WO2014040077A1 - Methods of treating muscular dystrophy - Google Patents

Methods of treating muscular dystrophy Download PDF

Info

Publication number
WO2014040077A1
WO2014040077A1 PCT/US2013/059074 US2013059074W WO2014040077A1 WO 2014040077 A1 WO2014040077 A1 WO 2014040077A1 US 2013059074 W US2013059074 W US 2013059074W WO 2014040077 A1 WO2014040077 A1 WO 2014040077A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
integrin
compound
subject
modulatory agent
Prior art date
Application number
PCT/US2013/059074
Other languages
French (fr)
Inventor
Dean Burkin
Ryan Wuebbles
Original Assignee
Board Of Regents Of The Nevada System Of Higher Education, On Behalf Of The Universisity Of Nevada, Reno
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 Board Of Regents Of The Nevada System Of Higher Education, On Behalf Of The Universisity Of Nevada, Reno filed Critical Board Of Regents Of The Nevada System Of Higher Education, On Behalf Of The Universisity Of Nevada, Reno
Priority to EP13835809.8A priority Critical patent/EP2892525B1/en
Priority to DK13835809.8T priority patent/DK2892525T3/en
Priority to CA2919516A priority patent/CA2919516C/en
Priority to US14/426,928 priority patent/US10028992B2/en
Priority to PL13835809T priority patent/PL2892525T3/en
Priority to JP2015531326A priority patent/JP2015527409A/en
Priority to EP18173735.4A priority patent/EP3406246A3/en
Priority to ES13835809T priority patent/ES2688023T3/en
Publication of WO2014040077A1 publication Critical patent/WO2014040077A1/en
Priority to US16/019,194 priority patent/US10398749B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • A61K31/36Compounds containing methylenedioxyphenyl groups, e.g. sesamin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • A61K31/37Coumarins, e.g. psoralen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4025Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil not condensed and containing further heterocyclic rings, e.g. cromakalim
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/4035Isoindoles, e.g. phthalimide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/4211,3-Oxazoles, e.g. pemoline, trimethadione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system having sulfur as a ring hetero atom, e.g. ticlopidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4402Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 2, e.g. pheniramine, bisacodyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • 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
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53831,4-Oxazines, e.g. morpholine ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • 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
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles

Definitions

  • This disclosure relates to the field of muscular dystrophy and in particular, to compositions and methods for treating muscular dystrophy, such as Duchenne muscular dystrophy, Fukuyama congenital muscular dystrophy or merosin deficient congenital muscular dystrophy type 1 A or ID.
  • ⁇ 7 ⁇ 1 integrin is also a major modifier of muscle disease progression in various genetic muscle diseases including various types of muscular dystrophy, such as Duchenne muscular dystrophy (DMD), Fukuyama congenital muscular dystrophy (FCMD) and merosin deficient congenital muscular dystrophy type 1A (MDCIA).
  • DMD Duchenne muscular dystrophy
  • FCMD Fukuyama congenital muscular dystrophy
  • MDCIA merosin deficient congenital muscular dystrophy type 1A
  • transcriptional regulation of the l integrin gene including such role in muscular dystrophy (e.g. , DMD, FCMD and/or MDCIA), remains poorly understood.
  • DMD Duchenne muscular dystrophy
  • X-chromosome-linked disease X-chromosome-linked disease and the most common form of muscular dystrophy. DMD affects 1 in 3500 live male births with patients suffering from chronic muscle degeneration and weakness. Clinical symptoms are first detected between the ages of 2 and 5 years and, by the time the patient is in their teens, the ability for independent ambulation is lost. Death typically occurs in the patient before they are 30 years old due to cardiopulmonary failure.
  • FCMD Fukuyama congenital muscular dystrophy
  • MDCIA is congential muscular dystrophies that are heritable neuromuscular disorders. MDCIA is characterized by muscle weakness at birth or in infancy. Affected infants will present with poor muscle tone and few movements. The quality of life and life span of the child is affected through progressive muscle wasting, respiratory compromise, and spinal rigidity. MDCIA is the most common and severe form of congenital muscular dystrophy, accounting for 30-40% of all congenital muscular dystrophy (CMD) diagnosed cases. MDCIA is characterized by congenital hypotonia, distinct joint contractures, and a lack of independent ambulation. Feeding tube placement and positive pressure ventilation is often required for the respiratory problems that occur.
  • CMD congenital muscular dystrophy
  • FCMD is caused by mutations in the fukutin gene, located at human chromosome 9q31. The disease is inherited in an autosomal recessive manner. FCMD is a type of Limb-Girdle muscular dystrophy. Currently there is no cure for DMD, FCMD or MDC1A.
  • the muscular dystrophies are a group of diverse, heritable neuromuscular disorders which represent a group of devastating neuromuscular diseases characterized by primary or secondary skeletal muscle involvement. Currently, there are no cures for such diseases.
  • ⁇ 7 ⁇ 1 integrin expression modulatory agents and methods of using such to treat a condition associated with impaired a7 integrin expression, such as muscular dystrophy.
  • a method for treating a subject with muscular dystrophy is disclosed.
  • the method includes administering an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent to the subject with muscular dystrophy, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent is ciclopirox ethanolamine, deferoxamine, 2,2- dipyridyl; 5a-cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, a compound provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I in U.S. Provisional Patent Application No.
  • ⁇ 7 ⁇ 1 integrin modulatory agent increases ⁇ 7 ⁇ 1 integrin expression or activity as compared to ⁇ 7 ⁇ 1 integrin expression or activity prior to treatment, thereby treating the subject with muscular dystrophy (such as MDC1A, MDC1D, LGMD, DMD, FCMD or FHMD).
  • the method includes administering an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent to the subject in need of muscle regeneration, repair or maintenance, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan- 3 ⁇ - ⁇ 1-6- ⁇ , Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, a compound provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S.
  • the present disclosure provides a method for increasing muscle regeneration in a subject.
  • geriatric subjects, subjects suffering from muscle disorders, and subjects suffering from muscle injury, including activity induced muscle injury, such as injury caused by exercise, may benefit from this embodiment.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered in a preventative manner, such as to prevent or reduce muscular damage or injury (such as activity or exercise induced injury).
  • muscular damage or injury such as activity or exercise induced injury.
  • the method includes contacting a cell with an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent includes ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a- cholestan-3 -ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, a compound provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S.
  • the methods of the present disclosure can include administering the ⁇ 7 ⁇ 1 integrin modulatory agent with one or more additional pharmacological substances, such as a therapeutic agent.
  • the additional therapeutic agent enhances the therapeutic effect of the ⁇ 7 ⁇ 1 integrin modulatory agent.
  • the therapeutic agent provides independent therapeutic benefit for the condition being treated.
  • the additional therapeutic agent is a component of the extracellular matrix, such as an integrin, dystrophin, dystroglycan, utrophin, or a growth factor.
  • the therapeutic agent reduces or enhances expression of a substance that enhances the formation or maintenance of the extracellular matrix.
  • the therapeutic agent is an additional ⁇ 7 ⁇ 1 integrin modulatory agent such as laminin-111, a laminin-111 fragment, valproic acid or a valproic acid analog.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is applied to a particular area of the subject to be treated.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent may be injected into a particular area to be treated, such as a muscle.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered such that it is distributed to multiple areas of the subject, such as systemic administration or regional administration.
  • a ⁇ 7 ⁇ 1 integrin modulatory agent can be administered by any suitable method, such as topically, parenterally (such as intravenously or intraperitoneally), or orally.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered systemically, such as through parenteral administration, such as stomach injection or peritoneal injection.
  • the disclosed methods generally have been described with respect to muscle regeneration, the disclosed methods also may be used to enhance repair or maintenance, or prevent damage to, other tissues and organs.
  • the methods of the present disclosure can be used to treat symptoms of muscular dystrophy stemming from effects to cells or tissue other than skeletal muscle, such as impaired or altered brain function, smooth muscles, or cardiac muscles.
  • FIG. 1A is two digital images of X-gal staining demonstrating that a7Pgal + " myoblasts express ⁇ - galactosidase which increases upon differentiation to myotubes.
  • FIG. IB is a digital image of a Western analysis of a7Pgal + " myoblasts differentiated from 0-72 hours shows a corresponding increase in both a7 integrin and ⁇ -galactosidase.
  • a-Tubulin was used as a loading control.
  • FIGS. 2A-2D demonstrate Laminin-111 increases a7 integrin levels in mouse and human muscle cells.
  • FIGS. 3A-3C demonstrate that intramuscular injection of laminin-111 prevents muscle disease in mdx mice.
  • (3A) Immunofluorescence of the TA muscles of control and laminin-111 treated mice confirm the absence dystrophin in mdx muscle treated with LAM-111 or PBS. Laminin-111 was not present in wild- type or PBS injected mdx muscle but was detected in the extracellular matrix of laminin-111-injected mdx muscle. Scale bar ⁇ .
  • (3B) Evans blue dye (EBD) uptake reveals mdx muscle injected with laminin- 111 exhibits reduced EBD uptake compared to control. Scale bar ⁇ .
  • H&E staining reveals that mdx muscle treated with laminin-111 contains few muscle fibers with centrally located nuclei and mononuclear cell infiltrate compared to control.
  • FIGS. 4A and 4B demonstrate Valproic Acid increases a7 integrin expression in muscle cells.
  • FIG. 5 shows the chemical structures for Ciclopirox, Deferoxamine and 2,2-dipyridyl.
  • FIGS. 6A and 6B show dose-response curves for Ciclopirox, Deferoxamine and 2,2-dipyridyl using a7Pgal + " myotubes and the FDG assay.
  • FIG. 7 shows the iron chelators 2,2-dipyridyl and Deferoxamine increase a7 protein in DMD myotubes.
  • DMD myotubes treated with either 2,2-dipyridyl or deferoxamine showed increased a7 integrin protein as determined by western analysis.
  • N 3 replicates, **P ⁇ 0.01; *P ⁇ 0.05.
  • FIG. 8 shows Cholestan increases a7 integrin promoter activity in mouse and DMD muscle cells.
  • a typical dose response was obtained using Cholestan in a7Pgal + " myotubes.
  • Treatment of DMD myotubes with cholestan resulted in increased a7 integrin protein compared to control.
  • FIG. 9 shows Compounds 1001, 1002 and 1003 activate a7 integrin promoter activity.
  • Typical dose response curves showing the fold increase in reporter activity vs drug dose were obtained for compounds 1001, 1002 and 1003 using a7Pgal + " myotubes.
  • FIG. 10 is an exemplary synthesis pathway for analogs of compounds 1002 and 1003.
  • FIG. 11 is a schematic illustrating two examples of muscular dystrophy in which enhanced l integrin is therapeutic.
  • Loss of dystrophin in DMD (A) or glycosylation of a dystroglycan in MDCID (B) results in defective membrane integrity and sarcolemma disruptures.
  • Enhancement of ⁇ 7 ⁇ 1 integrin improves membrane integrity, minimize sarcolemma ruptures and mitigate the progression of disease in DMD (B) and MDCID (D).
  • FIGS. 12A-12D are scatter plots from fluorescence-activated cell sorting (FACS) analyses demonstrating that a 24 hour treatment of a7pi-gal +/- myoblasts with lOOnM LAM-111 and a fluorescent ⁇ -gal substrate resulted increase a l integrin expression compared to PBS treatment.
  • FIGS. 14A-14U are digital images of sections of TA muscle from wild-type (A through G), PBS- treated mdx mice (H through N), and LAM-111 -treated mdx mice (O through U). Detection of dystrophin (A, H, O), LAM-111 (B, I, P), hematoxylin and eosin (C, J, Q), Evans blue dye uptake (D, K, R), al integrin (E, L, S), utrophin (F, M, T), and a-bungarotoxin (G, N, U).
  • FIGS. 14V and 14W show percentage of Evans blue dye (EBD) positive myofibers (Panel V) and percentage of centrally-located nuclei (CLN) (Panel W) in the TA muscle of wild-type (black bar), PBS- treated mdx (white bar), and LAM-111 treated (gray bar) mdx mice.
  • FIGS. 15A-15D shows the results of immunoblot detection and quantitation of skeletal muscle protein.
  • FIGS. 16A-16K are digital images showing intraperitoneal delivery of LAM-111 distributes throughout mdx skeletal and cardiac muscles. Immunofluorescence detection of LAM-111 in the heart (A, B, C), diaphragm (D, E, F, J, K), and gastrocnemius (G, H, ⁇ ) of wild-type (A, D, G), PBS-treated mdx (B, E, H, J), and LAM-111 treated mdx mice (C, F, I, K).
  • FIGS. 16A-16C 100X
  • FIGS. 16D-16I 63X.
  • FIGS. 17A-17C includes three bar graphs illustrating the blood chemistry following intraperitoneal delivery of LAM-111.
  • Serum creatine kinase (CK) activity (FIG. 17A), creatine (FIG. 17B) and Blood Urea Nitrogen (BUN) (FIG. 17C) in wild-type (black bars), PBS-treated mdx mice (white bars), and LAM-111- treated mdx mice (gray bars).
  • FIGS. 18A-18C shows- that pretreatment with LAM-111 protects mdx TA from eccentric exercise- induced damage.
  • n 4 mice per group.
  • FIG. 19 shows that LAM-111 ameliorates muscle disease in the dyW mouse model for MDC1A.
  • Animals were injected i.p. twice weekly with 1 mg/kg of LAM-111 beginning at 10 days of age. Tissues were harvested at 7 weeks of age.
  • Scale bar 20uM
  • FIGS. 20A-20C show that systemic LAM-111 decreases muscle pathology of dyW skeletal muscle
  • Multiple systemic doses of LAM-111 to dyW mice result in a decreased percentage of centrally nucleated myofibers (FIG. 20A), decreased percentage of Evans Blue dye positive myofibers (FIG. 20B), and decreased percentage of TUNEL positive myofibers (FIG. 20C).
  • PBS-treated dyW black
  • LAM-111- treated dyW gray
  • WT white mice.
  • FIGS. 21-28 are dose response graphs illustrating the effect of disclosed analogs on a7 integrin promoter activity. Typical dose response curves showing the fold increase in reporter activity vs drug dose were obtained for specific analogs using a7Pgal + " myotubes.
  • FIGS. 29A-29H illustrate the synthesis of compounds provided in Table 11.
  • FIG. 30 is a digital image illustrating the results of quantitative real-time PCR used to assess Itga7, Itgbl, and Lama2 transcript levels in C2C12 myoblasts and myotubes treated for 24 hours with DMSO control, 10 ⁇ MLS000683232-01 (IED-232), 10 ⁇ MLS001165937-01 (IED-937), Hydroxylpropyl- Beta-Cyclodextrin (HPBCD) control, or 12 ⁇ SU9516 in HPBCD.
  • * denotes a significant difference in relative transcript levels with ** p-value ⁇ 0.01 and *** p ⁇ 0.001.
  • FIG. 31 is a digital image of Western Blots and quantitative analysis of a7 Integrin and GAPDH protein levels in C2C12 myotubes treated for 48 hours with DMSO control, 10 ⁇ MLS000683232-01 (IED-232), Hydroxylpropyl-Beta-Cyclodextrin (HPBCD) control, or 12 ⁇ SU9516 in HPBCD. Bands were quantified using Image J software and then graphed as al Integrin protein levels relative to GAPDH protein levels. * denotes a significant difference in relative protein levels with ** p ⁇ 0.01.
  • FIG. 32 is an image of results (fluorescence relative to DMSO at various concentrations of the agent) from a screen using particular embodiments of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents.
  • ⁇ 7 ⁇ 1 integrin expression modulatory agents and methods of using such to treat a condition associated with impaired al integrin expression, such as muscular dystrophy.
  • the present disclosure is related to the subject matter disclosed in U.S. Provisional Patent Application No. 61/533,059, which is incorporated herein by reference in its entirety.
  • the compounds of the present disclosure may be as identified in and have the features described in Appendix I (Table 6), and Appendix II (Table 7) of U.S. Provisional Patent Application No. 61/798,479. The described features are considered to contribute to solving the technical problem underlying the invention of the present application.
  • the described features include, for example, ⁇ 7 ⁇ 1 integrin modulatory activity suitable for treating a subject with muscular dystrophy, enhancing ⁇ 7 ⁇ 1 integrin expression, preventing or reducing muscle injury or damage in a subject, enhancing muscle regeneration, repair, or maintenance in a subject, and combinations thereof.
  • a method for treating a subject with muscular dystrophy includes administering an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent to the subject with muscular dystrophy, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent is ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan ⁇ -ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No.
  • ⁇ 7 ⁇ 1 integrin modulatory agent increases ⁇ 7 ⁇ 1 integrin expression or activity as compared to ⁇ 7 ⁇ 1 integrin expression or activity prior to treatment, thereby treating the subject with muscular dystrophy.
  • a method for treating a subject with muscular dystrophy comprises administering an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent to the subject with muscular dystrophy, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, an agent having a formula selected from
  • each R 1 and R 2 independently is selected from Ci-ioalkyl, substituted Ci-ioalkyl, Ci-ioalkoxy, substituted Ci-ioalkoxy, acyl, acylamino, acyloxy, acylCi-ioalkyloxy, amino, substituted amino, aminoacyl, aminocarbonylCi-ioalkyl, aminocarbonylamino, aminodicarbonylamino, aminocarbonyloxy, aminosulfonyl, C6-i 5 aryl, substituted C6-isaryl, C6-i 5 aryloxy, substituted C6-i 5 aryloxy, C6-i 5 arylthio, substituted C6-isarylthio, carboxyl, carboxyester, (carboxyester)amino, (carboxyester)oxy, cyano, C3- 8 cycloalkyl, substituted C3- 8 cycloalkyl, (C3- 8 cycloalky
  • R 1 substituents together with the atom to which each is bound, may form ring selected from a C6-i 5 aryl, substituted C6-isaryl, C3- 8 cycloalkyl, substituted C3- 8 cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C 2 -iosubstituted heterocyclyl, and C 2 -ioheterocyclyloxy, substituted;
  • R 2 substituents together with the atom to which each is bound, may form ring selected from a C6-i 5 aryl, substituted C6-isaryl, C3- 8 cycloalkyl, substituted C3- 8 cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C 2 -iosubstituted heterocyclyl, and C 2 -ioheterocyclyloxy, substituted;
  • each of A, B, C, D, E, and F independently may be selected from carbon, nitrogen, oxygen, and sulfur;
  • n may be zero, 1, 2, 3, 4, or 5;
  • ⁇ 7 ⁇ 1 integrin modulatory agent increases ⁇ 7 ⁇ 1 integrin expression or activity as compared to ⁇ 7 ⁇ 1 integrin expression or activity prior to treatment, thereby treating the subject with muscular dystrophy.
  • Particular disclosed embodiments concerning one or more of the compounds provided in Tables 10, 11, 6 and/or 7 see Appendix I of U.S. Provisional Pat. App. No. 61/796,476 for Table 6 and Appendix II of U.S. Provisional Pat. App. No. 61/796,476 for Table 7).
  • the muscular dystrophy is merosin deficient congenital muscular dystrophy Type 1A (MDC1A), merosin deficient congenital muscular dystrophy Type ID (MDC1D), limb -girdle muscular dystrophy (LGMD), Duchenne muscular dystrophy (DMD), Fukuyama congenital muscular dystrophy (FCMD) or Facioscapulohumeral muscular dystrophy (FHMD).
  • MDC1A merosin deficient congenital muscular dystrophy Type 1A
  • MDC1D merosin deficient congenital muscular dystrophy Type ID
  • LGMD limb -girdle muscular dystrophy
  • DMD Duchenne muscular dystrophy
  • FCMD Fukuyama congenital muscular dystrophy
  • FHMD Facioscapulohumeral muscular dystrophy
  • the muscular dystrophy is DMD, MDC1A or FCMD.
  • the muscular dystrophy is DMD.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered with an additional therapeutic agent.
  • the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional ⁇ 7 ⁇ 1 integrin modulatory agent.
  • the additional ⁇ 7 ⁇ 1 integrin modulatory agent is laminin-111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
  • the method further includes selecting a subject with muscular dystrophy.
  • the selecting a subject with muscular dystrophy includes diagnosing the subject with muscular dystrophy prior to administering an effective amount of the ⁇ 7 ⁇ 1 integrin modulatory agent to the subject.
  • a method of enhancing muscle regeneration, repair, or maintenance in a subject is disclosed.
  • the method includes administering an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent to the subject with muscular dystrophy, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan ⁇ -ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064- 0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No.
  • ⁇ 7 ⁇ 1 integrin modulatory agent increases ⁇ 7 ⁇ 1 integrin expression or activity as compared to ⁇ 7 ⁇ 1 integrin expression or activity prior to treatment, thereby enhancing muscle regeneration, repair or maintenance in a subject.
  • the method includes administering the ⁇ 7 ⁇ 1 modulatory agent prior to the subject experiencing muscle damage or disease.
  • the method is a method of enhancing muscle maintenance in a subject.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered to the subject prior to the subject exercising.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered to a subject at risk of acquiring a muscle disease or damage, such as an elderly subject.
  • the method also includes selecting a subject in need of enhancing muscle regeneration, repair, or maintenance.
  • selecting a subject in need of enhancing muscle regeneration, repair, or maintenance includes diagnosing the subject with a condition characterized by impaired muscle regeneration prior to administering an effective amount of the ⁇ 7 ⁇ 1 integrin modulatory agent to the subject.
  • selecting a subject in need of enhancing muscle regeneration, repair, or maintenance comprises diagnosing the subject with a condition characterized by impaired production of a component of ⁇ 7 ⁇ 1 integrin prior to administering an effective amount of the ⁇ 7 ⁇ 1 integrin modulatory agent to the subject.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered with an additional therapeutic agent.
  • the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional ⁇ 7 ⁇ 1 integrin modulatory agent.
  • the additional ⁇ 7 ⁇ 1 integrin modulatory agent is laminin-111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
  • a method of prospectively preventing or reducing muscle injury or damage in a subject is disclosed.
  • the method includes administering an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent to the subject wherein the ⁇ 7 ⁇ 1 integrin modulatory agent includes ciclopirox
  • the subject is at risk of developing a muscle injury or damage.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered with an additional therapeutic agent.
  • the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional ⁇ 7 ⁇ 1 integrin modulatory agent.
  • the additional ⁇ 7 ⁇ 1 integrin modulatory agent is laminin-111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
  • a method of enhancing ⁇ 7 ⁇ 1 integrin expression includes contacting a cell with an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent includes ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069- 0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S.
  • the cell is a muscle cell.
  • the muscle cell is present in a mammal, and wherein contacting the cell with an agent comprises administering the agent to the mammal.
  • impermissible substitution patterns ⁇ e.g., methyl substituted with 5 different groups, pentavalent carbon, and the like.
  • impermissible substitution patterns are easily recognized by a person of ordinary skill in the art.
  • agents such as an agent that increases ⁇ 7 ⁇ 1 expression and/or treats one or more symptoms associated with muscular dystrophy, by any effective route.
  • routes of administration include, but are not limited to, injection (such as subcutaneous, intramuscular, intradermal, intraperitoneal, and intravenous), oral, sublingual, rectal, transdermal, intranasal, vaginal and inhalation routes.
  • Agent Any protein, nucleic acid molecule (including chemically modified nucleic acids), compound, antibody, small molecule, organic compound, inorganic compound, or other molecule of interest.
  • Agent can include a therapeutic agent, a diagnostic agent or a pharmaceutical agent.
  • a therapeutic or pharmaceutical agent is one that alone or together with an additional compound induces the desired response (such as inducing a therapeutic or prophylactic effect when administered to a subject, including treating a subject with a muscular dystrophy).
  • an agent can act directly or indirectly to alter the expression and/or activity of ⁇ 7 ⁇ 1.
  • a therapeutic agent significantly increases the expression and/or activity of ⁇ 7 ⁇ 1 (which is a muscular dystrophy associated molecule) thereby treating one or more signs or symptoms associated with muscular dystrophy.
  • An example of a therapeutic agent is one that can increase the expression and/or activity of the ⁇ 7 ⁇ 1 gene or gene product, for example as measured by a clinical response (such as a decrease in one or more signs or symptoms associated with the muscular dystrophy, an improvement in muscular health, regeneration, repair or maintenance of a muscle cell or tissue).
  • improving muscular health refers to an improvement in muscular health compared with a preexisting state or compared with a state which would occur in the absence of treatment. For example, improving muscular health may include enhancing muscle regeneration, maintenance, or repair. Improving muscular health may also include prospectively treating a subject to prevent or reduce muscular damage or injury.
  • Regeneration refers to the repair of cells or tissue, such as muscle cells or tissue (or organs) which includes muscle cells, following injury or damage to at least partially restore the muscle or tissue to a condition similar to which the cells or tissue existed before the injury or damage occurred. Regeneration also refers to facilitating repair of cells or tissue in a subject having a disease affecting such cells or tissue to eliminate or ameliorate the effects of the disease.
  • regeneration places the cells or tissue in the same condition or an improved physiological condition as before the injury or damage occurred or the condition which would exist in the absence of disease.
  • Maintenance of cells or tissue such as muscle cells or tissue (or organs) which includes muscle cells, refers to maintaining the cells or tissue in at least substantially the same physiological condition, such as maintaining such condition even in the presence of stimulus which would normally cause damage, injury, or disease.
  • Repair of cells or tissue, such as muscle cells or tissue (or organs) which includes muscle cells, refers to the physiological process of healing damage to the cells or tissue following damage or other trauma.
  • a “pharmaceutical agent” is a chemical compound or composition capable of inducing a desired therapeutic or prophylactic effect when administered to a subject, alone or in combination with another therapeutic agent(s) or pharmaceutically acceptable carriers.
  • a pharmaceutical agent significantly increases the expression and/or activity of ⁇ 7 ⁇ 1 thereby treating a condition or disease associated with decreased ⁇ 7 ⁇ 1 expression/activity, such as muscular dystrophy.
  • R a is selected from hydrogen, alkyl, aryl, and cycloalkyl.
  • alkyl-C(0)0- substituted alkyl-C(0)0-, aryl-C(0)0-, substituted aryl-C(0)0-, cycloalkyl-C(0)0-, substituted cycloalkyl-C(0)0-, heteroaryl-C(0)0-, substituted heteroaryl-C(0)0-, heterocyclyl-C(0)0-, and substituted heterocyclyl-C(0)0-.
  • Acylalkyloxy alkyl-C(0)alkylO-, substituted alkyl-C(0)alkylO-, aryl-C(0)alkylO-, substituted aryl-C(0)alkylO-, cycloalkyl-C(0)alkylO-, substituted cycloalkyl-C(0)alkylO-, heteroaryl-C(0)alkylO-, substituted heteroaryl-C(0)alkylO-, heterocyclyl-C(0)alkylO-, and substituted heterocyclyl-C(0)alkylO-.
  • Alkyl A saturated or unsaturated monovalent hydrocarbon having a number of carbon atoms ranging from one to ten (e.g., Ci-ioalkyl), which is derived from removing one hydrogen atom from one carbon atom of a parent compound (e.g., alkane, alkene, alkyne).
  • Ci-ioalkyl A saturated or unsaturated monovalent hydrocarbon having a number of carbon atoms ranging from one to ten (e.g., Ci-ioalkyl), which is derived from removing one hydrogen atom from one carbon atom of a parent compound (e.g., alkane, alkene, alkyne).
  • An alkyl group may be branched or straight-chain.
  • Alkenyl A unsaturated monovalent hydrocarbon having a number of carbon atoms ranging from one to ten (e.g., C2-ioalkenyl), which has at least one carbon-carbon double bond and is derived from removing one hydrogen atom from one carbon atom of a parent alkene.
  • An alkenyl group may be branched, straight-chain, cyclic, cis, or trans (e.g., E or Z).
  • Alkynyl A unsaturated monovalent hydrocarbon having a number of carbon atoms ranging from one to ten (e.g., C2-ioalkynyl), which has at least one carbon-carbon triple bond and is derived from removing one hydrogen atom from one carbon atom of a parent alkyne.
  • An alkynyl group may be branched, straight-chain, or cyclic.
  • Alkoxy -O-alkyl (e.g., methoxy, ethoxy, «-propoxy, isopropoxy, «-butoxy, i-butoxy, sec-butoxy, «-pentoxy).
  • Alkylthio -S-alkyl, wherein alkyl is as defined herein. This term also encompasses oxidized forms of sulfur, such as -S(0)-alkyl, or -S(0)2-alkyl.
  • Aminocarbonyl -C(0)N(R b )2, wherein each R b independently is selected from hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclyl, substituted heterocyclyl. Also, each R b may optionally be joined together with the nitrogen bound thereto to form a heterocyclyl or substituted heterocyclyl group, provided that both R b are not both hydrogen.
  • Aminocarbonylalkyl -alkylC(0)N(R b )2, wherein each R b independently is selected from hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclyl, substituted heterocyclyl. Also, each R b may optionally be joined together with the nitrogen bound thereto to form a heterocyclyl or substituted heterocyclyl group, provided that both R b are not both hydrogen.
  • Aminocarbonylamino -NR a C(0)N(R b )2, wherein R a and each R b are as defined herein.
  • Analog or Derivative A compound which is sufficiently homologous to a compound such that it has a similar functional activity for a desired purpose as the original compound.
  • Analogs or derivatives refers to a form of a substance, such as cholestan, which has at least one functional group altered, added, or removed, compared with the parent compound.
  • examples of an analog are provided in Table 11, for example.
  • “Functional group” refers to a radical, other than a hydrocarbon radical, that adds a physical or chemical property to a substance.
  • Aryl a monovalent aromatic carbocyclic group of from 6 to 15 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl), which condensed rings may or may not be aromatic provided that the point of attachment is through an atom of the aromatic aryl group.
  • Arylthio -S-aryl wherein aryl is as defined herein. This term also encompasses oxidized forms of sulfur, such as -S(0)-aryl, or -S(0)2-aryl.
  • Biological activity The beneficial or adverse effects of an agent on living matter. When the agent is a complex chemical mixture, this activity is exerted by the substance's active ingredient or pharmacophore, but can be modified by the other constituents. Activity is generally dosage-dependent and it is not uncommon to have effects ranging from beneficial to adverse for one substance when going from low to high doses. In one example, the agent significantly increases the biological activity of ⁇ 7 ⁇ 1 which reduces one or more signs or symptoms associated with the muscular dystrophy.
  • Contacting an agent with a cell can occur in vitro by adding the agent to isolated cells or in vivo by administering the agent to a subject.
  • Control A sample or standard used for comparison with a test sample, such as a biological sample obtained from a patient (or plurality of patients) without a particular disease or condition, such as a muscular dystrophy.
  • the control is a sample obtained from a healthy patient (or plurality of patients) (also referred to herein as a "normal" control), such as a normal biological sample.
  • the control is a historical control or standard value ⁇ e.g., a previously tested control sample or group of samples that represent baseline or normal values (e.g., expression values), such as baseline or normal values of a particular gene such as a ⁇ 7 ⁇ 1 gene, gene product in a subject without a muscular dystrophy).
  • control is a standard value representing the average value (or average range of values) obtained from a plurality of patient samples (such as an average value or range of values of the gene or gene products, such as the ⁇ 7 ⁇ 1 gene or gene products, in the subjects without a muscular dystrophy).
  • Carboxyl -COOH or salts thereof.
  • Carboxyester -C(0)0-alkyl, -C(0)0- substituted alkyl, -C(0)0-aryl, -C(0)0-substituted aryl, -C(0)0-cycloalkyl, -C(0)0-substituted cycloalkyl, -C(0)0-heteroaryl, -C(0)0-substituted heteroaryl, -C(0)0-heterocyclyl, and -C(0)0-substituted heterocyclyl.
  • (Carboxyester)amino -NR a -C(0)0-alkyl, -NR a -C(0)0- substituted alkyl, -NR a -C(0)0-aryl, -NR a -C(0)0-substituted aryl, -NR a -C(0)0-cycloalkyl, -NR a -C(0)0-substituted cycloalkyl, - NR a -C(0)0-heteroaryl, -NR a -C(0)0-substituted heteroaryl, -NR a -C(0)0-heterocyclyl, and - NR a -C(0)0-substituted heterocyclyl, wherein R a is as recited herein.
  • (Carboxyester)oxy -0-C(0)0-alkyl, -0-C(0)0- substituted alkyl, -0-C(0)0-aryl, - 0-C(0)0-substituted aryl, -0-C(0)0-cycloalkyl, -0-C(0)0-substituted cycloalkyl, -0-C(0)0-heteroaryl, -0-C(0)0-substituted heteroaryl, -0-C(0)0-heterocyclyl, and -0-C(0)0-substituted heterocyclyl.
  • Cycloalkyl cyclic alkyl (or alkenyl, or alkynyl) groups of from 3 to 10 carbon atoms having single or multiple cyclic rings including fused, bridged, and spiro ring systems (e.g., cyclopropyl, cyclobutyl, etc.).
  • (Cycloalkyl)thio -S-cycloalkyl. This term also encompasses oxidized forms of sulfur, such as - S(0)-cycloalkyl, or -S(0)2-cycloalkyl. Decrease: To reduce the quality, amount, or strength of something. In one example, a therapy decreases one or more symptoms associated with the muscular dystrophy, for example as compared to the response in the absence of the therapy.
  • Diagnosis The process of identifying a disease, such as muscular dystrophy, by its signs, symptoms and results of various tests. The conclusion reached through that process is also called "a diagnosis.” Forms of testing commonly performed include blood tests, medical imaging, urinalysis, and biopsy.
  • Effective amount An amount of agent that is sufficient to generate a desired response, such as reducing or inhibiting one or more signs or symptoms associated with a condition or disease. When administered to a subject, a dosage will generally be used that will achieve target tissue/cell concentrations. In some examples, an "effective amount" is one that treats one or more symptoms and/or underlying causes of any of a disorder or disease. In some examples, an "effective amount” is a therapeutically effective amount in which the agent alone with an additional therapeutic agent(s) (for example anti-pathogenic agents), induces the desired response such as treatment of a muscular dystrophy, such as DMD, FCMD or MDC1A.
  • an additional therapeutic agent(s) for example anti-pathogenic agents
  • it is an amount of an agent capable of increasing ⁇ 7 ⁇ 1 gene expression or activity by least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100% (elimination of the disease to a point beyond detection).
  • an effective amount is an amount of a pharmaceutical preparation that alone, or together with a pharmaceutically acceptable carrier or one or more additional therapeutic agents, induces the desired response.
  • a desired response is to increase the subject's survival time by slowing the progression of the disease, such as slowing the progression of muscular dystrophy.
  • the disease does not need to be completely inhibited for the pharmaceutical preparation to be effective.
  • a pharmaceutical preparation can decrease the progression of the disease by a desired amount, for example by at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, as compared to the progression typical in the absence of the pharmaceutical preparation.
  • Treatment can involve only slowing the progression of the disease temporarily, but can also include halting or reversing the progression of the disease permanently.
  • Effective amounts of the agents described herein can be determined in many different ways, such as assaying for a reduction in of one or more signs or symptoms associated with the muscular dystrophy in the subject or measuring the expression level of one or more molecules known to be associated with the muscular dystrophy. Effective amounts also can be determined through various in vitro, in vivo or in situ assays, including the assays described herein.
  • the disclosed therapeutic agents can be administered in a single dose, or in several doses, for example daily, during a course of treatment.
  • the effective amount can be dependent on the source applied (for example a nucleic acid molecule isolated from a cellular extract versus a chemically synthesized and purified nucleic acid), the subject being treated, the severity and type of the condition being treated, and the manner of administration.
  • Expression The process by which the coded information of a gene is converted into an operational, non-operational, or structural part of a cell, such as the synthesis of a protein.
  • Gene expression can be influenced by external signals. For instance, exposure of a cell to a hormone may stimulate expression of a hormone-induced gene. Different types of cells can respond differently to an identical signal.
  • Expression of a gene also can be regulated anywhere in the pathway from DNA to RNA to protein. Regulation can include controls on transcription, translation, RNA transport and processing, degradation of intermediary molecules such as mRNA, or through activation, inactivation, compartmentalization or degradation of specific protein molecules after they are produced. In an example, expression, such as expression of ⁇ 7 ⁇ 1 , can be regulated to treat one or more signs or symptoms associated with muscular dystrophy.
  • nucleic acid molecule can be altered relative to a normal (wild type) nucleic acid molecule.
  • Alterations in gene expression, such as differential expression include but are not limited to: (1) overexpression; (2) underexpression; or (3) suppression of expression.
  • Alternations in the expression of a nucleic acid molecule can be associated with, and in fact cause, a change in expression of the
  • Protein expression can also be altered in some manner to be different from the expression of the protein in a normal (wild type) situation. This includes but is not necessarily limited to: (1) a mutation in the protein such that one or more of the amino acid residues is different; (2) a short deletion or addition of one or a few (such as no more than 10-20) amino acid residues to the sequence of the protein; (3) a longer deletion or addition of amino acid residues (such as at least 20 residues), such that an entire protein domain or sub-domain is removed or added; (4) expression of an increased amount of the protein compared to a control or standard amount; (5) expression of a decreased amount of the protein compared to a control or standard amount; (6) alteration of the subcellular localization or targeting of the protein; (7) alteration of the temporally regulated expression of the protein (such that the protein is expressed when it normally would not be, or alternatively is not expressed when it normally would be); (8) alteration in stability of a protein through increased longevity in the time that the protein remains localized in a cell; and (
  • Controls or standards for comparison to a sample, for the determination of differential expression include samples believed to be normal (in that they are not altered for the desired characteristic, for example a sample from a subject who does not have muscular dystrophy, such as DMD, FCMD or MDC1A) as well as laboratory values (e.g. , range of values), even though possibly arbitrarily set, keeping in mind that such values can vary from laboratory to laboratory.
  • Laboratory standards and values can be set based on a known or determined population value and can be supplied in the format of a graph or table that permits comparison of measured, experimentally determined values.
  • Extracellular matrix An extracellular structure of a tissue or a layer thereof, including the arrangement, composition, and forms of one or more matrix components, such as proteins, including structural proteins such as collagen and elastin, proteins such as fibronectin and laminins, and proteoglycans.
