US20020099075A1 - Combination therapy - Google Patents

Combination therapy Download PDF

Info

Publication number
US20020099075A1
US20020099075A1 US10/052,320 US5232002A US2002099075A1 US 20020099075 A1 US20020099075 A1 US 20020099075A1 US 5232002 A US5232002 A US 5232002A US 2002099075 A1 US2002099075 A1 US 2002099075A1
Authority
US
United States
Prior art keywords
carbonyl
guanidine
pyrazole
inhibitor
cyclopropyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/052,320
Other languages
English (en)
Other versions
US6423705B1 (en
Inventor
Wayne Tracey
Roger Hill
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/052,320 priority Critical patent/US6423705B1/en
Application granted granted Critical
Publication of US6423705B1 publication Critical patent/US6423705B1/en
Publication of US20020099075A1 publication Critical patent/US20020099075A1/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines

Definitions

  • the invention relates to methods of reducing tissue damage resulting from ischemia using a combination, or a pharmaceutical composition of such combination, of a sodium/hydrogen exchange type-1 (NHE-1) inhibitor and a second compound selected from the group consisting of: (a) a complement modulator, (b) a metabolic modulator, (c) an antiapoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator.
  • NHE-1 sodium/hydrogen exchange type-1
  • the invention further provides kits directed to such combinations.
  • Ischemic injury particularly to that of the myocardium, can occur in out patients as well as in perioperative settings and can lead to the development of sudden death, myocardial infarction, or congestive heart failure.
  • myocardial infarction can occur in out patients as well as in perioperative settings and can lead to the development of sudden death, myocardial infarction, or congestive heart failure.
  • myocardial infarction can occur in out patients as well as in perioperative settings and can lead to the development of sudden death, myocardial infarction, or congestive heart failure.
  • myocardial ischemic injury particularly perioperative myocardial infarction.
  • Such therapy is anticipated to be lifesaving and reduce the need for hospitalization, enhance quality of life and reduce overall health care costs of high risk patients.
  • cardioprotection would reduce the incidence and progression of myocardial infarction and dysfunction occurring in these perioperative surgical settings.
  • cardioprotection would also decrease the incidence of cardiac morbidity and mortality due to myocardial infarction and dysfunction in “at risk” patients (i.e., those patients greater than 65 years of age, exercise intolerant, those suffering from coronary artery disease, diabetes mellitus, or hypertension, and the like) that require non-cardiac surgery.
  • NHE-1 inhibitors that inhibit the sodium/hydrogen exchange type-1 (NHE-1) transport system.
  • NHE-1 inhibitors in combination with certain other therapeutic agents.
  • EPO 0 918 515 discloses the use of NHE-1 inhibitors with blood pressure reducing agents, ACE-inhibitors, angiotensin receptors antagonists, fat level reducing agents, and HMG-CoA reductase inhibitors
  • CA 2,227,112 discloses the use of NHE-1 inhibitors with sodium-dependent bicarbonate/chloride exchanger (NCBE) inhibitors
  • CA 2,245,776 discloses the use of NHE-1 inhibitors with, inter alia, ⁇ -receptor blockers, calcium antagonists, loop diuretics, thiazide diuretics, potassium-sparing diuretics, aldosterone antagonists, cardiac glycosides, antiarrythmics, K ATP channel openers, K ATP channel blockers, and veratride-activatable sodium channel inhibitors; and commonly assigned PCT International Application Publication No.
  • WO 99/43663 discloses the use of NHE-1 inhibitors with, inter alia, adenosine, adenosine agonists, nitrates, platelet inhibitors, aspirin, dipyridamol, potassium chloride, clonidine, prazosin, and adenosine A 3 -receptor agonists.
  • kits, methods and pharmaceutical compositions of the instant invention it is believed that the administration of a combination of an NHE-1 inhibitor and a second compound selected from the group consisting of: (a) a complement modulator, (b) a metabolic modulator, (c) an anti-apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor, will afford greater therapeutic advantages than either of the combination components administered alone.
  • a complement modulator a complement modulator
  • a metabolic modulator e.g., a metabolic modulator
  • an anti-apoptotic agent e.g.,
  • the invention provdes methods of reducing tissue damage resulting from ischemia which comprise administering to a mammal in need of such reduction an effective amount of a combination, or a pharmaceutical composition comprising such combination, of an NHE-1 inhibitor and a second compound selected from the group consisting of: (a) a complement modulator, (b) a metabolic modulator, (c) an anti-apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator.
  • the invention further provides pharmaceutical compositions comprising an amount of an NHE-1 inhibitor; an amount of a second compound selected from the group consisting of (a) a complement modulator, (b) a metabolic modulator, (c) an anti-apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor; and, preferably, a pharmaceutically acceptable pharmaceutically acceptable carrier, vehicle, or diluent.
  • a second compound selected from the group consisting of (a) a complement modulator, (b) a metabolic modulator, (c) an anti-apoptotic agent, (
  • the invention further provides a kit comprising an amount of a sodium-hydrogen exchanger type-1 inhibitor, and a pharmaceutically acceptable carrier, vehicle, or diluent in a first unit dosage form; an amount of a second compound selected from the group consisting of (a) a complement modulator, (b) a metabolic modulator, (c) an anti-apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor, and a pharmaceutically acceptable carrier, vehicle, or diluent in a second unit dosage form; and a container.
  • a second compound selected from the
  • the invention provides methods of reducing tissue damage (e.g., substantially preventing tissue damage and/or inducing tissue protection) resulting from ischemia which methods comprise administering to a mammal (e.g., human male or female) in need of such reduction a therapeutically effective amount of a combination, or a pharmaceutical composition comprising such combination, of a sodium-hydrogen exchanger type 1 (NHE-1) inhibitor, and a second compound selected from the group consisting of (a) a complement modulator, (b) a metabolic modulator, (c) an anti-apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose
  • Preferred ischemic tissues that may be treated in accordance with the methods of the present invention comprise those tissues selected from the group consisting of brain, cardiac, liver, kidney, lung, gut, skeletal muscle, spleen, pancreas, nerve, spinal cord, retinal tissue, vasculature, and intestinal tissue.
  • An especially preferred tissue comprises cardiac tissue.
  • NHE-1 inhibitor may be employed in the methods and pharmaceutical compositions of the present invention, it is generally preferred that such inhibitor be selected from the group consisting of:
  • Z is carbon connected and is a five-membered, diaza, diunsaturated ring having two contiguous nitrogens, said ring optionally mono-, di-, or tri-substituted with up to three substituents independently selected from R 1 , R 2 and R 3 ;
  • Z is carbon connected and is a five-membered, triaza, diunsaturated ring, said ring optionally mono- or di-substituted with up to two substituents independently selected from R 4 and R 5 ;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently hydrogen, hydroxy(C 1 -C 4 )alkyl, (C 1 -C 4 )alkyl, (C 1 -C 4 )alkylthio, (C 3 -C 4 )cycloalkyl, (C 3 -C 7 )cycloalkyl(C 1 -C 4 )alkyl, (C 1 -C 4 )alkoxy, (C 1 -C 4 )alkoxy(C 1 -C 4 )alkyl, mono-N- or di-N,N-(C 1 -C 4 )alkylcarbamoyl, M or M(C 1 -C 4 )alkyl, any of said previous (C 1 -C 4 )alkyl moieties optionally having from one to nine fluorines; said (C 1 -C 4 )alkyl or (C 3 -C 4 )cycloalkyl
  • M is a partially saturated, fully saturated or fully unsaturated five to eight membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen, or, a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen;
  • said M is optionally substituted, on one ring if the moiety is monocyclic, or one or both rings if the moiety is bicyclic, on carbon or nitrogen with up to three substituents independently selected from R 6 , R 7 and R 8 , wherein one of R 6 , R 7 and R 8 is optionally a partially saturated, fully saturated, or fully unsaturated three to seven membered ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen optionally substituted with (C 1 -C 4 )alkyl and additionally R 6 , R 7 and R 8 are optionally hydroxy, nitro, halo, (C 1 -C 4 )alkoxy, (C 1 -C 4 )alkoxycarbonyl, (C 1 -C 4 )alkyl, formyl, (C 1 -C 4 )alkanoyl, (C 1 -C 4 )alkanoyloxy, (C 1 -C 4 )alkanoylamino,
