WO2008140814A1 - Methods for detecting cardiac damage - Google Patents

Methods for detecting cardiac damage Download PDF

Info

Publication number
WO2008140814A1
WO2008140814A1 PCT/US2008/006060 US2008006060W WO2008140814A1 WO 2008140814 A1 WO2008140814 A1 WO 2008140814A1 US 2008006060 W US2008006060 W US 2008006060W WO 2008140814 A1 WO2008140814 A1 WO 2008140814A1
Authority
WO
WIPO (PCT)
Prior art keywords
ctnl
ctnt
cardiac
patient
cardiac tissue
Prior art date
Application number
PCT/US2008/006060
Other languages
French (fr)
Inventor
Xinhua Yan
Anthony O Caggiano
Original Assignee
Acorda Therapeutics, Inc.
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 IN1227MUN2014 priority Critical patent/IN2014MN01227A/en
Priority to MX2014013196A priority patent/MX348082B/en
Application filed by Acorda Therapeutics, Inc. filed Critical Acorda Therapeutics, Inc.
Priority to MX2009012174A priority patent/MX2009012174A/en
Priority to CN200880021887.5A priority patent/CN101743474B/en
Priority to US12/451,397 priority patent/US8628929B2/en
Priority to BRPI0810631A priority patent/BRPI0810631A2/en
Priority to AU2008251822A priority patent/AU2008251822B2/en
Priority to CA2686959A priority patent/CA2686959C/en
Priority to JP2010507486A priority patent/JP5749490B2/en
Priority to RU2009145695/15A priority patent/RU2498309C2/en
Priority to EP08767674A priority patent/EP2153228A4/en
Publication of WO2008140814A1 publication Critical patent/WO2008140814A1/en
Priority to US14/153,695 priority patent/US9329171B2/en
Priority to US15/053,284 priority patent/US9757429B2/en
Priority to US15/674,053 priority patent/US10258667B2/en
Priority to US16/359,383 priority patent/US11071770B2/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5061Muscle cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1883Neuregulins, e.g.. p185erbB2 ligands, glial growth factor, heregulin, ARIA, neu differentiation factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6887Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids from muscle, cartilage or connective tissue
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4712Muscle proteins, e.g. myosin, actin, protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • G01N2800/324Coronary artery diseases, e.g. angina pectoris, myocardial infarction