  • the matrix may comprise fibrillic collagen, having a network of fibers.
  • the extracellular matrix is connected to cells through the costameric protein network.
  • Halogen or Halo fluoro, chloro, bromo, and iodo.
  • Heteroaryl an aromatic group of from 1 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur within the ring.
  • Such heteroaryl groups can have a single ring (e.g., pyridinyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl), wherein the condensed rings may or may not be aromatic and/or contain a heteroatom, provided that the point of attachment is through an atom of the aromatic heteroaryl group.
  • the nitrogen and/or sulfur ring atom(s) of the heteroaryl group are optionally oxidized to provide for the N-oxide (N ⁇ 0), sulfinyl, or sulfonyl moieties.
  • Heteroaryloxy -O-heteroaryl.
  • Heteroarylthio -S-heteroaryl. This term also encompasses oxidized forms of sulfur, such as - S(0)-heteroaryl, or -S(0)2-heteoaryl.
  • Heterocyclyl a saturated, unsaturated group, or combinations thereof, having a single ring or multiple condensed rings, including fused bridged and spiro ring systems, and having from 3 to 15 ring atoms, including 1 to 4 heteroatoms, selected from nitrogen, sulfur, or oxygen.
  • These groups may be substituted with one or more of the substituents disclosed herein for substituted aryl and/or substituted alkyl.
  • These groups encompass, for example, a saturated heterocyclyl fused with one or more aromatic hydrocarbons or heteroaryl groups.
  • Heterocyclyloxy -O-heterocycyl.
  • Heterocyclylthio -S-heterocycyl. This term also encompasses oxidized forms of sulfur, such as - S(0)-heterocyclyl, or -S(0)2-heterocyclyl.
  • R c may be selected from hydrogen, aminocarbonylalkyloxy, substituted aminocarbonylalkyloxy, aminocarbonylalkylamino, and substituted aminocarbonylalkylamino.
  • an agent increases the activity or expression of ⁇ 7 ⁇ 1 , for example relative to an absence of the agent.
  • an agent increases the activity or expression of ⁇ 7 ⁇ 1 by at least 10%, at least 20%, at least 50%, or even at least 90%, including between 10% to 95%, 20% to 80%, 30% to 70%, 40% to 50%, such as 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 98%, or 100%.
  • Such increases can be measured using the methods disclosed herein.
  • a therapy increases (also known as up-regulates) the expression of ⁇ 7 ⁇ 1 , such as an increase of at least 10%, at least 20%, at least 50%, or even at least 90% in ⁇ 7 ⁇ 1 expression, thereby treating/alleviating one or more signs or symptoms associated with muscular dystrophy.
  • an increase in expression refers to an increase in a ⁇ 7 ⁇ 1 gene product.
  • An ⁇ 7 ⁇ 1 gene product can be RNA (such as mRNA, rRNA, tRNA, and structural RNA) or protein.
  • Gene upregulation includes any detectable increase in the production of a ⁇ 7 ⁇ 1 gene product.
  • production of a ⁇ 7 ⁇ 1 gene product increases by at least 2-fold, for example at least 3-fold or at least 4-fold, as compared to a control (such an amount of gene expression in a normal cell).
  • a control is a relative amount of a7 gene expression or protein expression in a biological sample taken from a subject who does not have muscular dystrophy, such as DMD, FCMD or MDC1A.
  • Such increases can be measured using the methods disclosed herein.
  • "detecting or measuring expression of ⁇ 7 ⁇ 1" includes quantifying the amount of the gene, gene product or modulator thereof present in a sample. Quantification can be either numerical or relative.
  • Detecting expression of the gene, gene product or modulators thereof can be achieved using any method known in the art or described herein, such as by measuring nucleic acids by PCR (such as RT-PCR) and proteins by ELISA.
  • the change detected is an increase or decrease in expression as compared to a control, such as a reference value or a healthy control subject.
  • the detected increase or decrease is an increase or decrease of at least two-fold compared with the control or standard.
  • Controls or standards for comparison to a sample, for the determination of differential expression include samples believed to be normal (in that they are not altered for the desired characteristic, for example a sample from a subject who does not have muscular dystrophy, such as DMD, FCMD or MDC1A) as well as laboratory values (e.g., range of values), even though possibly arbitrarily set, keeping in mind that such values can vary from laboratory to laboratory.
  • samples believed to be normal in that they are not altered for the desired characteristic, for example a sample from a subject who does not have muscular dystrophy, such as DMD, FCMD or MDC1A
  • laboratory values e.g., range of values
  • Laboratory standards and values can be set based on a known or determined population value and can be supplied in the format of a graph or table that permits comparison of measured, experimentally determined values.
  • the increase or decrease is of a diagnostically significant amount, which refers to a change of a sufficient magnitude to provide a statistical probability of the diagnosis.
  • the level of expression in either a qualitative or quantitative manner can detect nucleic acid or protein.
  • Exemplary methods include microarray analysis, RT-PCR, Northern blot, Western blot, and mass spectrometry.
  • Inhibiting a disease or condition A phrase referring to reducing the development of a disease or condition, for example, in a subject who is at risk for a disease or who has a particular disease. Particular methods of the present disclosure provide methods for inhibiting muscular dystrophy. "
  • Integrin A cell surface transmembrane glycoprotein receptor. Integrins are involved in many biological processes such as wound healing, blood clot formation, gene regulation, and immune responses. Integrins can regulate tissue specific cell adhesion molecules. Integrins are heterodimeric non-covalently associated glycoproteins composed of two subunits. The subunits, which are designated a and beta, have approximate molecular weights of 150-180 kilodaltons and 90-110 kilodaltons, respectively.
  • the ⁇ 7 ⁇ 1 integrin is a major laminin receptor expressed in skeletal muscle.
  • the ⁇ 7 ⁇ 1 integrin plays a role in the development of neuromuscular and myotendinous junctions.
  • the ⁇ 7 ⁇ 1 integrin In the adult, the ⁇ 7 ⁇ 1 integrin is concentrated at junctional sites and found in extrajunctional regions where it mediates the adhesion of the muscle fibers to the extracellular matrix.
  • Mice that lack the l chain develop muscular dystrophy that affects the myotendinous junctions.
  • the absence of al integrin results in defective matrix deposition at the myotendinous junction.
  • Loss of the al integrin in ⁇ -sarcoglycan mice results in severe muscle pathology. Absence of the al integrin in mdx mice also results in severe muscular dystrophy, confirming that the ⁇ 7 ⁇ 1 integrin serves as a major genetic modifier for Duchenne and other muscular dystrophies.
  • al integrin may be important for muscle regeneration.
  • the ⁇ 7 ⁇ 1 integrin regulates myoblast migration to regions of myofiber formation. It has been found that MyoD (myogenic determination protein) transactivates al integrin gene expression in vitro, which would increase al integrin levels in activated satellite cells.
  • NM_001144116 human
  • NM_008398.2 mouse
  • Gene Accession No. NM_002211 for ⁇ integrin also known as CD29
  • Exemplary ⁇ 7 ⁇ 1 integrin modulatory agents are disclosed herein, such as in Tables 8, 9 and 10, including ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl, 5 a-cholestan-3p-ol- 6-one, Compound ID#1001, Compound ID# 1002, Compound ID# 1003, N032-0003, N066-0070, N069- 0071, N069-0075, 1080-0573 , N064-0028, N066-0053, or N069-0073 or analogs provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof.
  • a ⁇ 7 ⁇ 1 integrin -associated condition is a condition associated with altered ⁇ 7 ⁇ 1 integrin expression or activity, including muscular dystrophy, such as DMD, FCMD, LGMD, FHMD, Beckers muscular dystrophy and/or MDC1A.
  • Laminin Any of the family of glycoproteins that are typically involved in the formation and maintenance of extracellular matrices.
  • Laminin is a heterotrimers formed from an a chain, a ⁇ chain, and a ⁇ chain.
  • the various chains of a particular laminin can affect the properties of the molecule.
  • fragments, derivatives, or analogs of various laminins can be used, such as laminins having at least a portion at least substantially homologous to the laminin al chain.
  • a "fragment of laminin,” as used herein, refers to a portion of a substance, such as laminin.
  • a fragment may be, in some examples, a particular domain or chain of a protein.
  • particular embodiments of the present disclosure involve administering a fragment of laminin- 1 corresponding to at least a portion of (or all of) the laminin al chain. Fragments may be synthetic or may be derived from larger parent substances.
  • laminins may be administered as a mixture of laminins, including fragments, analogs, and derivatives thereof. Suitable methods for preparing analogs of laminin domains are disclosed in U.S. Patent No. 6,933,280, incorporated by reference herein to the extent not inconsistent with this disclosure.
  • the laminin materials or compositions of the present disclosure may be delivered as discrete molecules or may be complexed with, or conjugated to, another substance.
  • the laminin may be combined with a carrier, such as to aid in delivery of the laminin to a site of interest or to increase physiological uptake or incorporation of the laminin.
  • the laminin administered includes or consists of laminin- 1 (LAM-111), which includes the chains ⁇ .
  • the laminin administered includes or consists of laminin-2, which includes the chains ⁇ 2 ⁇ 1 ⁇ 1.
  • the laminin administered includes or consists of laminin-4, which includes the chains ⁇ 2 ⁇ 2 ⁇ 1.
  • Laminins may be obtained from any suitable source.
  • laminin- 1 may be obtained from placental tissue or from Engelbreth-Holm-Swarm murine sarcoma. Suitable methods of isolating various laminins are disclosed in U.S. Patent No. 5,444,158, incorporated by reference herein to the extent not inconsistent with the present disclosure.
  • Muscle Any myoblast, myocyte, myofiber, myotube or other structure composed of muscle cells. Muscles or myocytes can be skeletal, smooth, or cardiac. Muscle may also refer to, in particular implementations of the present disclosure, cells or other materials capable of forming myocytes, such as stem cells and satellite cells.
  • Muscular dystrophy A term used to refer to a group of genetic disorders that lead to progressive muscle weakness. Muscular dystrophy can result in skeletal muscle weakness and defects in skeletal muscle proteins, leading to a variety of impaired physiological functions. No satisfactory treatment of muscular dystrophy exists. Existing treatments typically focus on ameliorating the effects of the disease and improving the patient's quality of life, such as through physical therapy or through the provision of orthopedic devices.
  • Mutated genes associated with muscular dystrophy are responsible for encoding a number of proteins associated with the costameric protein network.
  • proteins include laminin-2, collagen, dystroglycan, integrins, caveolin-3, ankyrin, dystrophin, a-dystrobrevin, vinculin, plectin, BPAGlb, muscle LIM protein, desmin, actinin-associated LIM protein, a-actin, titin, telethonin, cypher, myotilin, and the sarcoglycan/sarcospan complex.
  • DMD muscular dystrophy
  • DMD is an X-linked recessive disorder characterized by a mutation in the gene that codes for dystrophin.
  • Dystrophin is a cytoskeletal protein about 430 kDa in size. This protein works to connect the cell's cytoskeleton and extracellular matrix.
  • the loss of dystrophin in DMD patients leads to a loss of muscle fiber attachment at the extracellular matrix during contraction, which ultimately leads to progressive fiber damage, membrane leakage and a loss of muscle function. Most patients die before they reach the age of 30 due to respiratory or cardiac failure.
  • Beckers muscular dystrophy (also known as Benign pseudohypertrophic muscular dystrophy) is related to DMD in that both result from a mutation in the dystrophin gene, but in DMD no functional dystrophin is produced making DMD much more severe than BMD.
  • BMD is an X-linked recessive inherited disorder characterized by slowly progressive muscle weakness of the legs and pelvis.
  • BMD is a type of dystrophinopathy, which includes a spectrum of muscle diseases in which there is insufficient dystrophin produced in the muscle cells, results in instability in the structure of muscle cell membrane. This is caused by mutations in the dystrophin gene, which encodes the protein dystrophin.
  • the pattern of symptom development of BMD is similar to DMD, but with a later, and much slower rate of progression.
  • Congenital muscular dystrophies are caused by gene mutations.
  • FCMD and MDC1A are examples of congenital muscular dystrophies.
  • MDC1A is a congential muscular dystrophy due to a genetic mutation in the LAMA2 gene which results in lack of or complete loss of laminin-a2 protein. This loss of laminin-a2 leads to an absence of laminins-211/221.
  • Laminins-211/221 are major components of the extracellular matrix and play a key role in muscle cell development. During muscle cell differentiation laminin binds to the ⁇ 7 ⁇ 1 integrin. Without laminin-a2, muscle fibers are unable to adhere to the basement membrane and myotubes undergo apotosis. Muscle regeneration also fails, leading to a loss of muscle repair and an increase in muscle fibrosis and inflammation. This chronic tissue injury is a major cause of morbidity and mortality in MDC1A.
  • Congenital Muscular Dystrophies CMD
  • Limb-Girdle muscular dystrophy LGMD
  • CMD Congenital Muscular Dystrophies
  • LGMD Limb-Girdle muscular dystrophy
  • MDC1A is a progressive muscle wasting disease that results in children being confined to a wheelchair, requiring ventilator assistance to breathe and premature death. Symptoms are detected at birth with poor muscle tone and "floppy" baby syndrome. DMD, BMD and LGMD are progressive muscle degenerative diseases usually diagnosed at 3-5 years of age when children show developmental delay including ability to walk and climb stairs. The disease is progressive and children are usually confined to a wheelchair in their teens and require ventilator assistance.
  • Fukuyama congenital muscular dystrophy is an inherited condition that predominantly affects the muscles, brain, and eyes.
  • Congenital muscular dystrophies are a group of genetic conditions that cause muscle weakness and wasting (atrophy) beginning very early in life. Fukuyama congenital muscular dystrophy affects the skeletal muscles, which are muscles the body uses for movement. The first signs of the disorder appear in early infancy and include a weak cry, poor feeding, and weak muscle tone
  • Fukuyama congenital muscular dystrophy Most children with Fukuyama congenital muscular dystrophy are never able to stand or walk, although some can sit without support and slide across the floor in a seated position. More than half of all affected children also experience seizures. Other signs and symptoms of Fukuyama congenital muscular dystrophy include impaired vision, other eye abnormalities, and slowly progressive heart problems after age 10. As the disease progresses, affected people may develop swallowing difficulties that can lead to a bacterial lung infection called aspiration pneumonia. Because of the serious medical problems associated with Fukuyama congenital muscular dystrophy, most people with the disorder live only into late childhood or adolescence.
  • Fukuyama congenital muscular dystrophy is seen almost exclusively in Japan, where it is the second most common form of childhood muscular dystrophy (after Duchenne muscular dystrophy). Fukuyama congenital muscular dystrophy has an estimated incidence of 2 to 4 per 100,000 Japanese infants.
  • Fukuyama congenital muscular dystrophy is caused by mutations in the FKTN gene which encodes fukutin.
  • the most common mutation in the FKTN gene reduces the amount of fukutin produced within cells.
  • a shortage of fukutin likely prevents the normal modification of a-dystroglycan, which disrupts that protein's normal function. Without functional a-dystroglycan to stabilize muscle cells, muscle fibers become damaged as they repeatedly contract and relax with use. The damaged fibers weaken and die over time, leading to progressive weakness and atrophy of the skeletal muscles.
  • Defective a-dystroglycan also affects the migration of neurons during the early development of the brain. Instead of stopping when they reach their intended destinations, some neurons migrate past the surface of the brain into the fluid-filled space that surrounds it. Because Fukuyama congenital muscular dystrophy involves a malfunction of a-dystroglycan, this condition is described as a dystroglycanopathy.
  • Facioscapulohumeral muscular dystrophy is a form of muscular dystrophy associated with progressive muscle weakness and loss of muscle tissue. Unlike DMD and BMD which mainly affect the lower body, FSHDaffects the upper body mainly the face, shoulder and upper arm muscles. However, it can affect muscles around the pelvis, hips, and lower leg. Symptoms for FSHDoften do not appear until age 10 - 26, but it is not uncommon for symptoms to appear much later. In some cases, symptoms never develop. Symptoms are usually mild and very slowly become worse.
  • Facial muscle weakness is common, and may include eyelid drooping, inability to whistle, decreased facial expression, depressed or angry facial expression, difficulty pronouncing words, shoulder muscle weakness (leading to deformities such as pronounced shoulder blades (scapular winging) and sloping shoulders), weakness of the lower, hearing loss and possible heart conditions.
  • compositions and formulations suitable for pharmaceutical delivery of one or more agents such as one or more ⁇ 7 ⁇ 1 modulatory agents.
  • parenteral formulations can include injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle.
  • pharmaceutical agents to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate, sodium lactate, potassium chloride, calcium chloride, and triethanolamine oleate.
  • Prodrug A compound that is transformed in vivo to yield a parent compound, for example, by hydrolysis in the gut or enzymatic conversion in blood.
  • Sample A biological specimen containing genomic DNA, RNA (including mRNA), protein, or combinations thereof, obtained from a subject. Examples include, but are not limited to, peripheral blood, urine, saliva, tissue biopsy, surgical specimen, and autopsy material.
  • a sample includes muscle biopsy, such as from a subject with DMD, FCMD, or MDC1A.
  • Signs or symptoms Any subjective evidence of disease or of a subject's condition, e.g., such evidence as perceived by the subject; a noticeable change in a subject's condition indicative of some bodily or mental state.
  • a "sign” is any abnormality indicative of disease, discoverable on examination or assessment of a subject.
  • a sign is generally an objective indication of disease.
  • Signs include, but are not limited to any measurable parameters such as tests for detecting muscular dystrophy, including measuring creatine kinase levels, electromyography (to determine if weakness is caused by destruction of muscle tissue rather than by damage to nerves) or immunohistochemistry/immunoblotting/immunoassay (e.g., ELISA) to measure muscular dystrophy-associated molecules, such as ⁇ 7 ⁇ 1 integrin.
  • ELISA immunohistochemistry/immunoblotting/immunoassay
  • reducing or inhibiting one or more symptoms or signs associated with muscular dystrophy includes increasing the activity or expression of ⁇ 7 ⁇ 1 integrin by a desired amount, for example by at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, as compared to the activity and/or expression in the absence of the treatment.
  • Symptoms of muscular dystrophy include, but are not limited to, muscle weakness and loss, difficulty running, difficulty hopping, difficulty jumping, difficulty walking, difficulty breathing, fatigue, skeletal deformities, muscle deformities (contractions of heels; pseudohypertrophy of calf muscles), heart disease (such as dilated cardiomyopathy), elevated creatine phosphokinase (CK) levels in blood or combinations thereof.
  • Subject Living multi-cellular vertebrate organisms, a category that includes human and non- human mammals.
  • Substituted Alkyl an alkyl (or alkenyl, or alkynyl) group having from 1 to 5 hydrogen atoms replaced with substituents selected alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, acylalkyloxy, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aminodicarbonylamino, aminocarbonylalkyl, aminosulfonyl, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, aminodiacylamino, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalky
  • Substituted Alkoxy -0-(substituted alkyl).
  • Substituted Alkylthio -S-(substituted alkyl). This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted alkyl, or -S(0)2-substituted alkyl.
  • each R b independently is selected from hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclyl, substituted heterocyclyl. Also, each R b may optionally be joined together with the nitrogen bound thereto to form a heterocyclyl or substituted heterocyclyl group, provided that both R b are not both hydrogen.
  • Substituted Aryl aryl groups having 1 to 5 hydrogens replaced with substituents independently selected from alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aminosulfonyl, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy, heteroarylthio, substituted heteroarylthio, heterocycly
  • Substituted Aryloxy -0-(substituted aryl).
  • Substituted Arylthio -S-(substituted aryl), wherein substituted aryl is as defined herein. This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted aryl, or -S(0)2-substituted aryl.
  • Cycloalkyl cycloalkyl ,cycloalkenyl, or cycloalkynyl group having from 1 to 5 substituents selected from the group consisting of oxo, alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aminosulfonyl, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, halo, hydroxy, heteroaryl, substituted heteroaryl
  • the aryl group may be substituted with 1 to 2, 1 to 3, or 1 to 4 of these groups, which are defined herein.
  • the cycloalkyl group may have multiple condensed rings (e.g. tetrahydronaphthyl or tetrahydroanthacenyl), provided that the point of attachment is through an atom of the nonaromatic ring.
  • Substituted (Cycloalkyl)thio refers to -S-(substituted cycloalkyl). This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted cycloalkyl, or -S(0)2-substituted cycloalkyl.
  • Substituted Heteroaryl heteroaryl groups that are substituted with from 1 to 5 substituents selected from the group consisting of the same group of substituents defined for substituted aryl.
  • Substituted Heteroarylthio -S-(substituted heteroaryl). This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted heteroaryl, or -S(0)2-substituted heteoaryl.
  • Substituted Heterocycyloxy -0-(substituted heterocyclyl) wherein the heterocyclyl group is substituted with one or more of the substituents recited for substituted alkyl.
  • Substituted Heterocycylthio -S-(substituted heterocycyl). This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted heterocyclyl, or -S(0)2-substituted heterocyclyl.
  • Sulfonyl -S02-alkyl, -SC -substituted alkyl, -S02-cycloalkyl, -S02-substituted cycloalkyl, -SO2- aryl, -S02-substituted aryl, -S02-heteroaryl, -SC -substituted heteroaryl, -S02-heterocyclyl, and -S02-substituted heterocyclyl.
  • Tissue An aggregate of cells, usually of a particular kind, together with their intercellular substance that form one of the structural materials of an animal and that in animals include connective tissue, epithelium, muscle tissue, and nerve tissue.
  • Treating a disease A therapeutic intervention that ameliorates a sign or symptom of a disease or pathological condition related to a muscular dystrophy, such as a sign or symptom of muscular dystrophy. Treatment can induce remission or cure of a condition or slow progression, for example, in some instances can include inhibiting the full development of a disease, for example preventing development of a muscular dystrophy. Prevention of a disease does not require a total absence of disease. For example, a decrease of at least 50% can be sufficient.
  • Treating a disease can be a reduction in severity of some or all clinical symptoms of the disease or condition, a reduction in the number of relapses of the disease or condition, an improvement in the overall health or well-being of the subject, by other parameters well known in the art that are specific to the particular disease or condition, and combinations of such factors.
  • a phrase that is used to describe any environment that permits the desired activity includes administering a disclosed agent to a subject sufficient to allow the desired activity.
  • the desired activity is increasing the expression or activity of ⁇ 7 ⁇ 1. ///.
  • the compound is effective in treating muscular dystrophy.
  • the compound is a small-molecule therapeutic.
  • the small- molecule therapeutic is a cyclic compound comprising a heteroatom-containing skeleton.
  • the small-molecule therapeutic is a cyclic compound comprising an all-carbon skeleton.
  • the cyclic compound comprising a heteroatom-containing skeleton has a formula illustrated below:
  • each R 1 independently is selected from Ci-ioalkyl, substituted Ci-ioalkyl, Ci-ioalkoxy, substituted Ci. loalkoxy, acyl, acylamino, acyloxy, acylCi-ioalkyloxy, amino, substituted amino, aminoacyl,
  • aminocarbonylCi-ioalkyl aminocarbonylamino, aminodicarbonylamino, aminocarbonyloxy, aminosulfonyl, C6-i 5 aryl, substituted C6-isaryl, C6-i 5 aryloxy, substituted C6-i 5 aryloxy, C6-i 5 arylthio, substituted C6-isarylthio, carboxyl, carboxyester, (carboxyester)amino, (carboxyester)oxy, cyano, C3- 8 cycloalkyl, substituted C3- 8 cycloalkyl, (C3- 8 cycloalkyl)oxy, substituted (C3-scycloalkyl)oxy, (C3- 8 cycloalkyl)thio, substituted (C3- 8 cycloalkyl)thio, halo, hydroxyl, Ci-ioheteroaryl, substituted Ci-ioheteroaryl, Ci-io
  • R 1 substituents together with the atom to which each is bound, may form ring selected from a C6-i 5 aryl, substituted C6-isaryl, C3- 8 cycloalkyl, substituted C3- 8 cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C 2 -iosubstituted heterocyclyl, and C 2 -ioheterocyclyloxy, substituted;
  • each of A, B, C, D, and E independently may be selected from carbon, nitrogen, oxygen, and sulfur;
  • n may be zero, 1, 2, 3, 4, or 5.
  • the cyclic compound comprising a heteroatom-containing moiety has a formula illustrated below:
  • each R 2 independently is selected from Ci-ioalkyl, substituted Ci-ioalkyl, Ci-ioalkoxy, substituted Ci. loalkoxy, acyl, acylamino, acyloxy, acylCi-ioalkyloxy, amino, substituted amino, aminoacyl,
  • aminocarbonylCi-ioalkyl aminocarbonylamino, aminodicarbonylamino, aminocarbonyloxy, aminosulfonyl, C6-i 5 aryl, substituted C6-isaryl, C6-i 5 aryloxy, substituted C6-i 5 aryloxy, C6-i 5 arylthio, substituted C6-isarylthio, carboxyl, carboxyester, (carboxyester)amino, (carboxyester)oxy, cyano, C3- 8 cycloalkyl, substituted C3- 8cycloalkyl, (C3-8cycloalkyl)oxy, substituted (C3-scycloalkyl)oxy, (C3-scycloalkyl)thio, substituted (C3- 8 cycloalkyl)thio, halo, hydroxyl, Ci-ioheteroaryl, substituted Ci-ioheteroaryl, Ci-ioheteroary
  • R 2 substituents together with the atom to which each is bound, may form ring selected from a C6-i 5 aryl, substituted C6-isaryl, C3- 8 cycloalkyl, substituted C3- 8 cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C 2 -iosubstituted heterocyclyl, and C 2 -ioheterocyclyloxy, substituted;
  • each of A, B, C, D, E, and F independently may be selected from carbon, nitrogen, oxygen, and sulfur;
  • n may be zero, 1, 2, 3, 4, or 5.
  • the cyclic compound comprising an all-carbon skeleton may have a general formula provided below:
  • R 3 and R 4 independently may be selected from hydroxyl, hydrogen, Ci-ioalkyl, substituted Ci- ioalkyl, carboxyl, acyl, aminoacyl, acylamino, amino, substituted amino, C6-i 5 aryl, substituted C6-isaryl, and Ci-ioalkoxy;
  • R 5 is selected from amino, substituted amino, oxo, hydroxyl, Ci-ioalkoxy, and imino; and n may be zero, 1, 2, 3, 4, or 5.
  • rings A and B are connected via the optional bonds to form a steroid-based skeleton.
  • R 5 may be bound to ring A via a double bond or a single bond, a feature that is indicated with the optional dashed bond in Formula 13. For example, if R 5 is amino, hydroxyl, substituted amino, or Ci-ioalkoxy, then R 5 is attached to ring A via a single bond, whereas if R 5 is oxo or imino, then R5 is attached to ring A via a double bond.
  • C6-i 5 aryl may be selected from phenyl, biphenyl, naphthalene, anthracene, and the like; substituted C6-i 5 aryl may be selected from phenyl, biphenyl, naphthalene, and anthracene substituted with one or more substituents as defined herein; Ci-ioalkyl may be selected from Ci.
  • loalkane, C 2 -ioalkene, and C 2 -ioalkyne are typically from methyl, ethyl, propyl, butyl, pentyl, hexyl, and the like; ethylene, propylene, butylene, and the like; and ethyne, propyne, butyne, and the like; substituted Ci-ioalkyl may be selected from Ci-ioalkane, C2-ioalkene, and C2-ioalkyne substituted with one or of the substituents as provided herein.
  • Exemplary embodiments concerning hetercyclyl and heteroaryl substitutents include, but are not limited to, epoxy, pyrrolyl, imidazole, pyrazole, pyridinyl, pyrazine, pyrimidine, oxanyl, thianyl, dioxanyl, dithianyl, coumarin, pyridazine, indolizine, isoindole, indolyl, indolinyl (or dihydroindole), indazole, purine, isoquinoline, quinoline, benzo[d]pyridazine, naphthyridine, quinoxaline, quinazoline,
  • Particular disclosed embodiments concern cyclic compounds comprising a five-membered heteroatom-containing skeleton having a formula selected from those provided below.
  • R 1 and n are as recited herein, and each X independently may be selected from carbon, oxygen, nitrogen, and sulfur.
  • the cyclic compound comprising a five-membered heteroatom-containing skeleton may have any one of the following formulas
  • Z may be selected from carbon and nitrogen
  • Y may be selected from nitrogen and oxygen
  • each X independently may be selected from nitrogen and carbon.
  • the dashed lines indicate variable bonds which may or may not be present, depending on the valency of the atom to which each variable bond is attached.
  • X typically is carbon, as a carbon atom can accommodate four bonds.
  • X may be nitrogen in such a compound; however, a person of ordinary skill in the art would recognize that the nitrogen atom would be positively charged due to the fact that its lone pairs are used to accommodate a fourth bond.
  • ring A is not connected with ring B and exists as an aryl compound having a formula illustrated below.
  • the compound may have any one of the following formulas
  • each Y 1 and Y 2 independently may be selected from oxygen, sulfur, and NR b wherein R b is as disclosed herein; and R 6 is selected from those substituents provided for R 1 .
  • Any of the compounds disclosed herein may also be used in an alternate chemical form, such as a pharmaceutically acceptable salt, a N-oxide, a prodrug, and/or a solvate.
  • the compound may be a pharmaceutically acceptable salt, such as a hydrochloride salt, a hydrobromide salt, a hydroiodide salt, a nitrate salt, a sulfate salt, a bisulfate salt, a phosphate salt, an acid phosphate salt, an isonicotinate salt, an acetate salt, a lactate salt, a salicylate salt, a citrate salt, a tartrate salt, an ascorbate salt, a succinate salt, a maleate salt, a fumarate salt, a gluconate salt, a saccharate salt, a formate salt, a benzoate salt, a glutamate salt, a methanesulfonate salt, an ethanesulfonate salt, a benzensulfonate salt, a />-toluenesulfonate salt, and combinations thereof.
  • a pharmaceutically acceptable salt such as a hydrochloride salt,
  • the ⁇ 7 ⁇ 1 integrin has been shown to be a major modifier of disease progression in patients with muscular dystrophy. Increased expression of the a7 integrin in muscle can alleviate muscle disease in mouse models of muscular dystrophy.
  • a muscle cell-based assay described in Example 1 below, the inventors identified the following molecules that up-regulate ⁇ 7 ⁇ 1 integrin expression in muscle:
  • the method includes administering an effective amount of a ⁇ 7 ⁇ 1 integrin modulatory agent to a subject with muscular dystrophy or suspected of having or developing muscular dystrophy, in which the agent increases the biological activity or expression of ⁇ 7 ⁇ 1 integrin and thereby, treating the muscular dystrophy in the subject.
  • the method of treatment inhibits or reduces one or more signs or symptoms associated with muscular dystrophy in the subject.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent includes one or more of the following molecules: ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, an analog of Compound ID# 1001, an analog of
  • Compound ID #1002 an analog of Compound ID #1003, an analog of cholestan (see Table 9), laminin-111, laminin-111 fragments, valproic acid, or a valproic acid analog.
  • Tables 8 and 9 and FIGS. 3A-3C, 4A-4B, 5, 6A-6B, and 7-9 provide the chemical structures and characterization data for such compounds.
  • Exemplary valproic acid analogs are disclosed in U.S. Patent Publication 2006/0223888 and International Patent Application No. 2010/080581, each of which is incorporated herein by reference in its entirety.
  • Table 11 provides analogs of disclosed Compound ID# 1001. Exemplary laminin-111 fragments are disclosed in U.S.
  • Patent Publication US-2009-0092587-A1 which is incorporated herein by reference in its entirety.
  • an analog of Compound ID #1002 or #1003 is synthesized according to the general synthesis pathway shown in FIG. 10.
  • an analog is synthesized according to the synthesis pathway shown in the Examples below.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is an
  • the ⁇ 7 ⁇ 1 integrin modulatory agent includes one or more molecules provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent may be selected from any one or more of the compounds within any one of Formulas 1-16, as provided herein.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents can alter the expression of nucleic acid sequences (such as DNA, cDNA, or mRNAs) and proteins of ⁇ 7 ⁇ 1 integrin. An increase in the expression or activity does not need to be 100% for the agent to be effective.
  • an agent can increase the expression or biological activity by a desired amount, for example by at least 10%, for example at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, including about 15% to about 98%, about 30% to about 95%, about 40% to about 80%, about 50% to about 70%, including about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%, as compared to activity or expression in a control.
  • Methods of assessing ⁇ 7 ⁇ 1 integrin expression and activity are known to those of ordinary skill in the art, including those described in the Examples below (e.g. , Western blot and ELISA assay with commercially available antibodies).
  • the subject is a human.
  • the method involves selecting a subject with muscular dystrophy.
  • a subject is selected for treatment following diagnosing the subject with muscular dystrophy.
  • the method can include diagnosing the subject as suffering from muscular dystrophy, such as DMD, MDC1A, MDC1D, LGMD, DMD, FCMD or FHMD.
  • Methods of diagnosing a subject with muscular dystrophy include, but are not limited to, muscle biopsies and measuring serum creatine kinase levels. Additionally, alterations in biomarker known to be associated with muscular dystrophy may be detected by measuring such levels in serum or urine sample.
  • the method involves diagnosing the subject as suffering from a disease, disorder, or condition characterized by a mutation in the gene encoding l integrin.
  • the method involves diagnosing the subject as suffering from a disease, disorder, or condition characterized by a decreased level of a l integrin expression.
  • Alterations in the expression can be measured at the nucleic acid level (such as by real time quantitative polymerase chain reaction or microarray analysis) or at the protein level (such as by Western blot analysis or ELISA). These methods are known to those of skill in the art.
  • the assay results, findings, diagnoses, predictions and/or treatment recommendations are recorded and communicated to technicians, physicians and/or patients, for example.
  • computers are used to communicate such information to interested parties, such as, patients and/or the attending physicians.
  • the therapy selected for administered is then based upon these results.
  • the results and/or related information is communicated to the subject by the subject's treating physician.
  • the results may be communicated directly to a test subject by any means of communication, including writing, such as by providing a written report, electronic forms of communication, such as email, or telephone. Communication may be facilitated by use of a computer, such as in case of email communications.
  • the communication containing results of a diagnostic test and/or conclusions drawn from and/or treatment recommendations based on the test may be generated and delivered automatically to the subject using a combination of computer hardware and software which will be familiar to artisans skilled in telecommunications.
  • a healthcare- oriented communications system is described in U.S. Pat. No.
  • identification of a subject as having muscular dystrophy results in the physician treating the subject, such as prescribing one or more disclosed ⁇ 7 ⁇ 1 agents for inhibiting or delaying one or more signs and symptoms associated with muscular dystrophy.
  • the dose or dosing regimen is modified based on the information obtained using the methods disclosed herein.
  • the method includes administering an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent to a subject in need of muscle regeneration, repair or maintenance, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent includes ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent includes one or more of the following molecules: ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan ⁇ -ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, an analog of Compound ID# 1001, an analog of
  • Compound ID #1002 an analog of Compound ID #1003, an analog of cholestan (see Table 9), laminin-111, laminin-111 fragments, valproic acid, or a valproic acid analog.
  • Tables 8 and 9 and FIGS. 3A-3C, 4A-4B, 5, 6A-6B, and 7-9 provide the chemical structures and characterization data for such compounds.
  • Exemplary valproic acid analogs are disclosed in U.S. Patent Publication 2006/0223888 and International Patent Application No. 2010/080581, each of which is incorporated herein by reference in its entirety.
  • Exemplary laminin-111 fragments are disclosed in U.S. Patent Publication US-2009-0092587-A1, which is incorporated herein by reference in its entirety.
  • an analog of Compound ID #1002 or #1003 is synthesized according to the general synthesis pathway shown in FIG. 10. In some examples, an analog is synthesized according to the synthesis pathway provided in the Examples below.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is an analog/derivative of any of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents which may be designed and synthesized according to the chemical principles known to one of ordinary skill in the art and identified as a ⁇ 7 ⁇ 1 integrin modulatory agent by methods known to those of ordinary skill in the art, including the muscle cell based assay described Example 1.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent includes one or more molecules provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents can increase the expression of nucleic acid sequences (such as DNA, cDNA, or mRNAs) and proteins of ⁇ 7 ⁇ 1 integrin. An increase in the expression or activity does not need to be 100% for the agent to be effective.
  • an agent can increase the expression or biological activity by a desired amount, for example by at least 10%, for example at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, including about 15% to about 98%, about 30% to about 95%, about 40% to about 80%, about 50% to about 70%, including about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%, as compared to activity or expression in a control.
  • Methods of assessing ⁇ 7 ⁇ 1 integrin expression and activity are known to those of ordinary skill in the art, including those described in the Examples below (e.g. , Western blot and ELISA assay with commercially available antibodies).
  • Muscle regeneration may benefit, for example, geriatric or other patient populations with reduced muscle repair capability, or simply speed the muscle repair process for otherwise physiologically unimpaired patients.
  • administration of a ⁇ 7 ⁇ 1 integrin modulatory agent can aid muscle repair, or reduction of muscle damage, in athletes or others having activity-induced muscle injury or damage.
  • muscle repair in patients suffering from muscle damage, such as through accident or injury can be augmented by administration of a ⁇ 7 ⁇ 1 integrin modulatory agent.
  • ⁇ 7 ⁇ 1 modulatory agent is administered prior to the subject experiencing muscle damage or disease. In some examples, the ⁇ 7 ⁇ 1 integrin modulatory agent is administered to the subject prior to the subject exercising.
  • the method further includes selecting a subject in need of enhancing muscle regeneration, repair, or maintenance.
  • selecting a subject in need of enhancing muscle regeneration, repair, or maintenance comprises diagnosing the subject with a condition characterized by impaired muscle regeneration prior to administering an effective amount of the ⁇ 7 ⁇ 1 integrin modulatory agent to the subject.
  • Methods for diagnosing and selecting a subject in need of muscle regeneration, repair or maintenance are known to those of ordinary skill in the art and include those provided described herein (including those in the Methods of Treatment of Muscular Dystrophy).