  • Especially preferred Formula (I) compounds are those compounds selected from the group consisting of:
  • the compounds of Formula (I), the prodrugs thereof, and the pharmaceutically acceptable salts of the compounds and prodrugs may be prepared as disclosed in the aforementioned, commonly-assigned PCT International Application Publication No. WO 99/43663, the disclosure of which is incorporated herein by reference.
  • the preferred NHE-1 inhibitor cariporide i.e. N-(aminoiminomethyl)-4-(1-methylethyl)-3-(methylsulfonyl)-benzamide
  • the preferred NHE-1 inhibitor eniporide i.e. N-(aminoiminomethyl)-2-methyl-5-(methylsulfonyl)-4-(1H-pyrrol-1-yl)-benzamide
  • U.S. Pat. No. 5,753,680 the disclosure of which is incorporated herein by reference.
  • the preferred NHE-1 inhibitor BIIB-513 i.e. N-(aminoiminomethyl)-4-(4-(2-furanylcarbonyl)-1-piperazinyl)-3-(methylsulfonyl)-benzamide, may be prepared as disclosed in U.S. Pat. No. 6,114,335, the disclosure of which is incorporated herein by reference.
  • the preferred NHE-1 inhibitor TY-12533 i.e. 6,7,8,9-tetrahydro-2-methyl-5H-cyclohepta[b]pyridine-3-carbonylguanidine, may be prepared as disclosed in PCT International Application Publication No. WO 98/39300, the disclosure of which is incorporated herein by reference.
  • the preferred NHE-1 inhibitor SM-15681 i.e. N-(aminoiminomethyl)-1-methyl-1H-indole-2-carboxamide, may be prepared as disclosed in EPO 0 708 091, the disclosure of which is incorporated herein by reference.
  • a complement modulator refers generally to an agent that modulates, i.e., regulates or inhibits, certain thermolabile substances, normally present in the serum, that are destructive to certain bacteria and other cells sensitized by a specific complement-fixing antibody known as antibody C.
  • Antibody C comprises a group of at least 20 disparate serum proteins, the activity of which is affected by a series of interactions resulting in enzymatic cleavages which can follow at least two distinct pathways.
  • the complement pathways contribute to myocardial ischemia-reperfusion injury in vivo, ostensibly via a combination of mechanisms including, but not limited to, stimulation of cytokine release from various cell types, expression of adhesion molecules, and neutrophil infiltration, of which all mechanisms directly elicit cell death. Accordingly, it is believed that the administration of a combination of an NHE-1 inhibitor and a complement modulator will afford greater protection from tissue damage resulting from ischemia than either agent administered alone.
  • complement modulator any complement modulator may be employed in the methods and pharmaceutical compositions of the instant invention, it is generally preferred that such complement modulator be selected from the group consisting of a C5a inhibitor, preferably L-747981, or a pharmaceutically acceptable salt thereof, a soluble complement receptor type 1 (sCR1) inhibitor, or an analog thereof, preferably amidinophenylpyruvic acid (APPA), and a Cl esterase inhibitor.
  • a C5a inhibitor preferably L-747981
  • sCR1 soluble complement receptor type 1
  • APPA amidinophenylpyruvic acid
  • the preferred C5a inhibitor L-747981 may be prepared according to known synthetic methods.
  • L-747981 to selectively bind to the C5a receptor is disclosed in Flanagan, K. L., et al., ACS, 210th Chicago: MEDI 085 (1995).
  • the preferred soluble complement receptor type 1 (sCR1) inhibitor amidinophenylpyruvic acid (APPA) may be obtained commercially.
  • the ability of a compound to inhibit C5a may be determined according to the methodology set forth in Vakeva, A. P., et al., Circulation, 97 (22), 2259-67 (1998).
  • the ability of a compound to inhibit complement activation at the sCR1 receptor may be determined according to the protocols described by Rittershaus, C.W., J. Biol. Chem., 274 (16), 11237-44 (1999).
  • the ability of a compound to inhibit Cl esterase may be determined according to the methodology disclosed in Benny, A. G., et al., Haematologia, 22(3),189-93 (1989).
  • metabolic modulator refers generally to any agent that serves to modulate, i.e., regulate, stimulate, or inhibit, one or more metabolic pathways.
  • metabolic modulators such as pyruvate dehydrogenase kinase inhibitors, for example, dichloroacetate (DCA), activate the myocardial dehydrogenase (PDC) complex, thus increasing glucose oxidation and decreasing fatty acid oxidation in the ischemic myocardium. Accordingly, reduction of ischemic tissue damage by treatment with a combination of an NHE-1 inhibitor and a metabolic modulator should elicit additional cardioprotective benefit.
  • DCA dichloroacetate
  • any metabolic modulator may be employed in the methods and pharmaceutical compositions of the instant invention, it is generally preferred that such modulator be selected from the group consisting of a pyruvate dehydrogenase complex up-regulator/activator, preferably dichloroacetate (DCA), a pyruvate dehydrogenase kinase inhibitor, preferably DCA, a malonyl CoA decarboxylase inhibitor, an acetyl CoA carboxylase activator, a partial fatty acid oxidation (pFOX) inhibitor, preferably ranolazine or trimetazidine, a 5′ AMP-activated protein kinase (AMPK) inhibitor, a carnitine palmitoyl transferase inhibitor, preferably etomoxir, and a fatty acid CoA synthase inhibitor, preferably triascin C.
  • DCA dichloroacetate
  • a pyruvate dehydrogenase kinase inhibitor
  • the preferred pFOX inhibitors ranolazine and trimetazidine may be prepared as disclosed in U.S. Pat. Nos. 4,567,264, and 4,663,325 respectively, which patents are incorporated herein in their entirety by reference.
  • the preferred carnitine palmitoyl transferase inhibitor etomoxir may be prepared as disclosed in U.S. Pat. No. 4,337,267, the disclosure of which is incorporated herein by reference.
  • the preferred fatty acid CoA synthase inhibitor triascin C, i.e. (2E,4E,7E)-undecatrienal-nitrosohydrazone may be prepared as disclosed in U.S. Pat. No. 4,297,096, the disclosure of which is incorporated herein by reference.
  • the ability of a compound to function as a pyruvate dehydrogenase complex up-regulator/activator, a pyruvate dehydrogenase kinase inhibitor, or a malonyl CoA decarboxylase inhibitor may be determined according to the methodologies disclosed in Stanley, W. C., et al., J. Mol. Cell. Cardiol., 28, 905-914 (1996).
  • the ability of a compound to function as an acetyl CoA carboxylase activator may be determined according to the protocols described in Belke, D. D., et al., Biochem. Biophys. Acta, 1391 (1), 25-36 (1998).
  • a compound to function as a partial fatty acid oxidation (pFOX) inhibitor may be determined according to the procedures of McCormack J. G., et al., J. Appl. Physiol., 81/2, 905-910 (1996), or Merrill G. F., et al., Am. J. Physiol., 273, E1107-1112 (1997).
  • the ability of a compound to function as a carnitine palmitoyl transferase inhibitor may be determined according to the methodology disclosed in Kudo, et al., J. Biol. Chem., 270, 17513-17520 (1995).
  • AMPK 5′ AMP-activated protein kinase
  • a reduction in absorbance at 620 nm indicates fatty acid CoA synthase inhibition.
  • an anti-apoptotic agent inhibits apoptosis, i.e., programmed cell death, the process by which certain cells self-destruct by fragmentation into membrane-bound particles which are subsequently phagocytized by other cells, for example, macrophages. Because apoptosis is known to occur during myocardial ischemia-reperfusion injury, it is believed that enzymatic inhibition of the apoptotic cascade with an anti-apoptic agent, in combination with treatment with an NHE-1 inhibitor, will confer greater protection from tissue damage resulting from ischemia than either agent administered alone.
  • caspase inhibitor refers to any agent that inhibits the activity of caspases, a salient family of enzymes involved in the induction of apoptosis in mammalian cells, for example, excessive apoptosis of cardiac myocytes during reperfusion, and neuronal cells during ischemia.
  • caspase inhibitors useful in the methods and pharmaceutical compositions of the instant invention, comprise those compounds selected from the group consisting of a compound of structural Formula (II)
  • R 1 and R 2 together with the nitrogen to which they are attached, form a 4- to 7-membered ring
  • R 3 and R 4 together with the nitrogen atom to which they are attached, form a 4- to 7-membered ring
  • R is benzoyl, or (C 1-6 )alkyl
  • R 1 is hydrogen, or (C 1-4 )alkyl
  • R 2 is (C 1-10 )alkyl, optionally substituted with aryl(C 1-4 )alkyl, optionally substituted heteroaryl(C 1-4 )alkyl, optionally substituted (C 3-7 )cycloalkyl, or R 1 and R 2 , together with the nitrogen to which they are attached, form a 3- to 10-membered ring which optionally contains an additional heteroatom selected from oxygen, nitrogen, or sulfur