Definitions

  • the invention relates to the field of medical diagnostics. More particularly, the invention is directed to a method for detecting heart damage in a patient. The invention also relates to methods for treatment of patients identified as having heart damage. The invention also pertains to methods for evaluating the efficacy of an ongoing therapeutic regimen designed to treat a damaged heart in a patient.
  • Heart failure is causally related to a number of conditions that damage the heart, including coronary heart disease, with or without a heart attack; hypertension; diseases, infections, or toxins that affect the heart muscle; and diseases of the heart valves.
  • the onset of heart failure can occur rapidly, over days to weeks, but more frequently develops slowly over the course of years, as the heart gradually and progressively weakens.
  • cytostatic agents used as chemotherapeutics for the treatment of various cancers frequently exhibit potentially lethal side effects, including cardiotoxicity.
  • Agents commonly used in cytostatic therapy include the anthracyclines daunorubicin and prodrugs thereof, zorubicin, doxorubicin (adriamycin) and epirubicin, and the synthetic antibiotic mitoxantrone.
  • Anthracyclines for example, represent a class of chemotherapeutic agents based on daunosamine and tetra-hydro-naphthacene-dione.
  • Cardiotoxicity associated with anthracycline use is correlated with the total dose administered and is frequently irreversible.
  • the cytostatic effects and cardiotoxicity of these compounds are due, at least in part, to alterations in membrane fluidity and permeability caused by anthracycline binding to components of the cell membrane. Free radical formation in the heart and accumulation of anthracycline metabolites are also thought to contribute to heart damage.
  • Cardiotoxicity often presents in electrocardiogram (EKG) abnormalities and arrhythmias or as cardiomyopathy, which may ultimately lead to congestive heart failure.
  • the invention is directed to providing novel diagnostic methods for screening patients to identify those exhibiting signs of heart damage. Patients so identified can then be treated with pharmaceutical preparations for the treatment of heart damage as described herein.
  • diagnostic methods for screening patients to identify those exhibiting signs of damage to the heart due to, for example, cardiotoxicity, hypertension, valvular disorders, myocardial infarction, viral myocarditis, or scleroderma are presented.
  • the invention is focused on identifying patients exhibiting cardiotoxicity resulting from chemotherapeutic intervention. Classification of such patients serves to identify a subgroup of patients in need of therapeutic intervention to alleviate short and long term effects of cardiotoxicity.
  • the subgroup of patients so identified can be treated with pharmaceutical preparations for the treatment of heart damage that occurs in connection with the use of cardiotoxic doses of medicaments or chemicals.
  • pharmaceutical preparations for the treatment of heart damage that occurs in connection with the use of cardiotoxic doses of medicaments or chemicals.
  • heart damage identified in a patient is due to an ongoing condition, such as, hypertension, valvular disorders, myocardial infarction, viral myocarditis, or scleroderma
  • appropriate pharmaceutical preparations can also be formulated to treat the patient with heart damage.
  • the present invention also encompasses a method for stratifying patients according to degree or type of heart damage, knowledge of which guides a skilled practitioner to choose appropriate therapeutic regimens.
  • the invention also includes a method whereby the efficacy of a therapeutic regimen is evaluated.
  • the novel methods of the invention are based on the discovery that changes in intracellular levels of cardiac troponin I (cTnl) and cardiac troponin T (cTnT) in intact cardiac tissue can be used as indicators for the presence of cardiac damage. More specifically, the present inventors have discovered that a decrease in intracellular cTnl and cTnT levels in intact cardiac tissue serves as a diagnostic marker to identify patients at risk for or experiencing cardiac damage. Cardiac tissue can be excised from a patient and tested in vitro or analyzed in vivo using molecular imaging protocols known in the art.
  • intracellular cTnl and cTnT levels determined for the patient's cardiac tissue are compared to those of control cardiac tissue that expresses wildtype or normal levels of intracellular cTnl and cTnT.
  • Reduced levels of intracellular cTnl and/or cTnT in a patient's cardiac tissue are readily determined by quantitating protein levels, which can be achieved using standard methods, and analyzing the results to determine if a statistically significant decrease in intracellular cTnl and cTnT levels is apparent in the patient's cardiac tissue relative to that of the control.
  • Patients showing evidence of reduced intracellular cTnl and/or cTnT levels are earmarked for treatment with appropriate compositions chosen to restore, at least in part, normal heart function as reflected in an increase in intracellular cTnl and cTnT levels or restoration of normal levels of intracellular cTnl and cTnT.
  • the control or normal intracellular levels of either cTnT or cTnl in cardiac tissue are established by determining the intracellular levels of either cTnT or cTnl in cardiac tissue of a patient with normal heart function.
  • the control or normal intracellular levels of either cTnT or cTnl in cardiac tissue are established by determining the intracellular levels of either cTnT or cTnl in cardiac tissue of a patient prior to onset of treatment capable of causing heart damage.
  • the heart damage is a result of cardiotoxicity, hypertension, valvular disorders, myocardial infarction, viral myocarditis, or scleroderma.
  • the cardiotoxicity is caused by treatment with a chemotherapeutic agent or radiation.
  • cTnl and cTnT levels in cardiac tissue of a treated patient it is also within the scope of the invention to evaluate the efficacy of a therapeutic regimen designed to at least partially restore normal heart function by measuring intracellular cTnl and cTnT levels in cardiac tissue of a treated patient.
  • an increase in intracellular cTnl and cTnT levels in cardiac tissue of a treated patient relative to those determined prior to treatment is a positive indicator that the treatment is acting to restore cardiac function.
  • intracellular levels of either cTnl or cTnT in cardiac tissue or intracellular levels of both cTnl and cTnT in cardiac tissue may be used as indicators of cardiac tissue activity and/or function.
  • the present invention pertains to animals, in general, and more particularly, to mammals, and even more particularly to humans.
  • the subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human.
  • the term "subject” or “patient” may be used to refer to a human.
  • the present invention also encompasses a combination therapeutic regimen wherein
  • GGF2 or an epidermal growth factor-like (EGFL) domain encoded by the neuregulin gene is administered in conjunction with a proteasome inhibitor to treat cardiac damage.
  • An exemplary proteasome inhibitor for use in the present invention is Proscript 519, which is a potent and selective proteasome inhibitor.
  • Other proteasome inhibitors of utility in the present invention include VelcadeTM and lactacystin. Additional proteasome inhibitors are known to those skilled in the art. Indeed, proteasome inhibitors are already used as therapeutic agents for the treatment of a number of diseases, including some cancers and neurodegenerative diseases.
  • GGF2 or an epidermal growth factor-like (EGFL) domain encoded by the neuregulin gene in the preparation of a medicament for administration to a patient identified by the present diagnostic methods as exhibiting damage to the heart.
  • the invention further encompasses the use of GGF2 or an epidermal growth factor-like (EGFL) domain encoded by the neuregulin gene in combination with a proteasome inhibitor in the preparation of a medicament for administration to a patient identified by the present diagnostic methods as exhibiting damage to the heart.
  • FIGS. IA-I C show survival graphs (A), histograms (B-C). and immunoblots (C).
  • A survival analysis
  • mice were injected with a single dose of doxorubicin [20mg/kg, intraperitoneally (i.p.)] with or without concomitant injection of NRGl (0.75mg/kg. s.c. daily).
  • NRGl 0.75mg/kg. s.c. daily
  • B serum creatine kinase
  • serum CK levels were measured in control, Dox-treated and Dox- NRGl treated mice four days after doxorubicin injection.
  • Figure 1C shows that NRGl injection alleviated doxorubicin-induced down-regulation of cTnl, cTnT and cTnC protein levels in mice.
  • Mice were treated with doxorubicin (20mg/kg, i.p.) with or without concomitant NRGl injection (0.75mg/kg, s.c. daily).
  • Protein levels of cTnl, cTnT and cTnC were measured by Western blot analysis five days after doxorubicin treatment.
  • Figures 2A-D show imunoblots probed to detect the indicated proteins.
  • Figure 2A reveals that NRGl alleviated doxorubicin-induced down-regulation of cTnl and cTnT protein levels in neonatal rat cardiomyocytes (RNCM).
  • RNCM neonatal rat cardiomyocytes
  • IuM doxorubicin
  • cTnl and cTnT protein levels were measured by Western blot analysis 48 hours after doxorubicin treatment.
  • Figure 2B shows that inhibition of erbB2 abolished the effects of NRGl on cTnl and cTnT.
  • RNCM were treated with doxorubicin (IuM) and NRGl (20ng/ml) in the presence or the absence or AG879 (lOuM) and AG1478 (lOuM). Protein levels of cTnl and cTnT were analyzed by Western blot analysis. As shown in Figure 2C, RNCM were treated with doxorubicin and NRGl in the presence of LY294002 (lOuM), Akti (5uM), PD98059 (5OuM) and Rapamycin (1OnM). cTnl and cTnT protein levels were analyzed by Western blot analysis.
  • Figure 2D shows that RNCM were treated with doxorubicin or doxorubicin+NRGl in the presence of cycloheximide (5ug/ml), Z-VAD (10OuM) or MGl 32 (lOuM). Protein levels of cTnl and cTnT were measured by Western blot analysis.
  • Figures 3A-3D show immunoblots (A, C, D), and histograms (B).
  • Figure 3A presents results wherein RNCM were treated with doxorubicin (IuM) in the presence of inhibitors for different caspases (2OuM). The protein levels of cTnl and cTnT were measured by Western blot analysis.
  • Figure 3B shows the effects of caspase activation in doxorubicin-treated RNCM. Cells were treated with Dox, Dox+NRGl or Dox+NRGl +LY. Caspase activation was analyzed by the caspase activation assay.
  • Figure 3 C shows that NRGl decreased doxorubicin-induced cytochrome c release.
  • RNCM were treated with Dox or Dox+NRGl .
  • Cytochrome c release was analyzed by cell fractionation and Western blot analysis.
  • Figure 3D reveals that NRGl decreased doxorubicin-induced ubiquitinylation of cTnl.
  • RNCM were treated with Dox or Dox+NRGl .
  • Cell lysates were immunoprecipated with cTnl antibody and probed with ubiquitin antibody.
  • Figures 4A-4B show ethidium bromide stained agarose gels (A) and immunoblots (B).
  • Figure 4 A reveals that NRG-I inhibited doxorubicin-induced down-regulation of mRNA levels of cTnl, cTnT and cardiac specific transcriptional factors.
  • RNCM were treated with Dox or Dox+NRGl .
  • mRNA levels of cTnl, cTnT, GATA4, MEF2c and NKX2.5 were analyzed by quantitative RT-PCR.
  • Figure 4B shows that NRGl inhibited doxorubicin-induced dephosphorylation of translational molecules.
  • RNCM were treated with Dox, Dox+NRGl or Dox+NRGl+LY.
  • the phosphorylation levels of mTOR, P70S6K, S6, 4EBP and EIF4G were analyzed by Western blot analysis.
  • Figures 5A-5C show a survival graph (A), histograms (B), and an immunoblot (C).
  • Figure 5A shows a survival analysis in doxorubicin-treated mice with cardiac myocyte-specific overexpression of a dominant negative PI3K (dnPDK).
  • Mice were treated with a single dose of doxorubicin (20mg/kg, i.p.) with or without concomitant treatment of NRGl (0.75mg/kg, s.c).
  • Figure 5B depicts hemodynamic measurements in doxorubicin-treated dnPI3K mice. Mice were treated with a single dose of doxorubicin (20mg/kg, i.p.). Hemodynamic measurements were performed six days after the doxorubicin treatment.
  • Figure 5C shows cTnl protein levels in dnPDK mice treated with Dox or
  • Figures 6A-D show amino acid and nucleic acid sequences of GGF2.
  • Figure 7 shows amino acid and nucleic acid sequences of EGFLl .
  • Figure 8 shows amino acid and nucleic acid sequences of EGFL2.
  • Figure 9 shows amino acid and nucleic acid sequences of EGFL3.
  • Figure 10 shows amino acid and nucleic acid sequences of EGFL4.
  • Figure 11 shows amino acid and nucleic acid sequences of EGFL5.
  • Figure 12 shows amino acid and nucleic acid sequences of EGFL6.
  • CK-MB creatine kinase
  • the rise in serum levels of CK, CK-MB, and the troponins is due to the release of these molecules following cardiac muscle cell death and serves, therefore, as a serum marker of necrosis.
  • CK, CK-MB, and the troponins As a heart muscle cell dies as a result of prolonged ischemia, for example, the cell membrane ruptures, releasing the cytosolic contents into the extracellular fluid space, from whence it enters the lymphatic system, and subsequently the bloodstream. Imaging tests, including echocardiogram and perfusion scintigraphy, may also be used in the context of diagnosis.
  • cardiac troponins are components of the contractile apparatus of myocardial cells.
  • Two cardiac troponins, cTnl and cTnT have been commercialized and detection of these markers has proven to be a reliable and specific assay for detection of minimal levels of myocardial damage.
  • the cardiac troponins like CK- MB, are released from dead cardiac muscle cells upon rupture of cell membranes, and are eventually detectable in the blood. Necrosis can occur as a result of a prolonged myocardial ischemia, but can also result from myocardial cell damage from other causes such as infection, trauma, or congestive heart failure.
  • the present invention differs from those procedures described in the prior art in a variety of aspects. At the outset, it is directed to measuring intracellular levels of cTnl and cTnT in intact cardiac tissue, rather than serum levels of these markers. Moreover, the present inventors have discovered that a decrease in intracellular cTnl and cTnT levels in intact cardiac tissue serves as a diagnostic marker to identify patients at risk for or experiencing cardiac damage. This approach stands in marked contrast to measurements of serum levels of these markers, an increase of which is indicative of heart damage.
  • an increase in serum levels of these markers is an acute or transient marker of heart damage
  • measurements of intracellular levels of cTnl and cTnT in intact cardiac tissue serves as a stable marker reflective of the condition of the heart.
  • identification of patients exhibiting a decrease in intracellular cTnl and cTnT levels in intact cardiac tissue also provides a screening method with which to stratify patients into categories for subsequent treatment. Patients showing evidence of reduced intracellular cTnl and cTnT levels are earmarked for treatment with appropriate compositions chosen to restore, at least in part, normal heart function as reflected in restoration of such.
  • An exemplary therapeutic agent for inclusion in such a composition is glial growth factor
  • GGF2 The amino acid and nucleic acid sequences of GGF2 are presented in Figures 6A-6D.
  • Therapeutic compositions may also include other exemplary polypeptides, such as epidermal growth factor-like (EGFL) domains encoded by the neuregulin gene, as shown in Figures 7-12, and described in United States Patent Number (USPN) 5,530,109, which is incorporated herein in its entirety.
  • EGFL epidermal growth factor-like domains encoded by the neuregulin gene
  • compositions containing the molecules or compounds of the invention can be administered for diagnostic and/or therapeutic treatments.
  • diagnostic applications compositions are administered to a patient to determine if the patient has cardiac damage and/or to stratify the patient with respect to prospective therapeutic regimens.
  • therapeutic applications compositions are administered to a patient diagnosed as having cardiac damage in an amount sufficient to treat the patient, thereby at least partially arresting the symptoms of the disease and its complications.
  • control sample of cardiac tissue refers to a sample of cardiac tissue for which intracellular levels of cTnl and cTnT are within normal range.
  • a normal or wildtype range of intracellular levels of cTnl and cTnT is established based on experiments such as those presented herein and known in the art wherein cardiac tissue of a subject having healthy heart function, as determined by a skilled practitioner, is used as the standard against which unknowns are compared. Standards, representative of normal hearts, may, for example, be procured from fresh autopsies performed on cadavers having no evidence of heart disease.
  • control or normal levels refers to levels established or determined as described herein and understood in the art to be within a range associated with healthy functionality.
  • healthy functionality refers to healthy heart function, which can be assessed by a skilled practitioner using standard procedures such as measuring systolic and diastolic blood pressure, measuring serum levels of indicator proteins such as CK. CK-MB, and the troponins, performing an EKG, and/or administering a stress test. A skilled practitioner would be aware of that which is generally considered a normal serum level of these proteins.
  • Troponin is also recognized as a sensitive and specific marker for cardiac injury. Indeed, detection of serum troponin I (sTnl) is considered to be more accurate than creatine kinase-MB concentrations for the diagnosis of MI and provides more useful prognostic information. Detection of sTnl also permits the early identification of those patients with acute coronary syndromes who are at an increased risk of death. sTnl is more sensitive than creatine kinase-MB concentrations for detection of minor ischemic myocardial injury in patients with small increases of total creatine kinase and avoids the high incidence of false diagnoses associated with the use of creatine kinase-MB as a diagnostic marker in perioperative MI.
  • cTnl and cTnT With respect to normal levels of intracellular cTnl and cTnT, such determinations are established by evaluating normal heart tissue using standards methods for determining protein levels such as those taught herein and known in the art. Decreased levels of intracellular cTnl and cTnT. such as those indicative of an injured or diseased heart, are determined as a decrease in the levels of these proteins relative to an established normal level. By way of example, a decrease of at least 50% in the level of cTnl and/or cTnT in heart tissue being tested for damage, relative to that of healthy heart tissue (normal control), serves as a positive indicator that the heart tissue being tested is damaged and a patient from whom the damaged tissue was removed would benefit from therapeutic intervention such as that taught herein.
  • a skilled practitioner would also be aware of the large body of scientific literature pertaining to the activity and levels of intracellular cTnl and cTnT in normal and diseased heart tissue. Examples of references that pertain to intracellular cTnl and cTnT in normal and diseased heart tissue include: Latif et al. (2007, J Heart Lung Transplant 26:230-235); Birks et al.
  • an increase in intracellular cTnl and cTnT levels demonstrates the therapeutic efficacy of the regimen and provides evidence that the regimen is promoting restoration of normal heart function.
  • preventing' " ' or "prevention” refers to a reduction in risk of acquiring or developing a disease or disorder (i.e.. causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to a disease-causing agent, or predisposed to the disease in advance of disease onset.
  • prophylaxis is related to "prevention", and refers to a measure or procedure the purpose of which is to prevent, rather than to treat or cure a disease.
  • prophylactic measures may include the administration of vaccines; the administration of low molecular weight heparin to hospital patients at risk for thrombosis due, for example, to immobilization; and the administration of an anti-malarial agent such as chloroquine, in advance of a visit to a geographical region where malaria is endemic or the risk of contracting malaria is high.
  • treating refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting the disease or reducing the manifestation, extent or severity of at least one of the clinical symptoms thereof).
  • treating refers to ameliorating at least one physical parameter, which may not be discernible by the subject.
  • 'treating' or 'treatment' refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