  • subjects may be selected based upon their life style (e.g., engaged in moderate to intense exercise or physical activities), age (e.g., elderly population at more risk of experiencing muscle degeneration or injury) or predisposition to muscle degeneration or injury (e.g., genetics or previous muscle injury).
  • life style e.g., engaged in moderate to intense exercise or physical activities
  • age e.g., elderly population at more risk of experiencing muscle degeneration or injury
  • predisposition to muscle degeneration or injury e.g., genetics or previous muscle injury.
  • the method includes administering an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent to the subject wherein the ⁇ 7 ⁇ 1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan ⁇ -ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, a compound provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S.
  • Provisional Pat. App. No. 61/796,476), and/or Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent increases ⁇ 7 ⁇ 1 integrin expression or activity as compared to ⁇ 7 ⁇ 1 integrin expression or activity prior to treatment, thereby prospectively preventing or reducing muscle injury or damage in the subject.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent includes one or more of the following molecules: ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan ⁇ -ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, an analog of Compound ID# 1001, an analog of
  • Compound ID #1002 an analog of Compound ID #1003, an analog of cholestan (see Table 9), laminin-111, laminin-111 fragments, valproic acid, or a valproic acid analog.
  • Tables 8 and 9 and FIGS. 3A-3C, 4A-4B, 5, 6A-6B, and 7-9 provide the chemical structures and characterization data for such compounds.
  • Exemplary valproic acid analogs are disclosed in U.S. Patent Publication 2006/0223888 and International Patent Application No. 2010/080581, each of which is incorporated herein by reference in its entirety.
  • Exemplary laminin-111 fragments are disclosed in U.S. Patent Publication US-2009-0092587-A1, which is incorporated herein by reference in its entirety.
  • an analog of Compound ID #1002 or #1003 is synthesized according to the general synthesis pathway shown in FIG. 10.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is an analog/derivative of any of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents which may be designed and synthesized according to the chemical principles known to one of ordinary skill in the art and identified as a ⁇ 7 ⁇ 1 integrin modulatory agent by methods known to those of ordinary skill in the art, including the muscle cell based assay described Example 1.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent includes one or more molecules provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents can increase the expression of nucleic acid sequences (such as DNA, cDNA, or mRNAs) and proteins of ⁇ 7 ⁇ 1 integrin. An increase in the expression or activity does not need to be 100% for the agent to be effective.
  • an agent can increase the expression or biological activity by a desired amount, for example by at least 10%, for example at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, including about 15% to about 98%, about 30% to about 95%, about 40% to about 80%, about 50% to about 70%, including about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%, as compared to activity or expression in a control.
  • Methods of assessing ⁇ 7 ⁇ 1 integrin expression and activity are known to those of ordinary skill in the art, including those described in the Examples below (e.g. , Western blot and ELISA assay with commercially available antibodies).
  • the method further includes selecting a subject at risk for developing a muscle injury or damage.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered to a subject prior to the subject exercising.
  • the method further includes selecting a subject at risk for developing a muscle injury or damage. Methods for selecting such s subject are known to those of ordinary skill in the art and include those provided described herein. As stated above, subjects may be selected based upon their life style (e.g., engaged in moderate to intense exercise or physical activities), age (elderly population at more risk of experiencing muscle degeneration or injury) or pre-disposition to muscle degeneration or injury (e.g., genetics or previous muscle injury).
  • these methods include contacting a cell with an effective amount of an ⁇ 7 ⁇ 1 integrin modulatory agent, wherein the ⁇ 7 ⁇ 1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a- cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S.
  • the cell is a muscle cell, such as a skeletal muscle cell.
  • the muscle cell is present in a mammal, and wherein contacting the cell with an agent comprises administering the agent to the mammal.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent includes one or more of the following molecules: ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan ⁇ -ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, an analog of Compound ID# 1001, an analog of
  • Compound ID #1002 an analog of Compound ID #1003, an analog of cholestan (see Table 9), laminin-111, laminin-111 fragments, valproic acid, or a valproic acid analog.
  • Tables 8-10 and FIGS. 3A-3C, 4A-4B, 5, 6A-6B, and 7-9 provide the chemical structures and characterization data for such compounds.
  • Exemplary valproic acid analogs are disclosed in U.S. Patent Publication 2006/0223888 and International Patent Application No. 2010/080581, each of which is incorporated herein by reference in its entirety.
  • Exemplary laminin-111 fragments are disclosed in U.S. Patent Publication US-2009-0092587-A1, which is incorporated herein by reference in its entirety.
  • an analog of Compound ID #1002 or #1003 is synthesized according to the general synthesis pathway shown in FIG. 10.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is an analog/derivative of any of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents which may be designed and synthesized according to the chemical principles known to one of ordinary skill in the art and identified as a ⁇ 7 ⁇ 1 integrin modulatory agent by methods known to those of ordinary skill in the art, including the muscle cell based assay described Example 1.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent includes one or more molecules provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents can increase the expression of nucleic acid sequences (such as DNA, cDNA, or mRNAs) and proteins of ⁇ 7 ⁇ 1 integrin. An increase in the expression or activity does not need to be 100% for the agent to be effective.
  • an agent can increase the expression or biological activity by a desired amount, for example by at least 10%, for example at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, including about 15% to about 98%, about 30% to about 95%, about 40% to about 80%, about 50% to about 70%, including about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%, as compared to activity or expression in a control.
  • Methods of assessing ⁇ 7 ⁇ 1 integrin expression and activity are known to those of ordinary skill in the art, including those described in the Examples below (e.g. , Western blot and ELISA assay with commercially available antibodies).
  • an effective amount of ⁇ 7 ⁇ 1 integrin modulatory agent is one when administered by a particular route and concentration induces the desired response (e.g. , treatment of muscular dystrophy, enhancing muscle regeneration, repair or maintenance, preventing or reducing muscle injury or damage, or enhancing ⁇ 7 ⁇ 1 integrin expression).
  • Methods of administration of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents are routine, and can be determined by a skilled clinician.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic substance are in general administered topically, nasally, intravenously, orally, intracranially,
  • ⁇ 7 ⁇ 1 integrin modulatory agents also may be administered to a subject using a combination of these techniques.
  • Suitable solid or liquid pharmaceutical preparation forms are, for example, aerosols,
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic agents of the present disclosure can be formulated into therapeutically-active pharmaceutical agents that can be administered to a subject parenterally or orally.
  • Parenteral administration routes include, but are not limited to epidermal, intraarterial, intramuscular (IM and depot IM), intraperitoneal (IP), intravenous ( ⁇ ), intrasternal injection or infusion techniques, intranasal (inhalation), intrathecal, injection into the stomach, subcutaneous injections (subcutaneous (SQ and depot SQ), transdermal, topical, and ophthalmic.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic agents can be mixed or combined with a suitable pharmaceutically acceptable excipients to prepare pharmaceutical agents.
  • compositions include, but are not limited to, alumina, aluminum stearate, buffers (such as phosphates), glycine, ion exchangers (such as to help control release of charged substances), lecithin, partial glyceride mixtures of saturated vegetable fatty acids, potassium sorbate, serum proteins (such as human serum albumin), sorbic acid, water, salts or electrolytes such as cellulose-based substances, colloidal silica, disodium hydrogen phosphate, magnesium trisilicate, polyacrylates, polyalkylene glycols, such as polyethylene glycol, polyethylene-polyoxypropylene-block polymers, polyvinyl pyrrolidone, potassium hydrogen phosphate, protamine sulfate, group 1 halide salts such as sodium chloride, sodium carboxymethylcellulose, waxes, wool fat, and zinc salts, for example. Liposomal suspensions may also be suitable as pharmaceutically acceptable carriers.
  • the resulting mixture may be a solid, solution, suspension, emulsion, or the like. These may be prepared according to methods known to those of ordinary skill in the art. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the agent in the selected carrier.
  • Pharmaceutical carriers suitable for administration of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic agents include any such carriers known to be suitable for the particular mode of administration.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic substance can also be mixed with other inactive or active materials that do not impair the desired action, or with materials that supplement the desired action, or have another action.
  • Methods for solubilizing may be used where the agents exhibit insufficient solubility in a carrier.
  • Such methods include, but are not limited to, dissolution in aqueous sodium bicarbonate, using cosolvents such as dimethylsulfoxide (DMSO), and using surfactants such as TWEEN® (ICI Americas, Inc., Wilmington, DE).
  • cosolvents such as dimethylsulfoxide (DMSO)
  • surfactants such as TWEEN® (ICI Americas, Inc., Wilmington, DE).
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic agents can be prepared with carriers that protect them against rapid elimination from the body, such as coatings or time-release formulations.
  • Such carriers include controlled release formulations, such as, but not limited to,
  • a disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic agent is included in the pharmaceutically acceptable carrier in an amount sufficient to exert a therapeutically useful effect, typically in an amount to avoid undesired side effects, on the treated subject.
  • therapeutically effective concentration may be determined empirically by testing the compounds in known in vitro and in vivo model systems for the treated condition. For example, mouse models of muscular dystrophy may be used to determine effective amounts or concentrations that can then be translated to other subjects, such as humans, as known in the art.
  • Injectable solutions or suspensions can be formulated, using suitable non-toxic, parenterally- acceptable diluents or solvents, such as 1,3-butanediol, isotonic sodium chloride solution, mannitol, Ringer's solution, saline solution, or water; or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid; a naturally occurring vegetable oil such as coconut oil, cottonseed oil, peanut oil, sesame oil, and the like; glycerine; polyethylene glycol; propylene glycol; or other synthetic solvent; antimicrobial agents such as benzyl alcohol and methyl parabens; antioxidants such as ascorbic acid and sodium bisulfite; buffers such as acetates, citrates, and phosphates; chelating agents such as ethylenediaminetetraacetic acid (EDTA); agents for the adjustment of tonicity
  • Parenteral preparations can be enclosed in ampoules, disposable syringes, or multiple dose vials made of glass, plastic, or other suitable material. Buffers, preservatives, antioxidants, and the like can be incorporated as required.
  • suitable carriers include physiological saline, phosphate-buffered saline (PBS), and solutions containing thickening and solubilizing agents such as glucose, polyethylene glycol, polypropyleneglycol, and mixtures thereof.
  • PBS phosphate-buffered saline
  • Liposomal suspensions, including tissue-targeted liposomes may also be suitable as pharmaceutically acceptable carriers.
  • one or more disclosed ⁇ 7 ⁇ 1 integrin modulatory agents, or other therapeutic agent may be made up into a cream, lotion, ointment, solution, or suspension in a suitable aqueous or nonaqueous carrier. Topical application can also be accomplished by transdermal patches or bandages which include the therapeutic substance. Additives can also be included, e.g., buffers such as sodium
  • metabisulphite or disodium edetate preservatives such as bactericidal and fungicidal agents, including phenyl mercuric acetate or nitrate, benzalkonium chloride, or chlorhexidine; and thickening agents, such as hypromellose.
  • the pharmaceutical agents can be prepared according to techniques well known in the art of pharmaceutical formulation and may contain a suspending agent, such as alginic acid or sodium alginate, bulking agent, such as microcrystalline cellulose, a viscosity enhancer, such as methylcellulose, and sweeteners/flavoring agents.
  • a suspending agent such as alginic acid or sodium alginate
  • bulking agent such as microcrystalline cellulose
  • a viscosity enhancer such as methylcellulose
  • sweeteners/flavoring agents can contain conventional additives such as suspending agents, e.g., gelatin, glucose syrup, hydrogenated edible fats, methyl cellulose, sorbitol, and syrup;
  • emulsifying agents e.g., acacia, lecithin, or sorbitan monooleate
  • non-aqueous carriers including edible oils, e.g., almond oil, fractionated coconut oil, oily esters such as glycerine, propylene glycol, or ethyl alcohol
  • preservatives such as methyl or propyl p-hydroxybenzoate or sorbic acid
  • conventional flavoring or coloring agents e.g., acacia, lecithin, or sorbitan monooleate
  • non-aqueous carriers including edible oils
  • almond oil, fractionated coconut oil, oily esters such as glycerine, propylene glycol, or ethyl alcohol
  • preservatives such as methyl or propyl p-hydroxybenzoate or sorbic acid
  • conventional flavoring or coloring agents e.g., conventional flavoring or coloring agents.
  • these agents can contain dicalcium phosphate, lactose, magnesium stearate, microcrystalline cellulose, and starch and/or other binders, diluents, disintegrants, excipients, extenders, and lubricants.
  • one or more disclosed ⁇ 7 ⁇ 1 integrin modulatory agents, or other therapeutic substances can be provided in a composition that protects it from the acidic environment of the stomach.
  • he disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic agents can be formulated with an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents, or other therapeutic agent can also be formulated in combination with an antacid or other such ingredient.
  • Oral compositions generally include an inert diluent or an edible carrier and can be compressed into tablets or enclosed in gelatin capsules.
  • an inert diluent or an edible carrier for the purpose of oral therapeutic administration, one or more of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents, or other therapeutic substances can be incorporated with excipients and used in the form of capsules, tablets, or troches.
  • Pharmaceutically compatible adjuvant materials or binding agents can be included as part of the composition.
  • the capsules, pills, tablets, troches, and the like can contain any of the following ingredients or compounds of a similar nature: a binder such as, but not limited to, acacia, corn starch, gelatin, gum tragacanth, polyvinylpyrrolidone, or sorbitol; a filler such as calcium phosphate, glycine, lactose, microcrystalline cellulose, or starch; a disintegrating agent such as, but not limited to, alginic acid and corn starch; a lubricant such as, but not limited to, magnesium stearate, polyethylene glycol, silica, or talc; a gildant, such as, but not limited to, colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; disintegrants such as potato starch; dispersing or wetting agents such as sodium lauryl sulfate; and a flavoring agent such as peppermint, methyl salicylate, or fruit flavoring.
  • dosage unit form When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier, such as a fatty oil.
  • dosage unit forms can contain various other materials that modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.
  • One or more of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents, or other therapeutic agent can also be administered as a component of an elixir, suspension, syrup, wafer, tea, chewing gum, or the like.
  • a syrup may contain, in addition to the active compounds, sucrose or glycerin as a sweetening agent and certain preservatives, dyes and colorings, and flavors.
  • the compounds When administered orally, the compounds can be administered in usual dosage forms for oral administration. These dosage forms include the usual solid unit dosage forms of tablets and capsules as well as liquid dosage forms such as solutions, suspensions, and elixirs. When the solid dosage forms are used, they can be of the sustained release type so that the compounds need to be administered less frequently.
  • one or more of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents and/or a therapeutic agent is injected into the stomach of a subject is incorporated systemically in the subject, such as in diverse muscle groups.
  • methods and compositions for administering therapeutic substances which include proteins include those discussed in Banga, Therapeutic Peptides and Proteins: Formulation, Processing, and Delivery Systems 2ed. (2005); Mahato, Biomaterials for Delivery and Targeting of Proteins and Nucleic Acids (2004); McNally, Protein Formulation and Delivery, 2ed. (2007); and Kumar et ah, "Novel Delivery Technologies for Protein and Peptide Therapeutics," Current Pharm. Biotech., 7:261-276 (2006); each of which is incorporated by reference herein to the extent not inconsistent with the present disclosure.
  • the effective amount of one or more of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents is administered as a single dose per time period, such as every three or four months, month, week, or day, or it can be divided into at least two unit dosages for administration over a period. Treatment may be continued as long as necessary to achieve the desired results. For instance, treatment may continue for about 3 or 4 weeks up to about 12-24 months or longer, including ongoing treatment.
  • the compound can also be administered in several doses intermittently, such as every few days (for example, at least about every two, three, four, five, or ten days) or every few weeks (for example at least about every two, three, four, five, or ten weeks).
  • Particular dosage regimens can be tailored to a particular subject, condition to be treated, or desired result.
  • an initial treatment regimen can be applied to arrest the condition.
  • Such initial treatment regimen may include administering a higher dosage of one or more of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents, or administering such material more frequently, such as daily.
  • a second treatment regimen may be applied, such as administering a lower dosage of one or more of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or administering such material less frequently, such as monthly, bi-monthly, quarterly, or semi-annually.
  • the second regimen may serve as a "booster" to restore or maintain a desired level of muscle regeneration.
  • Similar treatment regimens may be used for other subjects with reduced or impaired muscle regeneration capabilities, such as geriatric subjects.
  • the subject is typically treated a sufficient period of time before the exertion or injury in order to provide therapeutic effect.
  • the subject may be treated at least about 24 hours before the expected activity or potential injury, such as at least about 48 hours, about 72 hours, about 1 week, about 2 weeks, about three weeks, or about 4 weeks or more prior.
  • one or more of the disclosed ⁇ 7 ⁇ 1 integrin modulatory agents or other therapeutic substance can be applied directly to, or proximately to, the area to be treated.
  • the substance can be injected into or near the area.
  • the substance can be applied topically to the area to be treated.
  • Treatment is typically initiated prior to the injury to several weeks following the injury. In more specific implementations, the treatment is initiated between about 12 and about 72 hours following injury, such as between about 24 and about 48 hours following injury. In some cases, a single administration of the substance is effective to provide the desired therapeutic effect. In further examples, additional
  • administrations are provided in order to achieve the desired therapeutic effect.
  • Amounts effective for various therapeutic treatments of the present disclosure may, of course, depend on the severity of the disease and the weight and general state of the subject, as well as the absorption, inactivation, and excretion rates of the therapeutically-active compound or component, the dosage schedule, and amount administered, as well as other factors known to those of ordinary skill in the art. It also should be apparent to one of ordinary skill in the art that the exact dosage and frequency of administration will depend on the particular ⁇ 7 ⁇ 1 integrin modulatory agent, or other therapeutic substance being administered, the particular condition being treated, the severity of the condition being treated, the age, weight, general physical condition of the particular subject, and other medication the subject may be taking.
  • dosages used in vitro may provide useful guidance in the amounts useful for in vivo administration of the pharmaceutical composition, and animal models may be used to determine effective dosages for treatment of particular disorders.
  • animal models may be used to determine effective dosages that can then be translated to dosage amount for other subjects, such as humans, as known in the art.
  • dosage determination are described, e.g., in Gilman et ah, eds., Goodman And Gilman's: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press (1990); and Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa. (1990), each of which is herein incorporated by reference to the extent not inconsistent with the present disclosure.
  • the one or more disclosed ⁇ 7 ⁇ 1 integrin modulatory agents is administered to a subject in an amount sufficient to provide a dose of the agent of between about 10 fmol/g and about 500 nmol/g, such as between about 2 nmol/g and about 20 nmol/g or between about 2 nmol/g and about 10 nmol/g.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered to a subject in an amount sufficient to provide a dose of between about 0.01 ⁇ g/kg and about 1000 mg/kg or between about 0.1 mg/kg and about 1000 mg/kg, in particular examples this amount is provided per day or per week.
  • the disclosed ⁇ 7 ⁇ 1 integrin modulatory agent is administered to a subject in an amount sufficient to provide a dose of agent of between about 0.2 mg/kg and about 2 mg/kg.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered to a subject in an amount sufficient to provide a concentration of ⁇ 7 ⁇ 1 integrin modulatory agent in the administrated material of between about 5 nM and about 500 nM, such as between about 50 nM and about 200 nm, or about 100 nM.
  • the ⁇ 7 ⁇ 1 integrin modulatory agent is administered to a subject between about 500 ⁇ g/ml and about 1 ⁇ g/ml, such as about 300 ⁇ g/ml and about 3 ⁇ g/ml, about 200 ⁇ g/ml and about 20 ⁇ g/ml, including 500 ⁇ g/ml, 400 ⁇ g/ml, 300 ⁇ g/ml, 250 ⁇ g/ml, 200 ⁇ g/ml, 150 ⁇ g/ml, 100 ⁇ g/ml, 50 ⁇ g/ml, 25 ⁇ g/ml, 12.5 ⁇ g/ml, 6.25 ⁇ g/ml, 3.125 ⁇ g/ml, 2.5 ⁇ g/ml and 1.25 ⁇ g/ml.
  • Desired Response is provided to a subject between about 500 ⁇ g/ml and about 1 ⁇ g/ml, such as about 300 ⁇ g/ml and about 3 ⁇ g/ml, about 200 ⁇ g/ml and about 20
  • a desired response refers to an amount effective for lessening, ameliorating, eliminating, preventing, or inhibiting at least one symptom of a disease, disorder, or condition treated and may be empirically determined.
  • a desired response is muscle regeneration, reductions or prevention of muscle degeneration, promotion of muscle maintenance, reduction or prevention of muscle injury or damage, reduction or prevention in one more signs or symptoms associated with muscular dystrophy.
  • indicators of muscular health can be assessed through various means, including monitoring markers of muscle regeneration, such as transcription factors such as Pax7, Pax3, MyoD, MRF4, and myogenin. For example, increased expression of such markers can indicate that muscle regeneration is occurring or has recently occurred. Markers of muscle regeneration, such as expression of embryonic myosin heavy chain (eMyHC), can also be used to gauge the extent of muscle regeneration, maintenance, or repair. For example, the presence of eMyHC can indicate that muscle regeneration has recently occurred in a subject.
  • markers of muscle regeneration such as transcription factors such as Pax7, Pax3, MyoD, MRF4, and myogenin.
  • markers of muscle regeneration such as transcription factors such as Pax7, Pax3, MyoD, MRF4, and myogenin.
  • increased expression of such markers can indicate that muscle regeneration is occurring or has recently occurred.
  • Markers of muscle regeneration such as expression of embryonic myosin heavy chain (eMyHC)
  • eMyHC embryonic myosin heavy chain
  • Muscle cell regeneration, maintenance, or repair can also be monitored by determining the girth, or mean cross sectional area, of muscle cells or density of muscle fibers. Additional indicators of muscle condition include muscle weight and muscle protein content. Mitotic index (such as by measuring BrdU incorporation) and myogenesis can also be used to evaluate the extent of muscle regeneration.
  • the improvement in muscle condition, such as regeneration, compared with a control is at least about 10%, such as at least about 30%, or at least about 50% or more, including an at least 15%, at least 20%, at least 25%, at least 30%, at least 40%, at least 45%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, including a 10% to 90% decrease, 20% to 80% increase, 30% to 70% increase or a 40% to 60% increase (e.g., a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100%, 200% or more increase).
  • the subject can receive one or more other therapies.
  • the subject receives one or more treatments prior to administration of a disclosed ⁇ 7 ⁇ 1 modulatory agent.
  • therapies include, but are not limited to, laminin-111 protein therapy, which works to stabilize the sarcolemma and reduce muscle degeneration.
  • a source of muscle cells can be added to aid in muscle regeneration and repair.
  • satellite cells are administered to a subject in combination with laminin therapy.
  • Patent Publication 2006/0014287 provides methods of enriching a collection of cells in myogenic cells and administering those cells to a subject.
  • stem cells such as adipose-derived stem cells
  • Suitable methods of preparing and administering adipose-derived stem cells are disclosed in U.S. Patent Publication 2007/0025972, incorporated by reference herein to the extent not inconsistent with the present disclosure.
  • Additional cellular materials such as fibroblasts, can also be administered, in some examples.
  • ⁇ 7 ⁇ 1 modulatory agents such as a component of the extracellular matrix, such as an integrin, dystrophin, dystroglycan, utrophin, or a growth factor.
  • the additional therapeutic agent reduces or enhances expression of a substance that enhances the formation or maintenance of the extracellular matrix.
  • the additional substance can include aggrecan, angiostatin, cadherins, collagens
  • glycosaminoglycans such as heparin
  • glycoproteins such as dystroglycan
  • proteoglycans such as heparan sulfate
  • growth stimulants such as cytokines, polypeptides, and growth factors such as brain-derived neurotrophic factor (BDNF), CNF (ciliary neurotrophic factor), EGF (epidermal growth factor), FGF (fibroblast growth factor), glial growth factor (GGF), glial maturation factor (GMF) glial - derived neurotrophic factor (GDNF), hepatocyte growth factor (HGF), insulin, insulin-like growth factors, kerotinocyte growth factor (KGF), nerve growth factor (NGF), neurotropin-3 and -4, PDGF (platelet-derived growth factor), vascular endothelial growth factor (VEGF), and combinations thereof may be administered with one of the disclosed methods.
  • BDNF brain-derived neurotrophic factor
  • CNF ciliary neurotrophic factor
  • EGF epidermal growth factor
  • FGF fibroblast growth factor
  • GGF glial growth factor
  • GGF glial maturation factor
  • GGF glial - derived neurotrophic factor
  • Phases I, II, III, and IV Phases I, II, III, and IV.
  • Phase I trials are used to determine the best mode of administration (for example, by pill or by injection), the frequency of administration, and the toxicity for the compounds.
  • Phase I studies frequently include laboratory tests, such as blood tests and biopsies, to evaluate the effects of the potential therapeutic in the body of the patient.
  • a Phase I trial a small group of patients with a muscular disorder are treated with a specific dose of a disclosed ⁇ 7 ⁇ 1 modulatory agent.
  • the dose is typically increased group by group in order to determine the maximum tolerated dose (MTD) and the dose-limiting toxicities (DLT) associated with the compound. This process determines an appropriate dose to use in a subsequent Phase II trial.
  • MTD maximum tolerated dose
  • DLT dose-limiting toxicities
  • a Phase II trial can be conducted to further evaluate the effectiveness and safety of the disclosed ⁇ 7 ⁇ 1 modulatory agent.
  • a disclosed ⁇ 7 ⁇ 1 modulatory agent is administered to groups of patients with a muscular disorder using the dosage found to be effective in Phase I trials.
  • Phase III trials focus on determining how a disclosed ⁇ 7 ⁇ 1 modulatory agent compares to the standard, or most widely accepted, treatment.
  • patients are randomly assigned to one of two or more "arms".
  • one arm will receive the standard treatment (control group) and the other arm will receive a disclosed ⁇ 7 ⁇ 1 modulatory agent treatment (investigational group).
  • Phase ⁇ trials are used to further evaluate the long-term safety and effectiveness of a disclosed ⁇ 7 ⁇ 1 modulatory agent. Phase ⁇ trials are less common than Phase I, II and III trials and take place after a disclosed ⁇ 7 ⁇ 1 modulatory agent has been approved for standard use.
  • Participant eligibility criteria can range from general (for example, age, sex, type of disease) to specific (for example, type and number of prior treatments, disease characteristics, blood cell counts, organ function).
  • eligible patients have been diagnosed with a muscular disorder.
  • Eligibility criteria may also vary with trial phase.
  • Patients eligible for clinical trials can also be chosen based on objective measurement of a muscular disorder and failure to respond to other muscular disorder treatments. For example, in Phase I and II trials, the criteria often exclude patients who may be at risk from the investigational treatment because of abnormal organ function or other factors. In Phase II and III trials additional criteria are often included regarding disease type and stage, and number and type of prior treatments.
  • Phase I trials usually include 15 to 30 participants for whom other treatment options have not been effective.
  • Phase II trials typically include up to 100 participants who have already received drug therapy, but for whom the treatment has not been effective.
  • Phase III trials are often restricted based on the previous treatment received. Phase III trials usually include hundreds to thousands of participants. This large number of participants is necessary in order to determine whether there are true differences between the effectiveness of a disclosed ⁇ 7 ⁇ 1 modulatory agent and the standard treatment. Phase III can include patients ranging from those newly diagnosed with a muscular disorder to those with re-occurring signs and/or symptoms associated with a muscular disorder or a muscular disorder that did not respond to prior treatment.
  • clinical trials should be designed to be as inclusive as possible without making the study population too diverse to determine whether the treatment might be as effective on a more narrowly defined population.
  • the more diverse the population included in the trial the more applicable the results could be to the general population, particularly in Phase III trials. Selection of appropriate participants in each phase of clinical trial is considered to be within the ordinary skills of a worker in the art. Assessment of patients prior to treatment
  • Patients Prior to commencement of the study, several measures known in the art can be used to first classify the patients. Patients can first be assessed, for example by determining serum creatine kinase (CK) levels or other indicators of a muscle disorder, such as increased levels of muscle inflammation, apoptosis, muscle loss, myotube hypertrophy, and/or decreased myofibers stability and cell survival.
  • CK serum creatine kinase
  • a disclosed ⁇ 7 ⁇ 1 modulatory agent is typically administered to the trial participants orally.
  • a range of doses of the agent can be tested. Provided with information from preclinical testing, a skilled practitioner can readily determine appropriate dosages of agent for use in clinical trials.
  • a dose range is from about 100 ⁇ g/kg and about 5000 mg/kg of the subject's weight, such asl mg/kg and about 2000 mg/kg of the subject's weight, about 100 mg/kg and about 1500 mg/kg of the subject's weight, about 100 ⁇ g/kg and about 2000 mg/kg of the subject's weight, about 200 mg/kg and about 1000 mg/kg of the subject's weight, about 200 mg/kg and about 750 mg/kg of the subject's weight, about 250 mg/kg and about 500 mg/kg of the subject's weight, about 100 ⁇ and about 500 mM.
  • subjects are given a disclosed ⁇ 7 ⁇ 1 modulatory agent orally at 10 to 60 mg/kg of body weight per day.
  • 10- 15mg/kg of a disclosed ⁇ 7 ⁇ 1 modulatory agent is administered for two weeks and if well tolerated the dose is increased by 5-lOmg/kg/week to achieve optimal clinical response.
  • the daily dose does not exceed 60mg/kg of body weight and is given for a minimum of 6 months with liver function monitored every two weeks to monthly.
  • ⁇ 7 ⁇ 1 modulatory agent is monitored, for example, by chemical analysis of samples, such as blood, collected at regular intervals. For example, samples can be taken at regular intervals up until about 72 hours after the start of treatment.
  • samples can be placed on dry ice after collection and subsequently transported to a freezer to be stored at -70 °C until analysis can be conducted.
  • Samples can be prepared for analysis using standard techniques known in the art and the amount of the disclosed ⁇ 7 ⁇ 1 modulatory agent present can be determined, for example, by high-performance liquid chromatography (HPLC).
  • HPLC high-performance liquid chromatography
  • Pharmacokinetic data can be generated and analyzed in collaboration with an expert clinical pharmacologist and used to determine, for example, clearance, half-life and maximum plasma concentration.
  • the endpoint of a clinical trial is a measurable outcome that indicates the effectiveness of a compound under evaluation.
  • the endpoint is established prior to the commencement of the trial and will vary depending on the type and phase of the clinical trial.
  • Examples of endpoints include, for example, decline in serum CK levels, inflammation, apoptosis, and muscle loss. For example, at least a 10% reduction in serum CK levels indicates the patient is responsive to the treatment.
  • LacZ reporter gene in a7Pgal + ⁇ muscle cells reports the transcriptional activity of the a7 integrin promoter
  • a l integrin null mouse was produced in which exon 1 of the gene encoding the a 7 integrin was replaced by the LacZ reporter. In these mice, all the transcriptional regulatory elements of the a l integrin promoter were retained, allowing ⁇ -galactosidase to report expression of 7 integrin.
  • a primary myoblast cell line (designated a7Pgal + " ) isolated from 10 day old al +l ⁇ pups were analyzed for the ability of ⁇ - galactosidase to report al integrin expression.
  • a7Pgal + " myoblasts were differentiated and subjected to X-gal staining and western analysis (FIGS. 1 A and IB).
  • the activity of the al integrin promoter was measured by ⁇ -galactosidase cleavage of the non-fluorescent compound fluorescein di- ⁇ -D-galactopyranoside (FDG) to fluorescein.
  • FDG fluorescein di- ⁇ -D-galactopyranoside
  • This example describes multiple compounds identified as ⁇ 7 ⁇ 1 integrin expression enhancers.
  • Example 1 Using the muscle cell based assay described in Example 1 , the following compound libraries were screened: Prestwick Chemical and Microsource Spectrum Libraries from BioFocus DPI (Leiden Netherlands with facilities in UK, Basel, Heidelberg); the DrVERSet library (Chembridge Corp., San Diego, CA) and compounds from the ChemDiv library. Also evaluated was the effect of various isoforms of laminin and the ligand for ⁇ 7 ⁇ 1 integrin on integrin expression. Positive hits were subjected to dose-response analysis, western analysis and a myostatin counterscreen (a negative regulator of muscle growth). To quantify myostatin expression a western blot based assay was utilized (however, an ELISA assay is now available).
  • ⁇ 7 ⁇ gal+/- and C2C12 myotubes were treated with the optimal drug concentration (see Table 8 below) for 24 hours, conditioned media removed and subjected to western analysis using an anti-myostatin antibody (AB3239, Millipore).
  • an anti-myostatin antibody (AB3239, Millipore).
  • cells were treated with 200mM Dexamethasone, which has been shown to increase myostatin expression in C2C12 myotubes. The results indicate that at the ECioo
  • This example demonstrates that intramuscular injection of laminin-111 prevents muscular dystrophy in mdx mice.
  • this example describes studies particular to laminin-111 it is contemplated that similar studies can be performed based upon the methods described herein and the optimal concentrations of the particular ⁇ 7 ⁇ 1 intregin enhancer molecules provided in Table 8 for the other ⁇ 7 ⁇ 1 intregin enhancer molecules and similar effects on muscular dystrophy are predicted.
  • a7Pgal + " myoblasts were exposed to 0- 200nM laminin-111 for 24 hours.
  • the activity of the a7 integrin promoter was measured by ⁇ -galactosidase cleavage of the non-fluorescent compound fluorescein di-P-D-galactopyranoside (FDG) to fluorescein.
  • Fluorescence activated cell sorting FACS
  • a7Pgal + " myoblasts treated for 24 hours with lOOnM laminin-111 produced the maximal increase in a7 integrin promoter activity.
  • laminin-111 The ability of laminin-111 to increase a7 integrin expression was confirmed by western analysis using mouse and human DMD muscle cells (FIGS. 2A-2D). These data indicate that the mechanism by which laminin-111 increases a7 integrin expression is conserved between mouse and human muscle cells and suggests that laminin-111 is highly likely to increase ⁇ 7 ⁇ 1 integrin expression in the skeletal muscle of DMD patients.
  • laminin-111 protein Injection of laminin-111 protein into the mdx mouse model of DMD increased expression of a7 integrin, stabilized the sarcolemma, restored serum creatine kinase to wild-type levels and protected muscle from exercise induced damage.
  • Valproic Acid as a treatment for Muscular Dystrophy
  • This example describes studies indicating the ability of valproic acid to be used to treat muscular dystrophy.
  • Valproic acid is a branched chain fatty acid that is FDA approved for treating epilepsy and bipolar disorders. VPA activates Akt in neurons and promotes their survival is also known to have histone deacetylase (HDAC) inhibitor activity.
  • HDAC histone deacetylase
  • valproic acid activates a7 integrin expression in muscle cells. Valproic acid gave a dose-response curve and increased a7 integrin in C2C12 myotubes (FIGS. 4 A and 4B). Mdx/utr A mice treated with Valproic Acid showed reduced muscle disease, improved mobility, reduced fibrosis and activation of the Akt signaling pathway in muscle. These results indicate that valproic acid is a candidate for the treatment of DMD.
  • This example shows that ciclopirox, 2,2-dipyridyl and deferoxamine increase a7 integrin expression through a common pathway.
  • ciclopirox and deferoxamine are as activators of a7 integrin promoter activity using a7Pgal + " myotubes.
  • Both ciclopirox and deferoxamine are iron chelating drugs.
  • Ciclopirox was independently identified in two compound libraries and is FDA approved as an antibiotic and antifungal drug.
  • Deferoxamine is an FDA approved drug used to treat iron toxicity.
  • Typical dose-response curves were obtained for both ciclopirox and deferoxamine with EC50 of 0 ⁇ g/ml and ⁇ respectively (FIG. 5).
  • a dose response curve for 2,2-dipyridyl also an iron chelating molecule, but is not FDA approved is shown in FIGS. 6A and 6B.
  • MATINSPECTOR (Genomatix) for the presence of HIF-1 binding sites.
  • the consensus DNA sequence for HIF-1 binding in the hypoxia-response element is 5'-[-A/G]CGTG-3' flanked with or without a second consensus site 5'-[A/C]ACAG-3'.
  • Analysis of the a7 integrin promoter sequence revealed the presence of a HIF-1 binding site along with flanking sequences that promote HIF-1 binding.
  • MATINSPECTOR analysis gave these sequences a perfect score for HIF-1 binding.
  • DMD primary myotubes were exposed to the iron chelators 2,2-dipyridyl (31.25 ⁇ ) and deferoxamine (5 and 10 ⁇ ) for 132 hrs to determine if they increased a7 integrin. Protein was extracted from the cells and subjected to western blotting using antibodies against a7 integrin. a-tubulin was used as a loading control. Results showed that both 2,2-dipyridyl and deferoxamine increased al integrin in DMD myotubes (FIG. 7). These results indicate the mechanism(s) by which the iron chelators act to increase integrin expression are conserved between mouse and human muscle cells.
  • This Example demonstrates the ability of cholestan and specific cholestan analogs to increase al integrin expression in myoblasts and myotubes.
  • cholestan as an enhancer of al integrin expression as determined by the muscle cell based assay.
  • Cholestan is a plant-derived compound of unknown function and gave a typical dose-response curve using a7Pgal + " myotubes and increased a7 integrin protein in DMD myotubes (FIG. 8).
  • specific analogs of cholestan retained the ability to activate expression of ⁇ 7 ⁇ 1 integrin (see Table 9 below).
  • One hundred and nineteen analogs of cholestan were obtained from Chemical Diversity laboratories and assessed for their ability to activate the expression of the al integrin in a7betagal+/- myoblasts and myotubes.
  • Four of 119 analogs retained the ability to activate a l integrin expression in myoblasts and myotubes, and an additional 4 of 119 retained the ability to activate a l integrin expression in myoblasts only (Table 9).
  • FIG. 9 The dose-response curves to activate integrin expression by these compounds (as well as the chemical structures of such compounds) are provided in FIG. 9. These studies demonstrate the ability of such compounds to increase al integrin expression in muscle and support their use as agents to regulate al integrin modulated conditions, including muscular dystrophy. It is contemplated that analogs of compounds 1001, 1002 and 1003 could have similar effects. For example, it is contemplated that analogs of compounds 1001, 1002 and 1003 could be synthesized such as by the synthesis pathway provided in FIG. 10 and evaluated by the muscle cell base assay provided in Example 1 to determine their effects on al integrin expression in muscle.