  • R 3 and R 4 are (C 1-6 )alkyl, hydrogen, nitro, or halogen
  • R 5 is (C 1-6 )alkyl, hydrogen, arylalkyl, or heteroarylalkyl.
  • the preferred caspase inhibitors of structural Formula (II) may be prepared as described in PCT International Application Publication No. WO 99/65451.
  • the preferred caspase inhibitors of structural Formula (III) may be prepared as described in PCT International Application Publication No. WO 99/06367.
  • the ability of a compound to inhibit apoptosis may be determined according to the methodology of Wu, J. C., et al., Methods, 17 (4), 320-8 (1999).
  • the ability of an agent to function as a caspase inhibitor may be determined according to the methodologies disclosed in the aforementioned PCT International Application Publication Nos. WO 99/06367 and WO 99-65451.
  • nitric oxide synthase-related agent refers generally to any agent that regulates, i.e., inhibits, promotes, or enhances, the enzymatic formation of nitric oxide (NO) free-radical, a known mediator of cell-to-cell communication and potent vasodilator, which free-radical is produced by the nitric oxide synthase-catalyzed reaction of L-arginine with 2 O 2 and 1.5 NADPH.
  • NO nitric oxide
  • nitric oxide synthase iNOS
  • Type 3 inducible nitric oxide synthase
  • NO produced acutely by endothelial NOS (Type 2), or neuronal NOS (Type 1) during ischemia-reperfusion injury is also believed to be cardioprotective.
  • endothelial NOS (Type 2), or neuronal NOS (Type 1) during ischemia-reperfusion injury is also believed to be cardioprotective.
  • any nitric oxide synthase-related agent may be employed in the methods and pharmaceutical compositions of the present invention, it is generally preferred that such agent be selected from the group consisting of monophosphoryl lipid A, or an analog thereof, preferably RC-552 (MPL-C) or ONO-4007, a nitric oxide donor, preferably nipride, and a nitric oxide synthase inhibitor, preferably aminoguanidine or N(G)-monomethyl-L-arginine (L-NMMA).
  • MPL-C RC-552
  • ONO-4007 a nitric oxide donor
  • a nitric oxide synthase inhibitor preferably aminoguanidine or N(G)-monomethyl-L-arginine (L-NMMA).
  • the preferred monophosphoryl lipid A analog ONO-4077 i.e. (S)-2-deoxy-2-((1-oxo-3-((1-oxo-9-phenylnonyl)oxy)-tetradecyl)amino)-, 3-benzenenonanoate 4-(hydrogen sulfate)-D-glucose, may be prepared as disclosed in U.S. Pat. Nos. 5,294,723 and 5,733,927, the disclosures of which are incorporated herein by reference.
  • the preferred nitric oxide donor nipride i.e., sodium nitrosylpentacyanoferrate (II), may be prepared as described in Playfair, L., Proc.
  • the preferred nitric oxide synthase inhibitor aminoguanidine may be prepared as described in Smith, G. B. L., et al., J. Am. Chem. Soc., 57, 2730 (1935).
  • the preferred nitric oxide inhibitor N(G)-monomethyl-L-arginine (L-NMMA) may be obtained from commercial sources.
  • the NHE-1 inhibitor may further be employed in combination with an enzyme/protein modulator selected from the group consisting of a protein kinase C ⁇ activator, an endothelin converting enzyme inhibitor, preferably S-17162, a tissue-activated fibrinolytic (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, preferably KB-R7943, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor, preferably 3-aminobenzamide, or a compound, or a pharmaceutically acceptable salt, prodrug, active metabolite, or solvate thereof, of Formula (IV)
  • an enzyme/protein modulator selected from the group consisting of a protein kinase C ⁇ activator, an endothelin converting enzyme inhibitor, preferably S-17162, a tissue-activated fibrinolytic (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1)
  • R 1 is H, halogen, cyano, an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl group;
  • R 10 is hydrogen, an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl group;
  • R 100 and R 110 are each independently H or an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl group;
  • R 2 is H or alkyl
  • R 3 is H or alkyl
  • R 4 is H, halogen or alkyl
  • X is O or S
  • Y is (CR 5 R 6 )(CR 7 R 8 )n or N ⁇ C(R 5 ), where n is 0 or 1;
  • R 5 and R 6 are each independently H or an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl group; and
  • R 7 and R 8 are each independently H or an optionally substituted alky, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl group.
  • protein kinase C ⁇ refers to the cytoplasmic, calcium-activated family of kinases that are believed to be downstream mediators of, inter alia, the ischemic pre-conditioning pathway. Accordingly, because activators of this enzyme are cardioprotective, reduction of ischemic tissue damage by treatment with a combination of an NHE-1 inhibitor and a protein kinase C ⁇ activator should provide additional cardioprotective benefit.
  • Endothelin is known to contribute to myocardial ischemia-reperfusion injury and, therefore, by preventing endothelin production with an endothelin converting enzyme inhibitor, reduction of ischemic tissue damage by treatment with a combination of an NHE-1 inhibitor and an endothelin converting enzyme inhibitor should provide additional cardioprotective advantages.
  • Tissue-activated fibrinolytic inhibitors are known to be useful in the treatment of both deep venous thrombosis (DVT) and acute coronary syndrome (ACS), a syndrome that embraces, inter alia, ischemic attack. Accordingly, reduction of ischemic tissue damage by treatment with a combination of an NHE-1 inhibitor and a TAFI inhibitor should elicit additional cardioprotective benefit.
  • NCX-1 Na + /Ca +2 exchanger isoform-1
  • NHE-1 activity During cardiac reperfusion, Na + /Ca +2 exchanger isoform-1 (NCX-1) increases intracellular calcium levels, due to the increased intracellular sodium levels resulting from NHE-1 activity, which leads to contracture, arrhythmias, and cellular death.
  • NCX-1 inhibition is cardioprotective and, therefore, reduction of ischemic tissue damage by treatment with a combination of an NHE-1 inhibitor and an NCX-1 inhibitor should elicit additional cardioprotective benefit.
  • PARS/PARP is a DNA repair enzyme that is activated during myocardial ischemia/reperfusion injury in response to DNA single strand breaks.
  • the enzyme consumes intracellular NAD + and ATP pools, and slows the rate of glycolysis and mitochondrial respiration which can contribute to, or directly cause, cardiomyocyte dysfunction and/or death. Accordingly, PARS/PARP inhibitors are believed to be cardioprotective and, therefore, reduction of ischemic tissue damage by treatment with a combination of an NHE-1 inhibitor and a PARS/PARP inhibitor should elicit additional cardioprotective benefit.
  • the preferred endothelin converting enzyme inhibitor S-17162 i.e. N-(2,3-dihydroxypropylphosphonyl-(S)-Leu-(S)-Trp-OH, disodium salt, may be prepared as disclosed in U.S. Pat. Nos. 5,481,030, 5,591,728, and 5,608,045, the disclosures of which are hereby incorporated by reference in their entirety.
  • the preferred Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor KB-R7943, i.e., (2-(2-(4-(4-nitrobenzyloxy)-phenyl)-ethyl)-isothiourea methanesulfonate), may be prepared as described in PCT International Application Publication No. WO 97/09306.
  • the preferred poly (ADP ribose) synthetase (PARS/PARP) inhibitor 3-aminobenzamide is available commercially.
  • the preferred poly (ADP ribose) synthetase (PARS/PARP) inhibitors of Formula (IV) may be prepared as disclosed in PCT International Application Publication No. WO 2000/42040.
  • the ability of an agent to function as a protein kinase C ⁇ activator may be determined according to the protocol disclosed in Bowling, N., et al., Circulation, 99, 384-91 (1999).
  • the ability of an agent to function as an endothelin converting enzyme inhibitor may be determined according to Fassina, G., et al., Peptide Res., 6, 73-78 (1993).
  • the ability of an agent to function as a tissue-activated fibrinolytic inhibitor (TAFI) inhibitor may be determined according to Bajzar, L., et al., J. Biol. Chem., 270, 14477-14484 (1995).
  • NCX-1 Na + /Ca +2 exchanger isoform-1
  • NX-1 Na + /Ca +2 exchanger isoform-1
  • the ability of an agent to function as a poly (ADP ribose) synthetase (PARS/PARP) inhibitor may be determined according to the methodologies of Decker, P., et al., Clinical Cancer Research, 5, 1169-1172 (1999).
  • the NHE-1 inhibitor can be administered to a mammal at dosage levels in the range of from about 0.001 to about 100 mg/kg body weight per day.
  • dosage levels in the range of from about 0.001 to about 100 mg/kg body weight per day.
  • a dosage in the range of from about 0.01 to about 50 mg/kg body weight is typically preferred.
  • some variability in these general dosage ranges may be required depending upon the age and weight of the mammal being treated, the intended route of administration, the particular agent being administered, and the like.