  • Myocardial biopsy is a common procedure in which heart tissue is obtained from the heart through a catheter, during thoracotomy, or during open chest surgery. It is commonly used to diagnose the etiology of heart failure. The heart tissue is analyzed both histologically and biochemically. The results of these tests are useful in diagnosing the cause of the heart failure.
  • Veinot 2002, Can J Cardiol. 18(3):287-96
  • the results of the myocardial biopsy may indicate that heart failure is the result of such causes as scleroderma, viral myocarditis, drug toxicity or any number of causes of heart failure. This diagnosis will dictate what, if any, therapeutic intervention may be useful and should be employed.
  • Myocardial biopsy (or cardiac biopsy) is an invasive procedure, wherein a bioptome (a small catheter with a grasping device on the end), for example, may be used to obtain a small piece of heart muscle tissue that can be analyzed. Myocardial biopsy may be used to evaluate heart transplant rejection and to diagnose myocarditis (inflammation of the heart).
  • a local anesthetic is used to numb part of the neck or groin of a patient; a practitioner inserts a plastic introducer sheath (a short, hollow tube through which the catheter is placed) into a blood vessel in the numbed region; a bioptome is inserted through the sheath and threaded to the right ventricle of the patient; and samples are collected from the heart using the grasping device of the bioptome.
  • an x-ray camera is generally used to position the bioptome properly. Samples are about the size of the head of a pin.
  • Certain proteins have routinely been measured in the blood stream following a myocardial event to predict and diagnose if and the extent to which the myocardium has been damaged. These proteins include, but are not limited to, creatine kinase and troponin. Detecting specific sub-types of these and other proteins in the blood stream is diagnostic of release of the proteins from the myocardium and thus damage. Detection of these indicator proteins in the sera is used routinely in the acute setting where a cardiac event is suspected and have proven useful in determining the best treatment for the patient.
  • cardiac protein levels in the blood stream have no value in diagnosing or predicting heart failure more than a few days after a cardiac event, nor do they have any value in determining the etiology of heart disease or proper treatment course.
  • cardiac protein levels are not elevated in the blood except for immediately following a cardiac event, measurement of these proteins in the blood stream reveals little regarding the state of the myocardium.
  • the present invention describes the use of myocardial biopsy to predict which patients may be responsive to a cardioprotective, cardiorestorative and other heart failure therapy.
  • Myocardial troponin content measurement may be used to optimize dosing for individual patients that are being treated for heart disease with a cardiorestorative treatment.
  • myocardial troponin levels may indicate success of a cardioprotective therapy such as a neuregulin in the presence of ongoing disease. In this manner myocardial troponin levels may similarly be used to predict response to therapy and optimization of dosing.
  • Dosing for GGF2 or EGFL domains of neuregulin, for example, can be initiated at about
  • intracellular levels of cTnl and cTnT in cardiac tissue can be determined by methods described in the Examples presented herein and known in the art.
  • Cellular lysates of isolated cardiac tissue or precipitates derived therefrom may, for example, be analyzed using a variety of techniques including, but not limited to, immunoblot analysis, enzyme-linked immunosorbent assay (ELISA), and mass spectrometry.
  • ELISA enzyme-linked immunosorbent assay
  • Ml 1.7 high-affinity, cardiac-specific antibody
  • the detection limit of this assay is lower than that of first generation ELISA protocols that used the cross-reacting antibody I BlO (0.0123 ⁇ g/L versus 0.04 ⁇ g/L, respectively).
  • molecular imaging is, indeed, “broadly defined as the in vivo characterization and measurement of biological processes at the cellular and molecular level”. See Weissleder et al. (Radiology 219:316-333, 2001), which is incorporated herein in its entirety.
  • Nuclear imaging for example, which includes positron emission tomography (PET), micro- PET, single photon emission computed tomographic (SPECT), and planar imaging, generally involves visualizing an endogenous or expressed protein using specific radiopharmaceuticals as detection probes.
  • PET for example, is capable of producing a three-dimensional image or map of functional processes in the body which facilitates real time analyses of cellular components and molecular interactions within cells.
  • PET is used as both a medical and a research tool. With respect to medical applications, it is used extensively in clinical oncology to image tumors and detect metastases and in clinical diagnosis of a variety of brain diseases, especially those associated with dementia.
  • the ability to perform repeated PET analyses on a patient enables a skilled practitioner to compare results over time so as to evaluate, for example, disease progression or efficacy of a selected treatment protocol.
  • PET has also been used as a research tool to map normal human brain and heart function.
  • Imaging scans such as CT and MRI are well suited to visualize organic anatomic changes in the body, whereas, as indicated above, PET scanners, like SPECT and fMRI, have the resolution to detect changes on a molecular level, even in advance of changes evident on the anatomic level. Imaging scans such as PET achieve this end by using radiolabeled molecular probes that exhibit different rates of uptake, depending on the type and function of the tissue of interest. Alterations in regional blood flow in various anatomic structures can also be visualized and quantified with a PET scan. Some of the above scanning techniques can be used in combination, depending on compatibility of radioisotopes utilized, so as to provide more comprehensive information that improves the accuracy of the clinical assessment. This is discussed herein below with respect to SPECT imaging.
  • nuclear imaging techniques such as PET, micro-PET, SPECT, and planar imaging, are directed to visualizing an endogenous or expressed protein using specific radiopharmaceuticals as detection probes.
  • Imaging marker genes that encode intracellular enzymes and imaging marker genes that encode cell surface proteins or receptors have been used successfully in a variety of experimental systems to image specific molecules in vivo. The activity of an intracellular enzyme may also be assessed by labeling by-products indicative of the level and/or activity of a particular intracellular enzyme.
  • PET and microPET for example, use positron-labeled molecules to image processes involved in metabolism, cellular communication, and gene expression.
  • Kim Karl (Korean J Radiology 4:201-210, 2003), which is incorporated herein by reference in its entirety.
  • Radionuclides used in PET scanning are typically isotopes with short half lives such as
  • Radiotracers 1 1 C (-20 rain), 13 N (-10 min), 15 O ( ⁇ 2 min), and 18 F (-110 min).
  • These radionuclides are incorporated into compounds normally used by the body such as glucose, water or ammonia and then injected into the body to trace where they become distributed.
  • Such labeled compounds are known as radiotracers.
  • the short half-life of these radiotracers restricts clinical PET primarily to the use of tracers labeled with 18 F, which has a half life of 1 10 minutes and can be transported a reasonable distance before use, or to 82 Rb, which can be created in a portable generator and is used for myocardial perfusion studies.
  • SPECT is an example of a nuclear medicine tomographic imaging technique that uses gamma rays.
  • SPECT can provide true 3D information. This information is typically presented as cross-sectional slices through the patient, but can be modified with regard to presentation as required.
  • the image obtained by a gamma camera image is a 2-dimensional view of 3 -dimensional distribution of a radionuclide. Because SPECT acquisition is very similar to planar gamma camera imaging, the same radiopharmaceuticals may be used for either protocol.
  • SPECT can also be used for cardiac gated acquisitions.
  • gated myocardial SPECT can be used to obtain quantitative information about myocardial perfusion, thickness, and contractility of the myocardium during various parts of the cardiac cycle. It is also used to facilitate calculation of left ventricular ejection fraction, stroke volume, and cardiac output.
  • Myocardial perfusion imaging is an example of functional cardiac imaging, which is used for the diagnosis of ischemic heart disease. It is based on the principle that impaired or diseased myocardium receives less blood flow than normal myocardium under conditions of stress.
  • a cardiac specific radiopharmaceutical is administered, for example, 99m Tc-tetrofosmin (MyoviewTM, GE healthcare) or 99m Tc-sestamibi (Cardiolite®, Bristol-Myers Squibb), after which administration the heart rate is raised to induce myocardial stress.
  • enhanced heart rate is typically either exercise induced or pharmacologically induced with adenosine, dobutamine or dipyridamole.
  • SPECT imaging performed after induction of stress reveals the distribution of the radiopharmaceutical, and therefore the relative blood flow to the different regions of the myocardium. Diagnosis is made by comparing stress images to a subsequent set of images obtained at rest.
  • troponin is a structural component of the cytoskeleton and an enzyme
  • molecular imaging techniques are envisioned to measure intracellular cTnl and cTnT levels in vivo.
  • cardiac troponin T is present in myocytes at high concentrations, both in cytosolic and structurally-bound protein pools.
  • the cytosolic pool amounts to 6%, whereas the amount in myofibrils corresponds to 94% of the total troponin T mass in the cardiomyocyte.
  • labeling of either or both pools of troponin T will generate sufficient signal to be visualized with a reasonable degree of accuracy and resolution.
  • One potential approach envisioned for visualizing troponin complexes in cardiac tissue in vivo takes advantage of the calcium (Ca 2+ ) binding properties of this complex.
  • Tn troponin
  • TnC troponin-binding subunit of Tn
  • the level of force is regulated by the availability of myosin binding sites on the thin filament, which is controlled by the position of tropomyosin (Tm) on the surface of actin.
  • TnI The inhibitory subunit of Tn (TnI) binds to actin in the absence of Ca 2+ , anchoring Tm so as to inhibit myosin binding.
  • a skilled practitioner would envision utilizing a Ca 2+ radionuclide to visualize cytoskeletal components and, more particularly, troponin levels in cardiac tissue in vivo.
  • small molecules that specifically recognize either cTnl or cTnT may be labeled with radionuclides and administered to patients to visualize intracellular levels of these proteins.
  • a skilled practitioner could envision a variety of labeled probes of utility in the present method, including ligands, antibodies, and substrates of cTnl or cTnT and/or proteins that interact with cTnl or cTnT.
  • Radiopharmaceuticals such as those described in USPN 5,324,502 may also be used to advantage in the present method for imaging myocardial tissues.
  • such radiopharmaceuticals are prepared by forming lipophilic, cationic complexes of radioactive metal ions with metal chelating ligands comprising the Schiff base adducts of triamines and tetraamines with optionally substituted salicylaldehydes.
  • These lipophilic, cationic, radioactive complexes exhibit high uptake and retention in myocardial tissues.
  • Preferred gallium-68(III) complexes in accordance with this invention can be used to image the heart using positron emission tomography.
  • such radiopharmaceuticals may be used as a means for targeting uptake of agents that bind to the troponins to cardiac tissue.
  • compositions comprise a therapeutically effective amount of an agent, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc.
  • suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E. W. Martin, incorporated in its entirety by reference herein.
  • Such compositions contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide a form for proper administration to a subject.
  • the formulation should suit the mode of administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the compounds of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the amount of the compound of the invention which will be effective in the treatment of a heart damage can be determined by standard clinical techniques based on the present description.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances.
  • suitable dosage ranges for intravenous administration are generally about 20-500 micrograms of active compound per kilogram body weight.
  • Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight.
  • Suppositories generally contain active ingredient in the range of 0.5% to 10% by weight; oral formulations preferably contain 10% to 95% active ingredient.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • Various delivery systems are known and can be used to administer a compound of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu (1987) J. Biol. Chem. 262:4429-4432), and construction of a nucleic acid as part of a retroviral or other vector.
  • Methods of introduction can be enteral or parenteral and include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compounds may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • compositions of the invention may be desirable to administer locally, e.g., by local infusion during surgery, topical application, e.g., by injection, by means of a catheter, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • the compound or agent can be delivered in a vesicle, in particular a liposome (see Langer (1990) Science 249: 1527-1533; Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • the compound or agent can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14:201 ; Buchwald et al. (1980) Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321 :574).
  • polymeric materials can be used (see Medical Applications of Controlled Release. Langer and Wise (eds.). CRC Pres.. Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.). Wiley.
  • a controlled release system can be placed in proximity of the therapeutic target, i.e., damaged heart, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • the present inventors have previously reported that GGF2, a recombinant neuregulin- 1 , improves survival and cardiac function in doxorubicin-treated mice. As described herein, the present inventors have investigated whether GGF2 prevents doxorubicin-induced cardiac myofibril loss in vivo and in cardiomyocytes in vitro. The results presented herein have led to the novel discovery that the intracellular expression levels of particular cardiac proteins are useful indicators of normal heart function. More specifically, the present inventors have discovered that changes in the intracellular levels of cardiac troponin I (cTnl) and cardiac troponin T (cTnT) in intact cardiac tissue can be used as indicators of cardiac damage.
  • cTnl cardiac troponin I
  • cTnT cardiac troponin T
  • a decrease in intracellular cTnl and cTnT levels in intact cardiac tissue has been shown to be a useful diagnostic marker to identify patients at risk for or experiencing cardiac damage. Such patients are then selected for appropriate preventative or therapeutic intervention as described herein. Determination of intracellular cTnl and cTnT levels in intact cardiac tissue may also be used to advantage to evaluate the efficacy of ongoing therapeutic intervention, since restoration of normal intracellular cTnl and cTnT levels would serve as a positive indicator that the therapy was improving heart function or restorative of normal heart function.
  • C57BL/6 mice and Wistar rats were obtained from Charles River Laboratories.
  • Doxorubicin was obtained from Bedford laboratories. Glial growth factor 2 was a gift from Acorda Therapeutics, Inc. MG 132, cycloheximide and actinomycin were obtained from Sigma. LY294002 and PD 98059 were from Cell Signalling Technology. Antibodies were ordered from the following vendors: Troponin I, GAT A4 and Nkx2.5 were from Santa Cruz Biotechnology; ⁇ -sarcomeric actin, troponin T, troponin C, tropomyosin and cardiac troponin T were from Abeam; Desmin and ⁇ -actinin were from Sigma and cardiac troponin I were from GeneTex. MEM, Hank's solution and fetal bovine serum were obtained from Invitrogen. All other reagents for cell culture were obtained from Sigma.
  • Neonatal cardiac myocytes were dissociated as described previously (Okoshi et al.
  • ventricles from day 0-day 3 Wistar rats were dissociated in trypsin and Dnase II. Cells were washed and pre-plated in 100 mm dishes in MEM containing 5% fetal bovine serum. After 30 minutes, the myocytes were suspended in the same medium containing 0.1 mmol/L bromodeoxyuridine and then plated at the density of 500-1000 cells/mm 2 in 100 mm culture dishes. Forty-eight hours after dissociation, the medium was changed to serum-free MEM containing 0.1% BSA and cultured overnight before stimulation.
  • NRGl improved survival and cardiac function in doxorubicin-treated mice.
  • the body weight (BW), heart weight (HW) and left ventricular weight (LVW) were significantly decreased in both Dox-Placebo and Dox-NRGl mice compared with control (untreated) mice.
  • HW and LVW normalized by tibia length (HW/TL and LVW/TL) were significantly decreased in Dox-Placebo mice compared to control mice. These indices were not, however, different between Dox-NRGl and control mice.
  • LV systolic pressure (LVSP), cardiac output and dP/dt min were significantly decreased in Dox-Placebo mice compared with controls.
  • NRGl alleviated doxorubicin-induced down-regulation of cTnl and cTnT in the heart in vivo.
  • One of the mechanisms of doxorubicin-induced cardiotoxicity is loss of cardiac myofibrils.
  • the present inventors investigated whether NRGl injection in vivo inhibited doxorubicin-induced myofibril loss. Results presented herein demonstrate that the levels of cardiac structural proteins, ⁇ -sarcomeric actin, ⁇ -actinin, troponin T (TnT), troponin I (TnI), troponin C (TnC) and tropomyosin were significantly decreased in doxorubicin-treated hearts.
  • NRGl injection in vivo significantly increased the protein levels of cTnl, cTnT and cTnC in doxorubicin-injured hearts ( Figure 1C), but had no effects on the protein levels of ⁇ -sarcomeric actin. ⁇ -actinin and tropomyosin.
  • NRGl abolished doxorubicin-induced down-regulation of cTnl and cTnT proteins in cardiomyocytes in vitro.
  • doxorubicin significantly reduced the protein levels of cTnl and cTnT in NRCM; the presence of NRG-I, however, maintained the levels of these proteins in doxorubicin-treated cardiomyocytes.
  • NRGl inhibited doxorubicin-induced caspase and proteasome degradation of cTnl and cTnT in cardiomyocytes in vitro.
  • PI3K inhibitor LY294002 abolished these effects of NRGl .
  • the present inventors further demonstrated that doxorubicin induced increasing of cytochrome c release to the cytosol in NRCM. NRGl treatment, however, inhibited this effect of doxorubicin (Figure 3C).
  • This result in combination with the findings of caspase-3, 6 and 9 activations, suggested that doxorubicin increased mitochondrial outer membrane permeabilization, which may be responsible for the activation of caspase-3, 6 and 9, and NRGl blocked these effects of doxorubicin.
  • NRG-I inhibited doxorubicin-induced activation of both intrinsic and extrinsic caspase activation, which were responsible, at least in part, for the degradation of cTnl and cTnT.
  • doxorubicin-induced down-regulation of cTnl was blocked by MGl 32 (Figure 2D).
  • the present inventors asked whether doxorubicin increased proteasome degradation of cTnl and whether NRGl blocked this effect.
  • doxorubicin increased the ubiquitinylation of cTnl; NRGl treatment abolished this effect of doxorubicin.
  • NRGl decreased doxorubicin-induced proteasome degradation of cTnl.
  • NRGl alleviated doxorubicin-induced decrease in synthesis of cTnl and cTnT in cardiomyocytes in vitro.
  • the present inventors measured cTnl and cTnT protein levels in doxorubicin-treated WT and dnPBK mice. Without doxorubicin treatment, the protein levels of cTnl and cTnT were similar in WT and dnPBK hearts. Two-weeks after the doxorubicin injection, a decrease in cTnl protein levels was observed in dnPBK-Dox-Placebo treated hearts compared with non-treated dnPBK hearts. Surprisingly, NRGl treatment still abolished doxorubicin-induced down-regulation of cTnl in dnPBK hearts (dnPI3K-Dox-NRG, Figure 5C). No changes in cTnT protein levels were observed in the hearts of doxorubicin-treated mice compared to control mice at this point.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • General Physics & Mathematics (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cardiology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Physiology (AREA)
  • Rheumatology (AREA)
  • Diabetes (AREA)