  • Immunofluorescence analysis of both mdx treatment groups demonstrated enhancements of a7A and a7B integrin, and utrophin compared to wild-type animals, and LAM-111 treated mdx mice demonstrated a further increase in a7A and a7B integrin, and utrophin beyond PBS-treated mdx mice (FIGS. 14A-14W).
  • Densitometry of immunoblots from protein extracts of PBS and LAM-111 treated TA demonstrated that both mdx treatment groups showed a statistically significant increase in expression of a7A and a7B integrin and utrophin compared to wild-type animals, and treatment of mdx mice with LAM-111 resulted in a further 100% increase in a7A, a 50% increase in a7B and 33% increase in utrophin beyond PBS-treated mdx mice (FIGS. 15A-15D).
  • VPA is a candidate for the treatment of DMD and existing human safety data may expedite its development for treatment of DMD.
  • LAM-111 localizes to skeletal muscle of dy w mice (FIG. 19-top panels), but required twice weekly doses to improve muscle pathology (FIG. 19-bottom panels) and reduce the percentage of myofibers containing centrally nucleated myofibers (FIG. 20A), Evans blue dye (FIG. 20B), and a TUNEL (apoptosis) reaction (FIG. 20C).
  • FIG. 30 is a digital image illustrating the results of quantitative real-time PCR used to assess Itga7, Itgbl, and Lama2 transcript levels in C2C12 myoblasts and myotubes treated for 24 hours with DMSO control, 10 ⁇ MLS000683232-01 (IED-232), 10 ⁇ MLS001165937-01 (IED-937), Hydroxylpropyl- Beta-Cyclodextrin (HPBCD) control, or 12 ⁇ SU9516 in HPBCD.
  • FIG. 1 is a digital image illustrating the results of quantitative real-time PCR used to assess Itga7, Itgbl, and Lama2 transcript levels in C2C12 myoblasts and myotubes treated for 24 hours with DMSO control, 10 ⁇ MLS000683232-01 (IED-232), 10 ⁇ MLS001165937-01 (IED-937), Hydroxylpropyl- Beta-Cyclodextrin (HPBCD) control, or 12 ⁇ SU95
  • FIG. 31 is a digital image of Western Blots and quantitative analysis of a7 Integrin and GAPDH protein levels in C2C12 myotubes treated for 48 hours with DMSO control, 10 ⁇ MLS000683232-01 (IED-232), Hydroxylpropyl-Beta-Cyclodextrin (HPBCD) control, or 12 ⁇ SU9516 in HPBCD. Bands were quantified using Image J software and then graphed as l Integrin protein levels relative to GAPDH protein levels. * denotes a significant difference in relative protein levels with ** p ⁇ 0.01.
  • This example provides the structures (Table 11 below), exemplary synthesis reactions (FIGS. 29A- 29H) and characterization studies (see FIGS. 21-28) for additional compounds for increasing a l integrin expression in muscle.
  • the analogs were made in 5 mg quantities, salt form (e.g., hydrochloride salt), as a dry powder, at an at least 90% purity as measured by HPLC.
  • salt form e.g., hydrochloride salt
  • Results of screens performed with particular embodiments of the disclosed compounds are provided in FIG. 32.

Abstract

Disclosed herein are α7β1 integrin modulatory agents and methods of using such to treat conditions associated with decreased α7β1 integrin expression or activity, including muscular dystrophy. In one example, methods for treating a subject with muscular dystrophy are disclosed. The methods include administering an effective amount of an α7β1 integrin modulatory agent to the subject with muscular dystrophy, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby treating the subject with muscular dystrophy. Also disclosed are methods of enhancing muscle regeneration, repair, or maintenance in a subject and methods of enhancing α7β1 integrin expression by use of the disclosed α7β1 integrin modulatory agents. Methods of prospectively preventing or reducing muscle injury or damage in a subject are also disclosed.

Description

METHODS OF TREATING MUSCULAR DYSTROPHY
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Patent Application Nos. 61/699,189, filed on September 10, 2012, and 61/798,479, filed on March 15, 2013, and U.S. Patent Application No. 13/842,781 filed on March 15, 2013, each of which is herein incorporated by reference in its entirety.
FIELD
This disclosure relates to the field of muscular dystrophy and in particular, to compositions and methods for treating muscular dystrophy, such as Duchenne muscular dystrophy, Fukuyama congenital muscular dystrophy or merosin deficient congenital muscular dystrophy type 1 A or ID.
ACKNOWLEDGMENT OF GOVERNMENT SUPPORT
This invention was made with government support under Grant Numbers R43 AR060030, R21 NS058429-01, and R21 AR060769 awarded by the National Institutes of Health. The government has certain rights in the invention.
BACKGROUND
Mutations in the a7 integrin gene are responsible for congenital myopathy in man. The α7β 1 integrin is also a major modifier of muscle disease progression in various genetic muscle diseases including various types of muscular dystrophy, such as Duchenne muscular dystrophy (DMD), Fukuyama congenital muscular dystrophy (FCMD) and merosin deficient congenital muscular dystrophy type 1A (MDCIA). However, transcriptional regulation of the l integrin gene, including such role in muscular dystrophy (e.g. , DMD, FCMD and/or MDCIA), remains poorly understood.
Duchenne muscular dystrophy (DMD) is an X-chromosome-linked disease and the most common form of muscular dystrophy. DMD affects 1 in 3500 live male births with patients suffering from chronic muscle degeneration and weakness. Clinical symptoms are first detected between the ages of 2 and 5 years and, by the time the patient is in their teens, the ability for independent ambulation is lost. Death typically occurs in the patient before they are 30 years old due to cardiopulmonary failure.
Fukuyama congenital muscular dystrophy (FCMD) and MDCIA are congential muscular dystrophies that are heritable neuromuscular disorders. MDCIA is characterized by muscle weakness at birth or in infancy. Affected infants will present with poor muscle tone and few movements. The quality of life and life span of the child is affected through progressive muscle wasting, respiratory compromise, and spinal rigidity. MDCIA is the most common and severe form of congenital muscular dystrophy, accounting for 30-40% of all congenital muscular dystrophy (CMD) diagnosed cases. MDCIA is characterized by congenital hypotonia, distinct joint contractures, and a lack of independent ambulation. Feeding tube placement and positive pressure ventilation is often required for the respiratory problems that occur.
Patients afflicted with MDC1 A often die before they reach the age of ten years. FCMD is caused by mutations in the fukutin gene, located at human chromosome 9q31. The disease is inherited in an autosomal recessive manner. FCMD is a type of Limb-Girdle muscular dystrophy. Currently there is no cure for DMD, FCMD or MDC1A.
SUMMARY
The muscular dystrophies are a group of diverse, heritable neuromuscular disorders which represent a group of devastating neuromuscular diseases characterized by primary or secondary skeletal muscle involvement. Currently, there are no cures for such diseases.
Disclosed herein are α7β1 integrin expression modulatory agents and methods of using such to treat a condition associated with impaired a7 integrin expression, such as muscular dystrophy. In one embodiment, a method for treating a subject with muscular dystrophy is disclosed. The method includes administering an effective amount of an α7β1 integrin modulatory agent to the subject with muscular dystrophy, wherein the α7β1 integrin modulatory agent is ciclopirox ethanolamine, deferoxamine, 2,2- dipyridyl; 5a-cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, a compound provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I in U.S. Provisional Patent Application No. 61/798,479, filed on March 15, 2013, which is hereby incorporated by reference in its entirety), Table 7 (see Appendix II in U.S. Provisional Patent Application No. 61/798,479, filed on March 15, 2013, which is hereby incorporated by reference in its entirety), Table 13, or a combination thereof, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby treating the subject with muscular dystrophy (such as MDC1A, MDC1D, LGMD, DMD, FCMD or FHMD).
Also disclosed are methods of enhancing muscle regeneration, repair, or maintenance in a subject. In some embodiments, the method includes administering an effective amount of an α7β1 integrin modulatory agent to the subject in need of muscle regeneration, repair or maintenance, wherein the α7β1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan- 3β-ο1-6-οηε, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, a compound provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby enhancing muscle regeneration, repair or maintenance in a subject.
In a specific embodiment, the present disclosure provides a method for increasing muscle regeneration in a subject. For example, geriatric subjects, subjects suffering from muscle disorders, and subjects suffering from muscle injury, including activity induced muscle injury, such as injury caused by exercise, may benefit from this embodiment.
In yet further embodiments of the disclosed method, the α7β1 integrin modulatory agent is administered in a preventative manner, such as to prevent or reduce muscular damage or injury (such as activity or exercise induced injury). For example, geriatric subjects, subjects prone to muscle damage, or subjects at risk for muscular injury, such as athletes, may be treated in order to eliminate or ameliorate muscular damage, injury, or disease.
Further disclosed are methods of enhancing α7β1 integrin expression. In some embodiments, the method includes contacting a cell with an effective amount of an α7β1 integrin modulatory agent, wherein the α7β1 integrin modulatory agent includes ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a- cholestan-3 -ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, a compound provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof and increases α7β1 integrin expression in the treated cell relative to α7β1 integrin expression in an untreated cell, thereby enhancing α7β1 integrin expression.
The methods of the present disclosure can include administering the α7β1 integrin modulatory agent with one or more additional pharmacological substances, such as a therapeutic agent. In some aspects, the additional therapeutic agent enhances the therapeutic effect of the α7β1 integrin modulatory agent. In further aspects, the therapeutic agent provides independent therapeutic benefit for the condition being treated. In various examples, the additional therapeutic agent is a component of the extracellular matrix, such as an integrin, dystrophin, dystroglycan, utrophin, or a growth factor. In further examples, the therapeutic agent reduces or enhances expression of a substance that enhances the formation or maintenance of the extracellular matrix. In some examples, the therapeutic agent is an additional α7β1 integrin modulatory agent such as laminin-111, a laminin-111 fragment, valproic acid or a valproic acid analog.
In some examples, the α7β1 integrin modulatory agent is applied to a particular area of the subject to be treated. For example, the α7β1 integrin modulatory agent may be injected into a particular area to be treated, such as a muscle. In further examples, the α7β1 integrin modulatory agent is administered such that it is distributed to multiple areas of the subject, such as systemic administration or regional administration.
A α7β1 integrin modulatory agent, can be administered by any suitable method, such as topically, parenterally (such as intravenously or intraperitoneally), or orally. In a specific example, the α7β1 integrin modulatory agent is administered systemically, such as through parenteral administration, such as stomach injection or peritoneal injection.
Although the disclosed methods generally have been described with respect to muscle regeneration, the disclosed methods also may be used to enhance repair or maintenance, or prevent damage to, other tissues and organs. For example, the methods of the present disclosure can be used to treat symptoms of muscular dystrophy stemming from effects to cells or tissue other than skeletal muscle, such as impaired or altered brain function, smooth muscles, or cardiac muscles.
The foregoing and other features of the disclosure will become more apparent from the following detailed description, which proceeds with reference to the accompanying figures.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1A is two digital images of X-gal staining demonstrating that a7Pgal+ " myoblasts express β- galactosidase which increases upon differentiation to myotubes.
FIG. IB is a digital image of a Western analysis of a7Pgal+ " myoblasts differentiated from 0-72 hours shows a corresponding increase in both a7 integrin and β-galactosidase. a-Tubulin was used as a loading control.
FIGS. 2A-2D demonstrate Laminin-111 increases a7 integrin levels in mouse and human muscle cells. (2 A) Western blotting reveals increased levels of α7β integrin in laminin-11 1 treated myoblasts compared to controls. Cox-1 was used as a loading control. (2B) Quantitation shows a two-fold increase in α7β integrin in C2C12 myoblasts treated with laminin-111. (2C) Western blotting reveals increased α7β integrin in laminin-111 treated DMD myoblasts compared to control. Cox-1 was used as a loading control. (2D) Quantitation shows a 2-fold increase in α7β integrin in DMD myoblasts treated with laminin-111.
FIGS. 3A-3C demonstrate that intramuscular injection of laminin-111 prevents muscle disease in mdx mice. (3A) Immunofluorescence of the TA muscles of control and laminin-111 treated mice confirm the absence dystrophin in mdx muscle treated with LAM-111 or PBS. Laminin-111 was not present in wild- type or PBS injected mdx muscle but was detected in the extracellular matrix of laminin-111-injected mdx muscle. Scale bar = ΙΟμπι. (3B) Evans blue dye (EBD) uptake reveals mdx muscle injected with laminin- 111 exhibits reduced EBD uptake compared to control. Scale bar = ΙΟμπι. H&E staining reveals that mdx muscle treated with laminin-111 contains few muscle fibers with centrally located nuclei and mononuclear cell infiltrate compared to control. (3C) Quantitation reveals wild-type and mdx muscle treated with laminin- 111 contained significantly fewer EBD positive fibers and myofibers with centrally located nuclei compared to control. *P<0.05, **P<0.001, n=5 mice/group.
FIGS. 4A and 4B demonstrate Valproic Acid increases a7 integrin expression in muscle cells. (4A) Dose response curve for Valproic Acid using a7Pgal+ " myotubes. (4B) Valproic acid increases a7 integrin protein in C2C12 myotubes.
FIG. 5 shows the chemical structures for Ciclopirox, Deferoxamine and 2,2-dipyridyl.
FIGS. 6A and 6B show dose-response curves for Ciclopirox, Deferoxamine and 2,2-dipyridyl using a7Pgal+ "myotubes and the FDG assay. FIG. 7 shows the iron chelators 2,2-dipyridyl and Deferoxamine increase a7 protein in DMD myotubes. DMD myotubes treated with either 2,2-dipyridyl or deferoxamine showed increased a7 integrin protein as determined by western analysis. N=3 replicates, **P<0.01; *P<0.05.
FIG. 8 shows Cholestan increases a7 integrin promoter activity in mouse and DMD muscle cells. A typical dose response was obtained using Cholestan in a7Pgal+ " myotubes. Treatment of DMD myotubes with cholestan resulted in increased a7 integrin protein compared to control.
FIG. 9 shows Compounds 1001, 1002 and 1003 activate a7 integrin promoter activity. Typical dose response curves showing the fold increase in reporter activity vs drug dose were obtained for compounds 1001, 1002 and 1003 using a7Pgal+ " myotubes.
FIG. 10 is an exemplary synthesis pathway for analogs of compounds 1002 and 1003.
FIG. 11 is a schematic illustrating two examples of muscular dystrophy in which enhanced l integrin is therapeutic. Loss of dystrophin in DMD (A) or glycosylation of a dystroglycan in MDCID (B) results in defective membrane integrity and sarcolemma disruptures. Enhancement of α7β1 integrin improves membrane integrity, minimize sarcolemma ruptures and mitigate the progression of disease in DMD (B) and MDCID (D).
FIGS. 12A-12D are scatter plots from fluorescence-activated cell sorting (FACS) analyses demonstrating that a 24 hour treatment of a7pi-gal +/- myoblasts with lOOnM LAM-111 and a fluorescent β-gal substrate resulted increase a l integrin expression compared to PBS treatment. FACS of a7Pgal+/- myoblasts treated for 24 hours with PBS (A), PBS followed by fluorescent β-gal substrate FDG (Molecular Probes) (B), and lOOnM LAM-111 followed by FDG (C). (D) Peak fluorescence of a7betagal+/- myoblasts treated with PBS (red), PBS+FDG (blue), and LAM-111+FDG (green). Samples were run on the Beckman Coulter XL/MCI flow cytometer and analyzed using FlowJo software. X-axis: FITC fluorescence, (A-C); Y- axis: # of cells, (D); Y-axis: % maximal fluorescence.
FIGS. 13A-13D illustrate western blot studies in which protein extracts from C2C12 (A, B) and DMD (C, D) myoblasts were first treated with PBS or lOOnM LAM-111 and then subjected to western analysis of the a7B integrin and the Cox-1 loading standard. *=p<0.05.
FIGS. 14A-14U are digital images of sections of TA muscle from wild-type (A through G), PBS- treated mdx mice (H through N), and LAM-111 -treated mdx mice (O through U). Detection of dystrophin (A, H, O), LAM-111 (B, I, P), hematoxylin and eosin (C, J, Q), Evans blue dye uptake (D, K, R), al integrin (E, L, S), utrophin (F, M, T), and a-bungarotoxin (G, N, U).
FIGS. 14V and 14W show percentage of Evans blue dye (EBD) positive myofibers (Panel V) and percentage of centrally-located nuclei (CLN) (Panel W) in the TA muscle of wild-type (black bar), PBS- treated mdx (white bar), and LAM-111 treated (gray bar) mdx mice. n=5 mice per group, 1000 fibers were counted per animal, *=p<0.05 , **=p<0.001. FIGS. 15A-15D shows the results of immunoblot detection and quantitation of skeletal muscle protein. Four weeks after one intramuscular injection of the TA muscle with PBS or LAM-111, TA muscles were subjected to western analysis (A), followed by densitometry of a7A (B), a7B (C), and utrophin (D). Densitometry values were normalized to a Cox-1 standard. *=p<0.05.
FIGS. 16A-16K are digital images showing intraperitoneal delivery of LAM-111 distributes throughout mdx skeletal and cardiac muscles. Immunofluorescence detection of LAM-111 in the heart (A, B, C), diaphragm (D, E, F, J, K), and gastrocnemius (G, H, Γ) of wild-type (A, D, G), PBS-treated mdx (B, E, H, J), and LAM-111 treated mdx mice (C, F, I, K). FIGS. 16A-16C: 100X, FIGS. 16D-16I: 63X.
FIGS. 17A-17C includes three bar graphs illustrating the blood chemistry following intraperitoneal delivery of LAM-111. Serum creatine kinase (CK) activity (FIG. 17A), creatine (FIG. 17B) and Blood Urea Nitrogen (BUN) (FIG. 17C) in wild-type (black bars), PBS-treated mdx mice (white bars), and LAM-111- treated mdx mice (gray bars). n=5 mice per group, *=p<0.05 (59).
FIGS. 18A-18C shows- that pretreatment with LAM-111 protects mdx TA from eccentric exercise- induced damage. Uptake of Evans blue dye in the TA of the mdx mice pretreated with PBS (FIG. 18A) or LAM-111 (FIG. 18B) and exercised on a downhill treadmill 4 weeks later. Percentage uptake of Evans blue dye in the TA of the mdx mice pretreated with PBS or LAM-111 and remained sedentary or were exercised on a downhill treadmill 2 weeks later (FIG. 18C). n=4 mice per group. Scale bar = 200 um. **=p<0.001 (59).
FIG. 19 shows that LAM-111 ameliorates muscle disease in the dyW mouse model for MDC1A. Immunofluorescence detection of LAM-111 (top) and H&E staining (bottom) of the skeletal muscle of WT, PBS-treated dyW, and LAM-111 -treated dyW mice. Animals were injected i.p. twice weekly with 1 mg/kg of LAM-111 beginning at 10 days of age. Tissues were harvested at 7 weeks of age. Scale bar= 20uM
FIGS. 20A-20C show that systemic LAM-111 decreases muscle pathology of dyW skeletal muscle Multiple systemic doses of LAM-111 to dyW mice result in a decreased percentage of centrally nucleated myofibers (FIG. 20A), decreased percentage of Evans Blue dye positive myofibers (FIG. 20B), and decreased percentage of TUNEL positive myofibers (FIG. 20C). PBS-treated dyW (black), LAM-111- treated dyW (gray) and WT (white) mice. Animals were injected i.p. twice weekly with 1 mg/kg of LAM- 111 beginning at 10 days of age. Tissues were harvested at 7 weeks of age. *=p<0.05, **=p<0.001.
FIGS. 21-28 are dose response graphs illustrating the effect of disclosed analogs on a7 integrin promoter activity. Typical dose response curves showing the fold increase in reporter activity vs drug dose were obtained for specific analogs using a7Pgal+ " myotubes.
FIGS. 29A-29H illustrate the synthesis of compounds provided in Table 11.
FIG. 30 is a digital image illustrating the results of quantitative real-time PCR used to assess Itga7, Itgbl, and Lama2 transcript levels in C2C12 myoblasts and myotubes treated for 24 hours with DMSO control, 10 μΜ MLS000683232-01 (IED-232), 10 μΜ MLS001165937-01 (IED-937), Hydroxylpropyl- Beta-Cyclodextrin (HPBCD) control, or 12μΜ SU9516 in HPBCD. * denotes a significant difference in relative transcript levels with ** p-value <0.01 and *** p <0.001.
FIG. 31 is a digital image of Western Blots and quantitative analysis of a7 Integrin and GAPDH protein levels in C2C12 myotubes treated for 48 hours with DMSO control, 10 μΜ MLS000683232-01 (IED-232), Hydroxylpropyl-Beta-Cyclodextrin (HPBCD) control, or 12μΜ SU9516 in HPBCD. Bands were quantified using Image J software and then graphed as al Integrin protein levels relative to GAPDH protein levels. * denotes a significant difference in relative protein levels with ** p <0.01.
FIG. 32 is an image of results (fluorescence relative to DMSO at various concentrations of the agent) from a screen using particular embodiments of the disclosed α7β1 integrin modulatory agents.
DETAILED DESCRIPTION OF SEVERAL EMBODIMENTS
/. Overview of Several Embodiments
Disclosed herein are α7β1 integrin expression modulatory agents and methods of using such to treat a condition associated with impaired al integrin expression, such as muscular dystrophy. The present disclosure is related to the subject matter disclosed in U.S. Provisional Patent Application No. 61/533,059, which is incorporated herein by reference in its entirety. Furthermore, the compounds of the present disclosure may be as identified in and have the features described in Appendix I (Table 6), and Appendix II (Table 7) of U.S. Provisional Patent Application No. 61/798,479. The described features are considered to contribute to solving the technical problem underlying the invention of the present application. The described features include, for example, α7β1 integrin modulatory activity suitable for treating a subject with muscular dystrophy, enhancing α7β1 integrin expression, preventing or reducing muscle injury or damage in a subject, enhancing muscle regeneration, repair, or maintenance in a subject, and combinations thereof.
In one embodiment, a method for treating a subject with muscular dystrophy is disclosed. The method includes administering an effective amount of an α7β1 integrin modulatory agent to the subject with muscular dystrophy, wherein the α7β1 integrin modulatory agent is ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan^-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No.
61/796,476), Table 13, or a combination thereof, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby treating the subject with muscular dystrophy.
In some embodiments, a method for treating a subject with muscular dystrophy, comprises administering an effective amount of an α7β1 integrin modulatory agent to the subject with muscular dystrophy, wherein the α7β1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, an agent having a formula selected from
r
Figure imgf000009_0001
wherein each R1 and R2 independently is selected from Ci-ioalkyl, substituted Ci-ioalkyl, Ci-ioalkoxy, substituted Ci-ioalkoxy, acyl, acylamino, acyloxy, acylCi-ioalkyloxy, amino, substituted amino, aminoacyl, aminocarbonylCi-ioalkyl, aminocarbonylamino, aminodicarbonylamino, aminocarbonyloxy, aminosulfonyl, C6-i5aryl, substituted C6-isaryl, C6-i5aryloxy, substituted C6-i5aryloxy, C6-i5arylthio, substituted C6-isarylthio, carboxyl, carboxyester, (carboxyester)amino, (carboxyester)oxy, cyano, C3-8cycloalkyl, substituted C3- 8cycloalkyl, (C3-8cycloalkyl)oxy, substituted (C3-scycloalkyl)oxy, (C3-8cycloalkyl)thio, substituted (C3- 8cycloalkyl)thio, halo, hydroxyl, Ci-ioheteroaryl, substituted Ci-ioheteroaryl, Ci-ioheteroaryloxy, substituted Ci-ioheteroaryloxy, Ci-ioheteroarylthio, substituted Ci-ioheteroarylthio, C2-ioheterocyclyl, C2-iosubstituted heterocyclyl, C2-ioheterocyclyloxy, substituted C2-ioheterocyclyloxy, C2-ioheterocyclylthio, substituted C2- loheterocyclylthio, imino, oxo, sulfonyl, sulfonylamino, thiol, Ci-ioalkylthio, and substituted Ci-ioalkythio, thiocarbonyl; or
two R1 substituents, together with the atom to which each is bound, may form ring selected from a C6-i5aryl, substituted C6-isaryl, C3-8cycloalkyl, substituted C3-8cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C2-iosubstituted heterocyclyl, and C2-ioheterocyclyloxy, substituted;
two R2 substituents, together with the atom to which each is bound, may form ring selected from a C6-i5aryl, substituted C6-isaryl, C3-8cycloalkyl, substituted C3-8cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C2-iosubstituted heterocyclyl, and C2-ioheterocyclyloxy, substituted;
each of A, B, C, D, E, and F independently may be selected from carbon, nitrogen, oxygen, and sulfur; and
n may be zero, 1, 2, 3, 4, or 5; or
a combination of any of these compounds, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby treating the subject with muscular dystrophy. Particular disclosed embodiments concerning one or more of the compounds provided in Tables 10, 11, 6 and/or 7 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476 for Table 6 and Appendix II of U.S. Provisional Pat. App. No. 61/796,476 for Table 7). In some embodiments, the muscular dystrophy is merosin deficient congenital muscular dystrophy Type 1A (MDC1A), merosin deficient congenital muscular dystrophy Type ID (MDC1D), limb -girdle muscular dystrophy (LGMD), Duchenne muscular dystrophy (DMD), Fukuyama congenital muscular dystrophy (FCMD) or Facioscapulohumeral muscular dystrophy (FHMD).
In some particular embodiments, the muscular dystrophy is DMD, MDC1A or FCMD.
In one particular embodiment, the muscular dystrophy is DMD.
In some embodiments, the α7β1 integrin modulatory agent is administered with an additional therapeutic agent.
In some embodiments, the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional α7β1 integrin modulatory agent.
In some embodiments, the additional α7β1 integrin modulatory agent is laminin-111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
In some embodiments, the method further includes selecting a subject with muscular dystrophy.
In some embodiments, the selecting a subject with muscular dystrophy includes diagnosing the subject with muscular dystrophy prior to administering an effective amount of the α7β1 integrin modulatory agent to the subject.
In other embodiments, a method of enhancing muscle regeneration, repair, or maintenance in a subject is disclosed.
In some embodiments, the method includes administering an effective amount of an α7β1 integrin modulatory agent to the subject with muscular dystrophy, wherein the α7β1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan^-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064- 0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), and/or Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby enhancing muscle regeneration, repair or maintenance in a subject.
In some embodiments, the method includes administering the α7β1 modulatory agent prior to the subject experiencing muscle damage or disease.
In some embodiments, the method is a method of enhancing muscle maintenance in a subject.
In some embodiments, the α7β1 integrin modulatory agent is administered to the subject prior to the subject exercising.
In some embodiments, the α7β1 integrin modulatory agent is administered to a subject at risk of acquiring a muscle disease or damage, such as an elderly subject. In some embodiments, the method also includes selecting a subject in need of enhancing muscle regeneration, repair, or maintenance.
In some embodiments, selecting a subject in need of enhancing muscle regeneration, repair, or maintenance includes diagnosing the subject with a condition characterized by impaired muscle regeneration prior to administering an effective amount of the α7β1 integrin modulatory agent to the subject.
In some embodiments, selecting a subject in need of enhancing muscle regeneration, repair, or maintenance comprises diagnosing the subject with a condition characterized by impaired production of a component of α7β1 integrin prior to administering an effective amount of the α7β1 integrin modulatory agent to the subject.
In some embodiments, the α7β1 integrin modulatory agent is administered with an additional therapeutic agent.
In some embodiments, the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional α7β1 integrin modulatory agent.
In some embodiments, the additional α7β1 integrin modulatory agent is laminin-111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
In further embodiments, a method of prospectively preventing or reducing muscle injury or damage in a subject is disclosed.
In some embodiments, the method includes administering an effective amount of an α7β1 integrin modulatory agent to the subject wherein the α7β1 integrin modulatory agent includes ciclopirox
ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan^-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S.
Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby prospectively preventing or reducing muscle injury or damage in the subject.
In some embodiments, the subject is at risk of developing a muscle injury or damage.
In some embodiments, the α7β1 integrin modulatory agent is administered with an additional therapeutic agent.
In some embodiments, the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional α7β1 integrin modulatory agent.
In some embodiments, the additional α7β1 integrin modulatory agent is laminin-111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
In even further embodiments, a method of enhancing α7β1 integrin expression is provided. In some embodiments, the method includes contacting a cell with an effective amount of an α7β1 integrin modulatory agent, wherein the α7β1 integrin modulatory agent includes ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069- 0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof and increases α7β1 integrin expression in the treated cell relative to α7β1 integrin expression in an untreated cell, thereby enhancing α7β1 integrin expression.
In some embodiments, the cell is a muscle cell.
In some embodiments, the muscle cell is present in a mammal, and wherein contacting the cell with an agent comprises administering the agent to the mammal.
//. Terms
The following explanations of terms and methods are provided to better describe the present disclosure and to guide those of ordinary skill in the art in the practice of the present disclosure. The singular terms "a," "an," and "the" include plural referents unless context clearly indicates otherwise.
Similarly, the word "or" is intended to include "and" unless the context clearly indicates otherwise. The term "comprises" means "includes." Thus, "comprising A or B," means "including A, B, or A and B," without excluding additional elements.
It is further to be understood that all base sizes or amino acid sizes, and all molecular weight or molecular mass values, given for nucleic acids or polypeptides are approximate, and are provided for description. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below.
Unless otherwise explained, 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 disclosure belongs. Definitions of common terms in molecular biology may be found in Benjamin Lewin, Genes V, published by Oxford University Press, 1994 (ISBN 0-19-854287-9); Kendrew et al. (eds.), The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182-9); and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1-56081-569-8).
Unless indicated otherwise, the nomenclature of substituents that are not explicitly defined herein are arrived at by naming the terminal portion of the functionality followed by the adjacent functionality toward the point of attachment.
A person of ordinary skill in the art would recognize that the above definitions are not intended to include impermissible substitution patterns {e.g., methyl substituted with 5 different groups, pentavalent carbon, and the like). Such impermissible substitution patterns are easily recognized by a person of ordinary skill in the art.
All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. All sequences provided in the disclosed Genbank Accession numbers are incorporated herein by reference as available on August 11, 2011. In case of conflict, the present specification, including explanations of terms, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
In order to facilitate review of the various embodiments of this disclosure, the following
explanations of specific terms are provided:
Administration: To provide or give a subject one or more agents, such as an agent that increases α7β 1 expression and/or treats one or more symptoms associated with muscular dystrophy, by any effective route. Exemplary routes of administration include, but are not limited to, injection (such as subcutaneous, intramuscular, intradermal, intraperitoneal, and intravenous), oral, sublingual, rectal, transdermal, intranasal, vaginal and inhalation routes.
Agent: Any protein, nucleic acid molecule (including chemically modified nucleic acids), compound, antibody, small molecule, organic compound, inorganic compound, or other molecule of interest. Agent can include a therapeutic agent, a diagnostic agent or a pharmaceutical agent. A therapeutic or pharmaceutical agent is one that alone or together with an additional compound induces the desired response (such as inducing a therapeutic or prophylactic effect when administered to a subject, including treating a subject with a muscular dystrophy).
In some examples, an agent can act directly or indirectly to alter the expression and/or activity of α7β 1. In a particular example, a therapeutic agent significantly increases the expression and/or activity of α7β 1 (which is a muscular dystrophy associated molecule) thereby treating one or more signs or symptoms associated with muscular dystrophy. An example of a therapeutic agent is one that can increase the expression and/or activity of the α7β 1 gene or gene product, for example as measured by a clinical response (such as a decrease in one or more signs or symptoms associated with the muscular dystrophy, an improvement in muscular health, regeneration, repair or maintenance of a muscle cell or tissue).
"Improving muscular health" refers to an improvement in muscular health compared with a preexisting state or compared with a state which would occur in the absence of treatment. For example, improving muscular health may include enhancing muscle regeneration, maintenance, or repair. Improving muscular health may also include prospectively treating a subject to prevent or reduce muscular damage or injury. "Regeneration" refers to the repair of cells or tissue, such as muscle cells or tissue (or organs) which includes muscle cells, following injury or damage to at least partially restore the muscle or tissue to a condition similar to which the cells or tissue existed before the injury or damage occurred. Regeneration also refers to facilitating repair of cells or tissue in a subject having a disease affecting such cells or tissue to eliminate or ameliorate the effects of the disease. In more specific examples, regeneration places the cells or tissue in the same condition or an improved physiological condition as before the injury or damage occurred or the condition which would exist in the absence of disease. "Maintenance" of cells or tissue, such as muscle cells or tissue (or organs) which includes muscle cells, refers to maintaining the cells or tissue in at least substantially the same physiological condition, such as maintaining such condition even in the presence of stimulus which would normally cause damage, injury, or disease. "Repair" of cells or tissue, such as muscle cells or tissue (or organs) which includes muscle cells, refers to the physiological process of healing damage to the cells or tissue following damage or other trauma.
A "pharmaceutical agent" is a chemical compound or composition capable of inducing a desired therapeutic or prophylactic effect when administered to a subject, alone or in combination with another therapeutic agent(s) or pharmaceutically acceptable carriers. In a particular example, a pharmaceutical agent significantly increases the expression and/or activity of α7β 1 thereby treating a condition or disease associated with decreased α7β 1 expression/activity, such as muscular dystrophy.
Acyl: H-C(O)-, alkyl-C(O)-, substituted alkyl-C(O)-, cycloalkyl-C(O)-, substituted cycloalkyl-C(O)-, substituted aryl-C(O)-, heteroaryl-C(O)-, substituted heteroaryl-C(O)-, heterocyclyl-C(O)-, and substituted heterocyclyl-C(O)-.
Acylamino: -NRaC(0)alkyl, -NRaC(0)substituted alkyl, -NRaC(0)cycloalkyl, - NRaC(0)substituted cycloalkyl, -NRaC(0)cycloalkenyl, -NRaC(0)substituted cycloalkenyl, - NRaC(0)alkenyl, -NRaC(0)substituted alkenyl, -NRaC(0)alkynyl, -NRaC(0)substituted alkynyl,
-NRaC(0)aryl, -NRaC(0)substituted aryl, -NRaC(0)heteroaryl, -NRaC(0)substituted heteroaryl,
-NRaC(0)heterocyclyl, and -NRaC(0)substituted heterocyclyl, wherein Ra is selected from hydrogen, alkyl, aryl, and cycloalkyl.
Acyloxy: alkyl-C(0)0-, substituted alkyl-C(0)0-, aryl-C(0)0-, substituted aryl-C(0)0-, cycloalkyl-C(0)0-, substituted cycloalkyl-C(0)0-, heteroaryl-C(0)0-, substituted heteroaryl-C(0)0-, heterocyclyl-C(0)0-, and substituted heterocyclyl-C(0)0-.
Acylalkyloxy: alkyl-C(0)alkylO-, substituted alkyl-C(0)alkylO-, aryl-C(0)alkylO-, substituted aryl-C(0)alkylO-, cycloalkyl-C(0)alkylO-, substituted cycloalkyl-C(0)alkylO-, heteroaryl-C(0)alkylO-, substituted heteroaryl-C(0)alkylO-, heterocyclyl-C(0)alkylO-, and substituted heterocyclyl-C(0)alkylO-.
Alkyl: A saturated or unsaturated monovalent hydrocarbon having a number of carbon atoms ranging from one to ten (e.g., Ci-ioalkyl), which is derived from removing one hydrogen atom from one carbon atom of a parent compound (e.g., alkane, alkene, alkyne). An alkyl group may be branched or straight-chain.
Alkenyl: A unsaturated monovalent hydrocarbon having a number of carbon atoms ranging from one to ten (e.g., C2-ioalkenyl), which has at least one carbon-carbon double bond and is derived from removing one hydrogen atom from one carbon atom of a parent alkene. An alkenyl group may be branched, straight-chain, cyclic, cis, or trans (e.g., E or Z).
Alkynyl: A unsaturated monovalent hydrocarbon having a number of carbon atoms ranging from one to ten (e.g., C2-ioalkynyl), which has at least one carbon-carbon triple bond and is derived from removing one hydrogen atom from one carbon atom of a parent alkyne. An alkynyl group may be branched, straight-chain, or cyclic.
Alkoxy: -O-alkyl (e.g., methoxy, ethoxy, «-propoxy, isopropoxy, «-butoxy, i-butoxy, sec-butoxy, «-pentoxy).
Alkylthio: -S-alkyl, wherein alkyl is as defined herein. This term also encompasses oxidized forms of sulfur, such as -S(0)-alkyl, or -S(0)2-alkyl.
Amino: -NH2.
Aminocarbonyl: -C(0)N(Rb)2, wherein each Rb independently is selected from hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclyl, substituted heterocyclyl. Also, each Rb may optionally be joined together with the nitrogen bound thereto to form a heterocyclyl or substituted heterocyclyl group, provided that both Rb are not both hydrogen.
Aminocarbonylalkyl: -alkylC(0)N(Rb)2, wherein each Rb independently is selected from hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclyl, substituted heterocyclyl. Also, each Rb may optionally be joined together with the nitrogen bound thereto to form a heterocyclyl or substituted heterocyclyl group, provided that both Rb are not both hydrogen.
Aminocarbonylamino: -NRaC(0)N(Rb)2, wherein Ra and each Rb are as defined herein.
Aminodicarbonylamino: -NRaC(0)C(0)N(Rb)2, wherein Ra and each Rb are as defined herein.
Aminocarbonyloxy: -0-C(0)N(Rb)2, wherein each Rb independently is as defined herein.
Aminosulfonyl: -S02N(Rb)2, wherein each Rb independently is as defined herein.
Analog or Derivative: A compound which is sufficiently homologous to a compound such that it has a similar functional activity for a desired purpose as the original compound. Analogs or derivatives refers to a form of a substance, such as cholestan, which has at least one functional group altered, added, or removed, compared with the parent compound. In some examples, examples of an analog are provided in Table 11, for example. "Functional group" refers to a radical, other than a hydrocarbon radical, that adds a physical or chemical property to a substance.