  • the determination of dosage ranges and optimal dosages for a particular mammal is within the ability of one of ordinary skill in the art having the benefit of the instant disclosure.
  • the dosage of the complement modulator, metabolic modulator, anti-apoptotic agent, nitric oxide synthase-related agent, or enzyme/protein modulator will also be generally dependent upon a number of factors including the health of the mammal being treated, the extent of treatment desired, the nature and kind of concurrent therapy, if any, and the frequency of treatment and the nature of the effect desired.
  • dosage ranges of complement modulators, metabolic modulators, anti-apoptotic agents, nitric oxide synthase-related agents, and enzyme/protein modulators range from about 0.001 to about 250 mg/kg body weight per day.
  • a dosage in the range of from about 0.1 to about 25 mg/kg body weight is typically preferred.
  • some variability in this general dosage range may be required depending upon the age and weight of the subject being treated, the intended route of administration, the particular agent being administered and the like.
  • the determination of dosage ranges and optimal dosages for a particular mammal is also well within the ability of one of ordinary skill in the art having the benefit of the instant disclosure.
  • a complement modulator e.g., a metabolic modulator, an anti-apoptotic agent, a nitric oxide synthase-related agent, and an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an end
  • the NHE-1 inhibitor, and the second compound selected from the group consisting of a complement modulator, a metabolic modulator, an anti-apoptotic agent, a nitric oxide synthase-related agent, and an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor may be administered either separately or in the pharmaceutical composition comprising both. It is generally preferred that such administration be oral. However, if the subject being treated is unable to swallow, or oral administration is otherwise impaired or undesirable, parenteral or transdermal administration will be appropriate.
  • TAFI tissue-activated fibrinolytic inhibitor
  • NCX-1 Na + /Ca +2 exchanger isoform-1
  • PARS/PARP poly (ADP ribose) synthetase
  • TAFI tissue-activated fibrinolytic inhibitor
  • NCX-1 Na + /Ca +2 exchanger isoform-1
  • PARS/PARP poly (ADP ribose) synthetase
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier, vehicle, or diluent.
  • the combination of the NHE-1 inhibitor, and the second compound selected from the group consisting of a complement modulator, a metabolic modulator, an anti-apoptotic agent, a nitric oxide synthase-related agent, and an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor can be administered to a patient separately or together in any conventional oral, rectal, transdermal, parenteral, (for example, intravenous, intramuscular, or subcutaneous) intracisternal, intravaginal, intraperitoneal, intravesical, local (for example, powder, ointment or drop), or buccal, or nasal, dosage form .
  • a complement modulator for
  • compositions suitable for parenteral injection may comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions, or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous carriers, diluents, solvents, or vehicles include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. Prevention of microorganism contamination of the compositions can be accomplished with various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. Prolonged absorption of injectable pharmaceutical compositions can be brought about by the use of agents capable of delaying absorption, for example, aluminum monostearate and gelatin.
  • Solid dosage forms for oral administration include capsules, tablets, powders, and granules.
  • the active compound is admixed with at least one inert customary pharmaceutical excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, mannitol, and silicic acid;
  • binders as for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia;
  • humectants as for example, glycerol;
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate;
  • solution retarders as for example, paraffin;
  • absorption accelerators as for example, quaternary ammonium compounds;
  • compositions of a similar type may also be used as fillers in soft or hard filled gelatin capsules using such excipients as lactose or milk sugar, as well as high molecular weight polyethylene glycols, and the like.
  • Solid dosage forms such as tablets, dragees, capsules, and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may also contain opacifying agents, and can also be of such composition that they release the active compound or compounds in a delayed manner. Examples of embedding compositions that can be used are polymeric substances and waxes. The active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage form may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame seed oil, glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • inert diluents commonly used in the art, such as water or other solvents, solubilizing
  • the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the active compound, may further comprise suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, or mixtures of these substances, and the like.
  • suspending agents as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, or mixtures of these substances, and the like.
  • compositions for rectal administration preferably comprise suppositories, which can be prepared by mixing a compound of the present invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax, which are solid at ordinary room temperature, but liquid at body temperature, and therefore, melt in the rectal cavity thereby releasing the active component.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax, which are solid at ordinary room temperature, but liquid at body temperature, and therefore, melt in the rectal cavity thereby releasing the active component.
  • Dosage forms for topical administration of the NHE-1 inhibitor, and the second compound selected from the group consisting of a complement modulator, a metabolic modulator, an anti-apoptotic agent, a nitric oxide synthase-related agent, and an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor may comprise ointments, powders, sprays and inhalants.
  • the active agent or agents are admixed under sterile condition with a pharmaceutically acceptable carrier, and any preservatives, buffers, or propellants that may be required.
  • the combinations and pharmaceutical compositions comprising the combinations of the present invention are useful as therapeutants or prophylactic agents for diseases caused or aggravated by the acceleration of the sodium/hydrogen (Na+/H+) exchange transport system, for example, cardiovascular diseases [e.g., arteriosclerosis, hypertension, arrhythmia (e.g.
  • ischemic arrhythmia arrhythmia due to myocardial infarction, myocardial stunning, myocardial dysfunction, arrhythmia after PTCA or after thrombolysis, etc.
  • angina pectoris cardiac hypertrophy, myocardial infarction, heart failure (e.g., congestive heart failure, acute heart failure, cardiac hypertrophy, etc.), restenosis after PTCA, PTCI, shock (e.g., hemorrhagic shock, endotoxin shock, etc.)], renal diseases (e.g., diabetes mellitus, diabetic nephropathy, ischemic acute renal failure, etc.) organ disorders associated with ischemia or ischemic reperfusion [(e.g., heart muscle ischemic reperfusion associated disorders, acute renal failure, or disorders induced by surgical treatment such as coronary artery bypass grafting (CABG) surgeries, vascular surgeries, organ transplantation, non-cardiac surgeries or percutaneous transluminal coronary angioplasty (PTCA
  • compositions of this invention can also be used as an agent for myocardial protection during coronary artery bypass grafting (CABG) surgeries, vascular surgeries, percutaneous transluminal coronary angioplasty (PTCA), PTCI, organ transplantation, or non-cardiac surgeries.
  • CABG coronary artery bypass grafting
  • PTCA percutaneous transluminal coronary angioplasty
  • PTCI percutaneous transluminal coronary angioplasty
  • organ transplantation or non-cardiac surgeries.
  • the combinations and pharmaceutical compositions of this invention can be used to for myocardial protection before, during, or after coronary artery bypass grafting (CABG) surgeries, vascular surgeries, percutaneous transluminal coronary angioplasty (PTCA), organ transplantation, or non-cardiac surgeries.
  • CABG coronary artery bypass grafting
  • PTCA percutaneous transluminal coronary angioplasty
  • organ transplantation or non-cardiac surgeries.
  • the combinations and pharmaceutical compositions of this invention can be used for myocardial protection in patients presenting with ongoing cardiac (acute coronary syndromes, e.g., myocardial infarction or unstable angina) or cerebral ischemic events (e.g., stroke).