Abstract

The present invention relates to a method for detecting heart damage in a patient. The invention also relates to methods for treatment of patients identified as having heart damage. The invention further pertains to methods for evaluating the efficacy of an ongoing therapeutic regimen designed to treat a damaged heart in a patient.

Description

METHODS FOR DETECTING CARDIAC DAMAGE
CROSS REFERENCE TO RELATED APPLICATION
[0001| This application claims priority under 35 USC § 1 19(e) from U. S Provisional Application Serial No. 60/928,541 , filed May 10, 2007, which application is herein specifically incorporated by reference in its entirety.
FIELD OF THE INVENTION
[0002] The invention relates to the field of medical diagnostics. More particularly, the invention is directed to a method for detecting heart damage in a patient. The invention also relates to methods for treatment of patients identified as having heart damage. The invention also pertains to methods for evaluating the efficacy of an ongoing therapeutic regimen designed to treat a damaged heart in a patient.
BACKGROUND OF THE INVENTION
[0003] Several publications and patent documents are referenced in this application in order to more fully describe the state of the art to which this invention pertains. The disclosure of each of these publications and documents is incorporated by reference herein.
[0004] Heart failure is causally related to a number of conditions that damage the heart, including coronary heart disease, with or without a heart attack; hypertension; diseases, infections, or toxins that affect the heart muscle; and diseases of the heart valves. The onset of heart failure can occur rapidly, over days to weeks, but more frequently develops slowly over the course of years, as the heart gradually and progressively weakens.
[0005] Therapeutic intervention directed to reduction of cancer cell load in a patient frequently, if not always, is accompanied by a range of deleterious side effects. Indeed, cytostatic agents used as chemotherapeutics for the treatment of various cancers frequently exhibit potentially lethal side effects, including cardiotoxicity. Agents commonly used in cytostatic therapy include the anthracyclines daunorubicin and prodrugs thereof, zorubicin, doxorubicin (adriamycin) and epirubicin, and the synthetic antibiotic mitoxantrone. Anthracyclines, for example, represent a class of chemotherapeutic agents based on daunosamine and tetra-hydro-naphthacene-dione. These compounds are used to treat a variety of cancers, including leukemias and lymphomas, and solid tumors of the breast, uterus, ovary, and lung. In addition to the expected adverse reactions observed in patients undergoing chemotherapy, such as hair loss and nausea, therapeutic intervention involving anthracycline administration is complicated and limited by the marked cardiotoxicity of this class of compounds. Cardiotoxicity associated with anthracycline use is correlated with the total dose administered and is frequently irreversible. The cytostatic effects and cardiotoxicity of these compounds are due, at least in part, to alterations in membrane fluidity and permeability caused by anthracycline binding to components of the cell membrane. Free radical formation in the heart and accumulation of anthracycline metabolites are also thought to contribute to heart damage. Cardiotoxicity often presents in electrocardiogram (EKG) abnormalities and arrhythmias or as cardiomyopathy, which may ultimately lead to congestive heart failure.
SUMMARY OF THE INVENTION
[0006] The invention is directed to providing novel diagnostic methods for screening patients to identify those exhibiting signs of heart damage. Patients so identified can then be treated with pharmaceutical preparations for the treatment of heart damage as described herein. In a particular aspect of the invention, diagnostic methods for screening patients to identify those exhibiting signs of damage to the heart due to, for example, cardiotoxicity, hypertension, valvular disorders, myocardial infarction, viral myocarditis, or scleroderma are presented. In a particular aspect, the invention is focused on identifying patients exhibiting cardiotoxicity resulting from chemotherapeutic intervention. Classification of such patients serves to identify a subgroup of patients in need of therapeutic intervention to alleviate short and long term effects of cardiotoxicity. The subgroup of patients so identified can be treated with pharmaceutical preparations for the treatment of heart damage that occurs in connection with the use of cardiotoxic doses of medicaments or chemicals. Under circumstances wherein the heart damage identified in a patient is due to an ongoing condition, such as, hypertension, valvular disorders, myocardial infarction, viral myocarditis, or scleroderma, appropriate pharmaceutical preparations can also be formulated to treat the patient with heart damage.
[0007] The present invention also encompasses a method for stratifying patients according to degree or type of heart damage, knowledge of which guides a skilled practitioner to choose appropriate therapeutic regimens. The invention also includes a method whereby the efficacy of a therapeutic regimen is evaluated.
[0008] The novel methods of the invention are based on the discovery that changes in intracellular levels of cardiac troponin I (cTnl) and cardiac troponin T (cTnT) in intact cardiac tissue can be used as indicators for the presence of cardiac damage. More specifically, the present inventors have discovered that a decrease in intracellular cTnl and cTnT levels in intact cardiac tissue serves as a diagnostic marker to identify patients at risk for or experiencing cardiac damage. Cardiac tissue can be excised from a patient and tested in vitro or analyzed in vivo using molecular imaging protocols known in the art. [0009| Using either approach, intracellular cTnl and cTnT levels determined for the patient's cardiac tissue are compared to those of control cardiac tissue that expresses wildtype or normal levels of intracellular cTnl and cTnT. Reduced levels of intracellular cTnl and/or cTnT in a patient's cardiac tissue are readily determined by quantitating protein levels, which can be achieved using standard methods, and analyzing the results to determine if a statistically significant decrease in intracellular cTnl and cTnT levels is apparent in the patient's cardiac tissue relative to that of the control. Patients showing evidence of reduced intracellular cTnl and/or cTnT levels are earmarked for treatment with appropriate compositions chosen to restore, at least in part, normal heart function as reflected in an increase in intracellular cTnl and cTnT levels or restoration of normal levels of intracellular cTnl and cTnT.
[00010] In an embodiment of the present invention, the control or normal intracellular levels of either cTnT or cTnl in cardiac tissue are established by determining the intracellular levels of either cTnT or cTnl in cardiac tissue of a patient with normal heart function. In another embodiment of the present invention, the control or normal intracellular levels of either cTnT or cTnl in cardiac tissue are established by determining the intracellular levels of either cTnT or cTnl in cardiac tissue of a patient prior to onset of treatment capable of causing heart damage.
[00011] In an aspect of the present invention, the heart damage is a result of cardiotoxicity, hypertension, valvular disorders, myocardial infarction, viral myocarditis, or scleroderma. In a further aspect of the invention, the cardiotoxicity is caused by treatment with a chemotherapeutic agent or radiation.
[00012] It is also within the scope of the invention to evaluate the efficacy of a therapeutic regimen designed to at least partially restore normal heart function by measuring intracellular cTnl and cTnT levels in cardiac tissue of a treated patient. In accordance with the present invention, an increase in intracellular cTnl and cTnT levels in cardiac tissue of a treated patient relative to those determined prior to treatment is a positive indicator that the treatment is acting to restore cardiac function. [00013] It is to be understood that intracellular levels of either cTnl or cTnT in cardiac tissue or intracellular levels of both cTnl and cTnT in cardiac tissue may be used as indicators of cardiac tissue activity and/or function. This applies to all aspects of the invention, including methods directed to evaluating or diagnosing cardiac damage, methods directed to stratifying patients with respect to particular therapeutic regimens, and methods directed to evaluating efficacy of a therapeutic regimen. [00014] In accordance with the present invention decreased levels of cTnl and/or cTnT mRNA in cardiac tissue are also indicative of heart damage and may be used to stratify patient populations. Partial or complete restoration of normal cTnl and/or cTnT mRNA levels is also, therefore, a positive indicator of therapeutic efficacy as described above with respect to protein levels.
[00015J The present invention pertains to animals, in general, and more particularly, to mammals, and even more particularly to humans. Accordingly, the subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human. Accordingly, the term "subject" or "patient" may be used to refer to a human.
[00016] The present invention also encompasses a combination therapeutic regimen wherein
GGF2 or an epidermal growth factor-like (EGFL) domain encoded by the neuregulin gene is administered in conjunction with a proteasome inhibitor to treat cardiac damage. An exemplary proteasome inhibitor for use in the present invention is Proscript 519, which is a potent and selective proteasome inhibitor. Other proteasome inhibitors of utility in the present invention include Velcade™ and lactacystin. Additional proteasome inhibitors are known to those skilled in the art. Indeed, proteasome inhibitors are already used as therapeutic agents for the treatment of a number of diseases, including some cancers and neurodegenerative diseases.
[00017] Also encompassed by the present invention is the use of GGF2 or an epidermal growth factor-like (EGFL) domain encoded by the neuregulin gene in the preparation of a medicament for administration to a patient identified by the present diagnostic methods as exhibiting damage to the heart. The invention further encompasses the use of GGF2 or an epidermal growth factor-like (EGFL) domain encoded by the neuregulin gene in combination with a proteasome inhibitor in the preparation of a medicament for administration to a patient identified by the present diagnostic methods as exhibiting damage to the heart.
[00018] Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims. BRIEF DESCRIPTION OF THE DRAWINGS
[000191 Figures IA-I C show survival graphs (A), histograms (B-C). and immunoblots (C). For the survival analysis (A), mice were injected with a single dose of doxorubicin [20mg/kg, intraperitoneally (i.p.)] with or without concomitant injection of NRGl (0.75mg/kg. s.c. daily). Fourteen day survival was analyzed by the Kaplan-Meier method. With respect to a determination of serum creatine kinase (CK) levels (B), serum CK levels were measured in control, Dox-treated and Dox- NRGl treated mice four days after doxorubicin injection. Figure 1C shows that NRGl injection alleviated doxorubicin-induced down-regulation of cTnl, cTnT and cTnC protein levels in mice. Mice were treated with doxorubicin (20mg/kg, i.p.) with or without concomitant NRGl injection (0.75mg/kg, s.c. daily). Protein levels of cTnl, cTnT and cTnC were measured by Western blot analysis five days after doxorubicin treatment.
[00020] Figures 2A-D show imunoblots probed to detect the indicated proteins. Figure 2A reveals that NRGl alleviated doxorubicin-induced down-regulation of cTnl and cTnT protein levels in neonatal rat cardiomyocytes (RNCM). RNCM were treated with doxorubicin (IuM) in the presence or absence of NRGl (20ng/ml or 50ng/ml). cTnl and cTnT protein levels were measured by Western blot analysis 48 hours after doxorubicin treatment. Figure 2B shows that inhibition of erbB2 abolished the effects of NRGl on cTnl and cTnT. RNCM were treated with doxorubicin (IuM) and NRGl (20ng/ml) in the presence or the absence or AG879 (lOuM) and AG1478 (lOuM). Protein levels of cTnl and cTnT were analyzed by Western blot analysis. As shown in Figure 2C, RNCM were treated with doxorubicin and NRGl in the presence of LY294002 (lOuM), Akti (5uM), PD98059 (5OuM) and Rapamycin (1OnM). cTnl and cTnT protein levels were analyzed by Western blot analysis. Figure 2D shows that RNCM were treated with doxorubicin or doxorubicin+NRGl in the presence of cycloheximide (5ug/ml), Z-VAD (10OuM) or MGl 32 (lOuM). Protein levels of cTnl and cTnT were measured by Western blot analysis.
[00021] Figures 3A-3D show immunoblots (A, C, D), and histograms (B). Figure 3A presents results wherein RNCM were treated with doxorubicin (IuM) in the presence of inhibitors for different caspases (2OuM). The protein levels of cTnl and cTnT were measured by Western blot analysis. Figure 3B shows the effects of caspase activation in doxorubicin-treated RNCM. Cells were treated with Dox, Dox+NRGl or Dox+NRGl +LY. Caspase activation was analyzed by the caspase activation assay. Figure 3 C shows that NRGl decreased doxorubicin-induced cytochrome c release. RNCM were treated with Dox or Dox+NRGl . Cytochrome c release was analyzed by cell fractionation and Western blot analysis. Figure 3D reveals that NRGl decreased doxorubicin-induced ubiquitinylation of cTnl.
RNCM were treated with Dox or Dox+NRGl . Cell lysates were immunoprecipated with cTnl antibody and probed with ubiquitin antibody.
[00022] Figures 4A-4B show ethidium bromide stained agarose gels (A) and immunoblots (B).
Figure 4 A reveals that NRG-I inhibited doxorubicin-induced down-regulation of mRNA levels of cTnl, cTnT and cardiac specific transcriptional factors. RNCM were treated with Dox or Dox+NRGl . mRNA levels of cTnl, cTnT, GATA4, MEF2c and NKX2.5 were analyzed by quantitative RT-PCR.
Figure 4B shows that NRGl inhibited doxorubicin-induced dephosphorylation of translational molecules. RNCM were treated with Dox, Dox+NRGl or Dox+NRGl+LY. The phosphorylation levels of mTOR, P70S6K, S6, 4EBP and EIF4G were analyzed by Western blot analysis.
[00023] Figures 5A-5C show a survival graph (A), histograms (B), and an immunoblot (C).
Figure 5A shows a survival analysis in doxorubicin-treated mice with cardiac myocyte-specific overexpression of a dominant negative PI3K (dnPDK). Mice were treated with a single dose of doxorubicin (20mg/kg, i.p.) with or without concomitant treatment of NRGl (0.75mg/kg, s.c).
Fourteen-day survival was analyzed by the Kaplan-Meier method. Figure 5B depicts hemodynamic measurements in doxorubicin-treated dnPI3K mice. Mice were treated with a single dose of doxorubicin (20mg/kg, i.p.). Hemodynamic measurements were performed six days after the doxorubicin treatment. Figure 5C shows cTnl protein levels in dnPDK mice treated with Dox or
Dox+NRGl.
[00024] Figures 6A-D show amino acid and nucleic acid sequences of GGF2.
[00025] Figure 7 shows amino acid and nucleic acid sequences of EGFLl .
[00026] Figure 8 shows amino acid and nucleic acid sequences of EGFL2.
[00027] Figure 9 shows amino acid and nucleic acid sequences of EGFL3.
[00028] Figure 10 shows amino acid and nucleic acid sequences of EGFL4.
[00029] Figure 11 shows amino acid and nucleic acid sequences of EGFL5.
[00030] Figure 12 shows amino acid and nucleic acid sequences of EGFL6.
DETAILED DESCRIPTION OF THE INVENTION
[00031] Typically, when a patient arrives at a hospital complaining of chest pain, the following diagnostic steps are taken to evaluate the condition of the patient's heart, and determine the severity of any problems identified. To begin, the patient is interviewed to compile a comprehensive list of symptoms so that a health care professional can rule out non-heart related problems. Second, an electrocardiogram (BKG ) reading is taken, which records the electrical waves made by the heart. The EKG is an essential tool for determining the sev erity of chest pains associated with heart conditions and measuring the degree of damage to the heart. Blood tests arc also performed to detect elevated serum levels of certain factors, such as the troponins and creatine kinase (CK). and the more cardiac specific isoform of creatine kinase (CK-MB). which are indicative of heart damage. The rise in serum levels of CK, CK-MB, and the troponins is due to the release of these molecules following cardiac muscle cell death and serves, therefore, as a serum marker of necrosis. As a heart muscle cell dies as a result of prolonged ischemia, for example, the cell membrane ruptures, releasing the cytosolic contents into the extracellular fluid space, from whence it enters the lymphatic system, and subsequently the bloodstream. Imaging tests, including echocardiogram and perfusion scintigraphy, may also be used in the context of diagnosis.
[00032] The most specific markers of cardiac necrosis available are the cardiac troponins. These proteins are components of the contractile apparatus of myocardial cells. Two cardiac troponins, cTnl and cTnT, have been commercialized and detection of these markers has proven to be a reliable and specific assay for detection of minimal levels of myocardial damage. The cardiac troponins, like CK- MB, are released from dead cardiac muscle cells upon rupture of cell membranes, and are eventually detectable in the blood. Necrosis can occur as a result of a prolonged myocardial ischemia, but can also result from myocardial cell damage from other causes such as infection, trauma, or congestive heart failure.
[00033] The present invention differs from those procedures described in the prior art in a variety of aspects. At the outset, it is directed to measuring intracellular levels of cTnl and cTnT in intact cardiac tissue, rather than serum levels of these markers. Moreover, the present inventors have discovered that a decrease in intracellular cTnl and cTnT levels in intact cardiac tissue serves as a diagnostic marker to identify patients at risk for or experiencing cardiac damage. This approach stands in marked contrast to measurements of serum levels of these markers, an increase of which is indicative of heart damage. Moreover, an increase in serum levels of these markers is an acute or transient marker of heart damage, whereas measurements of intracellular levels of cTnl and cTnT in intact cardiac tissue serves as a stable marker reflective of the condition of the heart. In accordance with the present invention, identification of patients exhibiting a decrease in intracellular cTnl and cTnT levels in intact cardiac tissue also provides a screening method with which to stratify patients into categories for subsequent treatment. Patients showing evidence of reduced intracellular cTnl and cTnT levels are earmarked for treatment with appropriate compositions chosen to restore, at least in part, normal heart function as reflected in restoration of such.
[00034] An exemplary therapeutic agent for inclusion in such a composition is glial growth factor
2 (GGF2). The amino acid and nucleic acid sequences of GGF2 are presented in Figures 6A-6D. Therapeutic compositions may also include other exemplary polypeptides, such as epidermal growth factor-like (EGFL) domains encoded by the neuregulin gene, as shown in Figures 7-12, and described in United States Patent Number (USPN) 5,530,109, which is incorporated herein in its entirety. [00035] In order to more clearly set forth the parameters of the present invention, the following definitions are used:
[00036] As used in this specification and the appended claims, the singular forms "a", "an", and
"the" include plural references unless the context clearly dictates otherwise. Thus for example, reference to "the method" includes one or more methods, and/or steps of the type described herein and/or which will become apparent to those persons skilled in the art upon reading this disclosure. [00037] The compositions containing the molecules or compounds of the invention can be administered for diagnostic and/or therapeutic treatments. In diagnostic applications, compositions are administered to a patient to determine if the patient has cardiac damage and/or to stratify the patient with respect to prospective therapeutic regimens. In therapeutic applications, compositions are administered to a patient diagnosed as having cardiac damage in an amount sufficient to treat the patient, thereby at least partially arresting the symptoms of the disease and its complications. An amount adequate to accomplish this is defined as a "therapeutically effective amount or dose." Amounts effective for this use will depend on the severity of the disease and the weight and general state of the patient. [00038] As used herein, the phrase "control sample of cardiac tissue" refers to a sample of cardiac tissue for which intracellular levels of cTnl and cTnT are within normal range. A normal or wildtype range of intracellular levels of cTnl and cTnT is established based on experiments such as those presented herein and known in the art wherein cardiac tissue of a subject having healthy heart function, as determined by a skilled practitioner, is used as the standard against which unknowns are compared. Standards, representative of normal hearts, may, for example, be procured from fresh autopsies performed on cadavers having no evidence of heart disease.