Aryl: a monovalent aromatic carbocyclic group of from 6 to 15 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl), which condensed rings may or may not be aromatic provided that the point of attachment is through an atom of the aromatic aryl group.
Aryloxy -O-aryl.
Arylthio -S-aryl, wherein aryl is as defined herein. This term also encompasses oxidized forms of sulfur, such as -S(0)-aryl, or -S(0)2-aryl.
Biological activity: The beneficial or adverse effects of an agent on living matter. When the agent is a complex chemical mixture, this activity is exerted by the substance's active ingredient or pharmacophore, but can be modified by the other constituents. Activity is generally dosage-dependent and it is not uncommon to have effects ranging from beneficial to adverse for one substance when going from low to high doses. In one example, the agent significantly increases the biological activity of α7β 1 which reduces one or more signs or symptoms associated with the muscular dystrophy.
Contacting: Placement in direct physical association, including both a solid and liquid form.
Contacting an agent with a cell can occur in vitro by adding the agent to isolated cells or in vivo by administering the agent to a subject.
Control: A sample or standard used for comparison with a test sample, such as a biological sample obtained from a patient (or plurality of patients) without a particular disease or condition, such as a muscular dystrophy. In some embodiments, the control is a sample obtained from a healthy patient (or plurality of patients) (also referred to herein as a "normal" control), such as a normal biological sample. In some embodiments, the control is a historical control or standard value {e.g., a previously tested control sample or group of samples that represent baseline or normal values (e.g., expression values), such as baseline or normal values of a particular gene such as a α7β 1 gene, gene product in a subject without a muscular dystrophy). In some examples, the control is a standard value representing the average value (or average range of values) obtained from a plurality of patient samples (such as an average value or range of values of the gene or gene products, such as the α7β 1 gene or gene products, in the subjects without a muscular dystrophy).
Carboxyl: -COOH or salts thereof.
Carboxyester: -C(0)0-alkyl, -C(0)0- substituted alkyl, -C(0)0-aryl, -C(0)0-substituted aryl, -C(0)0-cycloalkyl, -C(0)0-substituted cycloalkyl, -C(0)0-heteroaryl, -C(0)0-substituted heteroaryl, -C(0)0-heterocyclyl, and -C(0)0-substituted heterocyclyl.
(Carboxyester)amino: -NRa-C(0)0-alkyl, -NRa-C(0)0- substituted alkyl, -NRa-C(0)0-aryl, -NRa-C(0)0-substituted aryl, -NRa-C(0)0-cycloalkyl, -NRa-C(0)0-substituted cycloalkyl, - NRa-C(0)0-heteroaryl, -NRa-C(0)0-substituted heteroaryl, -NRa-C(0)0-heterocyclyl, and - NRa-C(0)0-substituted heterocyclyl, wherein Ra is as recited herein.
(Carboxyester)oxy: -0-C(0)0-alkyl, -0-C(0)0- substituted alkyl, -0-C(0)0-aryl, - 0-C(0)0-substituted aryl, -0-C(0)0-cycloalkyl, -0-C(0)0-substituted cycloalkyl, -0-C(0)0-heteroaryl, -0-C(0)0-substituted heteroaryl, -0-C(0)0-heterocyclyl, and -0-C(0)0-substituted heterocyclyl.
Cyano: -CN.
Cycloalkyl: cyclic alkyl (or alkenyl, or alkynyl) groups of from 3 to 10 carbon atoms having single or multiple cyclic rings including fused, bridged, and spiro ring systems (e.g., cyclopropyl, cyclobutyl, etc.).
(Cycloalkyl)oxy: -O-cycloalkyl.
(Cycloalkyl)thio: -S-cycloalkyl. This term also encompasses oxidized forms of sulfur, such as - S(0)-cycloalkyl, or -S(0)2-cycloalkyl. Decrease: To reduce the quality, amount, or strength of something. In one example, a therapy decreases one or more symptoms associated with the muscular dystrophy, for example as compared to the response in the absence of the therapy.
Diagnosis: The process of identifying a disease, such as muscular dystrophy, by its signs, symptoms and results of various tests. The conclusion reached through that process is also called "a diagnosis." Forms of testing commonly performed include blood tests, medical imaging, urinalysis, and biopsy.
Effective amount: An amount of agent that is sufficient to generate a desired response, such as reducing or inhibiting one or more signs or symptoms associated with a condition or disease. When administered to a subject, a dosage will generally be used that will achieve target tissue/cell concentrations. In some examples, an "effective amount" is one that treats one or more symptoms and/or underlying causes of any of a disorder or disease. In some examples, an "effective amount" is a therapeutically effective amount in which the agent alone with an additional therapeutic agent(s) (for example anti-pathogenic agents), induces the desired response such as treatment of a muscular dystrophy, such as DMD, FCMD or MDC1A.
In particular examples, it is an amount of an agent capable of increasing α7β 1 gene expression or activity by least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100% (elimination of the disease to a point beyond detection).
In some examples, an effective amount is an amount of a pharmaceutical preparation that alone, or together with a pharmaceutically acceptable carrier or one or more additional therapeutic agents, induces the desired response.
In one example, a desired response is to increase the subject's survival time by slowing the progression of the disease, such as slowing the progression of muscular dystrophy. The disease does not need to be completely inhibited for the pharmaceutical preparation to be effective. For example, a pharmaceutical preparation can decrease the progression of the disease by a desired amount, for example by at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, as compared to the progression typical in the absence of the pharmaceutical preparation.
In another or additional example, it is an amount sufficient to partially or completely alleviate symptoms of the muscular dystrophy within the subject. Treatment can involve only slowing the progression of the disease temporarily, but can also include halting or reversing the progression of the disease permanently.
Effective amounts of the agents described herein can be determined in many different ways, such as assaying for a reduction in of one or more signs or symptoms associated with the muscular dystrophy in the subject or measuring the expression level of one or more molecules known to be associated with the muscular dystrophy. Effective amounts also can be determined through various in vitro, in vivo or in situ assays, including the assays described herein.
The disclosed therapeutic agents can be administered in a single dose, or in several doses, for example daily, during a course of treatment. However, the effective amount can be dependent on the source applied (for example a nucleic acid molecule isolated from a cellular extract versus a chemically synthesized and purified nucleic acid), the subject being treated, the severity and type of the condition being treated, and the manner of administration.
Expression: The process by which the coded information of a gene is converted into an operational, non-operational, or structural part of a cell, such as the synthesis of a protein. Gene expression can be influenced by external signals. For instance, exposure of a cell to a hormone may stimulate expression of a hormone-induced gene. Different types of cells can respond differently to an identical signal. Expression of a gene also can be regulated anywhere in the pathway from DNA to RNA to protein. Regulation can include controls on transcription, translation, RNA transport and processing, degradation of intermediary molecules such as mRNA, or through activation, inactivation, compartmentalization or degradation of specific protein molecules after they are produced. In an example, expression, such as expression of α7β 1 , can be regulated to treat one or more signs or symptoms associated with muscular dystrophy.
The expression of a nucleic acid molecule can be altered relative to a normal (wild type) nucleic acid molecule. Alterations in gene expression, such as differential expression, include but are not limited to: (1) overexpression; (2) underexpression; or (3) suppression of expression. Alternations in the expression of a nucleic acid molecule can be associated with, and in fact cause, a change in expression of the
corresponding protein.
Protein expression can also be altered in some manner to be different from the expression of the protein in a normal (wild type) situation. This includes but is not necessarily limited to: (1) a mutation in the protein such that one or more of the amino acid residues is different; (2) a short deletion or addition of one or a few (such as no more than 10-20) amino acid residues to the sequence of the protein; (3) a longer deletion or addition of amino acid residues (such as at least 20 residues), such that an entire protein domain or sub-domain is removed or added; (4) expression of an increased amount of the protein compared to a control or standard amount; (5) expression of a decreased amount of the protein compared to a control or standard amount; (6) alteration of the subcellular localization or targeting of the protein; (7) alteration of the temporally regulated expression of the protein (such that the protein is expressed when it normally would not be, or alternatively is not expressed when it normally would be); (8) alteration in stability of a protein through increased longevity in the time that the protein remains localized in a cell; and (9) alteration of the localized (such as organ or tissue specific or subcellular localization) expression of the protein (such that the protein is not expressed where it would normally be expressed or is expressed where it normally would not be expressed), each compared to a control or standard. Controls or standards for comparison to a sample, for the determination of differential expression, include samples believed to be normal (in that they are not altered for the desired characteristic, for example a sample from a subject who does not have muscular dystrophy, such as DMD, FCMD or MDC1A) as well as laboratory values (e.g. , range of values), even though possibly arbitrarily set, keeping in mind that such values can vary from laboratory to laboratory.
Laboratory standards and values can be set based on a known or determined population value and can be supplied in the format of a graph or table that permits comparison of measured, experimentally determined values.
Extracellular matrix: An extracellular structure of a tissue or a layer thereof, including the arrangement, composition, and forms of one or more matrix components, such as proteins, including structural proteins such as collagen and elastin, proteins such as fibronectin and laminins, and proteoglycans. The matrix may comprise fibrillic collagen, having a network of fibers. In some examples, the extracellular matrix is connected to cells through the costameric protein network.
Halogen or Halo: fluoro, chloro, bromo, and iodo.
Heteroaryl: an aromatic group of from 1 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur within the ring. Such heteroaryl groups can have a single ring (e.g., pyridinyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl), wherein the condensed rings may or may not be aromatic and/or contain a heteroatom, provided that the point of attachment is through an atom of the aromatic heteroaryl group. In one embodiment, the nitrogen and/or sulfur ring atom(s) of the heteroaryl group are optionally oxidized to provide for the N-oxide (N→0), sulfinyl, or sulfonyl moieties.
Heteroaryloxy: -O-heteroaryl.
Heteroarylthio: -S-heteroaryl. This term also encompasses oxidized forms of sulfur, such as - S(0)-heteroaryl, or -S(0)2-heteoaryl.
Heterocyclyl: a saturated, unsaturated group, or combinations thereof, having a single ring or multiple condensed rings, including fused bridged and spiro ring systems, and having from 3 to 15 ring atoms, including 1 to 4 heteroatoms, selected from nitrogen, sulfur, or oxygen. These groups may be substituted with one or more of the substituents disclosed herein for substituted aryl and/or substituted alkyl. These groups encompass, for example, a saturated heterocyclyl fused with one or more aromatic hydrocarbons or heteroaryl groups.
Heterocyclyloxy: -O-heterocycyl.
Heterocyclylthio: -S-heterocycyl. This term also encompasses oxidized forms of sulfur, such as - S(0)-heterocyclyl, or -S(0)2-heterocyclyl.
Hydroxyl or Hydroxy: -OH.
Imino: -N=RC wherein Rc may be selected from hydrogen, aminocarbonylalkyloxy, substituted aminocarbonylalkyloxy, aminocarbonylalkylamino, and substituted aminocarbonylalkylamino.
Increase: To enhance the quality, amount, or strength of something. In one example, an agent increases the activity or expression of α7β 1 , for example relative to an absence of the agent. In a particular example, an agent increases the activity or expression of α7β 1 by at least 10%, at least 20%, at least 50%, or even at least 90%, including between 10% to 95%, 20% to 80%, 30% to 70%, 40% to 50%, such as 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 98%, or 100%. Such increases can be measured using the methods disclosed herein.
In a particular example, a therapy increases (also known as up-regulates) the expression of α7β 1 , such as an increase of at least 10%, at least 20%, at least 50%, or even at least 90% in α7β 1 expression, thereby treating/alleviating one or more signs or symptoms associated with muscular dystrophy. In some examples, an increase in expression refers to an increase in a α7β 1 gene product. An α7β 1 gene product can be RNA (such as mRNA, rRNA, tRNA, and structural RNA) or protein.
Gene upregulation includes any detectable increase in the production of a α7β 1 gene product. In certain examples, production of a α7β 1 gene product increases by at least 2-fold, for example at least 3-fold or at least 4-fold, as compared to a control (such an amount of gene expression in a normal cell). In one example, a control is a relative amount of a7 gene expression or protein expression in a biological sample taken from a subject who does not have muscular dystrophy, such as DMD, FCMD or MDC1A. Such increases can be measured using the methods disclosed herein. For example, "detecting or measuring expression of α7β1" includes quantifying the amount of the gene, gene product or modulator thereof present in a sample. Quantification can be either numerical or relative. Detecting expression of the gene, gene product or modulators thereof can be achieved using any method known in the art or described herein, such as by measuring nucleic acids by PCR (such as RT-PCR) and proteins by ELISA. In primary embodiments, the change detected is an increase or decrease in expression as compared to a control, such as a reference value or a healthy control subject. In some examples, the detected increase or decrease is an increase or decrease of at least two-fold compared with the control or standard. Controls or standards for comparison to a sample, for the determination of differential expression, include samples believed to be normal (in that they are not altered for the desired characteristic, for example a sample from a subject who does not have muscular dystrophy, such as DMD, FCMD or MDC1A) as well as laboratory values (e.g., range of values), even though possibly arbitrarily set, keeping in mind that such values can vary from laboratory to laboratory.
Laboratory standards and values can be set based on a known or determined population value and can be supplied in the format of a graph or table that permits comparison of measured, experimentally determined values.
In other embodiments of the methods, the increase or decrease is of a diagnostically significant amount, which refers to a change of a sufficient magnitude to provide a statistical probability of the diagnosis. The level of expression in either a qualitative or quantitative manner can detect nucleic acid or protein. Exemplary methods include microarray analysis, RT-PCR, Northern blot, Western blot, and mass spectrometry.
Inhibiting a disease or condition: A phrase referring to reducing the development of a disease or condition, for example, in a subject who is at risk for a disease or who has a particular disease. Particular methods of the present disclosure provide methods for inhibiting muscular dystrophy. "
Integrin: A cell surface transmembrane glycoprotein receptor. Integrins are involved in many biological processes such as wound healing, blood clot formation, gene regulation, and immune responses. Integrins can regulate tissue specific cell adhesion molecules. Integrins are heterodimeric non-covalently associated glycoproteins composed of two subunits. The subunits, which are designated a and beta, have approximate molecular weights of 150-180 kilodaltons and 90-110 kilodaltons, respectively.
The α7β1 integrin is a major laminin receptor expressed in skeletal muscle. The α7β1 integrin plays a role in the development of neuromuscular and myotendinous junctions. In the adult, the α7β1 integrin is concentrated at junctional sites and found in extrajunctional regions where it mediates the adhesion of the muscle fibers to the extracellular matrix. Mice that lack the l chain develop muscular dystrophy that affects the myotendinous junctions. The absence of al integrin results in defective matrix deposition at the myotendinous junction. Loss of the al integrin in γ-sarcoglycan mice results in severe muscle pathology. Absence of the al integrin in mdx mice also results in severe muscular dystrophy, confirming that the α7β1 integrin serves as a major genetic modifier for Duchenne and other muscular dystrophies.
Mutations in the al gene are responsible for muscular dystrophy in humans. A screen of 117 muscle biopsies from patients with undefined muscle disease revealed 3 which lacked the al integrin chain and had reduced levels of βΐϋ integrin chain. These patients exhibited delayed developmental milestones and impaired mobility consistent with the role for the α7β1 integrin in neuromuscular and myotendinous junction development and function.
Several lines of evidence suggest the al integrin may be important for muscle regeneration. For example, during embryonic development, the α7β1 integrin regulates myoblast migration to regions of myofiber formation. It has been found that MyoD (myogenic determination protein) transactivates al integrin gene expression in vitro, which would increase al integrin levels in activated satellite cells.
Human, mouse and rat myoblast cell lines derived from satellite cells express high levels of al integrin. Elevated al integrin rnRNA and protein are detected in the skeletal muscle of 5 week old mdx mice, which correlates with the period of maximum muscle degeneration and regeneration. In addition, the α7β1 integrin associates with muscle specific βΐ -integrin binding protein (MIBP), which regulates laminin deposition in C2C12 myoblasts. Laminin provides an environment that supports myoblast migration and proliferation. Finally, enhanced expression of the al integrin in dystrophic skeletal muscle results in increased numbers of satellite cells. The sequences for α7β1 integrin subunits are publicly available on GenBank, see, for example Gene Accession No. NM_001144116 (human) and NM_008398.2 (mouse) for a l integrin, and Gene Accession No. NM_002211 for βΐ integrin (also known as CD29), each of which is herein incorporated by reference as available on September 8, 2011. Exemplary α7β1 integrin modulatory agents are disclosed herein, such as in Tables 8, 9 and 10, including ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl, 5 a-cholestan-3p-ol- 6-one, Compound ID#1001, Compound ID# 1002, Compound ID# 1003, N032-0003, N066-0070, N069- 0071, N069-0075, 1080-0573 , N064-0028, N066-0053, or N069-0073 or analogs provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof.
A α7β1 integrin -associated condition is a condition associated with altered α7β1 integrin expression or activity, including muscular dystrophy, such as DMD, FCMD, LGMD, FHMD, Beckers muscular dystrophy and/or MDC1A.
Laminin: Any of the family of glycoproteins that are typically involved in the formation and maintenance of extracellular matrices. Laminin is a heterotrimers formed from an a chain, a β chain, and a γ chain. The various chains of a particular laminin can affect the properties of the molecule. In some aspects of the present disclosure, fragments, derivatives, or analogs of various laminins can be used, such as laminins having at least a portion at least substantially homologous to the laminin al chain. A "fragment of laminin," as used herein, refers to a portion of a substance, such as laminin. A fragment may be, in some examples, a particular domain or chain of a protein. For example, particular embodiments of the present disclosure involve administering a fragment of laminin- 1 corresponding to at least a portion of (or all of) the laminin al chain. Fragments may be synthetic or may be derived from larger parent substances.
In some aspects, laminins may be administered as a mixture of laminins, including fragments, analogs, and derivatives thereof. Suitable methods for preparing analogs of laminin domains are disclosed in U.S. Patent No. 6,933,280, incorporated by reference herein to the extent not inconsistent with this disclosure.
The laminin materials or compositions of the present disclosure may be delivered as discrete molecules or may be complexed with, or conjugated to, another substance. For example, the laminin may be combined with a carrier, such as to aid in delivery of the laminin to a site of interest or to increase physiological uptake or incorporation of the laminin.
In specific examples, the laminin administered includes or consists of laminin- 1 (LAM-111), which includes the chains αΐβΐγΐ . In further examples, the laminin administered includes or consists of laminin-2, which includes the chains α2β1γ1. In yet further examples, the laminin administered includes or consists of laminin-4, which includes the chains α2β2γ1.
Laminins may be obtained from any suitable source. For example, laminin- 1 may be obtained from placental tissue or from Engelbreth-Holm-Swarm murine sarcoma. Suitable methods of isolating various laminins are disclosed in U.S. Patent No. 5,444,158, incorporated by reference herein to the extent not inconsistent with the present disclosure.
Muscle: Any myoblast, myocyte, myofiber, myotube or other structure composed of muscle cells. Muscles or myocytes can be skeletal, smooth, or cardiac. Muscle may also refer to, in particular implementations of the present disclosure, cells or other materials capable of forming myocytes, such as stem cells and satellite cells.
Muscular dystrophy: A term used to refer to a group of genetic disorders that lead to progressive muscle weakness. Muscular dystrophy can result in skeletal muscle weakness and defects in skeletal muscle proteins, leading to a variety of impaired physiological functions. No satisfactory treatment of muscular dystrophy exists. Existing treatments typically focus on ameliorating the effects of the disease and improving the patient's quality of life, such as through physical therapy or through the provision of orthopedic devices.
Mutated genes associated with muscular dystrophy are responsible for encoding a number of proteins associated with the costameric protein network. Such proteins include laminin-2, collagen, dystroglycan, integrins, caveolin-3, ankyrin, dystrophin, a-dystrobrevin, vinculin, plectin, BPAGlb, muscle LIM protein, desmin, actinin-associated LIM protein, a-actin, titin, telethonin, cypher, myotilin, and the sarcoglycan/sarcospan complex.
The most common form of muscular dystrophy is DMD, affecting 1 in 3,500 live male births. DMD is an X-linked recessive disorder characterized by a mutation in the gene that codes for dystrophin. Dystrophin is a cytoskeletal protein about 430 kDa in size. This protein works to connect the cell's cytoskeleton and extracellular matrix. The loss of dystrophin in DMD patients leads to a loss of muscle fiber attachment at the extracellular matrix during contraction, which ultimately leads to progressive fiber damage, membrane leakage and a loss of muscle function. Most patients die before they reach the age of 30 due to respiratory or cardiac failure.
Beckers muscular dystrophy (also known as Benign pseudohypertrophic muscular dystrophy) is related to DMD in that both result from a mutation in the dystrophin gene, but in DMD no functional dystrophin is produced making DMD much more severe than BMD. BMD is an X-linked recessive inherited disorder characterized by slowly progressive muscle weakness of the legs and pelvis. BMD is a type of dystrophinopathy, which includes a spectrum of muscle diseases in which there is insufficient dystrophin produced in the muscle cells, results in instability in the structure of muscle cell membrane. This is caused by mutations in the dystrophin gene, which encodes the protein dystrophin. The pattern of symptom development of BMD is similar to DMD, but with a later, and much slower rate of progression.
Congenital muscular dystrophies are caused by gene mutations. FCMD and MDC1A are examples of congenital muscular dystrophies. MDC1A is a congential muscular dystrophy due to a genetic mutation in the LAMA2 gene which results in lack of or complete loss of laminin-a2 protein. This loss of laminin-a2 leads to an absence of laminins-211/221. Laminins-211/221 are major components of the extracellular matrix and play a key role in muscle cell development. During muscle cell differentiation laminin binds to the α7β 1 integrin. Without laminin-a2, muscle fibers are unable to adhere to the basement membrane and myotubes undergo apotosis. Muscle regeneration also fails, leading to a loss of muscle repair and an increase in muscle fibrosis and inflammation. This chronic tissue injury is a major cause of morbidity and mortality in MDC1A.
Congenital Muscular Dystrophies (CMD) and Limb-Girdle muscular dystrophy (LGMD) are common forms of highly heterogeneous muscular dystrophies which can be distinguished by their age at onset. In CMD, onset of symptoms is at birth or within the first 6 months of life; in LGMD onset of symptoms is in late childhood, adolescence or even adult life. Inheritance in LGMD can be autosomal dominant (LGMD type 1) or autosomal recessive (LGMD type 2), CMD is recessively inherited. CMD and LGMD can overlap both clinically and genetically
MDC1A is a progressive muscle wasting disease that results in children being confined to a wheelchair, requiring ventilator assistance to breathe and premature death. Symptoms are detected at birth with poor muscle tone and "floppy" baby syndrome. DMD, BMD and LGMD are progressive muscle degenerative diseases usually diagnosed at 3-5 years of age when children show developmental delay including ability to walk and climb stairs. The disease is progressive and children are usually confined to a wheelchair in their teens and require ventilator assistance.
Fukuyama congenital muscular dystrophy (FCMD) is an inherited condition that predominantly affects the muscles, brain, and eyes. Congenital muscular dystrophies are a group of genetic conditions that cause muscle weakness and wasting (atrophy) beginning very early in life. Fukuyama congenital muscular dystrophy affects the skeletal muscles, which are muscles the body uses for movement. The first signs of the disorder appear in early infancy and include a weak cry, poor feeding, and weak muscle tone
(hypotonia). Weakness of the facial muscles often leads to a distinctive facial appearance including droopy eyelids (ptosis) and an open mouth. In childhood, muscle weakness and joint deformities (contractures) restrict movement and interfere with the development of motor skills such as sitting, standing, and walking. Fukuyama congenital muscular dystrophy also impairs brain development. People with this condition have a brain abnormality called cobblestone lissencephaly, in which the surface of the brain develops a bumpy, irregular appearance (like that of cobblestones). These changes in the structure of the brain lead to significantly delayed development of speech and motor skills and moderate to severe intellectual disability. Social skills are less severely impaired. Most children with Fukuyama congenital muscular dystrophy are never able to stand or walk, although some can sit without support and slide across the floor in a seated position. More than half of all affected children also experience seizures. Other signs and symptoms of Fukuyama congenital muscular dystrophy include impaired vision, other eye abnormalities, and slowly progressive heart problems after age 10. As the disease progresses, affected people may develop swallowing difficulties that can lead to a bacterial lung infection called aspiration pneumonia. Because of the serious medical problems associated with Fukuyama congenital muscular dystrophy, most people with the disorder live only into late childhood or adolescence.
Fukuyama congenital muscular dystrophy is seen almost exclusively in Japan, where it is the second most common form of childhood muscular dystrophy (after Duchenne muscular dystrophy). Fukuyama congenital muscular dystrophy has an estimated incidence of 2 to 4 per 100,000 Japanese infants.
Fukuyama congenital muscular dystrophy is caused by mutations in the FKTN gene which encodes fukutin. The most common mutation in the FKTN gene reduces the amount of fukutin produced within cells. A shortage of fukutin likely prevents the normal modification of a-dystroglycan, which disrupts that protein's normal function. Without functional a-dystroglycan to stabilize muscle cells, muscle fibers become damaged as they repeatedly contract and relax with use. The damaged fibers weaken and die over time, leading to progressive weakness and atrophy of the skeletal muscles.
Defective a-dystroglycan also affects the migration of neurons during the early development of the brain. Instead of stopping when they reach their intended destinations, some neurons migrate past the surface of the brain into the fluid-filled space that surrounds it. Because Fukuyama congenital muscular dystrophy involves a malfunction of a-dystroglycan, this condition is described as a dystroglycanopathy.
Facioscapulohumeral muscular dystrophy (FHMD) is a form of muscular dystrophy associated with progressive muscle weakness and loss of muscle tissue. Unlike DMD and BMD which mainly affect the lower body, FSHDaffects the upper body mainly the face, shoulder and upper arm muscles. However, it can affect muscles around the pelvis, hips, and lower leg. Symptoms for FSHDoften do not appear until age 10 - 26, but it is not uncommon for symptoms to appear much later. In some cases, symptoms never develop. Symptoms are usually mild and very slowly become worse. Facial muscle weakness is common, and may include eyelid drooping, inability to whistle, decreased facial expression, depressed or angry facial expression, difficulty pronouncing words, shoulder muscle weakness (leading to deformities such as pronounced shoulder blades (scapular winging) and sloping shoulders), weakness of the lower, hearing loss and possible heart conditions.
Oxo: (=0).
Pharmaceutically acceptable carriers: The pharmaceutically acceptable carriers (vehicles) useful in this disclosure are conventional. Remington's Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, PA, 19th Edition (1995), describes compositions and formulations suitable for pharmaceutical delivery of one or more agents, such as one or more α7β1 modulatory agents.
In general, the nature of the carrier will depend on the particular mode of administration being employed. For instance, parenteral formulations can include injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle. In addition to biologically-neutral carriers, pharmaceutical agents to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate, sodium lactate, potassium chloride, calcium chloride, and triethanolamine oleate.
Prodrug: A compound that is transformed in vivo to yield a parent compound, for example, by hydrolysis in the gut or enzymatic conversion in blood.
Sample (or biological sample): A biological specimen containing genomic DNA, RNA (including mRNA), protein, or combinations thereof, obtained from a subject. Examples include, but are not limited to, peripheral blood, urine, saliva, tissue biopsy, surgical specimen, and autopsy material. In one example, a sample includes muscle biopsy, such as from a subject with DMD, FCMD, or MDC1A.
Signs or symptoms: Any subjective evidence of disease or of a subject's condition, e.g., such evidence as perceived by the subject; a noticeable change in a subject's condition indicative of some bodily or mental state. A "sign" is any abnormality indicative of disease, discoverable on examination or assessment of a subject. A sign is generally an objective indication of disease. Signs include, but are not limited to any measurable parameters such as tests for detecting muscular dystrophy, including measuring creatine kinase levels, electromyography (to determine if weakness is caused by destruction of muscle tissue rather than by damage to nerves) or immunohistochemistry/immunoblotting/immunoassay (e.g., ELISA) to measure muscular dystrophy-associated molecules, such as α7β1 integrin. In one example, reducing or inhibiting one or more symptoms or signs associated with muscular dystrophy, includes increasing the activity or expression of α7β1 integrin by a desired amount, for example by at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, as compared to the activity and/or expression in the absence of the treatment. Symptoms of muscular dystrophy include, but are not limited to, muscle weakness and loss, difficulty running, difficulty hopping, difficulty jumping, difficulty walking, difficulty breathing, fatigue, skeletal deformities, muscle deformities (contractions of heels; pseudohypertrophy of calf muscles), heart disease (such as dilated cardiomyopathy), elevated creatine phosphokinase (CK) levels in blood or combinations thereof.
Subject: Living multi-cellular vertebrate organisms, a category that includes human and non- human mammals.
Substituted Alkyl: an alkyl (or alkenyl, or alkynyl) group having from 1 to 5 hydrogen atoms replaced with substituents selected alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, acylalkyloxy, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aminodicarbonylamino, aminocarbonylalkyl, aminosulfonyl, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, aminodiacylamino, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy, heteroarylthio, substituted heteroarylthio, heterocyclyl, substituted heterocyclyl, heterocyclyloxy, substituted heterocyclyloxy, heterocyclylthio, substituted heterocyclylthio, imino, oxo, sulfonylamino, nitro, SO3H, sulfonyl, thiol, imino, substituted imino, alkylthio, and substituted alkylthio. The alkyl may be substituted with 1 to 2, 1 to 3, or 1 to 4 of these groups, which are defined herein.
Substituted Alkoxy: -0-(substituted alkyl).
Substituted Alkylthio: -S-(substituted alkyl). This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted alkyl, or -S(0)2-substituted alkyl.
Substituted Amino: -N(Rb)2, wherein each Rb independently is selected from hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclyl, substituted heterocyclyl. Also, each Rb may optionally be joined together with the nitrogen bound thereto to form a heterocyclyl or substituted heterocyclyl group, provided that both Rb are not both hydrogen.
Substituted Aryl: aryl groups having 1 to 5 hydrogens replaced with substituents independently selected from alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aminosulfonyl, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy, heteroarylthio, substituted heteroarylthio, heterocyclyl, substituted heterocyclyl, heterocyclyloxy, substituted heterocyclyloxy, heterocyclylthio, substituted heterocyclylthio, nitro, SO3H, sulfonyl, thiol, alkylthio, and substituted alkylthio. The aryl group may be substituted with 1 to 2, 1 to 3, or 1 to 4 of these groups, which are defined herein.
Substituted Aryloxy: -0-(substituted aryl).
Substituted Arylthio: -S-(substituted aryl), wherein substituted aryl is as defined herein. This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted aryl, or -S(0)2-substituted aryl.
Substituted Cycloalkyl: cycloalkyl ,cycloalkenyl, or cycloalkynyl group having from 1 to 5 substituents selected from the group consisting of oxo, alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aminosulfonyl, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy, heteroarylthio, substituted heteroarylthio, heterocyclyl, substituted heterocyclyl, heterocyclyloxy, substituted heterocyclyloxy, heterocyclylthio, substituted heterocyclylthio, nitro, SO3H, sulfonyl, thiol, alkylthio, and substituted alkylthio. The aryl group may be substituted with 1 to 2, 1 to 3, or 1 to 4 of these groups, which are defined herein. In some embodiments, the cycloalkyl group may have multiple condensed rings (e.g. tetrahydronaphthyl or tetrahydroanthacenyl), provided that the point of attachment is through an atom of the nonaromatic ring.
Substituted (Cycloalkyl)oxy: -0-(substituted cycloalkyl).
Substituted (Cycloalkyl)thio: refers to -S-(substituted cycloalkyl). This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted cycloalkyl, or -S(0)2-substituted cycloalkyl.
Substituted Heteroaryl: heteroaryl groups that are substituted with from 1 to 5 substituents selected from the group consisting of the same group of substituents defined for substituted aryl.
Substituted Heteroaryloxy: -0-(substituted heteroaryl).
Substituted Heteroarylthio: -S-(substituted heteroaryl). This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted heteroaryl, or -S(0)2-substituted heteoaryl.
Substituted Heterocycyloxy: -0-(substituted heterocyclyl) wherein the heterocyclyl group is substituted with one or more of the substituents recited for substituted alkyl.
Substituted Heterocycylthio: -S-(substituted heterocycyl). This term also encompasses oxidized forms of sulfur, such as -S(0)-substituted heterocyclyl, or -S(0)2-substituted heterocyclyl.
Sulfonyl: -S02-alkyl, -SC -substituted alkyl, -S02-cycloalkyl, -S02-substituted cycloalkyl, -SO2- aryl, -S02-substituted aryl, -S02-heteroaryl, -SC -substituted heteroaryl, -S02-heterocyclyl, and -S02-substituted heterocyclyl.
Sulfonylamino: -NRaS02alkyl, -NRaS02substituted alkyl, -NRaS02cycloalkyl, - NRaS02substituted cycloalkyl, -NRaS02aryl, -NRaS02substituted aryl, -NRaS02heteroaryl, - NRaS02Substituted heteroaryl, -NRaS02heterocyclyl, -NRaS02Substituted heterocyclyl, wherein each Ra independently is as defined herein.
Thiol: -SH.
Thiocarbonyl: (=S)
Tissue: An aggregate of cells, usually of a particular kind, together with their intercellular substance that form one of the structural materials of an animal and that in animals include connective tissue, epithelium, muscle tissue, and nerve tissue.
Treating a disease: A therapeutic intervention that ameliorates a sign or symptom of a disease or pathological condition related to a muscular dystrophy, such as a sign or symptom of muscular dystrophy. Treatment can induce remission or cure of a condition or slow progression, for example, in some instances can include inhibiting the full development of a disease, for example preventing development of a muscular dystrophy. Prevention of a disease does not require a total absence of disease. For example, a decrease of at least 50% can be sufficient.
Treating a disease can be a reduction in severity of some or all clinical symptoms of the disease or condition, a reduction in the number of relapses of the disease or condition, an improvement in the overall health or well-being of the subject, by other parameters well known in the art that are specific to the particular disease or condition, and combinations of such factors.
Under conditions sufficient for: A phrase that is used to describe any environment that permits the desired activity. In one example, includes administering a disclosed agent to a subject sufficient to allow the desired activity. In particular examples, the desired activity is increasing the expression or activity of α7β1. ///. Compounds for Treating Muscular Dystrophy
Disclosed herein are compounds that may be used as α1β7 integrin modulatory agents in methods disclosed herein. In particular disclosed embodiments, the compound is effective in treating muscular dystrophy. The compound is a small-molecule therapeutic. In particular disclosed embodiments, the small- molecule therapeutic is a cyclic compound comprising a heteroatom-containing skeleton. In other disclosed embodiments, the small-molecule therapeutic is a cyclic compound comprising an all-carbon skeleton. In certain disclosed embodiments, the cyclic compound comprising a heteroatom-containing skeleton has a formula illustrated below:
Figure imgf000029_0001
Formula 1
wherein each R1 independently is selected from Ci-ioalkyl, substituted Ci-ioalkyl, Ci-ioalkoxy, substituted Ci. loalkoxy, acyl, acylamino, acyloxy, acylCi-ioalkyloxy, amino, substituted amino, aminoacyl,
aminocarbonylCi-ioalkyl, aminocarbonylamino, aminodicarbonylamino, aminocarbonyloxy, aminosulfonyl, C6-i5aryl, substituted C6-isaryl, C6-i5aryloxy, substituted C6-i5aryloxy, C6-i5arylthio, substituted C6-isarylthio, carboxyl, carboxyester, (carboxyester)amino, (carboxyester)oxy, cyano, C3-8cycloalkyl, substituted C3- 8cycloalkyl, (C3-8cycloalkyl)oxy, substituted (C3-scycloalkyl)oxy, (C3-8cycloalkyl)thio, substituted (C3- 8cycloalkyl)thio, halo, hydroxyl, Ci-ioheteroaryl, substituted Ci-ioheteroaryl, Ci-ioheteroaryloxy, substituted Ci-ioheteroaryloxy, Ci-ioheteroarylthio, substituted Ci-ioheteroarylthio, C2-ioheterocyclyl, C2-iosubstituted heterocyclyl, C2-ioheterocyclyloxy, substituted C2-ioheterocyclyloxy, C2-ioheterocyclylthio, substituted C2- loheterocyclylthio, imino, oxo, sulfonyl, sulfonylamino, thiol, Ci-ioalkylthio, and substituted Ci-ioalkythio, thiocarbonyl; or
two R1 substituents, together with the atom to which each is bound, may form ring selected from a C6-i5aryl, substituted C6-isaryl, C3-8cycloalkyl, substituted C3-8cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C2-iosubstituted heterocyclyl, and C2-ioheterocyclyloxy, substituted;
each of A, B, C, D, and E independently may be selected from carbon, nitrogen, oxygen, and sulfur; and
n may be zero, 1, 2, 3, 4, or 5.
In other embodiments, the cyclic compound comprising a heteroatom-containing moiety has a formula illustrated below:
Figure imgf000029_0002
Formula 2 wherein each R2 independently is selected from Ci-ioalkyl, substituted Ci-ioalkyl, Ci-ioalkoxy, substituted Ci. loalkoxy, acyl, acylamino, acyloxy, acylCi-ioalkyloxy, amino, substituted amino, aminoacyl,
aminocarbonylCi-ioalkyl, aminocarbonylamino, aminodicarbonylamino, aminocarbonyloxy, aminosulfonyl, C6-i5aryl, substituted C6-isaryl, C6-i5aryloxy, substituted C6-i5aryloxy, C6-i5arylthio, substituted C6-isarylthio, carboxyl, carboxyester, (carboxyester)amino, (carboxyester)oxy, cyano, C3-8cycloalkyl, substituted C3- 8cycloalkyl, (C3-8cycloalkyl)oxy, substituted (C3-scycloalkyl)oxy, (C3-scycloalkyl)thio, substituted (C3- 8cycloalkyl)thio, halo, hydroxyl, Ci-ioheteroaryl, substituted Ci-ioheteroaryl, Ci-ioheteroaryloxy, substituted Ci-ioheteroaryloxy, Ci-ioheteroarylthio, substituted Ci-ioheteroarylthio, C2-ioheterocyclyl, C2-iosubstituted heterocyclyl, C2-ioheterocyclyloxy, substituted C2-ioheterocyclyloxy, C2-ioheterocyclylthio, substituted C2- loheterocyclylthio, imino, oxo, sulfonyl, sulfonylamino, thiol, Ci-ioalkylthio, and substituted Ci-ioalkythio, thiocarbonyl; or
two R2 substituents, together with the atom to which each is bound, may form ring selected from a C6-i5aryl, substituted C6-isaryl, C3-8cycloalkyl, substituted C3-8cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C2-iosubstituted heterocyclyl, and C2-ioheterocyclyloxy, substituted;
each of A, B, C, D, E, and F independently may be selected from carbon, nitrogen, oxygen, and sulfur; and
n may be zero, 1, 2, 3, 4, or 5.
