 2006-04-13 2011-12-10 Актелион Фармасьютиклз Лтд Antagonists of endoteline receptor intended for early stage of idiopatic pulmonary fibrosis
US20080003635A1 (en) * 2006-06-12 2008-01-03 Intermune, Inc. High Throughput Collagen Synthesis Assay
WO2007147297A1 (en) * 2006-06-15 2007-12-27 Shanghai Genomics, Inc. The use of derviate of pyridone for preventing and treating radioactive injury of lungs
RU2008152171A (en) * 2006-07-05 2010-08-10 Интермьюн, Инк. (Us) NEW HEPATITIS C VIRAL REPLICATION INHIBITORS
CA2667654C (en) * 2006-12-18 2016-12-13 Intermune, Inc. Method of providing pirfenidone therapy to a patient
US20080287508A1 (en) * 2007-05-18 2008-11-20 Intermune, Inc. Altering pharmacokinetics of pirfenidone therapy
CN102099036B (en) * 2008-06-03 2015-05-27 英特芒尼公司 Compounds and methods for treating inflammatory and fibrotic disorders
US7566729B1 (en) * 2008-11-10 2009-07-28 Intermune, Inc. Modifying pirfenidone treatment for patients with atypical liver function
US7635707B1 (en) * 2008-11-10 2009-12-22 Intermune, Inc. Pirfenidone treatment for patients with atypical liver function

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050101581A1 (en) * 2002-08-28 2005-05-12 Reading Christopher L. Therapeutic treatment methods 2
WO2007075388A2 (en) * 2005-12-15 2007-07-05 X-Cell Medical Incorporated Methods of locally treating and preventing cardiac disorders
WO2008073395A2 (en) * 2006-12-11 2008-06-19 3D Matrix, Inc. Compositions and methods for cardiac tissue protection and regeneration

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2389227A4 *

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9062001B2 (en) 2007-06-20 2015-06-23 Auspex Pharmaceuticals, Inc. Substituted N-aryl pyridinones
US8383823B2 (en) 2007-06-20 2013-02-26 Auspex Pharmaceuticals Substituted N-aryl pyridinones
US8680123B1 (en) 2007-06-20 2014-03-25 Auspex Pharmaceuticals, Inc Substituted N-aryl pyridinones
US9504677B2 (en) 2007-06-20 2016-11-29 Auspex Pharmaceuticals, Inc. Substituted N-aryl pyridinones
US8969576B2 (en) 2007-06-20 2015-03-03 Auspex Pharmaceuticals, Inc. Substituted N-aryl pyridinones
US8969575B2 (en) 2007-06-20 2015-03-03 Auspex Pharmaceuticals, Inc. Substituted N-Aryl pyridinones
USRE47142E1 (en) 2008-06-03 2018-11-27 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
US9290450B2 (en) 2008-06-03 2016-03-22 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
US8969347B2 (en) 2008-06-03 2015-03-03 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
US8426407B2 (en) 2009-05-25 2013-04-23 Central South University Preparation of 1-(substituted aryl)-5-trifluoromethyl-2-(1H)pyridone compounds and salts thereof and their applications
US8377932B2 (en) 2009-05-25 2013-02-19 Central South University Preparation of 1-(substituted benzyl)-5-trifluoromethyl-2(1H)pyridone compounds and salts thereof and their applications
US10092552B2 (en) 2011-01-31 2018-10-09 Avalyn Pharma Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
US10105356B2 (en) 2011-01-31 2018-10-23 Avalyn Pharma Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
US9018232B2 (en) 2011-03-08 2015-04-28 Auspex Pharmaceuticals, Inc. Substituted N-aryl pyridinones
JP2014518880A (en) * 2011-05-25 2014-08-07 インターミューン, インコーポレイテッド Pirfenidone and antifibrotic therapy in selected patients
WO2013078283A1 (en) 2011-11-22 2013-05-30 Intermune, Inc. Methods of diagnosing and treating idiopathic pulmonary fibrosis
US9078918B2 (en) * 2012-05-08 2015-07-14 Northwestern University Inhibition of fibrosis and AF by TGF-beta inhibition in the posterior left atrium (PLA)
US20140037545A1 (en) * 2012-05-08 2014-02-06 Northwestern University Inhibition of fibrosis and af by tgf-beta inhibition in the posterior left atrium (pla)
US10376497B2 (en) 2012-10-02 2019-08-13 Intermune, Inc. Anti-fibrotic pyridinones
US9359379B2 (en) 2012-10-02 2016-06-07 Intermune, Inc. Anti-fibrotic pyridinones
WO2014055548A1 (en) * 2012-10-02 2014-04-10 Intermune, Inc. Anti-fibrotic pyridinones
US9675593B2 (en) 2012-10-02 2017-06-13 Intermune, Inc. Anti-fibrotic pyridinones
EA027120B1 (en) * 2012-10-02 2017-06-30 Интермьюн, Инк. Anti-fibrotic pyridinones
US10898474B2 (en) 2012-10-02 2021-01-26 Intermune, Inc. Anti-fibrotic pyridinones
US10028966B2 (en) 2014-01-10 2018-07-24 Avalyn Pharma Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
US9770443B2 (en) 2014-01-10 2017-09-26 Genoa Pharmaceuticals, Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
RU2692485C2 (en) * 2014-04-02 2019-06-25 Интермьюн, Инк. Antifibrous pyridinones
US10233195B2 (en) 2014-04-02 2019-03-19 Intermune, Inc. Anti-fibrotic pyridinones
US10544161B2 (en) 2014-04-02 2020-01-28 Intermune, Inc. Anti-fibrotic pyridinones
US11890275B2 (en) 2017-04-18 2024-02-06 Celgene Quanticel Research, Inc. Therapeutic compounds