[00039] Similarly, the term "control or normal levels" refers to levels established or determined as described herein and understood in the art to be within a range associated with healthy functionality. With respect to the present invention, healthy functionality refers to healthy heart function, which can be assessed by a skilled practitioner using standard procedures such as measuring systolic and diastolic blood pressure, measuring serum levels of indicator proteins such as CK. CK-MB, and the troponins, performing an EKG, and/or administering a stress test. A skilled practitioner would be aware of that which is generally considered a normal serum level of these proteins.
[00040] Various studies have been presented with respect to serum CK levels, for example, and general guidelines have been established. In one such study, for example, patients with suspected myocardial infarction (MI) who had a serum creatine kinase level of 280 IU/L or more were very likely to have had an MI; patients with a serum creatine kinase level of 80 to 279 IU/L were likely to have had an MI; patients with a creatine kinase level of 40 to 79 IU/L were less likely to have had an MI; and patients with a creatine kinase level of less than 40 IU/L were much less likely to have had an MI. With respect to that which is considered a normal serum level of CK or troponins, it is to be understood that different hospitals have established standards that vary slightly. Moreover, a skilled practitioner would be cognizant of the accepted standard in the particular clinical setting (e.g., particular hospital) in which the practitioner is working.
[00041] Troponin is also recognized as a sensitive and specific marker for cardiac injury. Indeed, detection of serum troponin I (sTnl) is considered to be more accurate than creatine kinase-MB concentrations for the diagnosis of MI and provides more useful prognostic information. Detection of sTnl also permits the early identification of those patients with acute coronary syndromes who are at an increased risk of death. sTnl is more sensitive than creatine kinase-MB concentrations for detection of minor ischemic myocardial injury in patients with small increases of total creatine kinase and avoids the high incidence of false diagnoses associated with the use of creatine kinase-MB as a diagnostic marker in perioperative MI. In one study, for example, patients with moderate elevations of serum troponin I (0.4-2.0 μg/L) had a significantly higher mortality rate and longer length of intensive care unit and hospital stays when compared with patients without similar elevations. Within the range of moderately elevated troponin concentrations, higher titers were associated with increasing mortality risk, longer hospital and intensive care unit stays, and a higher incidence of myocardial infarction. Treatment of patients exhibiting maximum serum troponin concentrations equal to or greater than 2 μg/L with β- blockers and aspirin improved their prognosis.
[00042] With respect to normal levels of intracellular cTnl and cTnT, such determinations are established by evaluating normal heart tissue using standards methods for determining protein levels such as those taught herein and known in the art. Decreased levels of intracellular cTnl and cTnT. such as those indicative of an injured or diseased heart, are determined as a decrease in the levels of these proteins relative to an established normal level. By way of example, a decrease of at least 50% in the level of cTnl and/or cTnT in heart tissue being tested for damage, relative to that of healthy heart tissue (normal control), serves as a positive indicator that the heart tissue being tested is damaged and a patient from whom the damaged tissue was removed would benefit from therapeutic intervention such as that taught herein. In a more particular example, a decrease of at least 75% in the level of cTnT in heart tissue being tested for damage, relative to that of healthy heart tissue (normal control), serves as a positive indicator that the heart tissue being tested is damaged and a patient from whom the damaged tissue was removed would benefit from therapeutic intervention such as that taught herein. [00043] A skilled practitioner would also be aware of the large body of scientific literature pertaining to the activity and levels of intracellular cTnl and cTnT in normal and diseased heart tissue. Examples of references that pertain to intracellular cTnl and cTnT in normal and diseased heart tissue include: Latif et al. (2007, J Heart Lung Transplant 26:230-235); Birks et al. ( 2005, Circulation 1 12(9 Suppl):I57-4); Day et al. (2006, Ann NY Acad Sci 1080:437-450); VanBuren et al. (2005, Heart Fail Rev 10: 199-209); de Jonge et al. (2005, Int J Cardiol 98:465-470); Wen et al. (2008, J Biol Chem Apr 22, Epub ahead of print); Li et al. (2008, Biochem Biophys Res Commun 369:88-99); Robinson et al. (2007, Circ Res 101 :1266-1273); Solzin et al. (2007, Biophys J 93:3917-3931); (Chen et al. (2007, Cardiovasc Toxicol 7:1 14-121); Solaro et al. (2007, Circ Res 101 :1 14-1 15); Kubo et al. (2007, J Am Coll Cardio 49:2419-2426); Day et al. (2007, J MoI Med 85:91 1-921); Milting et al. (2006, MoI Call Cardiol 41 :441-450); the entire contents of each of which is incorporated herein by reference. [00044] In another embodiment of the invention, an increase in intracellular cTnl and cTnT levels, as determined by assaying levels of the proteins before and during (or after) a therapeutic regimen, demonstrates the therapeutic efficacy of the regimen and provides evidence that the regimen is promoting restoration of normal heart function. Indeed, an increase in intracellular cTnl and cTnT levels may be used as a surrogate endpoint (i.e, a biomarker intended to substitute for a clinical endpoint) for improved heart function. If, however, intracellular cTnl and cTnT levels remain at a reduced level or are further reduced after therapeutic intervention, a skilled practitioner would reconsider the merit of the regimen with respect to the patient being treated and alter or potentially truncate the therapeutic regimen. [00045] As used herein, "preventing'"' or "prevention" refers to a reduction in risk of acquiring or developing a disease or disorder (i.e.. causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to a disease-causing agent, or predisposed to the disease in advance of disease onset.
[00046] The term "prophylaxis" is related to "prevention", and refers to a measure or procedure the purpose of which is to prevent, rather than to treat or cure a disease. Non-limiting examples of prophylactic measures may include the administration of vaccines; the administration of low molecular weight heparin to hospital patients at risk for thrombosis due, for example, to immobilization; and the administration of an anti-malarial agent such as chloroquine, in advance of a visit to a geographical region where malaria is endemic or the risk of contracting malaria is high.
[00047] The term "treating" or "treatment" of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting the disease or reducing the manifestation, extent or severity of at least one of the clinical symptoms thereof). In another embodiment, "treating" or "treatment" refers to ameliorating at least one physical parameter, which may not be discernible by the subject. In yet another embodiment, 'treating' or 'treatment' refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
Invasive procedures for Isolating Tissue
[00048] Myocardial biopsy is a common procedure in which heart tissue is obtained from the heart through a catheter, during thoracotomy, or during open chest surgery. It is commonly used to diagnose the etiology of heart failure. The heart tissue is analyzed both histologically and biochemically. The results of these tests are useful in diagnosing the cause of the heart failure. For review of myocardial biopsy in the clinical setting see Veinot (2002, Can J Cardiol. 18(3):287-96), the entire disclosure of which is incorporated herein by reference.
[00049] The results of the myocardial biopsy may indicate that heart failure is the result of such causes as scleroderma, viral myocarditis, drug toxicity or any number of causes of heart failure. This diagnosis will dictate what, if any, therapeutic intervention may be useful and should be employed. [00050] Myocardial biopsy (or cardiac biopsy) is an invasive procedure, wherein a bioptome (a small catheter with a grasping device on the end), for example, may be used to obtain a small piece of heart muscle tissue that can be analyzed. Myocardial biopsy may be used to evaluate heart transplant rejection and to diagnose myocarditis (inflammation of the heart).
[00051] Although a skilled practitioner would be aware of the basic protocol for myocardial biopsy using a catheter, it essentially involves the following: a local anesthetic is used to numb part of the neck or groin of a patient; a practitioner inserts a plastic introducer sheath (a short, hollow tube through which the catheter is placed) into a blood vessel in the numbed region; a bioptome is inserted through the sheath and threaded to the right ventricle of the patient; and samples are collected from the heart using the grasping device of the bioptome. During the procedure, an x-ray camera is generally used to position the bioptome properly. Samples are about the size of the head of a pin. When a sufficient number of samples have been collected, the catheter is removed and localized bleeding is controlled by firm pressure. Patients may be awake and conscious during the procedure. [00052] Certain proteins have routinely been measured in the blood stream following a myocardial event to predict and diagnose if and the extent to which the myocardium has been damaged. These proteins include, but are not limited to, creatine kinase and troponin. Detecting specific sub-types of these and other proteins in the blood stream is diagnostic of release of the proteins from the myocardium and thus damage. Detection of these indicator proteins in the sera is used routinely in the acute setting where a cardiac event is suspected and have proven useful in determining the best treatment for the patient. While useful in the acute setting, cardiac protein levels in the blood stream have no value in diagnosing or predicting heart failure more than a few days after a cardiac event, nor do they have any value in determining the etiology of heart disease or proper treatment course. As cardiac protein levels are not elevated in the blood except for immediately following a cardiac event, measurement of these proteins in the blood stream reveals little regarding the state of the myocardium. [00053] In contrast, the present invention describes the use of myocardial biopsy to predict which patients may be responsive to a cardioprotective, cardiorestorative and other heart failure therapy. Predicting which patient will respond to these therapies will improve treatment by helping to ensure that the correct patients receive specific therapies and, significantly, limit the number of patients that receive therapies that will have little value and thus only expose them to the risk of potentially serious side effects. Additionally, myocardial biopsy and determination of certain protein content may demonstrate which patients are responding to certain therapies and with whom treatment should be continued. [00054] The data presented herein show that, as predicted, immediately following cardiac challenge certain proteins, including creatine kinase and troponin can be detected in the blood stream. The data also demonstrate that one can measure a stable decrease in myocardial proteins from samples of myocardium taken long after the cardiac proteins in the blood stream have returned to normal low levels. These data also show that treatment of failing hearts with neuregulin can prevent the decline and restore the cardiac protein content of the diseased heart. This restoration correlates with improved survival and cardiac physiology.
[00055] Moreover, these data demonstrate that neuregulin can restore the troponin content of the myocardium following insult. Troponin is a key protein essential for the contractile properties of the heart. Restoration of the troponin content is thus important for restoration of normal cardiac physiology. The use of myocardial troponin content measurement in a myocardial biopsy to determine if reduced troponin myocardial content is a component of the reduced cardiac function will help predict which patients may respond to a cardiorestorative therapy such as administration of a neuregulin. Additionally, myocardial troponin content measurement may be used as an assay for determining which patients are responding to a cardiorestorative therapy such as a neuregulin. Knowing when patients are responding favorably to the therapy will help predict if and when such patients should be treated again, observed or removed from therapy. Myocardial troponin content measurement may be used to optimize dosing for individual patients that are being treated for heart disease with a cardiorestorative treatment. Similarly, in a chronic disease state in which continuous decline of cardiac troponin levels occur, maintenance of myocardial troponin levels may indicate success of a cardioprotective therapy such as a neuregulin in the presence of ongoing disease. In this manner myocardial troponin levels may similarly be used to predict response to therapy and optimization of dosing.
[00056] Dosing for GGF2 or EGFL domains of neuregulin, for example, can be initiated at about
100 mg/kg and dosing thereof titrated to higher levels based on patient response, including an assessment of intracellular levels of cTnl and cTnT in cardiac tissue of the patient. [00057] Once isolated, intracellular levels of cTnl and cTnT in cardiac tissue can be determined by methods described in the Examples presented herein and known in the art. Cellular lysates of isolated cardiac tissue or precipitates derived therefrom may, for example, be analyzed using a variety of techniques including, but not limited to, immunoblot analysis, enzyme-linked immunosorbent assay (ELISA), and mass spectrometry. Indeed, an ELISA protocol has been developed to detect cardiac- specific troponin T that utilizes a high-affinity, cardiac-specific antibody (Ml 1.7). The detection limit of this assay is lower than that of first generation ELISA protocols that used the cross-reacting antibody I BlO (0.0123 μg/L versus 0.04 μg/L, respectively). Methods for detecting Troponin I and T levels In Vivo
[00058] Technological advances in the field of molecular imaging have made possible noninvasive, high-resolution in vivo imaging techniques that enable clinicians to diagnose and evaluate therapeutic efficacy on a molecular and cellular level. The term molecular imaging is, indeed, "broadly defined as the in vivo characterization and measurement of biological processes at the cellular and molecular level". See Weissleder et al. (Radiology 219:316-333, 2001), which is incorporated herein in its entirety. Nuclear imaging, for example, which includes positron emission tomography (PET), micro- PET, single photon emission computed tomographic (SPECT), and planar imaging, generally involves visualizing an endogenous or expressed protein using specific radiopharmaceuticals as detection probes. PET, for example, is capable of producing a three-dimensional image or map of functional processes in the body which facilitates real time analyses of cellular components and molecular interactions within cells. PET is used as both a medical and a research tool. With respect to medical applications, it is used extensively in clinical oncology to image tumors and detect metastases and in clinical diagnosis of a variety of brain diseases, especially those associated with dementia. The ability to perform repeated PET analyses on a patient enables a skilled practitioner to compare results over time so as to evaluate, for example, disease progression or efficacy of a selected treatment protocol. PET has also been used as a research tool to map normal human brain and heart function.
[00059] Alternative methods of scanning include x-ray computed tomography (CT), magnetic resonance imaging (MRI), functional magnetic resonance imaging (fMRI), and ultrasound. Imaging scans such as CT and MRI are well suited to visualize organic anatomic changes in the body, whereas, as indicated above, PET scanners, like SPECT and fMRI, have the resolution to detect changes on a molecular level, even in advance of changes evident on the anatomic level. Imaging scans such as PET achieve this end by using radiolabeled molecular probes that exhibit different rates of uptake, depending on the type and function of the tissue of interest. Alterations in regional blood flow in various anatomic structures can also be visualized and quantified with a PET scan. Some of the above scanning techniques can be used in combination, depending on compatibility of radioisotopes utilized, so as to provide more comprehensive information that improves the accuracy of the clinical assessment. This is discussed herein below with respect to SPECT imaging.
[00060] As indicated above, nuclear imaging techniques such as PET, micro-PET, SPECT, and planar imaging, are directed to visualizing an endogenous or expressed protein using specific radiopharmaceuticals as detection probes. Imaging marker genes that encode intracellular enzymes and imaging marker genes that encode cell surface proteins or receptors have been used successfully in a variety of experimental systems to image specific molecules in vivo. The activity of an intracellular enzyme may also be assessed by labeling by-products indicative of the level and/or activity of a particular intracellular enzyme. PET and microPET, for example, use positron-labeled molecules to image processes involved in metabolism, cellular communication, and gene expression. Molecular imaging technologies are described in Kim (Korean J Radiology 4:201-210, 2003), which is incorporated herein by reference in its entirety.
[00061] Radionuclides used in PET scanning are typically isotopes with short half lives such as
1 1C (-20 rain), 13N (-10 min), 15O (~2 min), and 18F (-110 min). These radionuclides are incorporated into compounds normally used by the body such as glucose, water or ammonia and then injected into the body to trace where they become distributed. Such labeled compounds are known as radiotracers. The short half-life of these radiotracers restricts clinical PET primarily to the use of tracers labeled with 18F, which has a half life of 1 10 minutes and can be transported a reasonable distance before use, or to 82Rb, which can be created in a portable generator and is used for myocardial perfusion studies. [00062] SPECT is an example of a nuclear medicine tomographic imaging technique that uses gamma rays. It is very similar to conventional nuclear medicine planar imaging using a gamma camera. Unlike planar imaging, however, SPECT can provide true 3D information. This information is typically presented as cross-sectional slices through the patient, but can be modified with regard to presentation as required. The image obtained by a gamma camera image is a 2-dimensional view of 3 -dimensional distribution of a radionuclide. Because SPECT acquisition is very similar to planar gamma camera imaging, the same radiopharmaceuticals may be used for either protocol. If a patient is examined using an alternative type of nuclear medicine scan, for example, but the images are non-diagnostic, it may be possible to perform a subsequent imaging scan using SPECT by reconfiguring the camera for SPECT image acquisition while the patient remains on the table or moving the patient to a SPECT instrument. [00063] SPECT can also be used for cardiac gated acquisitions. To obtain differential information about the heart in various parts of its cycle, gated myocardial SPECT can be used to obtain quantitative information about myocardial perfusion, thickness, and contractility of the myocardium during various parts of the cardiac cycle. It is also used to facilitate calculation of left ventricular ejection fraction, stroke volume, and cardiac output. [00064] Myocardial perfusion imaging (MPI) is an example of functional cardiac imaging, which is used for the diagnosis of ischemic heart disease. It is based on the principle that impaired or diseased myocardium receives less blood flow than normal myocardium under conditions of stress. In brief, a cardiac specific radiopharmaceutical is administered, for example, 99mTc-tetrofosmin (Myoview™, GE healthcare) or 99mTc-sestamibi (Cardiolite®, Bristol-Myers Squibb), after which administration the heart rate is raised to induce myocardial stress. In accordance with standard practice, enhanced heart rate is typically either exercise induced or pharmacologically induced with adenosine, dobutamine or dipyridamole. SPECT imaging performed after induction of stress reveals the distribution of the radiopharmaceutical, and therefore the relative blood flow to the different regions of the myocardium. Diagnosis is made by comparing stress images to a subsequent set of images obtained at rest. [00065] In that troponin is a structural component of the cytoskeleton and an enzyme, molecular imaging techniques are envisioned to measure intracellular cTnl and cTnT levels in vivo. [00066] Indeed, cardiac troponin T is present in myocytes at high concentrations, both in cytosolic and structurally-bound protein pools. The cytosolic pool amounts to 6%, whereas the amount in myofibrils corresponds to 94% of the total troponin T mass in the cardiomyocyte. In view of the high levels normally present in myocytes, labeling of either or both pools of troponin T will generate sufficient signal to be visualized with a reasonable degree of accuracy and resolution. [00067] One potential approach envisioned for visualizing troponin complexes in cardiac tissue in vivo takes advantage of the calcium (Ca2+) binding properties of this complex. It is known that contraction is initiated by Ca2+ binding to troponin (Tn) in striated muscle, and more particularly by the Ca2+ -binding subunit of Tn (TnC), which leads to myosin binding to actin, force generation and shortening. The level of force is regulated by the availability of myosin binding sites on the thin filament, which is controlled by the position of tropomyosin (Tm) on the surface of actin. The inhibitory subunit of Tn (TnI) binds to actin in the absence of Ca2+, anchoring Tm so as to inhibit myosin binding. When Ca binds to TnC, it induces strong TnI-TnC interactions and weakens the TnI- actin interactions, resulting in increased mobility of Tm and exposure of strong myosin binding sites on actin. In addition, myosin binding to actin leads to crossbridge formation, which is thought to further displace Tm from blocking positions and is necessary for maximal activation of thin filaments in skeletal muscle.