In particular disclosed embodiments, the cyclic compound comprising an all-carbon skeleton may have a general formula provided below:
Figure imgf000030_0001
Formula 3
wherein R3 and R4 independently may be selected from hydroxyl, hydrogen, Ci-ioalkyl, substituted Ci- ioalkyl, carboxyl, acyl, aminoacyl, acylamino, amino, substituted amino, C6-i5aryl, substituted C6-isaryl, and Ci-ioalkoxy; R5 is selected from amino, substituted amino, oxo, hydroxyl, Ci-ioalkoxy, and imino; and n may be zero, 1, 2, 3, 4, or 5. A person of ordinary skill in the art will recognize that the dashed lines indicate optional bonds which may be present in certain compounds and not present in others.
In particular disclosed embodiments, rings A and B are connected via the optional bonds to form a steroid-based skeleton. In embodiments wherein rings A and B are connected, R5 may be bound to ring A via a double bond or a single bond, a feature that is indicated with the optional dashed bond in Formula 13. For example, if R5 is amino, hydroxyl, substituted amino, or Ci-ioalkoxy, then R5 is attached to ring A via a single bond, whereas if R5 is oxo or imino, then R5 is attached to ring A via a double bond. In particular disclosed embodiments, C6-i5aryl may be selected from phenyl, biphenyl, naphthalene, anthracene, and the like; substituted C6-i5aryl may be selected from phenyl, biphenyl, naphthalene, and anthracene substituted with one or more substituents as defined herein; Ci-ioalkyl may be selected from Ci. loalkane, C2-ioalkene, and C2-ioalkyne; more typically from methyl, ethyl, propyl, butyl, pentyl, hexyl, and the like; ethylene, propylene, butylene, and the like; and ethyne, propyne, butyne, and the like; substituted Ci-ioalkyl may be selected from Ci-ioalkane, C2-ioalkene, and C2-ioalkyne substituted with one or of the substituents as provided herein.
Exemplary embodiments concerning hetercyclyl and heteroaryl substitutents include, but are not limited to, epoxy, pyrrolyl, imidazole, pyrazole, pyridinyl, pyrazine, pyrimidine, oxanyl, thianyl, dioxanyl, dithianyl, coumarin, pyridazine, indolizine, isoindole, indolyl, indolinyl (or dihydroindole), indazole, purine, isoquinoline, quinoline, benzo[d]pyridazine, naphthyridine, quinoxaline, quinazoline,
benzopyridazine, pteridine, carbazole, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,7-tetrahydrobenzo[b]thiophene, thiazole, thiazolidine, thiophene, benzo[b]thiophene, morpholinyl, thiomorpholinyl, piperidinyl, pyrrolidinyl, piperazinyl, oxazolidinyl, oxazolyl, thiophenyl, isooxazolidinyl, and tetrahydrofuranyl.
Exemplary substituents wherein at least two R1 groups have been joined together include the following:
Figure imgf000031_0001
Particular disclosed embodiments concern cyclic compounds comprising a five-membered heteroatom-containing skeleton having a formula selected from those provided below.
Figure imgf000031_0002
Formula 4
Figure imgf000032_0001
Formula 5
Figure imgf000032_0002
Formula 6
With reference to Formulas 4-6, R1 and n are as recited herein, and each X independently may be selected from carbon, oxygen, nitrogen, and sulfur.
In yet other embodiments, the cyclic compound comprising a five-membered heteroatom-containing skeleton may have any one of the following formulas
Figure imgf000032_0003
Formula 7
Figure imgf000032_0004
Formula 8
Figure imgf000032_0005
Formula 9
Figure imgf000032_0006
Formula 10
Figure imgf000032_0007
Formula 11
Figure imgf000033_0001
32
Figure imgf000034_0001
Figure imgf000035_0001
Particular embodiments concern cyclic compounds comprising a six-membered heteroatom- containing skeleton having any one of the formulas provided below:
Figure imgf000035_0002
Formula 12
Figure imgf000035_0003
Formula 13
Figure imgf000035_0004
Formula 14
wherein R2 and n are as recited herein, Z may be selected from carbon and nitrogen, Y may be selected from nitrogen and oxygen, and each X independently may be selected from nitrogen and carbon. A person of ordinary skill in the art will recognize that the dashed lines indicate variable bonds which may or may not be present, depending on the valency of the atom to which each variable bond is attached. For example, if the variable bond indicated in Formula 11 is present, X typically is carbon, as a carbon atom can accommodate four bonds. X may be nitrogen in such a compound; however, a person of ordinary skill in the art would recognize that the nitrogen atom would be positively charged due to the fact that its lone pairs are used to accommodate a fourth bond.
Exemplary compounds are provided below solely as illustrative examples.
Figure imgf000036_0001
Figure imgf000037_0001
Formula 15
An exemplary compound is provided below.
Figure imgf000037_0002
In other disclosed embodiments of the compound comprising an all-carbon skeleton, ring A is not connected with ring B and exists as an aryl compound having a formula illustrated below.
Figure imgf000037_0003
Formula 16
Exemplary compounds are illustrated below.
Figure imgf000037_0004
In yet other embodiments, the compound may have any one of the following formulas
Figure imgf000037_0005
Formula 17
Figure imgf000038_0001
Formula 19
Figure imgf000038_0002
Formula 21
wherein each Y1 and Y2 independently may be selected from oxygen, sulfur, and NRb wherein Rb is as disclosed herein; and R6 is selected from those substituents provided for R1. Any of the compounds disclosed herein may also be used in an alternate chemical form, such as a pharmaceutically acceptable salt, a N-oxide, a prodrug, and/or a solvate. In particular disclosed embodiments, the compound may be a pharmaceutically acceptable salt, such as a hydrochloride salt, a hydrobromide salt, a hydroiodide salt, a nitrate salt, a sulfate salt, a bisulfate salt, a phosphate salt, an acid phosphate salt, an isonicotinate salt, an acetate salt, a lactate salt, a salicylate salt, a citrate salt, a tartrate salt, an ascorbate salt, a succinate salt, a maleate salt, a fumarate salt, a gluconate salt, a saccharate salt, a formate salt, a benzoate salt, a glutamate salt, a methanesulfonate salt, an ethanesulfonate salt, a benzensulfonate salt, a />-toluenesulfonate salt, and combinations thereof. Exemplary compounds are provided below in Tables 1-6A.
Table 1
Figure imgf000038_0003
R1 R1' R2 Y1
C(0)NH2 NH2 N[(CH2)o-5CH3]2 S Table 1
R1 R1' R2 Y1
C(0)N[(CH2)o-5CH3]2 NH2 N[(CH2)o-5CH3]2 S
C(0)OH NH2 N[(CH2)o-5CH3]2 S
C(0)OP(0)(OH)2 NH2 N[(CH2)o-5CH3]2 S
C(0)OC(0)C(CH3)3 NH2 N[(CH2)o-5CH3]2 S
C(0)OC(0)NMe2 NH2 N[(CH2)o-5CH3]2 S
C(0)0(CH2)o-5CH3 NH2 N[(CH2)o-5CH3]2 S
C(0)NH2 NH2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)N[(CH2)o-5CH3]2 NH2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)OH NH2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)OP(0)(OH)2 NH2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)OC(0)C(CH3)3 NH2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)OC(0)NMe2 NH2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)0(CH2)o-5CH3 NH2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)NH2 NH2 N[(CH2)o-5CH3]2 0
C(0)N[(CH2)o-5CH3]2 NH2 N[(CH2)o-5CH3]2 0
C(0)OH NH2 N[(CH2)o-5CH3]2 0
C(0)OP(0)(OH)2 NH2 N[(CH2)o-5CH3]2 0
C(0)OC(0)C(CH3)3 NH2 N[(CH2)o-5CH3]2 0 Table 1
R1 R1' R2 Y1
C(0)OC(0)NMe2 NH2 N[(CH2)o-5CH3]2 0
C(0)0(CH2)o-5CH3 NH2 N[(CH2)o-5CH3]2 0
C(0)NH2 NH2 N[(CH2)o-5CH3]2 NH
C(O)N[(CH2)0-5CH3]2 NH2 N[(CH2)o-5CH3]2 NH
C(0)OH NH2 N[(CH2)o-5CH3]2 NH
C(0)OP(0)(OH)2 NH2 N[(CH2)o-5CH3]2 NH
C(0)OC(0)C(CH3)3 NH2 N[(CH2)o-5CH3]2 NH
C(0)OC(0)NMe2 NH2 N[(CH2)o-5CH3]2 NH
C(0)0(CH2)o-5CH3 NH2 N[(CH2)o-5CH3]2 NH
C(0)NH2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 S
C(0)N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 S
C(0)OH N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 s
C(0)OP(0)(OH)2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 s
C(0)OC(0)C(CH3)3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 s
C(0)OC(0)NMe2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 s
C(0)0(CH2)o-5CH3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 s
C(0)NH2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 0
C(0)N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 0 Table 1
R1 R1' R2 Y1
C(0)OH N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 0
C(0)OP(0)(OH)2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 0
C(0)OC(0)C(CH3)3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 0
C(0)OC(0)NMe2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 0
C(0)0(CH2)o-5CH3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 0
C(0)NH2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)OH N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)OP(0)(OH)2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)OC(0)C(CH3)3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)OC(0)NMe2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)0(CH2)o-5CH3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N(CH2)o-5CH3
C(0)NH2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NH
C(0)N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NH
C(0)OH N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NH
C(0)OP(0)(OH)2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NH
C(0)OC(0)C(CH3)3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NH
C(0)OC(0)NMe2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NH Table 1
R1 R1' R2 Y1
C(0)0(CH2)o-5CH3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NH
C(0)NH2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NCH2OP(0)(OH)2
C(0)N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NCH2OP(0)(OH)2
C(0)OH N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NCH2OP(0)(OH)2
C(0)0(CH2)o-5CH3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NCH2OP(0)(OH)2
C(0)OP(0)(OH)2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NCH2OP(0)(OH)2
C(0)OC(0)C(CH3)3 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NCH2OP(0)(OH)2
C(0)OC(0)NMe2 N[(CH2)o-5CH3]2 N[(CH2)o-5CH3]2 NCH2OP(0)(OH)2
Figure imgf000042_0001
Figure imgf000043_0001
Table 2
Figure imgf000044_0001
R1 R2 Y1
C(0)NH(Ph)/?CH2SH (CH2)o-5CH3 NH
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SH (CH2)o-5CH3 NH
C(0)NH(Ph)/?CH2SMe (CH2)o-5CH3 NH
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SMe (CH2)o-5CH3 NH
C(O)N[(CH2)0-5CH3](Ph)/?CH2NH2 (CH2)o-5CH3 NH
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2NH2 (CH2)o-5CH3 NH
C(0)N[(CH2y5CH3](Ph)/?CH2OH (CH2)o-5CH3 NH
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OH (CH2)o-5CH3 NH
C(0)N[(CH2)o-5CH3](Ph)^CH2N[(CH2)o-5CH3]2 (CH2)o-5CH3 NH
C(O)N[CH2OP(O)(OH)2](Ph)^CH2N[(CH2)0-5CH3]2 (CH2)o-5CH3 NH
C(0)N[(CH2)o-5CH3](Ph)/?CH2OMe (CH2)o-5CH3 NH
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OMe (CH2)o-5CH3 NH
C(O)N[(CH2)0-5CH3](Ph)/?CH2SH (CH2)o-5CH3 NH
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SH (CH2)o-5CH3 NH
C(0)N[(CH2)o-5CH3](Ph)/?CH2SMe (CH2)o-5CH3 NH
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SMe (CH2)o-5CH3 NH
C(0)NH(Ph)/?CH2NHC(0)Oi-Bu (CH2)o-5CH3 0
Figure imgf000045_0001
Figure imgf000046_0001
Table 2
Figure imgf000047_0001
R1 R2 Y1
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SH (CH2)o-5CH3 0
C(O)N[(CH2)0-5CH3](Ph)/?CH2SMe (CH2)o-5CH3 0
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SMe (CH2)o-5CH3 0
C(0)NH(Ph)/?CH2NHC(0)Oi-Bu (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2NHC(0)Oi-Bu (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[CH2OP(0)(OH)2]C(0)Oi-Bu (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2N[CH2OP(0)(OH)2]C(0)Oi-Bu (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2NHC(0)OH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2NHC(0)OH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2NHC(0)OMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2NHC(0)OMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[CH2OP(0)(OH)2]C(0)OMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(O)N[(CH2)0-5CH3](Ph)/?CH2NHC(O)Oi-Bu (CH2)o-5CH3 NCH2OP(0)(OH)2
C(O)N[(CH2)0-5CH3](Ph)^CH2N[CH2OP(O)(OH)2]C(O)Oi-Bu (CH2)o-5CH3 NCH2OP(0)(OH)2
C(O)N[(CH2)0-5CH3](Ph)/?CH2NHC(O)OH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[(CH2)o-5CH3](Ph)^CH2N[CH2OP(0)(OH)2]C(0)OH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(O)N[(CH2)0-5CH3](Ph)^CH2NHC(O)OMe (CH2)o-5CH3 NCH2OP(0)(OH)2 Table 2
Figure imgf000048_0001
R1 R2 Y1
C(0)N[(CH2)o-5CH3](Ph)^CH2N[CH2OP(0)(OH)2]C(0)OMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2NH2 (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2NH2 (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2OH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(O)NH(Ph)/?CH2N[(CH2)0-5CH3]2 (CH2)o-5CH3 NCH2OP(0)(OH)2
C(O)N[CH2OP(O)(OH)2](Ph)^CH2N[(CH2)0-5CH3]2 (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2OMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2SH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2SMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(O)N[(CH2)0-5CH3](Ph)/?CH2NH2 (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2NH2 (CH2)o-5CH3 NCH2OP(0)(OH)2
C(O)N[(CH2)0-5CH3](Ph)/?CH2OH (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OH (CH2)o-5CH3 NCH2OP(0)(OH)2 Table 2
Figure imgf000049_0001
R1 R2 Y1
C(0)N[(CH2)o-5CH3](Ph)^CH2N[(CH2)o-5CH3]2 (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH20P(0)(OH)2](Ph)^CH2N[(CH2)o-5CH3]2 (CH2)o-5CH3 NCH2OP(0)(OH)2
(CH2)o-5CH3 NCH2OP(0)(OH)2
Figure imgf000049_0002
(CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)^CH2SH (CH2)o-5CH3 NCH2OP(0)(OH)2
Figure imgf000049_0003
(CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)N[CH2OP(0)(OH)2](Ph)^CH2SMe (CH2)o-5CH3 NCH2OP(0)(OH)2
C(0)NH(Ph)/?CH2NHC(0)Oi-Bu (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)^CH2NHC(0)Oi-Bu (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[CH20P(0)(OH)2]C(0)Oi-Bu (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)^CH2N[CH2OP(0)(OH)2]C(0)Oi-Bu (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)/?CH2NHC(0)OH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)^CH2NHC(0)OH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)/?CH2NHC(0)OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)^CH2NHC(0)OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[CH20P(0)(OH)2]C(0)OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[(CH2)o-5CH3](Ph)^CH2NHC(0)Oi-Bu (CH2)o-5CH3 N(CH2)o-5CH3 Table 2
Figure imgf000050_0001
R1 R2 Y1
C(0)N[(CH2)o-5CH3](Ph)^CH2N[CH2OP(0)(OH)2]C(0)Oi-Bu (CH2)o-5CH3 N(CH2)o-5CH3
C(O)N[(CH2)0-5CH3](Ph)/?CH2NHC(O)OH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[(CH2)o-5CH3](Ph)^CH2N[CH2OP(0)(OH)2]C(0)OH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[(CH2)o-5CH3](Ph)/?CH2NHC(0)OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(O)N[(CH2)0-5CH3](Ph)^CH2N[CH2OP(O)(OH)2]C(O)OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)/?CH2NH2 (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2NH2 (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)/?CH2OH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)/?CH2N[(CH2)o-5CH3]2 (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[(CH2)o-5CH3]2 (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)/?CH2OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)/?CH2SH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)NH(Ph)/?CH2SMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SMe (CH2)o-5CH3 N(CH2)o-5CH3 Table 2
ft 0
R1 R2 Y1
C(O)N[(CH2)0-5CH3](Ph)/?CH2NH2 (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2NH2 (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[(CH2)o-5CH3](Ph)/?CH2OH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[(CH2)o-5CH3](Ph)^CH2N[(CH2)o-5CH3]2 (CH2)o-5CH3 N(CH2)o-5CH3
C(O)N[CH2OP(O)(OH)2](Ph)^CH2N[(CH2)0-5CH3]2 (CH2)o-5CH3 N(CH2)o-5CH3
C(O)N[(CH2)0-5CH3](Ph)^CH2OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2OMe (CH2)o-5CH3 N(CH2)o-5CH3
C(O)N[(CH2)0-5CH3](Ph)^CH2SH (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SH (CH2)o-5CH3 N(CH2)o-5CH3
C(O)N[(CH2)0-5CH3](Ph)^CH2SMe (CH2)o-5CH3 N(CH2)o-5CH3
C(0)N[CH2OP(0)(OH)2](Ph)/?CH2SMe (CH2)o-5CH3 N(CH2)o-5CH3
Table 3
Figure imgf000051_0001
R1 R6 R1 R6
C(0)-Ph-/?-Br NHC(0)-3-bromo-5-pyridyl C(0)-Ph-/?-Br NHC(0)-3-fluoro-5-pyridyl Table 3
Figure imgf000052_0001
R1 R6 R1 R6
C(0)-Ph-/?-Cl NHC(0)-3-bromo-5-pyridyl C(0)-Ph-/?-Cl NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-/?-F NHC(0)-3-bromo-5-pyridyl C(0)-Ph-/?-F NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-/?-I NHC(0)-3-bromo-5-pyridyl C(0)-Ph-/?-I NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-/?-
NHC(0)-3-bromo-5-pyridyl C(0)-Ph-/?-CF3 NHC(0)-3-fluoro-5-pyridyl CF3
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-/?-Br C(0)-Ph-/?-Br
5-pyridyl fluoro-5-pyridyl
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-/?-Cl C(0)-Ph-/?-Cl
5-pyridyl fluoro-5-pyridyl
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-/?-F C(0)-Ph-/?-F
5-pyridyl fluoro-5-pyridyl
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-/?-I C(0)-Ph-/?-I
5-pyridyl fluoro-5-pyridyl
C(0)-Ph-/?- N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-/?-CF3
CF3 5-pyridyl fluoro-5-pyridyl
C(0)-Ph-o-Br NHC(0)-3-bromo-5-pyridyl C(0)-Ph-o-Br NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-o-Cl NHC(0)-3-bromo-5-pyridyl C(0)-Ph-o-Cl NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-o-F NHC(0)-3-bromo-5-pyridyl C(0)-Ph-o-F NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-o-I NHC(0)-3-bromo-5-pyridyl C(0)-Ph-o-I NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-o-
NHC(0)-3-bromo-5-pyridyl C(0)-Ph-o-CF3 NHC(0)-3-fluoro-5-pyridyl CF3
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-o-Br C(0)-Ph-o-Br
5-pyridyl fluoro-5-pyridyl Table 3
Figure imgf000053_0001
R1 R6 R1 R6
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-o-Cl C(0)-Ph-o-Cl
5-pyridyl fluoro-5-pyridyl
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-o-F C(0)-Ph-o-F
5-pyridyl fluoro-5-pyridyl
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-o-I C(0)-Ph-o-I
5-pyridyl fluoro-5-pyridyl
C(0)-Ph-o- N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-o-CF3
CF3 5-pyridyl fluoro-5-pyridyl
C(0)-Ph-m-Br NHC(0)-3-bromo-5-pyridyl C(0)-Ph-m-Br NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-m-Cl NHC(0)-3-bromo-5-pyridyl C(0)-Ph-m-Cl NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-m-F NHC(0)-3-bromo-5-pyridyl C(0)-Ph-m-F NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-m-I NHC(0)-3-bromo-5-pyridyl C(0)-Ph-m-I NHC(0)-3-fluoro-5-pyridyl
C(0)-Ph-m-
NHC(0)-3-bromo-5-pyridyl C(0)-Ph-m-CF3 NHC(0)-3-fluoro-5-pyridyl CF3
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-m-Br C(0)-Ph-m-Br
5-pyridyl fluoro-5-pyridyl
NH[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-m-Cl C(0)-Ph-m-Cl
5-pyridyl fluoro-5-pyridyl
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-m-F C(0)-Ph-m-F
5-pyridyl fluoro-5-pyridyl
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-m-I C(0)-Ph-m-I
5-pyridyl fluoro-5-pyridyl
C(0)-Ph-m- N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
C(0)-Ph-m-CF3
CF3 5-pyridyl fluoro-5-pyridyl
H NHC(0)-3-bromo-5-pyridyl H NHC(0)-3-fluoro-5-pyridyl Table 3
Figure imgf000054_0001
R1 R6 R1 R6
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
H H
5-pyridyl fluoro-5-pyridyl
(CH2)o-5CH3 NHC(0)-3-bromo-5-pyridyl (CH2)o-5CH3 NHC(0)-3-fluoro-5-pyridyl
N[CH2OP(0)(OH)2]C(0)-3-bromo- N[CH2OP(0)(OH)2]C(0)-3-
(CH2)o-5CH3 (CH2)o-5CH3
5-pyridyl fluoro-5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-/?-Br C(0)-Ph-/?-Br
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-/?-Cl C(0)-Ph-/?-Cl
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)0-5CH3]C(O)-3-fluoro-
C(0)-Ph-/?-F C(0)-Ph-/?-F
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)0-5CH3]C(O)-3-fluoro-
C(0)-Ph-/?-I C(0)-Ph-/?-I
pyridyl 5-pyridyl
C(0)-Ph-/?- N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)0-5CH3]C(O)-3-fluoro-
C(0)-Ph-/?-CF3
CF3 pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)0-5CH3]C(O)-3-fluoro-
C(0)-Ph-o-Br C(0)-Ph-o-Br
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)0-5CH3]C(O)-3-fluoro-
C(0)-Ph-o-Cl C(0)-Ph-o-Cl
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-o-F C(0)-Ph-o-F
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-o-I C(0)-Ph-o-I
pyridyl 5-pyridyl
C(0)-Ph-o- N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-o-CF3
CF3 pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)0-5CH3]C(O)-3-fluoro-
C(0)-Ph-m-Br C(0)-Ph-m-Br
pyridyl 5-pyridyl Table 3
Figure imgf000055_0001
R1 R6 R1 R6
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-m-Cl C(0)-Ph-m-Cl
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-m-F C(0)-Ph-m-F
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-m-I C(0)-Ph-m-I
pyridyl 5-pyridyl
C(0)-Ph-m- N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
C(0)-Ph-m-CF3
CF3 pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
H H
pyridyl 5-pyridyl
N[(CH2)o-5CH3]C(0)-3-bromo-5- N[(CH2)o-5CH3]C(0)-3-fluoro-
(CH2)o-5CH3 (CH2)o-5CH3
pyridyl 5-pyridyl
Table 4
N^N
R1 R2 R2' A
(CH2V5CH3 NH2 N02 Phenyl
(CH2)o-5CH3 N[(CH2)o-5CH3]2 N02 Phenyl
(CH2)o-5CH3 OH N02 Phenyl
(CH2)o-5CH3 OP(rO)(OH)2 N02 Phenyl
(CH2)o-5CH3 OC(0)C(CH3)3 N02 Phenyl
(CH2V5CH3 OC(0)NMe2 N02 Phenyl
Figure imgf000056_0001
Table 4
N^N
R1 R2 R2' A
H OC(0)NMe2 N02 Phenyl
H OMe N02 Phenyl
H SH N02 Phenyl
H SMe N02 Phenyl
H NH2 N02 None
H N[(CH2)o-5CH3]2 N02 None
H OH N02 None
H OP(0)(OH)2 N02 None
H OC(0)C(CH3)3 N02 None
H OC(0)NMe2 N02 None
H OMe N02 None
H SH N02 None
H SMe N02 None
Table 5
N^N
R1 R2 R2' A Table 5
N^N
R1 R2 R2' A
(CH2)o-5CH3 NH2 N02 Phenyl
(CH2)o-5CH3 N[(CH2)o-5CH3]2 N02 Phenyl
(CH2)o-5CH3 OH N02 Phenyl
(CH2)o-5CH3 OP(0)(OH)2 N02 Phenyl
(CH2)o-5CH3 OC(0)C(CH3)3 N02 Phenyl
(CH2)o-5CH3 OC(0)NMe2 N02 Phenyl
(CH2)o-5CH3 OMe N02 Phenyl
(CH2)o-5CH3 SH N02 Phenyl
(CH2)o-5CH3 SMe N02 Phenyl
(CH2)o-5CH3 NH2 N02 None
(CH2)o-5CH3 N[(CH2)o-5CH3]2 N02 None
(CH2)o-5CH3 OH N02 None
(CH2)o-5CH3 OP(0)(OH)2 N02 None
(CH2)o-5CH3 OC(0)C(CH3)3 N02 None
(CH2)o-5CH3 OC(0)NMe2 N02 None
(CH2)o-5CH3 OMe N02 None
(CH2)o-5CH3 SH N02 None Table 5
N^N
R1 R2 R2' A
(CH2)o-5CH3 SMe N02 None
H NH2 N02 Phenyl
H N[(CH2)o-5CH3]2 N02 Phenyl
H OH N02 Phenyl
H OP(0)(OH)2 N02 Phenyl
H OC(0)C(CH3)3 N02 Phenyl
H OC(0)NMe2 N02 Phenyl
H OMe N02 Phenyl
H SH N02 Phenyl
H SMe N02 Phenyl
H NH2 N02 None
H N[(CH2)o-5CH3]2 N02 None
H OH N02 None
H OP(0)(OH)2 N02 None
H OC(0)C(CH3)3 N02 None
H OC(0)NMe2 N02 None
H OMe N02 None
Figure imgf000060_0001
Figure imgf000060_0002
Table 6A
R1 Y1 Y2 R1 Y1 Y2
0(CH2)o-5CH3 o 0 0(CH2)o-5CH3 o NH
OH o 0 OH o NH
OP(0)(OH)2 0 0 OP(0)(OH)2 0 NH
OC(0)C(CH3)3 o 0 OC(0)C(CH3)3 o NH
OC(0) Me2 o 0 OC(0)NMe2 o NH
S(CH2)o-5CH3 o 0 S(CH2)o-5CH3 o NH
SH 0 0 SH 0 NH
NH2 0 0 NH2 0 NH
N[(CH2)o- N[(CH2)o- o 0 o NH 5CH3]2 5CH3]2
OCF3 o 0 OCF3 o NH
CF3 o 0 CF3 o NH
0(CH2)o-5CH3 NCH20P(0)(0H)2 NH 0(CH2)o-5CH3 NCH2OP(0)(OH)2 0
OH NCH20P(0)(0H)2 NH OH NCH2OP(0)(OH)2 0
OP(0)(OH)2 NCH20P(0)(0H)2 NH OP(0)(OH)2 NCH2OP(0)(OH)2 0
OC(0)C(CH3)3 NCH20P(0)(0H)2 NH OC(0)C(CH3)3 NCH2OP(0)(OH)2 0
OC(0) Me2 NCH20P(0)(0H)2 NH OC(0)NMe2 NCH2OP(0)(OH)2 0
S(CH2)o-5CH3 NCH20P(0)(0H)2 NH S(CH2)o-5CH3 NCH2OP(0)(OH)2 0 Table 6A
Figure imgf000062_0001
R1 Y1 Y2 R1 Y1 Y2
SH NCH2OP(0)(OH)2 NH SH NCH2OP(0)(OH)2 0
NH2 NCH2OP(0)(OH)2 NH NH2 NCH2OP(0)(OH)2 0
N[(CH2)o- N[(CH2)o-
NCH20P(0)(0H)2 NH NCH2OP(0)(OH)2 0 5CH3]2 5CH3]2
OCFs NCH20P(0)(0H)2 NH OCFs NCH2OP(0)(OH)2 0
CF3 NCH20P(0)(0H)2 NH CF3 NCH2OP(0)(OH)2 0
O(CH2)0-5CH3 NCH20P(0)(0H)2 NCH2OP(0)(OH)2 0(CH2)o-5CH3 O NCH20P(0)(0H)2
OH NCH20P(0)(0H)2 NCH2OP(0)(OH)2 OH O NCH2OP(0)(OH)2
OP(0)(OH)2 NCH20P(0)(0H)2 NCH2OP(0)(OH)2 OP(0)(OH)2 O NCH2OP(0)(OH)2
OC(0)C(CH3)3 NCH20P(0)(0H)2 NCH2OP(0)(OH)2 OC(0)C(CH3)3 0 NCH2OP(0)(OH)2
OC(0) Me2 NCH20P(0)(0H)2 NCH2OP(0)(OH)2 OC(0)NMe2 O NCH2OP(0)(OH)2
S(CH2)o-5CH3 NCH20P(0)(0H)2 NCH2OP(0)(OH)2 S(CH2)o-5CH3 O NCH2OP(0)(OH)2
SH NCH20P(0)(0H)2 NCH2OP(0)(OH)2 SH O NCH2OP(0)(OH)2
NH2 NCH2OP(0)(OH)2 NCH2OP(0)(OH)2 NH2 0 NCH2OP(0)(OH)2
N[(CH2)o- N[(CH2)o-
NCH20P(0)(0H)2 NCH2OP(0)(OH)2 0 NCH2OP(0)(OH)2 5CH3]2 5CH3]2
OCF3 NCH20P(0)(0H)2 NCH2OP(0)(OH)2 OCF3 0 NCH2OP(0)(OH)2
CF3 NCH20P(0)(0H)2 NCH2OP(0)(OH)2 CF3 0 NCH20P(0)(0H)2 Table 6A
R1 Y1 Y2 R1 Y1 Y2
0(CH2)o-5CH3 s 0 0(CH2)o-5CH3 s NH
OH s 0 OH s NH
OP(0)(OH)2 s 0 OP(0)(OH)2 s NH
OC(0)C(CH3)3 s 0 OC(0)C(CH3)3 s NH
OC(0) Me2 s 0 OC(0)NMe2 s NH
S(CH2)o-5CH3 s 0 S(CH2)o-5CH3 s NH
SH s 0 SH s NH
NH2 s 0 NH2 s NH
N[(CH2)o- N[(CH2)o- s 0 s NH 5CH3]2 5CH3]2
OCF3 s 0 OCF3 s NH
CF3 s 0 CF3 s NH
0(CH2)o-5CH3 s NCH2OP(0)(OH)2 0(CH2)o-5CH3 s S
OH s NCH2OP(0)(OH)2 OH s S
OP(0)(OH)2 s NCH2OP(0)(OH)2 OP(0)(OH)2 s S
OC(0)C(CH3)3 s NCH2OP(0)(OH)2 OC(0)C(CH3)3 s S
OC(0) Me2 s NCH2OP(0)(OH)2 OC(0)NMe2 s S
S(CH2)o-5CH3 s NCH2OP(0)(OH)2 S(CH2)o-5CH3 s S Table 6A
R1 Y1 Y2 R1 Y1 Y2
SH s NCH2OP(0)(OH)2 SH s s
NH2 s NCH2OP(0)(OH)2 NH2 s s
N[(CH2)o- N[(CH2)o- s NCH2OP(0)(OH)2 s s 5CH3]2 5CH3]2
OCF3 s NCH2OP(0)(OH)2 OCF3 s s
CF3 s NCH2OP(0)(OH)2 CF3 s s
///. Methods of Use
I. Methods of Treating Muscular Dystrophy
The α7β 1 integrin has been shown to be a major modifier of disease progression in patients with muscular dystrophy. Increased expression of the a7 integrin in muscle can alleviate muscle disease in mouse models of muscular dystrophy. By use of a muscle cell-based assay (described in Example 1 below), the inventors identified the following molecules that up-regulate α7β 1 integrin expression in muscle:
laminin-111; valproic acid; ciclopirox ethanolamine; deferoxamine; 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one; Compound ID# 1001; Compound ID# 1002; Compound ID #1003; and analogs of cholestan (see Table 9). Based on these observations, methods of treatment of muscular dystrophy by increasing the expression or activity of α7β 1 integrin are disclosed.
In particular, methods are disclosed herein for treating muscular dystrophy, such as DMD, FCMD, LGMD, FHMD, BMD, MDC1A or MDC1D. In one example, the method includes administering an effective amount of a α7β1 integrin modulatory agent to a subject with muscular dystrophy or suspected of having or developing muscular dystrophy, in which the agent increases the biological activity or expression of α7β 1 integrin and thereby, treating the muscular dystrophy in the subject. In some example, the method of treatment inhibits or reduces one or more signs or symptoms associated with muscular dystrophy in the subject.
In some examples, the α7β 1 integrin modulatory agent includes one or more of the following molecules: ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, an analog of Compound ID# 1001, an analog of
Compound ID #1002, an analog of Compound ID #1003, an analog of cholestan (see Table 9), laminin-111, laminin-111 fragments, valproic acid, or a valproic acid analog. Tables 8 and 9 and FIGS. 3A-3C, 4A-4B, 5, 6A-6B, and 7-9 provide the chemical structures and characterization data for such compounds. Exemplary valproic acid analogs are disclosed in U.S. Patent Publication 2006/0223888 and International Patent Application No. 2010/080581, each of which is incorporated herein by reference in its entirety. Table 11 provides analogs of disclosed Compound ID# 1001. Exemplary laminin-111 fragments are disclosed in U.S. Patent Publication US-2009-0092587-A1, which is incorporated herein by reference in its entirety. In some examples, an analog of Compound ID #1002 or #1003 is synthesized according to the general synthesis pathway shown in FIG. 10. In some examples, an analog is synthesized according to the synthesis pathway shown in the Examples below. In further examples, the α7β 1 integrin modulatory agent is an
analog/derivative of any of the disclosed α7β 1 integrin modulatory agents which may be designed and synthesized according to the chemical principles known to one of ordinary skill in the art and identified as a α7β 1 integrin modulatory agent by methods known to those of ordinary skill in the art, including the muscle cell based assay described Example 1. For examples, in some examples, the α7β 1 integrin modulatory agent includes one or more molecules provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof. In further embodiments, the α7β 1 integrin modulatory agent may be selected from any one or more of the compounds within any one of Formulas 1-16, as provided herein.
The disclosed α7β 1 integrin modulatory agents can alter the expression of nucleic acid sequences (such as DNA, cDNA, or mRNAs) and proteins of α7β 1 integrin. An increase in the expression or activity does not need to be 100% for the agent to be effective. For example, an agent can increase the expression or biological activity by a desired amount, for example by at least 10%, for example at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, including about 15% to about 98%, about 30% to about 95%, about 40% to about 80%, about 50% to about 70%, including about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%, as compared to activity or expression in a control. Methods of assessing α7β 1 integrin expression and activity are known to those of ordinary skill in the art, including those described in the Examples below (e.g. , Western blot and ELISA assay with commercially available antibodies).
In a particular example, the subject is a human.
In additional aspects, the method involves selecting a subject with muscular dystrophy. In some example, a subject is selected for treatment following diagnosing the subject with muscular dystrophy. For example, the method can include diagnosing the subject as suffering from muscular dystrophy, such as DMD, MDC1A, MDC1D, LGMD, DMD, FCMD or FHMD.
Methods of diagnosing a subject with muscular dystrophy are known to those of skillin the art and include, but are not limited to, muscle biopsies and measuring serum creatine kinase levels. Additionally, alterations in biomarker known to be associated with muscular dystrophy may be detected by measuring such levels in serum or urine sample.
In a further implementation, the method involves diagnosing the subject as suffering from a disease, disorder, or condition characterized by a mutation in the gene encoding l integrin. In another
implementation, the method involves diagnosing the subject as suffering from a disease, disorder, or condition characterized by a decreased level of a l integrin expression.
Alterations in the expression can be measured at the nucleic acid level (such as by real time quantitative polymerase chain reaction or microarray analysis) or at the protein level (such as by Western blot analysis or ELISA). These methods are known to those of skill in the art.
In some examples, following the measurement of the expression levels of a l integrin expression or serum creatine kinase levels, the assay results, findings, diagnoses, predictions and/or treatment recommendations are recorded and communicated to technicians, physicians and/or patients, for example. In certain embodiments, computers are used to communicate such information to interested parties, such as, patients and/or the attending physicians. The therapy selected for administered is then based upon these results.
In one embodiment, the results and/or related information is communicated to the subject by the subject's treating physician. Alternatively, the results may be communicated directly to a test subject by any means of communication, including writing, such as by providing a written report, electronic forms of communication, such as email, or telephone. Communication may be facilitated by use of a computer, such as in case of email communications. In certain embodiments, the communication containing results of a diagnostic test and/or conclusions drawn from and/or treatment recommendations based on the test, may be generated and delivered automatically to the subject using a combination of computer hardware and software which will be familiar to artisans skilled in telecommunications. One example of a healthcare- oriented communications system is described in U.S. Pat. No. 6,283,761; however, the present disclosure is not limited to methods which utilize this particular communications system. In certain embodiments of the methods of the disclosure, all or some of the method steps, including the assaying of samples, diagnosing of diseases, and communicating of assay results or diagnoses, may be carried out in diverse (e.g., foreign) jurisdictions.