Also Published As

Publication number Publication date
IL213526A0 (en) 2011-07-31
MX2011007854A (en) 2011-08-15
CL2011001811A1 (en) 2011-11-11
EP2389227A1 (en) 2011-11-30
EP2389227A4 (en) 2012-08-08
AU2010206543A1 (en) 2011-07-07
AP2011005824A0 (en) 2011-08-31
JP2012515800A (en) 2012-07-12
TN2011000364A1 (en) 2013-03-27
US20100190731A1 (en) 2010-07-29
CN102292124A (en) 2011-12-21
CA2747251A1 (en) 2010-07-29
NI201100147A (en) 2011-11-09
KR20110114684A (en) 2011-10-19
BRPI1006979A2 (en) 2016-04-12
SG172981A1 (en) 2011-08-29
US20110218515A1 (en) 2011-09-08

Similar Documents

Publication Publication Date Title
US20110218515A1 (en) Methods for Treating Acute Myocardial Infarctions and Associated Disorders
US20120046321A1 (en) Methods of treating atrial fibrillation with p38 inhibitor compounds
JP5723889B2 (en) How to treat atrial fibrillation
KR20110042344A (en) Use of dronedarone for the preparation of a medicament for the prevention of stroke or transient ischemic attack
Sugiyama et al. Effects of Nonsedating Antihistamine, Astemizole, on thein SituCanine Heart Assessed by Cardiohemodynamic and Monophasic Action Potential Monitoring
JP6522845B1 (en) Non-obstructive hypertrophic cardiomyopathy therapeutic agent
AU2010277725B2 (en) Otamixaban for treatment of elderly and renal impaired non-ST elevation myocardial infarction patients
JPH11509830A (en) Use of retinoids for manufacturing a medicament for treating restenosis
WO2017185142A1 (en) Method for preventing and/or treating atrial fibrillation
JP4658919B2 (en) Combination of phenylcarboxylic acid amide and IKr channel blocker and its use to treat atrial arrhythmia
SK286984B6 (en) Use of derivatives of 3(2H)-pyridazinone for preparation of medicament for treatment of erectile dysfunction
AU2009327374A1 (en) Treatment of major adverse cardiac events and acute coronary syndrome using secretory phospholipase A2 (sPLA2) inhibitor or sPLA2 inhibitor combination therapies
US20140364417A1 (en) Method of treating atrial fibrillation
JP2008519065A (en) Dosing regimes for ion channel modulating compounds
AU2022219157A1 (en) Compositions and methods for treating arrhythmogenic cardiomyopathy
KR20220100544A (en) Pharmaceutical composition and treatment method for the treatment of arrhythmias through PI3K/Akt-mediated late sodium current inhibition
CN114099679A (en) New application of sodium-glucose cotransporter 2 inhibitor
TW201219037A (en) Prophylactic and/or therapeutic agent against lymphedema
Fukunari et al. Y-26763 protects the canine heart from a stunning injury through opening of the KATP channels
JPWO2008126398A1 (en) Antitumor agent for continuous intravenous administration containing cytidine derivative
KR20030072404A (en) Medicinal composition for treatment of chronic cardiac failure
JP2004503586A (en) How to treat sepsis
EP0810862A2 (en) Use of vinyl carboxylic acid compounds for the treatment or prevention of transient ischaemic attacks
CN104363906A (en) Desethylamiodarone compositions

Legal Events

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

Ref document number: 201080005590.7

Country of ref document: CN

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

Ref document number: 10734007

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2747251

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010206543

Country of ref document: AU

Ref document number: 593583

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2010206543

Country of ref document: AU

Date of ref document: 20100126

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12011501488

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2011548228

Country of ref document: JP

Ref document number: MX/A/2011/007854

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011001811

Country of ref document: CL

WWE Wipo information: entry into national phase

Ref document number: 5985/DELNP/2011

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 20117019794

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12349

Country of ref document: GE

Ref document number: 2010734007

Country of ref document: EP

Ref document number: 201170978

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: a201109441

Country of ref document: UA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI1006979

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI1006979

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110726