[00068] In view of the above properties, a skilled practitioner would envision utilizing a Ca2+ radionuclide to visualize cytoskeletal components and, more particularly, troponin levels in cardiac tissue in vivo. Alternatively, small molecules that specifically recognize either cTnl or cTnT may be labeled with radionuclides and administered to patients to visualize intracellular levels of these proteins. A skilled practitioner could envision a variety of labeled probes of utility in the present method, including ligands, antibodies, and substrates of cTnl or cTnT and/or proteins that interact with cTnl or cTnT.
[00069] Radiopharmaceuticals such as those described in USPN 5,324,502 may also be used to advantage in the present method for imaging myocardial tissues. As described in USPN 5,324,502, such radiopharmaceuticals are prepared by forming lipophilic, cationic complexes of radioactive metal ions with metal chelating ligands comprising the Schiff base adducts of triamines and tetraamines with optionally substituted salicylaldehydes. These lipophilic, cationic, radioactive complexes exhibit high uptake and retention in myocardial tissues. Preferred gallium-68(III) complexes in accordance with this invention can be used to image the heart using positron emission tomography. In an aspect of the present invention, such radiopharmaceuticals may be used as a means for targeting uptake of agents that bind to the troponins to cardiac tissue.
Pharmaceutical Compositions
[00070] The present invention also provides pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of an agent, and a pharmaceutically acceptable carrier. In a particular embodiment, the term "pharmaceutically acceptable" means approved by a regulatory agency of the federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
[00071] Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin, incorporated in its entirety by reference herein. Such compositions contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide a form for proper administration to a subject. The formulation should suit the mode of administration.
[00072] In a particular embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
[00073] The compounds of the invention can be formulated as neutral or salt forms.
Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
[00074] The amount of the compound of the invention which will be effective in the treatment of a heart damage can be determined by standard clinical techniques based on the present description. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances. However, suitable dosage ranges for intravenous administration are generally about 20-500 micrograms of active compound per kilogram body weight. Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 1 mg/kg body weight. Suppositories generally contain active ingredient in the range of 0.5% to 10% by weight; oral formulations preferably contain 10% to 95% active ingredient. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. [00075J Various delivery systems are known and can be used to administer a compound of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu (1987) J. Biol. Chem. 262:4429-4432), and construction of a nucleic acid as part of a retroviral or other vector. Methods of introduction can be enteral or parenteral and include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The compounds may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, it may be desirable to introduce the pharmaceutical compositions of the invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
[00076] In a specific embodiment, it may be desirable to administer the pharmaceutical compositions of the invention locally, e.g., by local infusion during surgery, topical application, e.g., by injection, by means of a catheter, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. [00077] In another embodiment, the compound or agent can be delivered in a vesicle, in particular a liposome (see Langer (1990) Science 249: 1527-1533; Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
[00078] In yet another embodiment, the compound or agent can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14:201 ; Buchwald et al. (1980) Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321 :574). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release. Langer and Wise (eds.). CRC Pres.. Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.). Wiley. New York (1984); Ranger and Peppas. J.. 1983, Macromol. Sci. Rev. Macromol. Chem. 23:61 ; see also Levy et al. (1985) Science 228: 190; During et al. (1989) Ann. Neurol. 25:351 ; Howard et al. (1989) J. Neurosurg. 71 : 105). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, i.e., damaged heart, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)). Other controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533). [00079] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, particular methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
EXAMPLE I
[00080] The present inventors have previously reported that GGF2, a recombinant neuregulin- 1 , improves survival and cardiac function in doxorubicin-treated mice. As described herein, the present inventors have investigated whether GGF2 prevents doxorubicin-induced cardiac myofibril loss in vivo and in cardiomyocytes in vitro. The results presented herein have led to the novel discovery that the intracellular expression levels of particular cardiac proteins are useful indicators of normal heart function. More specifically, the present inventors have discovered that changes in the intracellular levels of cardiac troponin I (cTnl) and cardiac troponin T (cTnT) in intact cardiac tissue can be used as indicators of cardiac damage. In a particular aspect of the invention, a decrease in intracellular cTnl and cTnT levels in intact cardiac tissue has been shown to be a useful diagnostic marker to identify patients at risk for or experiencing cardiac damage. Such patients are then selected for appropriate preventative or therapeutic intervention as described herein. Determination of intracellular cTnl and cTnT levels in intact cardiac tissue may also be used to advantage to evaluate the efficacy of ongoing therapeutic intervention, since restoration of normal intracellular cTnl and cTnT levels would serve as a positive indicator that the therapy was improving heart function or restorative of normal heart function. Materials and Methods Materials
[00081] C57BL/6 mice and Wistar rats were obtained from Charles River Laboratories.
Doxorubicin was obtained from Bedford laboratories. Glial growth factor 2 was a gift from Acorda Therapeutics, Inc. MG 132, cycloheximide and actinomycin were obtained from Sigma. LY294002 and PD 98059 were from Cell Signalling Technology. Antibodies were ordered from the following vendors: Troponin I, GAT A4 and Nkx2.5 were from Santa Cruz Biotechnology; α-sarcomeric actin, troponin T, troponin C, tropomyosin and cardiac troponin T were from Abeam; Desmin and α-actinin were from Sigma and cardiac troponin I were from GeneTex. MEM, Hank's solution and fetal bovine serum were obtained from Invitrogen. All other reagents for cell culture were obtained from Sigma.
Animal models
[00082] Eight to ten week old C57BL/6 male mice were used for analyses wherein heart samples were isolated. A subacute doxorubicin cardiotoxicity model was used for this study. Mice were treated with a single dose of doxorubicin (20 mg/kg, i.p.). Twenty-four hours before, on the day, and every day after doxorubicin treatment, mice were treated with either GGF2 (0.75mg/kg, s.c.) or placebo (formulation buffer for GGF2). Mice were sacrificed 4.5 days after doxorubicin treatment. Heart samples (n=3-4 per group) were collected and snap-frozen in liquid nitrogen. [00083] With respect to those analyses wherein cardiac function was measured (as detailed below), three-month old C57BL/6 mice were treated with a single dose of doxorubicin (20 mg/kg, i.p.). Glial growth factor 2 (GGF2-Dox, 0.75 mg/kg, s.c, n=74) or placebo (the buffer used to dissolve GGF2, placebo-Dox, n=73) was injected into mice one day before and once daily following doxorubicin treatment. Mice without doxorubicin treatment were used as controls (n=20). Cardiac function was assessed by direct left ventricular (LV) catheterization four days and two weeks after doxorubicin treatment. Two-week survival was analyzed by the Kaplan-Meier methods.
Neonatal cardiac myocyte culture
[00084] Neonatal cardiac myocytes were dissociated as described previously (Okoshi et al.
Journal of Cardiac Failure. 2004; 10:51 1-518). In brief, ventricles from day 0-day 3 Wistar rats were dissociated in trypsin and Dnase II. Cells were washed and pre-plated in 100 mm dishes in MEM containing 5% fetal bovine serum. After 30 minutes, the myocytes were suspended in the same medium containing 0.1 mmol/L bromodeoxyuridine and then plated at the density of 500-1000 cells/mm2 in 100 mm culture dishes. Forty-eight hours after dissociation, the medium was changed to serum-free MEM containing 0.1% BSA and cultured overnight before stimulation.
NRGl improved survival and cardiac function in doxorubicin-treated mice.
[00085] The present results reveal that two-weeks after doxorubicin treatment, survival in doxorubicin treated mice was significantly decreased compared with non-treated control mice. Concomitant treatment with NRGl (Dox-NRGl), however, significantly improved survival in doxorubicin-injured mice compared to placebo-treated mice (Dox-Placebo) (Figure IA). Cardiac function was further assessed five days after doxorubicin injection, at which point, survival started to decline in doxorubicin-treated mice. As shown in Table 1 , the body weight (BW), heart weight (HW) and left ventricular weight (LVW) were significantly decreased in both Dox-Placebo and Dox-NRGl mice compared with control (untreated) mice. HW and LVW normalized by tibia length (HW/TL and LVW/TL) were significantly decreased in Dox-Placebo mice compared to control mice. These indices were not, however, different between Dox-NRGl and control mice. LV systolic pressure (LVSP), cardiac output and dP/dt min were significantly decreased in Dox-Placebo mice compared with controls. In contrast, these indicators of heart function were not significantly different between Dox-NRGl mice and control mice, indicating an improvement of cardiac systolic function in NRGl (Dox-NRGl) treated mice compared with Placebo treated mice (Dox-Placebo). Serum creatine kinase (CK) levels, an index of cardiac injury, were also assessed to provide an additional read-out of heart function. As shown in Figure IB, the CK level was significantly increased in both Dox-Placebo and Dox-NRGl mice compared to control mice. The CK level was significantly lower in Dox-NRGl mice, however, as compared with Dox-Placebo mice. These results demonstrated that NRG- 1 treatment significantly improved survival and cardiac systolic function in doxorubicin-injured mice.
[00086] Table 1. Hemodynamic measurements in Dox+Placebo and Dox+NRGl treated C57BL/6 mice.
Figure imgf000024_0001
*p<0.05; tp<0.01 vs. Control.
NRGl alleviated doxorubicin-induced down-regulation of cTnl and cTnT in the heart in vivo. [00087] One of the mechanisms of doxorubicin-induced cardiotoxicity is loss of cardiac myofibrils. As described herein, the present inventors investigated whether NRGl injection in vivo inhibited doxorubicin-induced myofibril loss. Results presented herein demonstrate that the levels of cardiac structural proteins, α-sarcomeric actin, α-actinin, troponin T (TnT), troponin I (TnI), troponin C (TnC) and tropomyosin were significantly decreased in doxorubicin-treated hearts. NRGl injection in vivo significantly increased the protein levels of cTnl, cTnT and cTnC in doxorubicin-injured hearts (Figure 1C), but had no effects on the protein levels of α-sarcomeric actin. α-actinin and tropomyosin.
NRGl abolished doxorubicin-induced down-regulation of cTnl and cTnT proteins in cardiomyocytes in vitro.
[00088] To further study the mechanism of how NRGl inhibited doxorubicin-induced down- regulation of cTnl and cTnT, the present inventors conducted in vitro studies using neonatal rat cardiomyocyte culture (NRCM). As shown in Figure 2A, doxorubicin significantly reduced the protein levels of cTnl and cTnT in NRCM; the presence of NRG-I, however, maintained the levels of these proteins in doxorubicin-treated cardiomyocytes. These results further demonstrated that these effects of NRGl were blocked by inhibitors for erbB2, PI3K, Akt, mTOR or ERK (Figure 2B and 2C), but were not blocked by inhibitors for erbB4 (Figure 2B), p38 or PKC. The instant results also showed that the preventative and/or restorative effects of NRGl were blocked by cycloheximide, a protein translation inhibitor (Figure 2D), but not by actinomycin D, a transcription inhibitor. Doxorubicin-induced down- regulation of cTnl and cTnT was abolished by Z-VAD, a pan-caspase inhibitor, and MGl 32, a proteasome inhibitor (Figure 2D), but not by bafilomycin Al, a lysosome inhibitor.
NRGl inhibited doxorubicin-induced caspase and proteasome degradation of cTnl and cTnT in cardiomyocytes in vitro.
[00089] The maintenance of the level of a protein in the cell is a dynamic process. It depends on the rate of synthesis and degradation of a protein. The present inventors explored the mechanism whereby doxorubicin decreased the protein levels of cTnl and cTnT to determine if this effect is caused by increasing their degradation and/or by decreasing their synthesis and whether NRGl blocked any of these effects of doxorubicin.
[00090] To investigate whether NRGl inhibited doxorubicin-induced caspase degradation of cTnl and cTnT, the present inventors first identified the specific caspases that were responsible for doxorubicin-induced degradation of cTnl and cTnT. Cardiomyocytes were treated with doxorubicin in the presence of a specific caspase inhibitor. As shown in Figure 3 A, inhibitors for caspase-3, caspase-6 or caspase-9 (intrinsic pathway) blocked doxorubicin-induced down-regulation of both cTnl and cTnT. In addition, down-regulation of cTnl was blocked by inhibitors for caspase- 10 (extrinsic pathway), caspase-2, caspase- 13 or caspase-5. On the other hand, the down-regulation of cTnT was also blocked by caspase-2 and caspase-13. The present inventors then tested whether doxorubicin activated and whether NRGl inhibited the activation of these caspases. An in vitro caspase activation assay revealed that doxorubicin significantly increased the activation of caspase 3, 6 and 9 as well as caspase- 10, 2, and 5. NRGl treatment of cardiomyocytes significantly inhibited doxorubicin-induced activation of these caspases (Figure 3B). PI3K inhibitor LY294002 abolished these effects of NRGl . The present inventors further demonstrated that doxorubicin induced increasing of cytochrome c release to the cytosol in NRCM. NRGl treatment, however, inhibited this effect of doxorubicin (Figure 3C). This result, in combination with the findings of caspase-3, 6 and 9 activations, suggested that doxorubicin increased mitochondrial outer membrane permeabilization, which may be responsible for the activation of caspase-3, 6 and 9, and NRGl blocked these effects of doxorubicin. These results demonstrated that NRG-I inhibited doxorubicin-induced activation of both intrinsic and extrinsic caspase activation, which were responsible, at least in part, for the degradation of cTnl and cTnT.
[00091] The present inventors further demonstrated that doxorubicin-induced down-regulation of cTnl was blocked by MGl 32 (Figure 2D). In short, the present inventors asked whether doxorubicin increased proteasome degradation of cTnl and whether NRGl blocked this effect. As shown in Figure 3D, doxorubicin increased the ubiquitinylation of cTnl; NRGl treatment abolished this effect of doxorubicin. This result demonstrated that NRGl decreased doxorubicin-induced proteasome degradation of cTnl.
NRGl alleviated doxorubicin-induced decrease in synthesis of cTnl and cTnT in cardiomyocytes in vitro.
[00092] To further test whether doxorubicin decreased the transcription of cTnl and cTnT and whether NRGl reversed this effect of doxorubicin, the mRNA levels of these proteins in Dox-Placebo and Dox-NRGl treated cardiomyocytes were measured. As shown in Figure 4A, doxorubicin decreased the mRNA of both cTnl and cTnT. On the other hand, NRGl maintained the mRNA level of cTnl and cTnT in doxorubicin-treated cardiomycoytes. In addition, NRGl maintained the mRNA level of GATA4 and slightly increased the mRNA level of MEF2c and Nkx2.5 (Figure 4A), which are transcriptional factors important for cardiac specific gene transcription.
[00093] The present results showed that NRG- Ts effects on cTnl and cTnT were blocked by cycloheximide (Figure 2D), suggesting that NRGl increased the translation of these proteins. We assessed the activation of several translation machineries and related signaling pathways in Dox-Placebo and Dox-NRG l treated cardiomyocytes. As shown in Figure 4B, 48 hours after the treatment, the phosphorylation levels of mTOR (Ser 2448), P70S6K(Thr421/Ser424), S6(Ser240/244) and eIF4G (Serl 108) were decreased in Dox-Placebo, but were maintained in Dox-NRGl treated cardiomycoytes. LY294002 blocked these effects of NRG-I . These results suggest that NRG, via PBK, maintained the activation of protein translational machineries in doxorubicin-treated cardiomyocytes, which may be responsible for maintaining the protein levels of cTnl and cTnT.
[00094] These results demonstrated that NRGl alleviated doxorubicin-induced down-regulation of cTnl and cTnT via multiple mechanisms, which include inhibition of the activation of intrinsic and extrinsic caspases, as well as inflammatory activated caspases, inhibition of the ubiquitinylation of cTnl, and an increase in transcription and the activation of translation signaling and machineries. These results also suggested that PBK played a major role for NRGl in maintaining cTnl and cTnT levels in cardiomyocytes.
[00095] To further investigate the role of PBK in mediating NRG l's cardiac protective effects in vivo, the present inventors used transgenic mice with cardiac myocyte- specific overexpression of a dominant negative PBK and treated them with doxorubicin as described above. As shown in Figure 5 A, the survival rate was decreased in dnPBK-Dox-Placebo mice compared with WT-Dox-Placebo mice. NRGl (WT-Dox-NRG) treatment improved survival in doxorubicin injured WT (WT-Dox-Placebo) mice (67% vs. 33%). Intriguingly, the magnitude of this improvement was greater than that observed in C57BL/6 male mice (Figure IA). The present results further showed that this improvement of the survival rate was dampened in doxorubicin-treated dnPBK mice (dnPBK-Dox-NRG: 56% vs. WT-Dox- NRG: 67%).
[00096] Cardiac function was also evaluated in these mice. As shown in Figure 5B, LVSP, dP/dt max and dP/dt min, as well as cardiac output were more severely depressed in dnPBK-Dox-Placebo mice compared with dnPBK control than WT-Dox-Placebo mice compared with WT mice, indicating more severe cardiac dysfunction in dnPBK-Dox-Placebo mice.
[00097] The present inventors measured cTnl and cTnT protein levels in doxorubicin-treated WT and dnPBK mice. Without doxorubicin treatment, the protein levels of cTnl and cTnT were similar in WT and dnPBK hearts. Two-weeks after the doxorubicin injection, a decrease in cTnl protein levels was observed in dnPBK-Dox-Placebo treated hearts compared with non-treated dnPBK hearts. Surprisingly, NRGl treatment still abolished doxorubicin-induced down-regulation of cTnl in dnPBK hearts (dnPI3K-Dox-NRG, Figure 5C). No changes in cTnT protein levels were observed in the hearts of doxorubicin-treated mice compared to control mice at this point.
[00098] These results demonstrate that GGF2 specifically maintains TnT and TnI protein levels in doxorubicin-injured hearts. Moreover, these findings reveal that GGF2 increases survival of doxorubicin-treated mice and this is associated with an improvement in cardiac function as evident in mice treated with both doxorubicin and GGF2.
[00099] While certain of the preferred embodiments of the present invention have been described and specifically exemplified above, it is not intended that the invention be limited to such embodiments.
Various modifications may be made thereto without departing from the scope and spirit of the present invention, as set forth in the following claims.