In several embodiments, identification of a subject as having muscular dystrophy, such as DMD, LGMD, FHMD, BMD, FCMD, MDC1D or MDC1A, results in the physician treating the subject, such as prescribing one or more disclosed α7β1 agents for inhibiting or delaying one or more signs and symptoms associated with muscular dystrophy. In additional embodiments, the dose or dosing regimen is modified based on the information obtained using the methods disclosed herein.
II. Methods of enhancing muscle regeneration, repair, or maintenance
Also disclosed are methods of enhancing muscle regeneration, repair or maintenance in a subject. In some examples, the method includes administering an effective amount of an α7β1 integrin modulatory agent to a subject in need of muscle regeneration, repair or maintenance, wherein the α7β1 integrin modulatory agent includes ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby enhancing muscle regeneration, repair or maintenance in a subject.
In some examples, the α7β 1 integrin modulatory agent includes one or more of the following molecules: ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan^-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, an analog of Compound ID# 1001, an analog of
Compound ID #1002, an analog of Compound ID #1003, an analog of cholestan (see Table 9), laminin-111, laminin-111 fragments, valproic acid, or a valproic acid analog. Tables 8 and 9 and FIGS. 3A-3C, 4A-4B, 5, 6A-6B, and 7-9 provide the chemical structures and characterization data for such compounds. Exemplary valproic acid analogs are disclosed in U.S. Patent Publication 2006/0223888 and International Patent Application No. 2010/080581, each of which is incorporated herein by reference in its entirety. Exemplary laminin-111 fragments are disclosed in U.S. Patent Publication US-2009-0092587-A1, which is incorporated herein by reference in its entirety. In some examples, an analog of Compound ID #1002 or #1003 is synthesized according to the general synthesis pathway shown in FIG. 10. In some examples, an analog is synthesized according to the synthesis pathway provided in the Examples below. In further examples, the α7β1 integrin modulatory agent is an analog/derivative of any of the disclosed α7β1 integrin modulatory agents which may be designed and synthesized according to the chemical principles known to one of ordinary skill in the art and identified as a α7β 1 integrin modulatory agent by methods known to those of ordinary skill in the art, including the muscle cell based assay described Example 1. For examples, in some examples, the α7β 1 integrin modulatory agent includes one or more molecules provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof.
The disclosed α7β 1 integrin modulatory agents can increase the expression of nucleic acid sequences (such as DNA, cDNA, or mRNAs) and proteins of α7β 1 integrin. An increase in the expression or activity does not need to be 100% for the agent to be effective. For example, an agent can increase the expression or biological activity by a desired amount, for example by at least 10%, for example at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, including about 15% to about 98%, about 30% to about 95%, about 40% to about 80%, about 50% to about 70%, including about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%, as compared to activity or expression in a control. Methods of assessing α7β 1 integrin expression and activity are known to those of ordinary skill in the art, including those described in the Examples below (e.g. , Western blot and ELISA assay with commercially available antibodies).
Muscle regeneration may benefit, for example, geriatric or other patient populations with reduced muscle repair capability, or simply speed the muscle repair process for otherwise physiologically unimpaired patients. In particular implementations, administration of a α7β1 integrin modulatory agent can aid muscle repair, or reduction of muscle damage, in athletes or others having activity-induced muscle injury or damage. In yet further implementations, muscle repair in patients suffering from muscle damage, such as through accident or injury, can be augmented by administration of a α7β1 integrin modulatory agent.
In some examples, α7β1 modulatory agent is administered prior to the subject experiencing muscle damage or disease. In some examples, the α7β1 integrin modulatory agent is administered to the subject prior to the subject exercising.
In some examples, the method further includes selecting a subject in need of enhancing muscle regeneration, repair, or maintenance. For example, in some instances, selecting a subject in need of enhancing muscle regeneration, repair, or maintenance comprises diagnosing the subject with a condition characterized by impaired muscle regeneration prior to administering an effective amount of the α7β1 integrin modulatory agent to the subject. Methods for diagnosing and selecting a subject in need of muscle regeneration, repair or maintenance are known to those of ordinary skill in the art and include those provided described herein (including those in the Methods of Treatment of Muscular Dystrophy). As stated above, subjects may be selected based upon their life style (e.g., engaged in moderate to intense exercise or physical activities), age (e.g., elderly population at more risk of experiencing muscle degeneration or injury) or predisposition to muscle degeneration or injury (e.g., genetics or previous muscle injury).
in. Methods of prospectively preventing or reducing muscle injury or damage
Also disclosed are methods prospectively preventing or reducing muscle injury or damage in a subject. In some embodiments, the method includes administering an effective amount of an α7β1 integrin modulatory agent to the subject wherein the α7β1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan^-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, a compound provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), and/or Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby prospectively preventing or reducing muscle injury or damage in the subject.
In some examples, the α7β 1 integrin modulatory agent includes one or more of the following molecules: ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan^-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, an analog of Compound ID# 1001, an analog of
Compound ID #1002, an analog of Compound ID #1003, an analog of cholestan (see Table 9), laminin-111, laminin-111 fragments, valproic acid, or a valproic acid analog. Tables 8 and 9 and FIGS. 3A-3C, 4A-4B, 5, 6A-6B, and 7-9 provide the chemical structures and characterization data for such compounds. Exemplary valproic acid analogs are disclosed in U.S. Patent Publication 2006/0223888 and International Patent Application No. 2010/080581, each of which is incorporated herein by reference in its entirety. Exemplary laminin-111 fragments are disclosed in U.S. Patent Publication US-2009-0092587-A1, which is incorporated herein by reference in its entirety. In some examples, an analog of Compound ID #1002 or #1003 is synthesized according to the general synthesis pathway shown in FIG. 10. In further examples, the α7β 1 integrin modulatory agent is an analog/derivative of any of the disclosed α7β 1 integrin modulatory agents which may be designed and synthesized according to the chemical principles known to one of ordinary skill in the art and identified as a α7β 1 integrin modulatory agent by methods known to those of ordinary skill in the art, including the muscle cell based assay described Example 1. For examples, in some examples, the α7β 1 integrin modulatory agent includes one or more molecules provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof.
The disclosed α7β 1 integrin modulatory agents can increase the expression of nucleic acid sequences (such as DNA, cDNA, or mRNAs) and proteins of α7β 1 integrin. An increase in the expression or activity does not need to be 100% for the agent to be effective. For example, an agent can increase the expression or biological activity by a desired amount, for example by at least 10%, for example at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, including about 15% to about 98%, about 30% to about 95%, about 40% to about 80%, about 50% to about 70%, including about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%, as compared to activity or expression in a control. Methods of assessing α7β 1 integrin expression and activity are known to those of ordinary skill in the art, including those described in the Examples below (e.g. , Western blot and ELISA assay with commercially available antibodies).
In some examples, the method further includes selecting a subject at risk for developing a muscle injury or damage. In some examples, the α7β1 integrin modulatory agent is administered to a subject prior to the subject exercising. In some examples, the method further includes selecting a subject at risk for developing a muscle injury or damage. Methods for selecting such s subject are known to those of ordinary skill in the art and include those provided described herein. As stated above, subjects may be selected based upon their life style (e.g., engaged in moderate to intense exercise or physical activities), age (elderly population at more risk of experiencing muscle degeneration or injury) or pre-disposition to muscle degeneration or injury (e.g., genetics or previous muscle injury).
iv. Methods of enhancing α7β1 integrin expression
Also disclosed herein are methods of enhancing α7β1 integrin expression. In some examples, these methods include contacting a cell with an effective amount of an α7β1 integrin modulatory agent, wherein the α7β1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a- cholestan-3p-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069-0073, 1080-0573, or any one of the compounds provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or a combination thereof and increases α7β1 integrin expression in the treated cell relative to α7β1 integrin expression in an untreated cell, thereby enhancing α7β1 integrin expression. In some examples, the cell is a muscle cell, such as a skeletal muscle cell. In some examples, the muscle cell is present in a mammal, and wherein contacting the cell with an agent comprises administering the agent to the mammal.
In some examples, the α7β 1 integrin modulatory agent includes one or more of the following molecules: ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a-cholestan^-ol-6-one, Compound ID# 1001, Compound ID# 1002, Compound ID #1003, an analog of Compound ID# 1001, an analog of
Compound ID #1002, an analog of Compound ID #1003, an analog of cholestan (see Table 9), laminin-111, laminin-111 fragments, valproic acid, or a valproic acid analog. Tables 8-10 and FIGS. 3A-3C, 4A-4B, 5, 6A-6B, and 7-9 provide the chemical structures and characterization data for such compounds. Exemplary valproic acid analogs are disclosed in U.S. Patent Publication 2006/0223888 and International Patent Application No. 2010/080581, each of which is incorporated herein by reference in its entirety. Exemplary laminin-111 fragments are disclosed in U.S. Patent Publication US-2009-0092587-A1, which is incorporated herein by reference in its entirety. In some examples, an analog of Compound ID #1002 or #1003 is synthesized according to the general synthesis pathway shown in FIG. 10. In further examples, the α7β 1 integrin modulatory agent is an analog/derivative of any of the disclosed α7β 1 integrin modulatory agents which may be designed and synthesized according to the chemical principles known to one of ordinary skill in the art and identified as a α7β 1 integrin modulatory agent by methods known to those of ordinary skill in the art, including the muscle cell based assay described Example 1. In some examples, the α7β 1 integrin modulatory agent includes one or more molecules provided herein, such as in Table 10, Table 11, Table 6 (see Appendix I of U.S. Provisional Pat. App. No. 61/796,476), Table 7 (see Appendix II of U.S. Provisional Pat. App. No. 61/796,476), Table 13, or combinations thereof. In some examples, the disclosed α7β 1 integrin modulatory agents can increase the expression of nucleic acid sequences (such as DNA, cDNA, or mRNAs) and proteins of α7β 1 integrin. An increase in the expression or activity does not need to be 100% for the agent to be effective. For example, an agent can increase the expression or biological activity by a desired amount, for example by at least 10%, for example at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, or even at least 100%, including about 15% to about 98%, about 30% to about 95%, about 40% to about 80%, about 50% to about 70%, including about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98% or about 100%, as compared to activity or expression in a control. Methods of assessing α7β 1 integrin expression and activity are known to those of ordinary skill in the art, including those described in the Examples below (e.g. , Western blot and ELISA assay with commercially available antibodies).
Administration of an Effective amount of an α7β1 integrin modulatory agent
For any of the disclosed methods, an effective amount of α7β 1 integrin modulatory agent is one when administered by a particular route and concentration induces the desired response (e.g. , treatment of muscular dystrophy, enhancing muscle regeneration, repair or maintenance, preventing or reducing muscle injury or damage, or enhancing α7β 1 integrin expression).
i. Administration routes, formulations and concentrations
Methods of administration of the disclosed α7β 1 integrin modulatory agents are routine, and can be determined by a skilled clinician. The disclosed α7β 1 integrin modulatory agents or other therapeutic substance are in general administered topically, nasally, intravenously, orally, intracranially,
intramuscularly, parenterally or as implants, but even rectal or vaginal use is possible in principle. The disclosed α7β 1 integrin modulatory agents also may be administered to a subject using a combination of these techniques.
Suitable solid or liquid pharmaceutical preparation forms are, for example, aerosols,
(micro)capsules, creams, drops, drops or injectable solution in ampoule form, emulsions, granules, powders, suppositories, suspensions, syrups, tablets, coated tablets, and also preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as binders, coating agents, disintegrants, flavorings, lubricants, solubilizers, sweeteners, or swelling agents are customarily used as described above. The pharmaceutical agents are suitable for use in a variety of drug delivery systems. For a brief review of various methods for drug delivery, see Langer, "New Methods of Drug Delivery," Science 249: 1527-1533 (1990), incorporated by reference herein to the extent not inconsistent with the present disclosure.
The disclosed α7β 1 integrin modulatory agents or other therapeutic agents of the present disclosure can be formulated into therapeutically-active pharmaceutical agents that can be administered to a subject parenterally or orally. Parenteral administration routes include, but are not limited to epidermal, intraarterial, intramuscular (IM and depot IM), intraperitoneal (IP), intravenous (Γν), intrasternal injection or infusion techniques, intranasal (inhalation), intrathecal, injection into the stomach, subcutaneous injections (subcutaneous (SQ and depot SQ), transdermal, topical, and ophthalmic.
The disclosed α7β 1 integrin modulatory agents or other therapeutic agents can be mixed or combined with a suitable pharmaceutically acceptable excipients to prepare pharmaceutical agents.
Pharmaceutically acceptable excipients include, but are not limited to, alumina, aluminum stearate, buffers (such as phosphates), glycine, ion exchangers (such as to help control release of charged substances), lecithin, partial glyceride mixtures of saturated vegetable fatty acids, potassium sorbate, serum proteins (such as human serum albumin), sorbic acid, water, salts or electrolytes such as cellulose-based substances, colloidal silica, disodium hydrogen phosphate, magnesium trisilicate, polyacrylates, polyalkylene glycols, such as polyethylene glycol, polyethylene-polyoxypropylene-block polymers, polyvinyl pyrrolidone, potassium hydrogen phosphate, protamine sulfate, group 1 halide salts such as sodium chloride, sodium carboxymethylcellulose, waxes, wool fat, and zinc salts, for example. Liposomal suspensions may also be suitable as pharmaceutically acceptable carriers.
Upon mixing or addition of one or more disclosed α7β 1 integrin modulatory agents and/or or other therapeutic agents, the resulting mixture may be a solid, solution, suspension, emulsion, or the like. These may be prepared according to methods known to those of ordinary skill in the art. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the agent in the selected carrier. Pharmaceutical carriers suitable for administration of the disclosed α7β 1 integrin modulatory agents or other therapeutic agents include any such carriers known to be suitable for the particular mode of administration. In addition, the disclosed α7β 1 integrin modulatory agents or other therapeutic substance can also be mixed with other inactive or active materials that do not impair the desired action, or with materials that supplement the desired action, or have another action.
Methods for solubilizing may be used where the agents exhibit insufficient solubility in a carrier. Such methods are known and include, but are not limited to, dissolution in aqueous sodium bicarbonate, using cosolvents such as dimethylsulfoxide (DMSO), and using surfactants such as TWEEN® (ICI Americas, Inc., Wilmington, DE).
The disclosed α7β 1 integrin modulatory agents or other therapeutic agents can be prepared with carriers that protect them against rapid elimination from the body, such as coatings or time-release formulations. Such carriers include controlled release formulations, such as, but not limited to,
microencapsulated delivery systems. A disclosed α7β 1 integrin modulatory agents or other therapeutic agent is included in the pharmaceutically acceptable carrier in an amount sufficient to exert a therapeutically useful effect, typically in an amount to avoid undesired side effects, on the treated subject. The
therapeutically effective concentration may be determined empirically by testing the compounds in known in vitro and in vivo model systems for the treated condition. For example, mouse models of muscular dystrophy may be used to determine effective amounts or concentrations that can then be translated to other subjects, such as humans, as known in the art.
Injectable solutions or suspensions can be formulated, using suitable non-toxic, parenterally- acceptable diluents or solvents, such as 1,3-butanediol, isotonic sodium chloride solution, mannitol, Ringer's solution, saline solution, or water; or suitable dispersing or wetting and suspending agents, such as sterile, bland, fixed oils, including synthetic mono- or diglycerides, and fatty acids, including oleic acid; a naturally occurring vegetable oil such as coconut oil, cottonseed oil, peanut oil, sesame oil, and the like; glycerine; polyethylene glycol; propylene glycol; or other synthetic solvent; antimicrobial agents such as benzyl alcohol and methyl parabens; antioxidants such as ascorbic acid and sodium bisulfite; buffers such as acetates, citrates, and phosphates; chelating agents such as ethylenediaminetetraacetic acid (EDTA); agents for the adjustment of tonicity such as sodium chloride and dextrose; and combinations thereof. Parenteral preparations can be enclosed in ampoules, disposable syringes, or multiple dose vials made of glass, plastic, or other suitable material. Buffers, preservatives, antioxidants, and the like can be incorporated as required. Where administered intravenously, suitable carriers include physiological saline, phosphate-buffered saline (PBS), and solutions containing thickening and solubilizing agents such as glucose, polyethylene glycol, polypropyleneglycol, and mixtures thereof. Liposomal suspensions, including tissue-targeted liposomes, may also be suitable as pharmaceutically acceptable carriers.
For topical application, one or more disclosed α7β 1 integrin modulatory agents, or other therapeutic agent may be made up into a cream, lotion, ointment, solution, or suspension in a suitable aqueous or nonaqueous carrier. Topical application can also be accomplished by transdermal patches or bandages which include the therapeutic substance. Additives can also be included, e.g., buffers such as sodium
metabisulphite or disodium edetate; preservatives such as bactericidal and fungicidal agents, including phenyl mercuric acetate or nitrate, benzalkonium chloride, or chlorhexidine; and thickening agents, such as hypromellose.
If the disclosed α7β 1 integrin modulatory agent, or other therapeutic agent is administered orally as a suspension, the pharmaceutical agents can be prepared according to techniques well known in the art of pharmaceutical formulation and may contain a suspending agent, such as alginic acid or sodium alginate, bulking agent, such as microcrystalline cellulose, a viscosity enhancer, such as methylcellulose, and sweeteners/flavoring agents. Oral liquid preparations can contain conventional additives such as suspending agents, e.g., gelatin, glucose syrup, hydrogenated edible fats, methyl cellulose, sorbitol, and syrup;
emulsifying agents, e.g., acacia, lecithin, or sorbitan monooleate; non-aqueous carriers (including edible oils), e.g., almond oil, fractionated coconut oil, oily esters such as glycerine, propylene glycol, or ethyl alcohol; preservatives such as methyl or propyl p-hydroxybenzoate or sorbic acid; and, if desired, conventional flavoring or coloring agents. When formulated as immediate release tablets, these agents can contain dicalcium phosphate, lactose, magnesium stearate, microcrystalline cellulose, and starch and/or other binders, diluents, disintegrants, excipients, extenders, and lubricants.
If oral administration is desired, one or more disclosed α7β 1 integrin modulatory agents, or other therapeutic substances can be provided in a composition that protects it from the acidic environment of the stomach. For example, he disclosed α7β 1 integrin modulatory agents or other therapeutic agents can be formulated with an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine. The disclosed α7β1 integrin modulatory agents, or other therapeutic agent can also be formulated in combination with an antacid or other such ingredient.
Oral compositions generally include an inert diluent or an edible carrier and can be compressed into tablets or enclosed in gelatin capsules. For the purpose of oral therapeutic administration, one or more of the disclosed α7β 1 integrin modulatory agents, or other therapeutic substances can be incorporated with excipients and used in the form of capsules, tablets, or troches. Pharmaceutically compatible adjuvant materials or binding agents can be included as part of the composition.
The capsules, pills, tablets, troches, and the like can contain any of the following ingredients or compounds of a similar nature: a binder such as, but not limited to, acacia, corn starch, gelatin, gum tragacanth, polyvinylpyrrolidone, or sorbitol; a filler such as calcium phosphate, glycine, lactose, microcrystalline cellulose, or starch; a disintegrating agent such as, but not limited to, alginic acid and corn starch; a lubricant such as, but not limited to, magnesium stearate, polyethylene glycol, silica, or talc; a gildant, such as, but not limited to, colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; disintegrants such as potato starch; dispersing or wetting agents such as sodium lauryl sulfate; and a flavoring agent such as peppermint, methyl salicylate, or fruit flavoring.
When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier, such as a fatty oil. In addition, dosage unit forms can contain various other materials that modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents. One or more of the disclosed α7β 1 integrin modulatory agents, or other therapeutic agent can also be administered as a component of an elixir, suspension, syrup, wafer, tea, chewing gum, or the like. A syrup may contain, in addition to the active compounds, sucrose or glycerin as a sweetening agent and certain preservatives, dyes and colorings, and flavors.
When administered orally, the compounds can be administered in usual dosage forms for oral administration. These dosage forms include the usual solid unit dosage forms of tablets and capsules as well as liquid dosage forms such as solutions, suspensions, and elixirs. When the solid dosage forms are used, they can be of the sustained release type so that the compounds need to be administered less frequently.
In some examples, one or more of the disclosed α7β 1 integrin modulatory agents and/or a therapeutic agent is injected into the stomach of a subject is incorporated systemically in the subject, such as in diverse muscle groups. Examples of methods and compositions for administering therapeutic substances which include proteins include those discussed in Banga, Therapeutic Peptides and Proteins: Formulation, Processing, and Delivery Systems 2ed. (2005); Mahato, Biomaterials for Delivery and Targeting of Proteins and Nucleic Acids (2004); McNally, Protein Formulation and Delivery, 2ed. (2007); and Kumar et ah, "Novel Delivery Technologies for Protein and Peptide Therapeutics," Current Pharm. Biotech., 7:261-276 (2006); each of which is incorporated by reference herein to the extent not inconsistent with the present disclosure.
In some implementations, the effective amount of one or more of the disclosed α7β 1 integrin modulatory agents is administered as a single dose per time period, such as every three or four months, month, week, or day, or it can be divided into at least two unit dosages for administration over a period. Treatment may be continued as long as necessary to achieve the desired results. For instance, treatment may continue for about 3 or 4 weeks up to about 12-24 months or longer, including ongoing treatment. The compound can also be administered in several doses intermittently, such as every few days (for example, at least about every two, three, four, five, or ten days) or every few weeks (for example at least about every two, three, four, five, or ten weeks).
Particular dosage regimens can be tailored to a particular subject, condition to be treated, or desired result. For example, when the methods of the present disclosure are used to treat muscular dystrophy or similar conditions, an initial treatment regimen can be applied to arrest the condition. Such initial treatment regimen may include administering a higher dosage of one or more of the disclosed α7β 1 integrin modulatory agents, or administering such material more frequently, such as daily. After a desired therapeutic result has been obtained, such as a desired level of muscle regeneration, a second treatment regimen may be applied, such as administering a lower dosage of one or more of the disclosed α7β 1 integrin modulatory agents or administering such material less frequently, such as monthly, bi-monthly, quarterly, or semi-annually. In such cases, the second regimen may serve as a "booster" to restore or maintain a desired level of muscle regeneration. Similar treatment regimens may be used for other subjects with reduced or impaired muscle regeneration capabilities, such as geriatric subjects.
When particular methods of the present disclosure are used to prevent or mitigate muscle damage, such as damage caused by exertion or injury, the subject is typically treated a sufficient period of time before the exertion or injury in order to provide therapeutic effect. For example, the subject may be treated at least about 24 hours before the expected activity or potential injury, such as at least about 48 hours, about 72 hours, about 1 week, about 2 weeks, about three weeks, or about 4 weeks or more prior.
When embodiments of the method of the present disclosure are used to prevent or treat a muscle injury, one or more of the disclosed α7β 1 integrin modulatory agents or other therapeutic substance can be applied directly to, or proximately to, the area to be treated. For example, the substance can be injected into or near the area. In further examples, the substance can be applied topically to the area to be treated.
Treatment is typically initiated prior to the injury to several weeks following the injury. In more specific implementations, the treatment is initiated between about 12 and about 72 hours following injury, such as between about 24 and about 48 hours following injury. In some cases, a single administration of the substance is effective to provide the desired therapeutic effect. In further examples, additional
administrations are provided in order to achieve the desired therapeutic effect.
Amounts effective for various therapeutic treatments of the present disclosure may, of course, depend on the severity of the disease and the weight and general state of the subject, as well as the absorption, inactivation, and excretion rates of the therapeutically-active compound or component, the dosage schedule, and amount administered, as well as other factors known to those of ordinary skill in the art. It also should be apparent to one of ordinary skill in the art that the exact dosage and frequency of administration will depend on the particular α7β 1 integrin modulatory agent, or other therapeutic substance being administered, the particular condition being treated, the severity of the condition being treated, the age, weight, general physical condition of the particular subject, and other medication the subject may be taking. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in vivo administration of the pharmaceutical composition, and animal models may be used to determine effective dosages for treatment of particular disorders. For example, mouse models of muscular dystrophy may be used to determine effective dosages that can then be translated to dosage amount for other subjects, such as humans, as known in the art. Various considerations in dosage determination are described, e.g., in Gilman et ah, eds., Goodman And Gilman's: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press (1990); and Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa. (1990), each of which is herein incorporated by reference to the extent not inconsistent with the present disclosure.
In specific examples, the one or more disclosed α7β 1 integrin modulatory agents is administered to a subject in an amount sufficient to provide a dose of the agent of between about 10 fmol/g and about 500 nmol/g, such as between about 2 nmol/g and about 20 nmol/g or between about 2 nmol/g and about 10 nmol/g. In additional examples, the α7β 1 integrin modulatory agent is administered to a subject in an amount sufficient to provide a dose of between about 0.01 μg/kg and about 1000 mg/kg or between about 0.1 mg/kg and about 1000 mg/kg, in particular examples this amount is provided per day or per week. In another example, the disclosed α7β 1 integrin modulatory agent is administered to a subject in an amount sufficient to provide a dose of agent of between about 0.2 mg/kg and about 2 mg/kg. In further examples, the α7β 1 integrin modulatory agent is administered to a subject in an amount sufficient to provide a concentration of α7β 1 integrin modulatory agent in the administrated material of between about 5 nM and about 500 nM, such as between about 50 nM and about 200 nm, or about 100 nM. In other examples, the α7β 1 integrin modulatory agent is administered to a subject between about 500 μg/ml and about 1 μg/ml, such as about 300 μg/ml and about 3 μg/ml, about 200 μg/ml and about 20 μg/ml, including 500 μg/ml, 400 μg/ml, 300 μg/ml, 250 μg/ml, 200 μg/ml, 150 μg/ml, 100 μg/ml, 50 μg/ml, 25 μg/ml, 12.5 μg/ml, 6.25 μg/ml, 3.125 μg/ml, 2.5 μg/ml and 1.25 μg/ml. ii. Desired Response
One or more disclosed α7β1 integrin modulatory agents and/or additional therapeutic agents are administered by a specific route and/or concentration to generate a desired response. In some examples, a desired response refers to an amount effective for lessening, ameliorating, eliminating, preventing, or inhibiting at least one symptom of a disease, disorder, or condition treated and may be empirically determined. In various embodiments of the present disclosure, a desired response is muscle regeneration, reductions or prevention of muscle degeneration, promotion of muscle maintenance, reduction or prevention of muscle injury or damage, reduction or prevention in one more signs or symptoms associated with muscular dystrophy.
In particular, indicators of muscular health, such as muscle cell regeneration, maintenance, or repair, can be assessed through various means, including monitoring markers of muscle regeneration, such as transcription factors such as Pax7, Pax3, MyoD, MRF4, and myogenin. For example, increased expression of such markers can indicate that muscle regeneration is occurring or has recently occurred. Markers of muscle regeneration, such as expression of embryonic myosin heavy chain (eMyHC), can also be used to gauge the extent of muscle regeneration, maintenance, or repair. For example, the presence of eMyHC can indicate that muscle regeneration has recently occurred in a subject.
Muscle cell regeneration, maintenance, or repair can also be monitored by determining the girth, or mean cross sectional area, of muscle cells or density of muscle fibers. Additional indicators of muscle condition include muscle weight and muscle protein content. Mitotic index (such as by measuring BrdU incorporation) and myogenesis can also be used to evaluate the extent of muscle regeneration.
In particular examples, the improvement in muscle condition, such as regeneration, compared with a control is at least about 10%, such as at least about 30%, or at least about 50% or more, including an at least 15%, at least 20%, at least 25%, at least 30%, at least 40%, at least 45%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, including a 10% to 90% decrease, 20% to 80% increase, 30% to 70% increase or a 40% to 60% increase (e.g., a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100%, 200% or more increase).
Hi. Additional treatments or therapeutic agents
In particular examples, prior to, during, or following administration of an effective amount of an agent that reduces or inhibits one or more signs or symptoms associated with muscular dystrophy, the subject can receive one or more other therapies. In one example, the subject receives one or more treatments prior to administration of a disclosed α7β1 modulatory agent. Examples of such therapies include, but are not limited to, laminin-111 protein therapy, which works to stabilize the sarcolemma and reduce muscle degeneration. In some examples, a source of muscle cells can be added to aid in muscle regeneration and repair. In some aspects of the present disclosure, satellite cells are administered to a subject in combination with laminin therapy. U.S. Patent Publication 2006/0014287, incorporated by reference herein to the extent not inconsistent with the present disclosure, provides methods of enriching a collection of cells in myogenic cells and administering those cells to a subject. In further aspects, stem cells, such as adipose-derived stem cells, are administered to the subject. Suitable methods of preparing and administering adipose-derived stem cells are disclosed in U.S. Patent Publication 2007/0025972, incorporated by reference herein to the extent not inconsistent with the present disclosure. Additional cellular materials, such as fibroblasts, can also be administered, in some examples.
Additional therapeutic agents include agents which enhance the effect of the disclosed
α7β1 modulatory agents, such as a component of the extracellular matrix, such as an integrin, dystrophin, dystroglycan, utrophin, or a growth factor. In some examples, the additional therapeutic agent reduces or enhances expression of a substance that enhances the formation or maintenance of the extracellular matrix. In some examples, the additional substance can include aggrecan, angiostatin, cadherins, collagens
(including collagen I, collagen III, or collagen IV), decorin, elastin, enactin, endostatin, fibrin, fibronectin, osteopontin, tenascin, thrombospondin, vitronectin, and combinations thereof. Biglycans,
glycosaminoglycans (such as heparin), glycoproteins (such as dystroglycan), proteoglycans (such as heparan sulfate), and combinations thereof can also be administered.
In some examples, growth stimulants such as cytokines, polypeptides, and growth factors such as brain-derived neurotrophic factor (BDNF), CNF (ciliary neurotrophic factor), EGF (epidermal growth factor), FGF (fibroblast growth factor), glial growth factor (GGF), glial maturation factor (GMF) glial - derived neurotrophic factor (GDNF), hepatocyte growth factor (HGF), insulin, insulin-like growth factors, kerotinocyte growth factor (KGF), nerve growth factor (NGF), neurotropin-3 and -4, PDGF (platelet-derived growth factor), vascular endothelial growth factor (VEGF), and combinations thereof may be administered with one of the disclosed methods.
IV. Clinical Trials
To obtain regulatory approval for the use of one or more of the disclosed α7β1 modulatory agents to treat a muscular disorder, clinical trials are performed. As is known in the art, clinical trials progress through phases of testing, which are identified as Phases I, II, III, and IV.
Initially the disclosed α7β1 modulatory agent is evaluated in a Phase I trial. Typically Phase I trials are used to determine the best mode of administration (for example, by pill or by injection), the frequency of administration, and the toxicity for the compounds. Phase I studies frequently include laboratory tests, such as blood tests and biopsies, to evaluate the effects of the potential therapeutic in the body of the patient. For a Phase I trial, a small group of patients with a muscular disorder are treated with a specific dose of a disclosed α7β1 modulatory agent. During the trial, the dose is typically increased group by group in order to determine the maximum tolerated dose (MTD) and the dose-limiting toxicities (DLT) associated with the compound. This process determines an appropriate dose to use in a subsequent Phase II trial. A Phase II trial can be conducted to further evaluate the effectiveness and safety of the disclosed α7β1 modulatory agent. In Phase II trials, a disclosed α7β1 modulatory agent is administered to groups of patients with a muscular disorder using the dosage found to be effective in Phase I trials.
Phase III trials focus on determining how a disclosed α7β1 modulatory agent compares to the standard, or most widely accepted, treatment. In Phase III trials, patients are randomly assigned to one of two or more "arms". In a trial with two arms, for example, one arm will receive the standard treatment (control group) and the other arm will receive a disclosed α7β1 modulatory agent treatment (investigational group).
Phase Γ trials are used to further evaluate the long-term safety and effectiveness of a disclosed α7β1 modulatory agent. Phase Γ trials are less common than Phase I, II and III trials and take place after a disclosed α7β1 modulatory agent has been approved for standard use.
Eligibility of Patients for Clinical Trials
Participant eligibility criteria can range from general (for example, age, sex, type of disease) to specific (for example, type and number of prior treatments, disease characteristics, blood cell counts, organ function). In one embodiment, eligible patients have been diagnosed with a muscular disorder. Eligibility criteria may also vary with trial phase. Patients eligible for clinical trials can also be chosen based on objective measurement of a muscular disorder and failure to respond to other muscular disorder treatments. For example, in Phase I and II trials, the criteria often exclude patients who may be at risk from the investigational treatment because of abnormal organ function or other factors. In Phase II and III trials additional criteria are often included regarding disease type and stage, and number and type of prior treatments.
Phase I trials usually include 15 to 30 participants for whom other treatment options have not been effective. Phase II trials typically include up to 100 participants who have already received drug therapy, but for whom the treatment has not been effective.
Participation in Phase III trials is often restricted based on the previous treatment received. Phase III trials usually include hundreds to thousands of participants. This large number of participants is necessary in order to determine whether there are true differences between the effectiveness of a disclosed α7β1 modulatory agent and the standard treatment. Phase III can include patients ranging from those newly diagnosed with a muscular disorder to those with re-occurring signs and/or symptoms associated with a muscular disorder or a muscular disorder that did not respond to prior treatment.
One skilled in the art will appreciate that clinical trials should be designed to be as inclusive as possible without making the study population too diverse to determine whether the treatment might be as effective on a more narrowly defined population. The more diverse the population included in the trial, the more applicable the results could be to the general population, particularly in Phase III trials. Selection of appropriate participants in each phase of clinical trial is considered to be within the ordinary skills of a worker in the art. Assessment of patients prior to treatment
Prior to commencement of the study, several measures known in the art can be used to first classify the patients. Patients can first be assessed, for example by determining serum creatine kinase (CK) levels or other indicators of a muscle disorder, such as increased levels of muscle inflammation, apoptosis, muscle loss, myotube hypertrophy, and/or decreased myofibers stability and cell survival.
Administration of a disclosed α7β1 modulatory agent in Clinical Trials
A disclosed α7β1 modulatory agent is typically administered to the trial participants orally. A range of doses of the agent can be tested. Provided with information from preclinical testing, a skilled practitioner can readily determine appropriate dosages of agent for use in clinical trials. In one embodiment, a dose range is from about 100 μg/kg and about 5000 mg/kg of the subject's weight, such asl mg/kg and about 2000 mg/kg of the subject's weight, about 100 mg/kg and about 1500 mg/kg of the subject's weight, about 100 μg/kg and about 2000 mg/kg of the subject's weight, about 200 mg/kg and about 1000 mg/kg of the subject's weight, about 200 mg/kg and about 750 mg/kg of the subject's weight, about 250 mg/kg and about 500 mg/kg of the subject's weight, about 100 μπι and about 500 mM. In some embodiments, subjects are given a disclosed α7β1 modulatory agent orally at 10 to 60 mg/kg of body weight per day. For example, 10- 15mg/kg of a disclosed α7β1 modulatory agent is administered for two weeks and if well tolerated the dose is increased by 5-lOmg/kg/week to achieve optimal clinical response. In some examples, the daily dose does not exceed 60mg/kg of body weight and is given for a minimum of 6 months with liver function monitored every two weeks to monthly.
Pharmacokinetic monitoring
To fulfill Phase I criteria, distribution of the disclosed α7β1 modulatory agent is monitored, for example, by chemical analysis of samples, such as blood, collected at regular intervals. For example, samples can be taken at regular intervals up until about 72 hours after the start of treatment.
If analysis is not conducted immediately, the samples can be placed on dry ice after collection and subsequently transported to a freezer to be stored at -70 °C until analysis can be conducted. Samples can be prepared for analysis using standard techniques known in the art and the amount of the disclosed α7β1 modulatory agent present can be determined, for example, by high-performance liquid chromatography (HPLC). Pharmacokinetic data can be generated and analyzed in collaboration with an expert clinical pharmacologist and used to determine, for example, clearance, half-life and maximum plasma concentration. Monitoring of Patient Outcome
The endpoint of a clinical trial is a measurable outcome that indicates the effectiveness of a compound under evaluation. The endpoint is established prior to the commencement of the trial and will vary depending on the type and phase of the clinical trial. Examples of endpoints include, for example, decline in serum CK levels, inflammation, apoptosis, and muscle loss. For example, at least a 10% reduction in serum CK levels indicates the patient is responsive to the treatment. The following examples are provided to illustrate certain particular features and/or embodiments. These examples should not be construed to limit the invention to the particular features or embodiments described.
EXAMPLES Example 1
LacZ reporter gene in a7Pgal+ ~ muscle cells reports the transcriptional activity of the a7 integrin promoter
This Example shows that the LacZ reporter gene in a7Pgal+ " muscle cells faithfully reports the transcriptional activity of the l integrin promoter and can be used screen for α7β1 enhancer molecules.
An a l integrin null mouse was produced in which exon 1 of the gene encoding the a 7 integrin was replaced by the LacZ reporter. In these mice, all the transcriptional regulatory elements of the a l integrin promoter were retained, allowing β -galactosidase to report expression of 7 integrin. A primary myoblast cell line (designated a7Pgal+ ") isolated from 10 day old al+l~ pups were analyzed for the ability of β - galactosidase to report al integrin expression. a7Pgal+ " myoblasts were differentiated and subjected to X-gal staining and western analysis (FIGS. 1 A and IB).
These results demonstrate that β-galactosidase expression in a7Pgal+ " muscle cells increased upon myogenic differentiation consistent with the expression pattern of al integrin in myoblasts and myotubes. These results confirm that the LacZ reporter gene in a7Pgal+ " muscle cells faithfully reports the
transcriptional activity of the al integrin promoter. The activity of the al integrin promoter was measured by β-galactosidase cleavage of the non-fluorescent compound fluorescein di- β-D-galactopyranoside (FDG) to fluorescein. This assay was used to screen compound libraries to identify molecules that up-regulate al integrin expression in muscle.
Example 2
Identification of compounds that promote α7β1 integrin expression
This example describes multiple compounds identified as α7β1 integrin expression enhancers.