  • ongoing cardiac acute coronary syndromes, e.g., myocardial infarction or unstable angina
  • cerebral ischemic events e.g., stroke
  • the combinations and pharmaceutical compositions of this invention can be used for chronic myocardial protection in patients with diagnosed coronary heart disease (e.g., previous myocardial infarction or unstable angina) or patients who are at high risk for myocardial infarction (age greater than 65 and two or more risk factors for coronary heart disease).
  • diagnosed coronary heart disease e.g., previous myocardial infarction or unstable angina
  • patients who are at high risk for myocardial infarction e.g., age greater than 65 and two or more risk factors for coronary heart disease.
  • Such assays may also provide a means whereby the activities of the combinations and pharmaceutical compositions of this invention can be compared with the activities of other known compounds. The results of these comparisons are useful for determining dosage levels in mammals, including humans, for the treatment of such diseases.
  • kits comprises an amount of a sodium-hydrogen exchanger type-1 (NHE-1) inhibitor, and a pharmaceutically acceptable carrier, vehicle, or diluent in a first unit dosage form; an amount of a second compound selected from the group consisting of (a) a complement modulator, (b) a metabolic modulator, (c) an anti-apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP rib
  • the container is employed to contain the separate components and may comprise, for example, a divided bottle or a divided foil packet, however, the separate compositions may also be contained within a single, undivided container.
  • the kit will also include directions for the administration of the separate components.
  • the kit form is particularly advantageous when the separate components are administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage levels, or when titration of the individual components of the combination is desired by the prescribing physician.
  • Blister packs are well known in the packaging industry and are used widely for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally comprise a sheet of relatively rigid material covered with a foil of a preferably transparent plastic material. During the packaging process, recesses (e.g., blisters) are formed in the plastic foil. The recesses generally conform to the size and shape of the tablets or capsules to be contained therein. Next, the tablets or capsules are placed in the recesses and the sheet of relatively rigid material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed.
  • recesses e.g., blisters
  • the tablets or capsules are captively retained and sealed inside the recesses between the plastic foil and the sheet.
  • the strength of the sheet is such that the tablets or capsules may be removed from the blister pack by the application of manual pressure on the outer surface of the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed through the formed opening.
  • a memory aid on the pack for example, in the form of numbers or similar indicia proximate to the tablets or capsules whereby the indicia correspond to the days of the regimen which the dosage form so specified is to be ingested.
  • An additional example of such a memory aid is a calendar printed on the pack, for example, as follows: “First Week, Monday, Tuesday, . . . etc . . . Second Week, Monday, Tuesday, . . . ” etc.
  • a “daily dose” can be a single tablet or capsule, or multiple tablets or capsules, or tablets or capsules to be ingested on a given day.
  • a daily dose of the sodium-hydrogen exchanger type 1 (NHE-1) inhibitor can consist of a single tablet or capsule, while a daily dose of the second compound, selected from the group consisting of (a) a compliment modulator, (b) a metabolic modulator, (c) an anti-apoptotic agent, (d) a nitric oxide synthase-related agent, and (e) an enzyme/protein modulator selected from the group consisting of a protein kinase C activator, an endothelin converting enzyme inhibitor, a tissue-activated fibrinolytic inhibitor (TAFI), a Na + /Ca +2 exchanger isoform-1 (NCX-1) inhibitor, and a poly (ADP ribose) synthetase (PARS/PARP) inhibitor, can consist of multiple tablets or capsules, and vice
  • a pack designed to dispense the daily doses one at a time in the order of their intended use is provided.
  • the pack is equipped with a memory aid, so as to further facilitate compliance with the dosage regimen.
  • a memory aid comprises a mechanical counter that indicates the number of daily doses to be dispensed.
  • Another example of such a memory aid comprises a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date and time that the last daily dose has been taken and/or reminds the patient when the next dose is to be taken.
  • fibroblasts stably expressing human NHE-1 (Counillon, L. et al., Mol. Pharmacol., 44:1041-1045 (1993) are plated onto collagen coated 96 well plates (50,000/well) and grown to confluence in growth media (DMEM high glucose, 10% fetal bovine serum, 50 u/ml penicillin and streptomycin). Confluent plates are incubated for 30 min at 37° C.
  • BCECF pH sensitive fluorescent probe
  • BCECF loaded cells are incubated for 30 min at 37° C. in acid loading media (70 mM choline chloride, 50 mM NHCl 4 , 5 mM KCl, 1 mM MgCl 2 , 1.8 mM CaCl 2 , 5 mM glucose, 10 mM HEPES, pH 7.5), and then placed in a Fluorescent Imaging Plate Reader (Molecular Devices, CA). BCECF fluorescence is monitored using excitation and emission wavelengths of 485 nM and 525 nM, respectively.
  • acid loading media 70 mM choline chloride, 50 mM NHCl 4 , 5 mM KCl, 1 mM MgCl 2 , 1.8 mM CaCl 2 , 5 mM glucose, 10 mM HEPES, pH 7.5
  • BCECF fluorescence is monitored using excitation and emission wavelengths of 485 nM and 525 nM, respectively.
  • Intracellular acidification is initiated via rapid replacement of acid loading media with recovery media (120 mM NaCl, 5 mM KCl, 1 mM MgCl 2 , 1.8 mM CaCl 2 , 5 mM glucose, 10 mM HEPES, pH 7.5) ⁇ test compound, and NHE-mediated recovery of intracellular pH is monitored as the subsequent time-dependent increase BCECF fluorescence.
  • the potency of human NHE-1 inhibitors is calculated as the concentration that reduces recovery of intracellular pH by 50% (IC 50 ).
  • the in vitro test described below demonstrates that the combinations and pharmaceutical compositions of the invention can also pharmacologically induce cardioprotection, i.e., reduced myocardial infarct size, when administered to a rabbit isolated heart.
  • cardioprotection i.e., reduced myocardial infarct size
  • the effects of the combinations and pharmaceutical compositions are compared to ischemic preconditioning and the A1 adenosine agonist, PIA (N 6 -1-(phenyl-2R-isopropyl)adenosine), that has been shown to pharmacologically induce cardioprotection in the rabbit isolated heart (Tracey, et aL, Cardiovasc. Res., 33: 410-415, 1997).
  • PIA N 6 -1-(phenyl-2R-isopropyl)adenosine
  • the heart is removed from the chest and rapidly ( ⁇ 30 sec) mounted on a Langendorff apparatus.
  • the heart is retrogradely perfused in a non-recirculating manner with a modified Krebs solution (NaCl 118.5 mM, KCl 4.7 mM, Mg SO 4 1.2 mM, KH 2 PO 4 1.2 mM, NaHCO 3 24.8 mM, CaCl 2 2.5 mM, and glucose 10 mM), at a constant pressure of 80 mmHg and a temperature of 37° C.
  • Perfusate pH is maintained at 7.4-7.5 by bubbling with 95% O 2/5 % CO 2 .
  • Heart temperature is tightly controlled by using heated reservoirs for the physiological solution and water jacketing around both the perfusion tubing and the isolated heart.
  • Heart rate and left ventricular pressures are determined via a latex balloon which is inserted in the left ventricle and connected by stainless steel tubing to a pressure transducer.
  • the intraventricular balloon is inflated to provide a systolic pressure of 80-100 mmHg, and a diastolic pressure ⁇ 10 mm Hg.
  • Total coronary flow is also continuously monitored using an in-line flow probe and normalized for heart weight.
  • the heart is allowed to equilibrate for 30 min, over which time the heart must show stable left ventricular pressures within the parameters outlined above. If the heart rate falls below 180 bpm at any time prior to the 30 min. period of regional ischemia, the heart is paced at about 200 bpm for the remainder of the experiment. Ischemic preconditioning is induced by total cessation of cardiac perfusion (global ischemia) for 5 min, followed by reperfusion for 10 min. The regional ischemia is provided by tightening the snare around the coronary artery branch. Following the 30 min regional ischemia, the snare is released and the heart reperfused for an additional 120 min.