Claims

IN THE CLAIMS:
1. A method for screening a population of patients to identify a subpopulation of patients exhibiting heart damage, the method comprising: detecting intracellular levels of cardiac troponin T (cTnT) or cardiac troponin I (cTnl) in cardiac tissue of a patient in the population, wherein a decrease in intracellular levels of either cTnT or cTnl in cardiac tissue of the patient relative to control or normal intracellular levels of either cTnT or cTnl, respectively, is indicative of heart damage and classifies the patient as a member of the subpopulation of patients exhibiting heart damage.
2. The method of claim 1 , wherein detecting intracellular levels of cTnT or cTnl in cardiac tissue is performed in vitro.
3. The method of claim 1, wherein detecting intracellular levels of cTnT or cTnl in cardiac tissue is performed in vivo.
4. The method of claim 1, wherein the heart damage is a result of cardiotoxicity, hypertension, valvular disorders, myocardial infarction, viral myocarditis, or scleroderma.
5. The method of claim 4, wherein the cardiotoxicity is caused by treatment with a chemotherapeutic agent or other therapeutic agent, radiation, or agents used for non-therapeutic, recreational purposes.
6. The method of claim 1, wherein the control or normal intracellular levels of either cTnT or cTnl in cardiac tissue are established by determining the intracellular levels of either cTnT or cTnl in cardiac tissue of a patient with normal heart function.
7. The method of claim 1, wherein the control or normal intracellular levels of either cTnT or cTnl in cardiac tissue are established by determining the intracellular levels of either cTnT or cTnl in cardiac tissue of the patient prior to onset of treatment capable of causing heart damage.
8. The method of claim 1 , wherein the patient is a mammal.
9. The method of claim 8, wherein the mammal is a human.
10. The method of claim 1 , further comprising treating patients in the subpopulation of patients exhibiting heart damage with a composition comprising at least one therapeutic agent.
1 1. The method of claim 10, wherein the at least one therapeutic agent is glial growth factor 2 (GGF2).
12. A method for diagnosing a patient suspected of having cardiac damage, the method comprising: detecting intracellular levels of cardiac troponin T (cTnT) or cardiac troponin I (cTnl) in cardiac tissue of the patient and comparing the intracellular levels of cTnT or cTnl in cardiac tissue detected for the patient to those of a control sample of cardiac tissue, wherein a decrease in intracellular levels of either cTnT or cTnl in cardiac tissue of the patient relative to those of the control sample of cardiac tissue is indicative of cardiac damage in the patient and identifies the patient as a candidate for therapeutic intervention to treat the heart damage.
13. The method of claim 12, wherein the control or normal intracellular levels of either cTnT or cTnl in cardiac tissue are established by determining the intracellular levels of either cTnT or cTnl in cardiac tissue of a patient with normal heart function.
14. The method of claim 12, wherein the patient is a mammal.
15. The method of claim 14, wherein the mammal is a human.
16. A method for determining efficacy of a therapeutic regimen, the method comprising: a) detecting intracellular levels of cardiac troponin T (cTnT) or cardiac troponin I (cTnl) in cardiac tissue of a patient prior to initiation of the therapeutic regimen; b) detecting intracellular levels of cTnT or cTnl in cardiac tissue of the patient after initiation of the therapeutic regimen; and
PCT/US2008/006060 2007-05-10 2008-05-12 Methods for detecting cardiac damage WO2008140814A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
JP2010507486A JP5749490B2 (en) 2007-05-10 2008-05-12 Method for detecting heart failure
CA2686959A CA2686959C (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage
MX2009012174A MX2009012174A (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage.
MX2014013196A MX348082B (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage.
US12/451,397 US8628929B2 (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage
BRPI0810631A BRPI0810631A2 (en) 2007-05-10 2008-05-12 methods for cardiac damage detection
RU2009145695/15A RU2498309C2 (en) 2007-05-10 2008-05-12 Methods for detecting cardiac disorders
IN1227MUN2014 IN2014MN01227A (en) 2007-05-10 2008-05-12
CN200880021887.5A CN101743474B (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage
AU2008251822A AU2008251822B2 (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage
EP08767674A EP2153228A4 (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage
US14/153,695 US9329171B2 (en) 2007-05-10 2014-01-13 Methods for detecting cardiac damage
US15/053,284 US9757429B2 (en) 2007-05-10 2016-02-25 Methods for detecting cardiac damage
US15/674,053 US10258667B2 (en) 2007-05-10 2017-08-10 Methods for detecting cardiac damage
US16/359,383 US11071770B2 (en) 2007-05-10 2019-03-20 Methods for detecting cardiac damage