  • Pharmacological cardioprotection is induced by infusing the combination or pharmaceutical composition at predetermined concentrations, starting 30 min prior to the 30 min regional ischemia, and continuing until the end of the 120 min reperfusion period. Hearts which receive test combinations or pharmaceutical compositions do not undergo the period of ischemic preconditioning. A reference compound, PIA (25 nM) is perfused through hearts (which do not receive the test combination or pharmaceutical composition) for a 5 min period which ends 10 min before the 30 min regional ischemia.
  • PIA 25 nM
  • the coronary artery snare is tightened, and a 0.5% suspension of fluorescent zinc cadmium sulfate particles (1-10 ⁇ m) Duke Scientific Corp.(Palo Alto, Calif.) is perfused through the heart; this stains all of the myocardium, except that area-at-risk for infarct development (area-at-risk).
  • the heart is removed from the Langendorff apparatus, blotted dry, wrapped in aluminum foil and stored overnight at ⁇ 20° C. The next day, the heart is sliced into 2 mm transverse sections from the apex to the top of the ventricles.
  • the slices are stained with 1% triphenyl tetrazolium chloride (TTC) in phosphate-buffered saline for 20 min at 37° C. Since TTC reacts with living tissue (containing NAD-dependent dehydrogenases), this stain differentiates between living (red stained) tissue, and dead tissue (unstained infarcted tissue).
  • TTC triphenyl tetrazolium chloride
  • the infarcted area (no stain) and the area-at-risk (no fluorescent particles) are calculated for each slice of left ventricle using a precalibrated image analyzer.
  • the data is expressed as the ratio of infarct area vs. area-at-risk (%IA/AAR). All data are expressed as mean ⁇ SE and compared statistically using a Mann-Whitney non-parametric test with a Bonferroni correction for multiple comparisons. Significance is considered as p ⁇ 0.05.
  • Cardioprotection as indicated by a reduction in infarcted myocardium, can be induced pharmacologically using intravenously administered adenosine receptor agonists in intact, anesthetized rabbits studied as an in situ model of myocardial ischemic preconditioning (Liu et al., Circulation 84:350-356, 1991).
  • Protocol Once arterial pressure and heart rate have been stable for at least 30 min. the test is started. Ischemic preconditioning is induced by occluding the coronary artery for 5 min followed by a 10 min. reperfusion. Pharmacological preconditioning is induced by infusing test combination or pharmaceutical composition over, for example 5 min. and allowing 10 min. before further intervention or by infusing the adenosine agonist, PIA (0.25 mg/kg). Following ischemic preconditioning, pharmacological preconditioning or no conditioning (unconditioned, vehicle control) the artery is occluded for 30 minutes and then reperfused for two hours to induce myocardial infarction. The combination or pharmaceutical composition and PIA are dissolved in saline or other suitable vehicle and delivered at 1 to 5 mg/kg, respectively.
  • Staining (Liu et al., Circulation 84:350-356, 1991): At the end of the 2 hour reperfusion period, the hearts are quickly removed, hung on a Langendorff apparatus, and flushed for 1 minute with normal saline heated to body temperature (38° C.). The silk suture used as the snare is then tied tightly to reocclude the artery and a 0.5% suspension of fluorescent zinc cadmium sulphate particles (1-10 pm) Duke Scientific Corp. (Palo Alto, CA) is infused with the perfusate to stain all of the myocardium except the area at risk (nonfluorescent ventricle). The hearts are then quickly frozen and stored overnight at ⁇ 20° C.
  • the hearts are cut into 2 mm slices and stained with 1% triphenyl tetrazolium chloride (TTC). Since TTC reacts with living tissue, this stain differentiates between living (red stained) tissue, and dead tissue (unstained infarcted tissue).
  • TTC triphenyl tetrazolium chloride
  • the infarcted area (no stain) and the area at risk (no fluorescent particles) are calculated for each slice of left ventricle using a pre-calibrated image analyzer.
  • the data is expressed as the ratio of infarct area vs. area at risk (%IA/AAR). All data are expressed as Mean ⁇ SEM and compared statistically using single factor ANOVA or Mann Whitney non-parametric test. Significance is considered as p ⁇ 0.05.
  • the combinations and pharmaceutical compositions of this invention can be tested for their utility in reducing or preventing ischemic injury in non-cardiac tissues, for example, the brain, or the liver, utilizing procedures reported in the scientific literature.
  • the combinations and pharmaceutical compositions of this invention in such tests can be administered by the preferred route and vehicle of administration and at the preferred time of administration either prior to the ischemic episode, during the ischemic episode, following the ischemic episode (reperfusion period) or during any of the below-mentioned experimental stages.
  • Body temperature can be monitored by a rectal thermometer, and the animals can be maintained normothermic, if necessary, by external heating.
  • the animals next undergo subtemporal craniectomy to expose the main trunk of the left middle cerebral artery (MCA) under an operating microscope, and the exposed artery is occluded with microbipolar coagulation to generate large ischemic lesions in the cerebral cortex and basal ganglia.
  • MCA left middle cerebral artery
  • the rats are deeply anesthetized with 2% halothane and a thoracotomy is performed to infuse heparinized saline into the left ventricle.
  • the effluent is collected via an incision of the right atrium.
  • the saline washout is followed by approximately 200 ml of a 40% formaldehyde, glacial acetic acid and absolute methanol solution (FAM; 1:1:8, v/v/v), then the animals are decapitated and the head is stored in fixative for 24 hours.
  • the brain is then removed, dissected, embedded in paraffin wax, and sectioned (approximately 100 sections 0.2 mm per brain). The sections are then stained with hematoxylin-eosin or with a combination of cresyl violet and Luxol fast blue, and examined by light microscopy to identify and quantitate the ischemic damage using a precalibrated image analyzer.
  • the ischemic volumes and areas are expressed in absolute units (mm 3 and mm 2 ) and as a percentage of the total region examined.
  • the effect of the combinations and pharmaceutical compositions of this invention to reduce ischemic brain damage induced by MCA occlusion is noted based on a reduction in the area or volume of relative or absolute ischemic damage in the brain sections from the rats in the treatment group compared to brain sections from rats in a placebo-treated control group.
  • the liver is extirpated and placed in an environmental chamber maintained at constant temperature (37° C.), then perfused through the portal vein at a constant pressure of 15 cm H 2 O with a modified, hemoglobin-free Krebs-Henseleit buffer (in mM: 118 NaCl, 4.7 KCl, 27 NaHCO 3 , 2.5 CaCl 2 , 1.2 MgSO 4 , 1.2 KH 2 PO 4 , 0.05 EDTA, and 11 mM glucose, plus 300 U heparin).
  • the pH of the perfusate is maintained at 7.4 by gassing the buffer with 95% O 2 -5% CO 2 .
  • Each liver is perfused at a flow rate of 20 ml/min in a single-pass manner for a 30 min washout and equilibration period (preischemic period), followed by a 2 hour period of global ischemia, and then a 2 hour period of reperfusion under conditions identical to the preischemic period. Aliquots (20 ml) of the perfusate are collected during the preischemic period, immediately after the occlusive ischemic period, and every 30 min of the 2 hour reperfusion period.
  • the perfusate samples are assayed for the appearance of hepatocellular enzymes, for example, aspartate amino-transferase (AST), alanine amino-transferase (ALT), and lactate dehydrogenase (LDH), which are taken to quantitatively reflect the degree of ischemic liver tissue damage during the procedure.
  • AST, ALT, and LDH activities in the perfusate can be determined by several methods, for example, by the reflectometry method using an automatic Kodak Ektachem 500 analyzer reported by Nakano, et al. (Hepatology 1995;22:539-545).
  • the effect of the combinations and pharmaceutical compositions of this invention in reducing ischemic liver damage induced by occlusion is noted based on a reduction in the release of hepatocellular enzymes immediately following the occlusive period and/or during the postischemic-reperfusion period in the perfused livers from the rats in the treatment group compared to perfused livers from rats in a placebo-treated control group.
US10/052,320 2001-01-25 2002-01-17 Combination therapy Expired - Fee Related US6423705B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/052,320 US6423705B1 (en) 2001-01-25 2002-01-17 Combination therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US26417301P 2001-01-25 2001-01-25
US10/052,320 US6423705B1 (en) 2001-01-25 2002-01-17 Combination therapy

Publications (2)

Publication Number Publication Date
US6423705B1 US6423705B1 (en) 2002-07-23
US20020099075A1 true US20020099075A1 (en) 2002-07-25

Family

ID=26730463

Family Applications (1)

Application Number Title Priority Date Filing Date
US10/052,320 Expired - Fee Related US6423705B1 (en) 2001-01-25 2002-01-17 Combination therapy

Country Status (1)

Country Link
US (1) US6423705B1 (US20020099075A1-20020725-C00001.png)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004064854A1 (en) * 2003-01-17 2004-08-05 Korea Institute Of Science And Technology Method for enhancing learning and memory by suppressing the activity of ncx2 protein
US20040198821A1 (en) * 2002-01-29 2004-10-07 Hwang Cheng Shine Reduction of hair growth
WO2005079803A1 (en) * 2004-02-13 2005-09-01 Pfizer Products, Inc. Compounds for treatment of cardiovascular diseases
US20060120971A1 (en) * 2004-10-08 2006-06-08 Signum Pharmaceuticals, Inc. Agents and methods for administration to the central nervous system
US20070026079A1 (en) * 2005-02-14 2007-02-01 Louis Herlands Intranasal administration of modulators of hypothalamic ATP-sensitive potassium channels
US20070093434A1 (en) * 2003-02-13 2007-04-26 Luciano Rossetti Regulation of food intake and glucose production by modulation of long-chain fatty acyl-coa levels in the hypothalamus
US20090137665A1 (en) * 2004-03-24 2009-05-28 Mccullough Louise D Novel method of neuroprotection by pharmacological inhibition of amp-activated protein kinase
WO2017007917A1 (en) * 2015-07-07 2017-01-12 Mast Therapeutics, Inc. Polyoxyethylene/polyoxypropylene copolymers and fibrinolytic inhibitors, uses thereof and compositions
CN107661491A (zh) * 2010-09-15 2018-02-06 赛尔戴克斯治疗公司 用可溶性I型补体受体(sCR1)治疗慢性肾病

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE60229416D1 (de) 2001-10-25 2008-11-27 Univ Emory Katheter für modifizierte Perfusion
US20070160645A1 (en) * 2001-10-25 2007-07-12 Jakob Vinten-Johansen PostConditioning System And Method For The Reduction Of Ischemic-Reperfusion Injury In The Heart And Other Organs
WO2003061700A1 (fr) * 2002-01-25 2003-07-31 Taisho Pharmaceutical Co.,Ltd. Medicaments servant au traitement de nephropathies chroniques
WO2003068263A1 (fr) * 2002-02-15 2003-08-21 Taisho Pharmaceutical Co.,Ltd. Remede contre l'hypertension
IL165304A0 (en) * 2002-05-21 2006-01-15 Cv Therapeutics Inc Method of treating diabetes
US8822473B2 (en) * 2002-05-21 2014-09-02 Gilead Sciences, Inc. Method of treating diabetes
US6693133B1 (en) * 2002-10-07 2004-02-17 University Of Alberta Methods of cardioprotection using dichloroacetate in combination with an inotrope
US20040202726A1 (en) * 2003-04-10 2004-10-14 Deshay Samuel L. Topical blood pressure composition
US20050209141A1 (en) * 2003-10-17 2005-09-22 Silver Randi B Mast cell-derived renin
EP1793830A2 (en) * 2004-09-22 2007-06-13 Pfizer, Inc. Therapeutic combinations comprising poly(adp-ribose) polymerases inhibitor
CA2592142A1 (en) * 2004-12-22 2006-06-29 Emory University Therapeutic adjuncts to enhance the organ protective effects of postconditioning
US7922669B2 (en) 2005-06-08 2011-04-12 Cardiac Pacemakers, Inc. Ischemia detection using a heart sound sensor
US7774057B2 (en) 2005-09-06 2010-08-10 Cardiac Pacemakers, Inc. Method and apparatus for device controlled gene expression for cardiac protection
US7885710B2 (en) 2005-12-23 2011-02-08 Cardiac Pacemakers, Inc. Method and apparatus for tissue protection against ischemia using remote conditioning
WO2007116243A2 (en) * 2006-04-10 2007-10-18 Mintails Limited Method for treating fibromyalgia and related conditions
US20080193530A1 (en) * 2007-02-13 2008-08-14 Brent Blackburn Use of ranolazine for the treatment of non-coronary microvascular diseases
EP2117508A1 (en) * 2007-02-13 2009-11-18 CV Therapeutics Inc. Intravenous solutions comprising ranolazine
US20080248112A1 (en) * 2007-02-13 2008-10-09 Brent Blackburn Use of ranolazine for the treatment of coronary microvascular diseases
US20090111826A1 (en) * 2007-02-13 2009-04-30 Louis Lange Use of ranolazine for the treatment of cardiovascular diseases
US20080233191A1 (en) * 2007-03-22 2008-09-25 Brent Blackburn Use of ranolazine for elevated brain-type natriuretic peptide
ES2402675T3 (es) * 2007-05-31 2013-05-07 Gilead Palo Alto, Inc. Ranolazina para el péptido natriurético de tipo cerebral a niveles elevados
CN101336914B (zh) * 2007-07-03 2011-12-28 常州高新技术产业开发区三维工业技术研究所有限公司 一种缩小心肌梗死面积的药物组合物及其应用
US10517839B2 (en) * 2008-06-09 2019-12-31 Cornell University Mast cell inhibition in diseases of the retina and vitreous
US20110224606A1 (en) * 2010-03-10 2011-09-15 Shibaji Shome Method and apparatus for remote ischemic conditioning during revascularization
SG11201403538TA (en) 2011-12-31 2014-09-26 Beigene Ltd Fused tetra or penta-cyclic pyridophthalazinones as parp inhibitors
NO2797921T3 (US20020099075A1-20020725-C00001.png) 2011-12-31 2018-02-03
US9480666B2 (en) 2014-08-16 2016-11-01 Tobias Deuse Compositions and methods for inhibiting intimal hyperplasia
US11612615B2 (en) * 2017-04-27 2023-03-28 The University Of South Alabama Combined administration of cysteine-aspartic protease inhibitors with P2Y12 receptor antagonists protects the heart against myocardial infarction