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US92854107P 2007-05-10 2007-05-10
US60/928,541 2007-05-10

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/451,397 A-371-Of-International US8628929B2 (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage
US14/153,695 Continuation US9329171B2 (en) 2007-05-10 2014-01-13 Methods for detecting cardiac damage

Publications (1)

Publication Number Publication Date
WO2008140814A1 true WO2008140814A1 (en) 2008-11-20

Family

ID=40002559

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/006060 WO2008140814A1 (en) 2007-05-10 2008-05-12 Methods for detecting cardiac damage

Country Status (11)

Country Link
US (5) US8628929B2 (en)
EP (1) EP2153228A4 (en)
JP (5) JP5749490B2 (en)
CN (2) CN101743474B (en)
AU (1) AU2008251822B2 (en)
BR (1) BRPI0810631A2 (en)
CA (2) CA2965208C (en)
IN (1) IN2014MN01227A (en)
MX (2) MX2009012174A (en)
RU (1) RU2498309C2 (en)
WO (1) WO2008140814A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011119836A1 (en) * 2010-03-24 2011-09-29 Massachusetts Institute Of Technology Methods and compositions for cardioprotection and cardioregeneration
WO2012025355A1 (en) * 2010-08-26 2012-03-01 Roche Diagnostics Gmbh Use of biomarkers in the assessment of the early transition from arterial hypertension to heart failure
US9198952B2 (en) 2008-09-22 2015-12-01 The Brigham And Women's Hospital, Inc. Compositions of and methods of using ligand dimers
RU2585391C1 (en) * 2014-12-19 2016-05-27 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования Чеченский государственный университет Method for immunological prediction of small myocardial damage
US9498515B2 (en) 2008-08-15 2016-11-22 Acorda Therapeutics, Inc. Compositions and methods for treatment during non-acute periods following CNS neurological injury
US20180296642A1 (en) * 2015-09-25 2018-10-18 Douglas B. Sawyer Methods for treating cardiac injury
RU2709458C2 (en) * 2017-12-22 2019-12-18 Марина Александровна Чичкова Method for optimization of early immunological diagnostics of complications of percutaneous coronary intervention in patients with ihd
RU2749552C1 (en) * 2020-08-03 2021-06-15 Федеральное государственное бюджетное научное учреждение "Томский национальный исследовательский медицинский центр Российской академии наук" (Томский НИМЦ) Method for selecting patients with long-term persistent atrial fibrillation for effective catheter treatment
US11071770B2 (en) 2007-05-10 2021-07-27 Acorda Therapeutics, Inc. Methods for detecting cardiac damage
RU2760242C1 (en) * 2021-03-01 2021-11-23 федеральное государственное бюджетное учреждение "Национальный медицинский исследовательский центр имени В.А. Алмазова" Министерства здравоохранения Российской Федерации Method for assessing myocardial damage and risk of complications development after cardiac surgery

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8609620B2 (en) * 2008-11-28 2013-12-17 Zensun (Shanghai) Science & Technology Ltd. Neuregulin peptides and their use
US8652788B2 (en) * 2009-12-02 2014-02-18 Abbott Laboratories Assay for diagnosis of cardiac myocyte damage
US8835120B2 (en) * 2009-12-02 2014-09-16 Abbott Laboratories Assay for cardiac troponin-T (cTnT)
AU2014407088B2 (en) 2014-09-26 2021-09-23 Somalogic Operating Co., Inc. Cardiovascular risk event prediction and uses thereof
RU2648529C1 (en) * 2016-12-27 2018-03-26 Федеральное государственное бюджетное образовательное учреждение высшего образования "Астраханский государственный медицинский университет" Министерства здравоохранения Российской Федерации (ФГБОУ ВО Астраханский ГМУ Минздрава России) Estimation method of the efficiency of medical therapy in adult patients with acute heart failure after operations in bare heart, receiving treatment with the method of extracorporal membrane oxygenation
RU2722111C1 (en) * 2019-12-30 2020-05-26 Федеральное государственное бюджетное образовательное учреждение высшего образования "Самарский государственный медицинский университет" Министерства здравоохранения Российской Федерации Method of managing patients with chronic lymphatic leukemia in a polychemotherapy according to a fcr scheme aimed at preventing cardiotoxicity
CN113238060B (en) * 2021-05-08 2022-10-11 迈克生物股份有限公司 Kit for predicting or diagnosing myocarditis

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6635249B1 (en) * 1999-04-23 2003-10-21 Cenes Pharmaceuticals, Inc. Methods for treating congestive heart failure

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5530109A (en) 1991-04-10 1996-06-25 Ludwig Institute For Cancer Research DNA encoding glial mitogenic factors
US5716930A (en) 1991-04-10 1998-02-10 Ludwig Institute For Cancer Research Glial growth factors
US7115554B1 (en) * 1993-05-06 2006-10-03 Acorda Therapeutics, Inc. Methods of increasing myotube formation or survival or muscle cell mitogenesis differentiation or survival using neuregulin GGF III
US5324502A (en) 1992-04-02 1994-06-28 Purdue Research Foundation Radiopharmaceuticals for imaging the heart
US6092695A (en) 1992-05-11 2000-07-25 Cytologix Corporation Interchangeable liquid dispensing cartridge pump
US6653249B1 (en) * 2000-05-25 2003-11-25 Penn Emblem Corporation Emblem having calendered fabric layer
GB0029154D0 (en) * 2000-11-30 2001-01-17 Lee Helen Signal enhancement with multiple labelled-antibodies
AU2002304965A1 (en) 2002-05-24 2003-12-12 Zensun (Shanghai) Sci-Tech.Ltd Neuregulin based methods and compositions for treating viral myocarditis and dilated cardiomyopathy
CN1267590C (en) * 2004-06-11 2006-08-02 清华大学 Preparing method for growth factor slow-releasing system for tissue repair
CN1715926B (en) * 2004-07-02 2011-08-17 上海泽生科技开发有限公司 Use of neuregulin mutant
WO2007011757A1 (en) 2005-07-14 2007-01-25 Myogen, Inc. Use of inhibitors of the ubiquitin proteasome pathway for increasing contractility of the heart
US7776817B2 (en) 2005-09-02 2010-08-17 Morehouse School Of Medicine Neuregulins for prevention and treatment of damage from acute assault on vascular and neuronal tissue and as regulators of neuronal stem cell migration
MX2009012174A (en) 2007-05-10 2010-03-30 Acorda Therapeutics Inc Methods for detecting cardiac damage.
DE102007061117A1 (en) 2007-12-19 2009-06-25 Robert Bosch Gmbh Electrical contact
US20130143805A1 (en) 2010-08-13 2013-06-06 Georgetown University Ggf2 and methods of use

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6635249B1 (en) * 1999-04-23 2003-10-21 Cenes Pharmaceuticals, Inc. Methods for treating congestive heart failure

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HERMAN E.H. ET AL.: "Use of cardiac troponin T levels as an indicator of doxorubicin-induced cardiotoxicity", CANCER RES., vol. 58, no. 2, 15 January 1998 (1998-01-15), pages 195 - 197, XP002584434 *
See also references of EP2153228A4 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11071770B2 (en) 2007-05-10 2021-07-27 Acorda Therapeutics, Inc. Methods for detecting cardiac damage
US10668131B2 (en) 2008-08-15 2020-06-02 Acorda Therapeutics, Inc. Compositions and methods for treatment during non-acute periods following CNS neurological injury
US9498515B2 (en) 2008-08-15 2016-11-22 Acorda Therapeutics, Inc. Compositions and methods for treatment during non-acute periods following CNS neurological injury
US9198952B2 (en) 2008-09-22 2015-12-01 The Brigham And Women's Hospital, Inc. Compositions of and methods of using ligand dimers
US9029328B2 (en) 2010-03-24 2015-05-12 The Brigham And Women's Hospital, Inc. Methods for cardioprotection and cardioregeneration with dimers of EGF family ligands
WO2011119836A1 (en) * 2010-03-24 2011-09-29 Massachusetts Institute Of Technology Methods and compositions for cardioprotection and cardioregeneration
US10942175B2 (en) 2010-08-26 2021-03-09 Roche Diagnostics Operations, Inc. Use of biomarkers in the assessment of the early transition from arterial hypertension to heart failure
WO2012025355A1 (en) * 2010-08-26 2012-03-01 Roche Diagnostics Gmbh Use of biomarkers in the assessment of the early transition from arterial hypertension to heart failure
RU2585391C1 (en) * 2014-12-19 2016-05-27 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования Чеченский государственный университет Method for immunological prediction of small myocardial damage
US20180296642A1 (en) * 2015-09-25 2018-10-18 Douglas B. Sawyer Methods for treating cardiac injury
US20210401938A1 (en) * 2015-09-25 2021-12-30 Douglas B. Sawyer Methods for treating cardiac injury
RU2709458C2 (en) * 2017-12-22 2019-12-18 Марина Александровна Чичкова Method for optimization of early immunological diagnostics of complications of percutaneous coronary intervention in patients with ihd
RU2749552C1 (en) * 2020-08-03 2021-06-15 Федеральное государственное бюджетное научное учреждение "Томский национальный исследовательский медицинский центр Российской академии наук" (Томский НИМЦ) Method for selecting patients with long-term persistent atrial fibrillation for effective catheter treatment
RU2760242C1 (en) * 2021-03-01 2021-11-23 федеральное государственное бюджетное учреждение "Национальный медицинский исследовательский центр имени В.А. Алмазова" Министерства здравоохранения Российской Федерации Method for assessing myocardial damage and risk of complications development after cardiac surgery

Also Published As

Publication number Publication date
JP5998171B2 (en) 2016-09-28
US9757429B2 (en) 2017-09-12
JP2017026622A (en) 2017-02-02
US20160263192A1 (en) 2016-09-15
JP2014122923A (en) 2014-07-03
JP5749490B2 (en) 2015-07-15
EP2153228A4 (en) 2010-08-04
AU2008251822B2 (en) 2014-02-06
RU2009145695A (en) 2011-06-20
MX348082B (en) 2017-05-25
AU2008251822A1 (en) 2008-11-20
US8628929B2 (en) 2014-01-14
CA2686959A1 (en) 2008-11-20
CN101743474B (en) 2015-06-03
US20180028610A1 (en) 2018-02-01
JP2010527000A (en) 2010-08-05
CN104614510B (en) 2017-07-04
JP2019010123A (en) 2019-01-24
US20140378377A1 (en) 2014-12-25
US9329171B2 (en) 2016-05-03
EP2153228A1 (en) 2010-02-17
CA2965208A1 (en) 2008-11-20
BRPI0810631A2 (en) 2017-05-09
US20190269759A1 (en) 2019-09-05
US11071770B2 (en) 2021-07-27
US10258667B2 (en) 2019-04-16
RU2498309C2 (en) 2013-11-10
CN104614510A (en) 2015-05-13
JP6636402B2 (en) 2020-01-29
JP6978553B2 (en) 2021-12-08
JP2020176138A (en) 2020-10-29
CN101743474A (en) 2010-06-16
MX2009012174A (en) 2010-03-30
CA2965208C (en) 2022-06-14
US20110085976A1 (en) 2011-04-14
IN2014MN01227A (en) 2015-07-03
CA2686959C (en) 2017-06-13

Similar Documents

Publication Publication Date Title
US11071770B2 (en) Methods for detecting cardiac damage
US5407653A (en) Evaluation of the multidrug resistance phenotype
US9575074B2 (en) Method for evaluating atherosclerotic lesion, and kit
JPH0662896A (en) Method of using spermidine/spermine n1-acetyl transferase as prognostic indicator and/or tumor response marker
Jong et al. Anthracycline-induced cardiotoxicity: From pathobiology to identification of molecular targets for nuclear imaging
AU2014202390B2 (en) Methods for detecting cardiac damage
US7695903B2 (en) Low-density lipoprotein receptor related protein-1 (LRP-1)in clearance of alzheimer&#39;s amyloid-beta peptide from the central nervous system
Ede et al. Serum copeptin level can be a helpful biomarker in evaluation of myocardial perfusion scintigraphy results
KR102469657B1 (en) Method for screening biomarkers diagnosing cardiovascular diseases, biomarkers screened thereby and method for diagnosing cardiovascular diseases using the same
Dali et al. Study of Short-term toxicity of Cyclophosphamide and Doxorubicin on the Cardiovascular System in women with Breast cancer treated at the center of radiation and chemotherapy at Tishreen University Hospital
KR101936502B1 (en) Biomarkers for diagnosis and prognosis of Hepatoma and uses thereof
Murphy et al. The need for early markers in Alzheimer’s disease

Legal Events

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

Ref document number: 200880021887.5

Country of ref document: CN

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

Ref document number: 08767674

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2686959

Country of ref document: CA

Ref document number: 2010507486

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2008251822

Country of ref document: AU

Ref document number: 2094/MUMNP/2009

Country of ref document: IN

Ref document number: MX/A/2009/012174

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2008251822

Country of ref document: AU

Date of ref document: 20080512

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2009145695

Country of ref document: RU

Ref document number: 2008767674

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12451397

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0810631

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20091110