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2988487A (en) * 1957-08-29 1961-06-13 Rutgers Res And Educational Fo Process of treating keratinaceous material and a keratinase produced thereby
US3751222A (en) * 1971-12-13 1973-08-07 Colgate Palmolive Co A process of cleaning cloth
US5171682A (en) * 1988-03-31 1992-12-15 North Carolina State University Purified Bacillus licheniformis PWD-1 keratinase

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19622222A1 (de) 1996-06-03 1997-12-04 Hoechst Ag Verwendung von Inhibitoren des zellulären Na·+·/H·+·-Exchangers (NHE) zur Herstellung eines Medikament zur Normalisierung der Serumlipide
EP0855392A3 (de) 1997-01-22 2000-01-05 Hoechst Aktiengesellschaft Fünfgliedrige Heterocyclen mit Biphenylsulfonylsubstitution, Verfahren zu ihrer Herstellung, ihre Verwendung als Medikament oder Diagnostikum sowie sie enthaltendes Medikament
DE19737224A1 (de) 1997-08-27 1999-03-18 Hoechst Marion Roussel De Gmbh Pharmazeutisches Kombinationspräparat aus einem Inhibitor des Natrium-Wasserstoff-Austauschers und einem Arzneimittel zur Behandlung von Herz-Kreislauferkrankungen
BR9908332A (pt) 1998-02-27 2000-11-07 Pfizer Prod Inc Derivados de n-[carbonil(anel di ou triaza diinsaturado de cinco membros substituìdo)] derivados de guanidina para tratamento de esquemia

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2988487A (en) * 1957-08-29 1961-06-13 Rutgers Res And Educational Fo Process of treating keratinaceous material and a keratinase produced thereby
US3751222A (en) * 1971-12-13 1973-08-07 Colgate Palmolive Co A process of cleaning cloth
US5171682A (en) * 1988-03-31 1992-12-15 North Carolina State University Purified Bacillus licheniformis PWD-1 keratinase

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040198821A1 (en) * 2002-01-29 2004-10-07 Hwang Cheng Shine Reduction of hair growth
WO2004064854A1 (en) * 2003-01-17 2004-08-05 Korea Institute Of Science And Technology Method for enhancing learning and memory by suppressing the activity of ncx2 protein
US20070093434A1 (en) * 2003-02-13 2007-04-26 Luciano Rossetti Regulation of food intake and glucose production by modulation of long-chain fatty acyl-coa levels in the hypothalamus
US7705016B2 (en) 2003-02-13 2010-04-27 Albert Einstein College Of Medicine Of Yeshiva University Regulation of food intake by modulation of long-chain fatty acyl-CoA levels in the hypothalamus
WO2005079803A1 (en) * 2004-02-13 2005-09-01 Pfizer Products, Inc. Compounds for treatment of cardiovascular diseases
US20090137665A1 (en) * 2004-03-24 2009-05-28 Mccullough Louise D Novel method of neuroprotection by pharmacological inhibition of amp-activated protein kinase
US8293791B2 (en) 2004-03-24 2012-10-23 Fasgen, Llc Method of neuroprotection by pharmacological inhibition of AMP-activated protein kinase
US20060120971A1 (en) * 2004-10-08 2006-06-08 Signum Pharmaceuticals, Inc. Agents and methods for administration to the central nervous system
US20070026079A1 (en) * 2005-02-14 2007-02-01 Louis Herlands Intranasal administration of modulators of hypothalamic ATP-sensitive potassium channels
CN107661491A (zh) * 2010-09-15 2018-02-06 赛尔戴克斯治疗公司 用可溶性I型补体受体(sCR1)治疗慢性肾病
WO2017007917A1 (en) * 2015-07-07 2017-01-12 Mast Therapeutics, Inc. Polyoxyethylene/polyoxypropylene copolymers and fibrinolytic inhibitors, uses thereof and compositions

Also Published As

Publication number Publication date
US6423705B1 (en) 2002-07-23

Similar Documents

Publication Publication Date Title
US6423705B1 (en) Combination therapy
US6294538B1 (en) Compounds for treating and preventing diabetic complications
AU733304B2 (en) Combination of an aldose reductase inhibitor and a glycogen phosphorylase inhibitor
CA2964760C (en) Composition for prevention or treatment of ischemic cardiac disease,comprising inhibitor against age-albumin synthesis or release of mononuclear phagocyte system cells as active ingredient
US20090306160A1 (en) Methods for improving drug disposition
Hock et al. Peptide leukotriene receptor antagonism in myocardial ischaemia and reperfusion
EP1226830A2 (en) Combination therapy involving a sodium-hydrogen exchanger type 1 or NHE-1 inhibitor for use in treatment of ischemia
KR20120078756A (ko) Amp-활성화 단백질 키나아제의 약리학적 저해에 의한 신규한 신경보호 방법
Munch-Ellingsen et al. 5-HD abolishes ischemic preconditioning independently of monophasic action potential duration in the heart
EA027810B1 (ru) Гетероциклические карбоксамиды для лечения вирусных заболеваний
AU2001288798B2 (en) Use of 20-HETE synthesizing enzyme inhibitors as therapy for cerebral vascular diseases
US5932581A (en) Method of reducing tissue damage associated with ischemia
US5700803A (en) Method for reducing infarct size in subjects afflicted with ischemic heart disease
JP2001500493A (ja) 心臓線維芽細胞の増殖および心線維症を抑制する方法
WO2022141328A1 (zh) 硫代咪唑烷酮药物在治疗covid-19疾病中的用途
KR20070006497A (ko) 로지글리타존을 함유하는 패혈성 신장 장해 치료용 약제 조성물
US6852733B2 (en) Sodium-hydrogen exchanger type 1 inhibitor
MXPA00003243A (en) Compounds for treating and preventing diabetic complications
MXPA97001527A (en) Procedure to reduce tissue damage associated with isque

Legal Events

Date Code Title Description
REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20060723