Using the muscle cell based assay described in Example 1 , the following compound libraries were screened: Prestwick Chemical and Microsource Spectrum Libraries from BioFocus DPI (Leiden Netherlands with facilities in UK, Basel, Heidelberg); the DrVERSet library (Chembridge Corp., San Diego, CA) and compounds from the ChemDiv library. Also evaluated was the effect of various isoforms of laminin and the ligand for α7β 1 integrin on integrin expression. Positive hits were subjected to dose-response analysis, western analysis and a myostatin counterscreen (a negative regulator of muscle growth). To quantify myostatin expression a western blot based assay was utilized (however, an ELISA assay is now available). α7βgal+/- and C2C12 myotubes were treated with the optimal drug concentration (see Table 8 below) for 24 hours, conditioned media removed and subjected to western analysis using an anti-myostatin antibody (AB3239, Millipore). As a positive control, cells were treated with 200mM Dexamethasone, which has been shown to increase myostatin expression in C2C12 myotubes. The results indicate that at the ECioo
concentrations used, none of the lead compounds increased myostatin expression.
From these studies nine molecules were identified that increased a7 integrin using our muscle cell- based assay as summarized in Table 8.
Table 8: Molecules identified as enhances of a7 integrin expression in muscle
Figure imgf000082_0001
Example 3
Intramuscular injection of laminin-111 prevents muscular dystrophy in mdx mice
This example demonstrates that intramuscular injection of laminin-111 prevents muscular dystrophy in mdx mice. Although this example describes studies particular to laminin-111 it is contemplated that similar studies can be performed based upon the methods described herein and the optimal concentrations of the particular α7β1 intregin enhancer molecules provided in Table 8 for the other α7β1 intregin enhancer molecules and similar effects on muscular dystrophy are predicted.
The ability of laminin to regulate a7 integrin expression, a7Pgal+ " myoblasts were exposed to 0- 200nM laminin-111 for 24 hours. The activity of the a7 integrin promoter was measured by β-galactosidase cleavage of the non-fluorescent compound fluorescein di-P-D-galactopyranoside (FDG) to fluorescein. Fluorescence activated cell sorting (FACS) demonstrated that a7Pgal+ " myoblasts treated for 24 hours with lOOnM laminin-111 produced the maximal increase in a7 integrin promoter activity. These results indicate laminin-111 promotes expression of a7 integrin in isolated mouse muscle cells. The ability of laminin-111 to increase a7 integrin expression was confirmed by western analysis using mouse and human DMD muscle cells (FIGS. 2A-2D). These data indicate that the mechanism by which laminin-111 increases a7 integrin expression is conserved between mouse and human muscle cells and suggests that laminin-111 is highly likely to increase α7β 1 integrin expression in the skeletal muscle of DMD patients.
To determine if laminin-111 prevented muscle pathology in mdx mice, Evans blue dye (EBD) uptake and Hemotoxylin and Eosin (H&E) staining were performed on cryosections from PBS and laminin- 111 injected TA muscle (FIG. 3A). Analysis revealed that mdx muscles injected with laminin-111 had a 12- fold reduction in the percentage of fibers positive for EBD compared to the contralateral controls (FIGS. 3B & 3C). In addition, mdx muscles injected with laminin-111 showed a 4-fold decrease in the percentage of muscle fibers with centrally located nuclei (FIG. 3C). These results indicate intramuscular injection of laminin-111 protein dramatically increased sacrolemmal integrity and reduced myofiber degeneration.
Injection of laminin-111 protein into the mdx mouse model of DMD increased expression of a7 integrin, stabilized the sarcolemma, restored serum creatine kinase to wild-type levels and protected muscle from exercise induced damage. These findings demonstrate that laminin-111 is a highly potent therapeutic for the mdx mouse model of DMD and represents a paradigm for the systemic delivery of extracellular matrix proteins as therapies for genetic diseases.
Example 4
Valproic Acid as a treatment for Muscular Dystrophy
This example describes studies indicating the ability of valproic acid to be used to treat muscular dystrophy.
Valproic acid (VP A) is a branched chain fatty acid that is FDA approved for treating epilepsy and bipolar disorders. VPA activates Akt in neurons and promotes their survival is also known to have histone deacetylase (HDAC) inhibitor activity. Using our muscle cell-based assay we identified that valproic acid activates a7 integrin expression in muscle cells. Valproic acid gave a dose-response curve and increased a7 integrin in C2C12 myotubes (FIGS. 4 A and 4B). Mdx/utrA mice treated with Valproic Acid showed reduced muscle disease, improved mobility, reduced fibrosis and activation of the Akt signaling pathway in muscle. These results indicate that valproic acid is a candidate for the treatment of DMD.
Example 5
Ciclopirox, Deferoxamine and 2,2-dipyridyl increase a7 integrin expression
This example shows that ciclopirox, 2,2-dipyridyl and deferoxamine increase a7 integrin expression through a common pathway.
As stated in Example 2 ciclopirox and deferoxamine are as activators of a7 integrin promoter activity using a7Pgal+ " myotubes. Both ciclopirox and deferoxamine are iron chelating drugs. Ciclopirox was independently identified in two compound libraries and is FDA approved as an antibiotic and antifungal drug. Deferoxamine is an FDA approved drug used to treat iron toxicity. Typical dose-response curves were obtained for both ciclopirox and deferoxamine with EC50 of 0^g/ml and ΙΟμΜ respectively (FIG. 5). A dose response curve for 2,2-dipyridyl (also an iron chelating molecule, but is not FDA approved) is shown in FIGS. 6A and 6B.
These results suggest ciclopirox, deferoxamine and 2,2-dipyridyl act through a common pathway to activate a7 integrin expression. Ciclopirox, deferoxamine and 2,2-dipyridyl have been shown to increase stability of the transcription factor hypoxia inducible factor- 1 (HIF-1) by preventing its breakdown. To determine if a7 integrin expression was responsive to HIF-1, bioinformatic analysis was performed on the a7 integrin promoter. A 2.8 kb fragment of the mouse a7 integrin promoter was analyzed using
MATINSPECTOR (Genomatix) for the presence of HIF-1 binding sites. The consensus DNA sequence for HIF-1 binding in the hypoxia-response element is 5'-[-A/G]CGTG-3' flanked with or without a second consensus site 5'-[A/C]ACAG-3'. Analysis of the a7 integrin promoter sequence revealed the presence of a HIF-1 binding site along with flanking sequences that promote HIF-1 binding. MATINSPECTOR analysis gave these sequences a perfect score for HIF-1 binding. These results suggest ciclopirox, deferoxamine and 2,2-dipyridyl may act to increase a7 integrin gene expression by inhibiting proteosomal breakdown of HIF- 1 in muscle cells resulting in increased cellular levels of HIF-1 protein. The overall result would increase a7 integrin protein on the surface of muscle cells increasing membrane stability. Further studies to determine if these drugs can experimentally increase HIF-1 protein levels in muscle cells need to be undertaken to add support to this mechanism of drug action. Interestingly, increased HIF-1 levels are associated with increased angiogenesis. Increasing HIF-1 levels in dystrophic muscle may not only increase membrane stability through increased a7 integrin expression, but increased muscle vascularization, improving blood flow and reducing the ischemia associated with dystrophic muscle.
DMD primary myotubes were exposed to the iron chelators 2,2-dipyridyl (31.25 μΜ) and deferoxamine (5 and 10 μΜ) for 132 hrs to determine if they increased a7 integrin. Protein was extracted from the cells and subjected to western blotting using antibodies against a7 integrin. a-tubulin was used as a loading control. Results showed that both 2,2-dipyridyl and deferoxamine increased al integrin in DMD myotubes (FIG. 7). These results indicate the mechanism(s) by which the iron chelators act to increase integrin expression are conserved between mouse and human muscle cells.
Example 6
Cholestan (5a-cholestan-3P-ol-6-one) and Cholestan analogs effect on al integrin expression
This Example demonstrates the ability of cholestan and specific cholestan analogs to increase al integrin expression in myoblasts and myotubes.
The studies described in Example 2 identified cholestan as an enhancer of al integrin expression as determined by the muscle cell based assay. Cholestan is a plant-derived compound of unknown function and gave a typical dose-response curve using a7Pgal+ " myotubes and increased a7 integrin protein in DMD myotubes (FIG. 8). In addition, specific analogs of cholestan retained the ability to activate expression of α7β1 integrin (see Table 9 below). One hundred and nineteen analogs of cholestan were obtained from Chemical Diversity laboratories and assessed for their ability to activate the expression of the al integrin in a7betagal+/- myoblasts and myotubes. Four of 119 analogs retained the ability to activate a l integrin expression in myoblasts and myotubes, and an additional 4 of 119 retained the ability to activate a l integrin expression in myoblasts only (Table 9).
Table 9: Analogs of cholestan that retained the ability to activate expression of the al integrin
Figure imgf000085_0001
Example 7
Compounds# 1001, 1002 & 1003 increased a7 integrin expression in muscle
Using the muscle cell based assay to screen the DIVERSet library of compounds (as described in Example 2), three compounds designed 1001, 1002 and 1003 all increased a7 integrin expression in muscle. Compound Nos. 1001, 1002 and 1003 are commercially available from ChemBridge Corporation (San Diego, CA). Compound No. 1001 is 3-methyl-2-[(2-oxo-2-phenylethyl)thio]-3H- spiro[benzo[h]quinazoline-5,l"-cyclopentan]-4(6H)-one (MW=417). Compound No. 1002 is l-{2-[3-(4- methyl-l-piperazinyl)propoxy] Phenyl} -1-propanone hydrochloride (MW=327). Compound 1003 is 1 {2-[3- (l-piperidinyl)propoxy}phenyl}-l-propanone hydrochloride (MW=312).
The dose-response curves to activate integrin expression by these compounds (as well as the chemical structures of such compounds) are provided in FIG. 9. These studies demonstrate the ability of such compounds to increase al integrin expression in muscle and support their use as agents to regulate al integrin modulated conditions, including muscular dystrophy. It is contemplated that analogs of compounds 1001, 1002 and 1003 could have similar effects. For example, it is contemplated that analogs of compounds 1001, 1002 and 1003 could be synthesized such as by the synthesis pathway provided in FIG. 10 and evaluated by the muscle cell base assay provided in Example 1 to determine their effects on al integrin expression in muscle.
Example 8
Laminin-111 effects on al Integrin Expression and Muscular Dystrophy
The studies described in this example demonstrate the effectiveness of LAM- 111 to increase al integrin levels and ameliorate the symptoms of disease in mdx and dyw mice. This data is included to demonstrate that enhancement of the al integrin is possible in vivo and as a therapeutic for muscular dystrophy. Thus, any of the disclosed α7β1 enhancers are believed to have similar in vivo effects as LAM- 111 and thus, useful therapeutic agents for muscular dystrophy.
FACS analysis demonstrated that a 24 hour treatment of a7betapi-gal +/- myoblasts with lOOnM LAM-111 and a fluorescent β-gal substrate resulted in a nearly 10-fold increase in al integrin expression compared to PBS treatment (FIGS. 12A-12D). Immunoblot analysis demonstrated that a 24 hour treatment of C2C12 or DMD myoblasts with lOOnM LAM-111 resulted in an approximately 2-fold increase in a7B integrin expression compared to PBS treatment (FIGS. 13A-13D). Overall, these results demonstrated that application of LAM-111 to myogenic cells results in an enhancement of al integrin protein expression that acted at the level of transcription or mRNA stability. It is contemplated that the identified parent scaffolds (Tables 8 and 9) increase al integrin expression through a similar mechanism.
One intramuscular dose of lOOnM (lOOul) of purified LAM-111 into the tibialis anterior (TA) of 10 day old mdx mice resulted in distribution of LAM-111 to all myoflbers, and resulted in a substantial reduction in the number of centrally nucleated and Evans blue dye (EBD) positive myofibers (FIGS. 14A- 14W). These studies demonstrate that intramuscular delivery of LAM-111 protects mdx myofibers from degeneration.
Immunofluorescence analysis of both mdx treatment groups demonstrated enhancements of a7A and a7B integrin, and utrophin compared to wild-type animals, and LAM-111 treated mdx mice demonstrated a further increase in a7A and a7B integrin, and utrophin beyond PBS-treated mdx mice (FIGS. 14A-14W). Densitometry of immunoblots from protein extracts of PBS and LAM-111 treated TA demonstrated that both mdx treatment groups showed a statistically significant increase in expression of a7A and a7B integrin and utrophin compared to wild-type animals, and treatment of mdx mice with LAM-111 resulted in a further 100% increase in a7A, a 50% increase in a7B and 33% increase in utrophin beyond PBS-treated mdx mice (FIGS. 15A-15D). Characterization of mdx/utro -I- dKO mouse expressing transgenic (rat) a7X2B integrin demonstrated that the 150% increase in a7X2B could fully account for the amelioration of disease, and is consistent with the presented data demonstrating that the enhanced expression of l integrin in LAM-111 treated mdx mice likely accounts for the observed therapeutic effect.
Four weeks following a single systemic dose (i.p) of 1.0 mg/kg LAM-111 to mdx mice, immunofluorescence analysis demonstrated continued localization of LAM-111 around all cardiac myofibers, and myofibers of the diaphragm and gastrocnemius (FIGS. 16A-16K). LAM-111 was absent from wild-type muscles and PBS-treated mdx mice (59). These studies demonstrate that systemic delivery of LAM-111 prevents sarcolemmal disruption of mdx myofibers.
Systemic treatment of mdx mice with LAM-111 demonstrated a near normalization of serum creatine kinase activity compared to PBS-treated mdx mice, and LAM-111-treated mdx mice showed no significant change in serum creatine or blood urea nitrogen (BUN) (FIGS. 17A-17C). These data suggest that a single systemic dose of LAM-111 provided a body- wide stabilization of sarcolemma integrity without affecting kidney function.
Ten day old mdx mice pretreated with a single systemic dose of PBS or 1.0 mg/kg LAM-111, and four weeks later subjected to eccentric downhill treadmill exercise, injection of Evan's blue dye and sacrifice 24 hours later were completely protected from sarcolemmal ruptures (FIGS. 18A-18C). These data suggest that a systemic delivery of LAM-111 protects myofibers from damaging eccentric exercise. It is contemplated that systemic delivery of the other disclosed α7β1 integrin expression enhancers would have similar protective effects on myofibers during exercise.
Treatment of mdx/utro -/- dKO mice with valproic acid resulted in activation of the AKT signaling pathway in muscle, improved mobility, reduced fibrosis, and reduced overall muscle disease. These results indicate that VPA is a candidate for the treatment of DMD and existing human safety data may expedite its development for treatment of DMD.
Systemically delivered LAM-111 localizes to skeletal muscle of dyw mice (FIG. 19-top panels), but required twice weekly doses to improve muscle pathology (FIG. 19-bottom panels) and reduce the percentage of myofibers containing centrally nucleated myofibers (FIG. 20A), Evans blue dye (FIG. 20B), and a TUNEL (apoptosis) reaction (FIG. 20C). Although body weights of LAM-111-treated dyw mice were not significantly different from PBS-treated dyw mice, PBS-treated dyw mice became moribund at 7 weeks of age (5.5 weeks post-injection) and had to be sacrificed while LAM-111-treated dyw mice sacrificed at 7 weeks of age as age matched controls were in much better health. These studies demonstrate that systemic delivery of LAM-111 prevents myofiber degeneration of dyw myofibers. It is contemplated that systemic delivery of the other disclosed α7β1 integrin expression enhancers would have similar effects on myofiber degeneration.
FIG. 30 is a digital image illustrating the results of quantitative real-time PCR used to assess Itga7, Itgbl, and Lama2 transcript levels in C2C12 myoblasts and myotubes treated for 24 hours with DMSO control, 10 μΜ MLS000683232-01 (IED-232), 10 μΜ MLS001165937-01 (IED-937), Hydroxylpropyl- Beta-Cyclodextrin (HPBCD) control, or 12μΜ SU9516 in HPBCD. FIG. 31 is a digital image of Western Blots and quantitative analysis of a7 Integrin and GAPDH protein levels in C2C12 myotubes treated for 48 hours with DMSO control, 10 μΜ MLS000683232-01 (IED-232), Hydroxylpropyl-Beta-Cyclodextrin (HPBCD) control, or 12μΜ SU9516 in HPBCD. Bands were quantified using Image J software and then graphed as l Integrin protein levels relative to GAPDH protein levels. * denotes a significant difference in relative protein levels with ** p <0.01.
Example 9
Additional Compounds for increasing al integrin expression in muscle
This example provides the structures (Table 11 below), exemplary synthesis reactions (FIGS. 29A- 29H) and characterization studies (see FIGS. 21-28) for additional compounds for increasing a l integrin expression in muscle. In some examples, the analogs were made in 5 mg quantities, salt form (e.g., hydrochloride salt), as a dry powder, at an at least 90% purity as measured by HPLC. Other exemplary compounds are provided below in Table 10. Results of screens performed with particular embodiments of the disclosed compounds are provided in FIG. 32.
Table 10: Exemplary Compounds
Figure imgf000088_0001
Potency: 1.122 Potency: 3.5481 Efficacy: 198.146 Efficacy: 228.085 Compound 1 Compound 2
Figure imgf000089_0001
Potency: 2.8184
Potency: 2.2387 Efficacy: 129.298 Potency: 1.122 Efficacy: 193.433 Compound 4 Efficacy: 92.4838 Compound 3 Compound 5
Figure imgf000089_0002
Potency: 4.4668 Potency: 2.8184
Potency: 1.122
Efficacy: 94.9208 Efficacy: 111.685 Efficacy: 128.12
Compound 7 Compound 8 Compound 6
Figure imgf000089_0003
Potency: 2.8184 Potency: 3.9811
Potency: 3.1623
Efficacy: 122.703 Efficacy: 101.893
Efficacy: 102.022
Compound 9 Compound 11
Compound 10
Figure imgf000089_0004
Potency: 4.4668 Potency: 2.2387
Potency: 2.8184
Efficacy: 121.996 Efficacy: 84.7736 Efficacy: 92.9057
Compound 13 Compound 14 Compound 12
Figure imgf000090_0001
Potency: 3.1623 Potency: 2.5119
Potency: 2.2387
Efficacy: 91.3808 Efficacy: 96.1948
Efficacy: 115.349
Compound 15 Compound 17
Compound 16
Figure imgf000090_0002
.579
Potency: 10 Efficacy: 81.6138 Efficacy: 101 Efficacy: 1751.56 Compound 19 Compound 20 Compound 18
Figure imgf000090_0003
Potency: 3.9811
Efficacy: 83.4252
Potency: 5.6234 Efficacy: 85.1623 Efficacy: 92.2825 Compound 22
Compound 23 Compound 21
Figure imgf000091_0001
Potency: 2.2387 Potency: 1.9953
Potency: 3.1623 Efficacy: 82.8995 Efficacy: 95.7363 Efficacy: 76.9771 Compound 25 Compound 26 Compound 24
Figure imgf000091_0002
Potency: 1.9953 Potency: 2.8184
Efficacy: 81.8506 Potency: 3.9811
Efficacy: 92.0171
Compound 27 Efficacy: 81.1658
Compound 28
Compound 29
Figure imgf000091_0003
Potency: 2.8184
Potency: 2.5119
Potency: 3.9811 Efficacy: 88.796
Efficacy: 86.4354
Efficacy: 92.7977 Compound 32
Compound 31
Compound 30
Figure imgf000091_0004
Potency: 1.9953 Potency: 1.4125 Potency: 2.5119 Efficacy: 83.3258 Efficacy: 70.4537 Efficacy: 72.7515 Compound 33 Compound 34
Compound 35
Figure imgf000092_0001
Efficacy: 75.7047
Compound 38
Potency: 2.5119
Compound 36
Efficacy: 77.0458
Compound 37
Figure imgf000092_0002
Potency: 14.1254
Potency: 14.1254
Potency: 3.5481 Efficacy: 447.862
Efficacy: 436.691
Efficacy: 78.5404 Compound 41
Compound 40
Compound 39
Figure imgf000092_0003
Compound 42 Compound 43
Potency: 11.2202 Efficacy:328.277 Compound 44
Figure imgf000093_0001
Potency: 7.9433 Efficacy: 141.317
Potency: 14.1254
Efficacy: 231.415 Compound 47 Efficacy: 426.535
Compound 46
Compound 45
Figure imgf000093_0002
Potency: 7.0795 Potency: 7.9433 Efficacy: 214.145 Potency: 5.6234 Efficacy: 181.323 Compound 48 Efficacy: 150.998 Compound 50
Compound 49
Figure imgf000093_0003
Efficacy: 273.425 Potency: 4.4668
Compound 51 Efficacy: 165.293 Potency: 7.0795
Compound 52 Efficacy: 163.55
Compound 53
Figure imgf000093_0004
Potency: 12.5893 Efficacy: 88.764 Potency: 12.5893 Efficacy: 271.246 Compound 56 Efficacy: 222.708 Compound 55
Compound 54
Potency: 4.4668
Figure imgf000094_0001
Efficacy: 115.732 Potency: 3.5481
Compound 57 Efficacy: 137.991 Potency: 10
Compound 58 Efficacy: 188.981
Compound 59
Figure imgf000094_0002
Potency: 4.4668 Potency: 5.0119
Potency: 7.0795 Efficacy: 128.904 Efficacy: 140.322
Efficacy: 186.257 Compound 60 Compound 61
Compound 62
Figure imgf000094_0003
Potency: 10
Potency: 5.0119 Potency: 19.9526 Efficacy: 200.43
Efficacy: 134.919 Efficacy: 491.985 Compound 63
Compound 64 Compound 65
Figure imgf000094_0004
Potency: 12.5893
Potency: 12.5893 Potency: 10
Efficacy: 239.617 Efficacy: 215.25 Efficacy: 157.616
Compound 68 Compound 66 Compound 67
Potency: 3.9811
Figure imgf000095_0001
Efficacy: 115.234
Efficacy: 90.5081
Potency: 7.9433 Compound 70
Compound 69
Efficacy: 124.512 Compound 71
Figure imgf000095_0002
Efficacy: 109.274 Efficacy: 138.663
Compound 72 Potency: 12.5893 Compound 74
Efficacy: 170.328
Compound 73
Figure imgf000095_0003
Potency: 4.4668
Potency: 3.9811 Potency: 6.3096
Efficacy: 121.35 Efficacy: 71.4431 Efficacy: 114.425
Compound 77 Compound 75 Compound 76
Figure imgf000096_0001
Potency: 10 Potency: 10
Potency: 4.4668
Efficacy: 152.941 Efficacy: 175.516
Efficacy: 85.208
Compound 78 Compound 80
Compound 79
Figure imgf000096_0002
Efficacy: 213.226 Potency: 3.1623 Efficacy: 175.054 Compound 81 Efficacy: 96.4736 Compound 83
Compound 82
Figure imgf000096_0003
Potency: 6.3096
Potency: 5.0119
Efficacy: 122.242 Potency: 4.4668
Efficacy: 99.705
Compound 84 Efficacy: 94.2018
Compound 85
Compound 86
Figure imgf000096_0004
Potency: 5.0119 Potency: 8.9125
Efficacy: 66.4058 Efficacy: 88.9864 Potency: 5.0119 Compound 87 Efficacy: 86.1979
Compound 88
Compound 89
Figure imgf000097_0001
Compound 90 Potency: 4.4668
Potency: 11.2202
Efficacy: 75.211 Efficacy: 139.097
Compound 92 Compound 91
Figure imgf000097_0002
Efficacy: 98.2729
Potency: 11.2202 Potency: 10
Efficacy: 117.921 Efficacy: 128.349 Compound 95 Compound 93 Compound 94
Figure imgf000097_0003
Efficacy: 83.068 Efficacy: 78.6154
Potency: 10 Compound 96 Compound 97
Efficacy: 125.681 Compound 98
Figure imgf000098_0001
Potency: 10 Potency: 8.9125 Efficacy: 121.073 Potency: 3.9811 Efficacy: 131.53 Compound 99 Efficacy: 95.5318 Compound 101
Compound 100
Figure imgf000098_0002
Potency: 6.3096
Potency: 12.5893 Potency: 10
Efficacy: 119.897
Efficacy: 149.767 Efficacy: 137.595
Compound 103
Compound 102 Compound 104
Figure imgf000098_0003
Potency: 10
Efficacy: 96.3714 Potency: 7.0795 Potency: 4.4668 Compound 105 Efficacy: 81.3792 Efficacy: 95.5906
Compound 106 Compound 107
Figure imgf000098_0004
Compound 108 Efficacy: 166.495 Potency: 12.5893
Compound 109 Efficacy: 180.783 Compound 110
Figure imgf000099_0001
Potency: 7.0795 Potency: 5.6234 Potency: 10 Efficacy: 99.2205 Efficacy: 73.9187 Efficacy: 111.758 Compound 111 Compound 112 Compound 113
Figure imgf000099_0002
Potency: 7.0795 Potency: 8.9125
Potency: 11.2202
Efficacy: 85.5436 Efficacy: 104.576 Efficacy: 138.65
Compound 115 Compound 116 Compound 114
P
Figure imgf000099_0003
Efficacy: 88.998 Efficacy: 127.904
Potency: 5.0119
Compound 117 Compound 119
Efficacy: 82.7908
Compound 118
Figure imgf000099_0004
Potency: 8.9125
Potency: 10 Potency: 10
Efficacy: 92.062
Efficacy: 114.827 Efficacy: 127.132
Compound 121
Compound 120 Compound 122
Figure imgf000100_0001
Potency: 11.2202
Potency: 8.9125 Potency: 8.9125 Efficacy: 136.36 Efficacy: 92.918 Efficacy: 123.416 Compound 125 Compound 123 Compound 124
Figure imgf000100_0002
Potency: 8.9125 Potency: 12.5893
Potency: 14.1254 Efficacy: 124.439 Efficacy: 171.242
Efficacy: 138.869 Compound 126 Compound 127
Compound 128
Figure imgf000100_0003
Potency: 14.1254
Efficacy: 119.808
Potency: 11.2202 Efficacy: 175.002
Compound 130
Efficacy: 145.936 Compound 131 Compound 129
Figure imgf000100_0004
Efficacy: 100.712 Efficacy: 95.4818
Potency: 12.5893 Compound 133 Compound 134 Efficacy: 120.609
Compound 132
Figure imgf000101_0001
Efficacy: 91.1222 Potency: 8.9125
Compound 135 Efficacy: 89.1179 Potency: 10
Compound 136 Efficacy: 81.023
Compound 137
Figure imgf000101_0002
Potency: 11.2202 Potency: 10
Potency: 11.2202
Efficacy: 135.363 Efficacy: 81.9056
Efficacy: 67.3145
Compound 138 Compound 140
Compound 139
Figure imgf000101_0003
Potency: 10 Efficacy: 76.5877
Potency: 8.9125
Efficacy: 100.669 Compound 143
Efficacy: 89.9548
Compound 141 Compound 142
Figure imgf000102_0001
Potency: lOEfficacy:
Efficacy: 94.0383
69.9943 Potency: 10 Compound 144
Compound 145 Efficacy: 96.4095
Compound 146
Figure imgf000102_0002
Efficacy: 90.1504 Efficacy: 138.741
Potency: 8.9125 Compound 147 Compound 148
Efficacy: 84.3701 Compound 149
Figure imgf000102_0003
Efficacy: 97.2406
Potency: 10
Potency: 10 Compound 152 Efficacy: 67.6116
Efficacy: 91.5246
Compound 150
Compound 151
Figure imgf000103_0001
Potency: 7.9433 Potency: 10
Potency: 3.1623
Efficacy: 95.7207 Efficacy: 93.0585
Efficacy: 93.8685
Compound 153 Compound 155
Compound 154
Figure imgf000103_0002
Potency: 10
Potency: 10 Efficacy: 70.1776
Efficacy: 71.256 Efficacy: 67.8359 Compound 157
Compound 158 Compound 156
Figure imgf000103_0003
Potency: 11.2202
Potency: 11.2202
Efficacy: 109.576 Potency: 3.9811
Efficacy: 103.065
Compound 159 Efficacy: 97.9916
Compound 160
Compound 161
Figure imgf000104_0001
Efficacy: 87.0645 Potency: 10 Compound 162 Potency: 10 Efficacy: 90.988
Efficacy: 79.1483 Compound 164 Compound 163
Figure imgf000104_0002
Potency: 7.0795
Potency: 11.2202 Potency: 12.5893
Efficacy: 86.8199 Efficacy: 128.695 Efficacy: 93.7937
Compound 167 Compound 165 Compound 166
Figure imgf000104_0003
Potency: 8.9125 Potency: 10
Potency: 10
Efficacy: 83.6415 Efficacy: 84.3344
Efficacy: 84.5587
Compound 168 Compound 170
Compound 169
Figure imgf000104_0004
Potency: 11.2202
Efficacy: 65.8043
Potency: 10 Efficacy: 85.4536
Compound 172
Efficacy: 59.61 Compound 173 Compound 171
Figure imgf000105_0001
Potency: 11.2202
Potency: 5.6234 Potency: 10 Efficacy: 104.045
Efficacy: 116.69 Efficacy: 115.567 Compound 174
Compound 175 Compound 176
Figure imgf000105_0002
Efficacy: 70.6131
Potency: 11.2202 Potency: 5.6234 Compound 177
Efficacy: 107.982 Efficacy: 84.0774 Compound 178 Compound 179
Figure imgf000105_0003
Potency: 10
Potency: 12.5893 Potency: 10 Efficacy: 77.6845
Efficacy: 80.5239 Efficacy: 99.1066 Compound 180
Compound 181 Compound 182
Figure imgf000105_0004
Potency: 10
Potency: 10 Potency: 10
Efficacy: 86.1315
Efficacy: 98.6809 Efficacy: 117.72
Compound 184
Compound 183 Compound 185
Figure imgf000106_0001
Potency: 3.9811 Potency: 10 Efficacy: 93.5129 Potency: 8.9125
Efficacy: 88.0312 Efficacy: 120.104
Compound 186 Compound 188
Compound 187
Figure imgf000106_0002
Potency: 11.2202
Potency: 8.9125 Efficacy: 116.431 Efficacy: 115.598 Efficacy: 76.0346 Compound 190 Compound 191 Compound 189
Figure imgf000106_0003
Potency: 11.2202 Potency: 8.9125
Potency: 11.2202 Efficacy: 89.7146 Efficacy: 76.8665 Efficacy: 74.0117 Compound 193 Compound 194 Compound 192
Figure imgf000106_0004
Potency: 10 Potency: 10
Potency: 8.9125 Efficacy: 99.4569 Efficacy: 67.0912
Efficacy: 83.2038 Compound 195 Compound 196
Compound 197
Figure imgf000107_0001
Efficacy: 78.0861 Potency: 12.5893
Potency: 8.9125 Compound 198 Efficacy: 96.0151
Efficacy: 98.6316 Compound 199
Compound 200
Potency: 10 Potency: 7.0795
Figure imgf000107_0002
Efficacy: 83.9426 Efficacy: 106.69
Potency: 11.2202 Compound 202 Compound 203 Efficacy: 94.7942
Compound 201
Figure imgf000107_0003
Potency: 10 Efficacy: 98.0553 Potency: 11.2202 Efficacy: 70.2778 Compound 205 Efficacy: 77.9394 Compound 204 Compound 206
Figure imgf000108_0001
Potency: 10
Efficacy: 75.3009 Potency: 10 Potency: 15.8489 Compound 207 Efficacy: 83.7154 Efficacy: 130.232
Compound 208 Compound 209
Figure imgf000108_0002
Potency: 11.2202
Potency: 12.5893 Potency: 14.1254
Efficacy: 109.939
Efficacy: 101.354 Efficacy: 119.538
Compound 211
Compound 210 Compound 212
Potency: 11.2202
Figure imgf000108_0003
Efficacy: 110.24
Compound 213 Potency: 11.2202 Compound 215
Efficacy: 94.2928
Compound 214
Figure imgf000108_0004
Potency: 10 Compound 217 Potency: 10 Efficacy: 90.7389 Efficacy: 58.0835 Compound 216 Compound 218
Figure imgf000109_0001
Potency: 11.2202
Potency: 11.2202 Potency: 11.2202 Efficacy: 59.5046 Efficacy: 83.9475 Efficacy: 86.9112 Compound 221 Compound 219 Compound 220
Figure imgf000109_0002
Potency: 12.5893
Efficacy: 56.9925
Compound 222
Table 11. Additional Exemplary Analogs
Figure imgf000110_0001
Compound 226(PRTH-002-F3) Compound 227 (PRTH-002-F5) Compound 228(PRTH-002-F6)
Figure imgf000110_0002
Compound 235
Compound 236 Compound 237
Figure imgf000111_0001
Compound 238 Compound 239 Compound 240(PRTH-002-F18)
Figure imgf000111_0002
Compound 241(PRTH-002-F19) Compound 242(PRTH-002-F21) Compound 243(PRTH-002-F20)
Compound 246(PRTH-002-F25)
Compound 245(PRTH-002-F23)
Compound 248(PRTH-002-F27) Compound 249(PRTH-002-F26)
Figure imgf000111_0003
Compound 250(PRTH-002-F28) Compound 251(PRTH-002-F29) Compound 252(PRTH-002-F30)
Figure imgf000112_0001
Compound 255(PRTH-002-F33)
Figure imgf000112_0002
Compound 258(PRTH-002-F36)
Figure imgf000112_0003
Compound 265 Compound 266 Compound 267
Figure imgf000113_0001
Compound 268 Compound 269
Table 12: Compound data
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000134_0001
Table 13: Results of particular embodiments in a a7+/" β-galactosidase assay.
Myotubes Myoblasts
40 uM 20 uM 10 uM 5 uM 1 uM 0.5 uM 40 uM 20 uM 10 uM 5 uM 1 uM 0.5 uM
0.135097 -0.13765 0.088 0.131633 0.033562 -0.0099
-0.29359 -0.31225 0.000591 0.071212 0.120068 -0.0152
0.011146 0.105576 0.373206 0.136432 0.031877 0.09422
-0.01563 -0.11816 0.089582 -0.03653 0.143592 0.057735
-0.11833 -0.04634 0.224019 0.012626 0.013996 0.07412
-0.13314 -0.2661 0.015256 -0.01977 0.013544 0.058626
-0.39149 -0.34143 0.134272 -0.157 0.066681 0.19903
-0.73405 -0.80511 -0.09235 0.04228 0.105211 0.006429
-0.12084 0.034604 0.264805 -0.02798 0.191188 0.17557 -0.25864 -0.31055 0.143282 0.19487 0.093218 0.06985
-0.24096 -0.25938 -0.12962 -0.19329 -0.15624 -0.1640
-0.2363 -0.28934 0.069762 0.001992 -0.06381 -0.13448
-0.75439 -0.51266 -0.25424 -0.12378 -0.03019 0.00673
-0.48225 -0.46526 -0.07782 0.090296 0.076333 0.09119
-0.15754 -0.23372 0.17046 0.062866 0.074199 0.21320
-0.34197 -0.4266 0.411821 0.400466 0.095524 0.172476
Figure imgf000135_0001
-0.12374 -0.02233 -0.06162 0.033033 0.019
■0.05014 -0.19448 0.135882 0.020076 -0.00083 -0.09933
0.198076 0.073533 0.458502 0.080717 0.082
■0.19824 -0.10019 -0.03138 0.649735 0.189224 0.15034
-0.65552 -0.13552 -0.03271 0.219322 0.221
■0.64093 -0.47479 -0.19573 -0.0494 0.180279 0.124844
0.215514 -0.03174 0.064915 0.135859 0.239
■0.23454 -0.15982 -0.05758 0.003264 0.262184 0.143442
-0.97306 -0.67646 -0.37656 0.222464 0.32
■0.92153 -0.76037 -0.30617 -0.09555 0.172088 0.138136
-0.39314 -0.12838 0.489673 0.143293 0.239
-0.2285 -0.3633 -0.07457 0.210712 0.137762 0.05381
-0.56078 -0.0702 -0.06784 0.038475 0.11
■0.33888 -0.47086 -0.05869 -0.07294 -0.08157 0.151
-0.09429 -0.03841 -0.01619 0.110062 0.209
■0.24428 -0.22569 0.095108 -0.03061 0.183361 0.258605
Figure imgf000136_0001
0.75277 -0.53309 -0.235 -0.10771 0.162123 0.
-0.37051 -0.24637 0.085559 0.146743 0.185925 -0.03985
-0.01341 -0.04001 0.038455 0.032026 0.041953 0.1
-0.27634 -0.15988 0.075267 0.048683 -0.11773 0.088184
0.188681 0.094472 -0.04766 0.015444 0.069116 0.06
-0.02803 0.014943 -0.00108 -0.02688 0.046048 0.092701
-0.35363 -0.43224 -0.7131 -0.39614 0.291572 0.2
-0.16397 0.0827 -0.36966 -0.34528 0.120102 0.266095
0.073674 0.101056 0.01243 0.079658 0.157036 0.06
-0.06707 -0.09449 0.01619 0.416848 0.073286 0.121796
0.083597 0.111292 -0.07816 0.843176 0.286217 0.28
-0.15716 -0.0871 0.005171 0.251369 0.279442 0.299517
0.074611 0.053868 -0.00558 0.009615 0.283453 0.11
-0.27114 -0.18133 -0.04013 -0.07324 0.060347 0.109646
0.018465 0.046507 0.042902 0.032767 0.18767 0.02
-0.0349 -0.0234 0.035774 0.00127 0.078993 0.11132
-0.28127 -0.25903 -0.22238 -0.06804 0.206728 0.14 -°-10004 -0.07223 -0.08652 -0.13541 0.166782 0.264735
Figure imgf000137_0001
-0.2221 -0.10276 -0.05318 0.012391 0.233656 0.1168
-0.25453 -0.13518 0.071763 -0.02107 0.267231 0.198007
-0.01244 0.021361 -0.00196 0.0782 0.155732 0.0394
-0.15172 -0.05815 0.095093 0.068865 0.050449 0.234537
-0.08865 -0.12747 -0.06761 -0.05162 -0.06237 -0.064
-0.18457 0.048471 -0.12383 -0.09085 0.159592 0.072803
-0.17575 0.847681 -0.00591 0.072202 0.049692 0.0252
0.02169 0.010905 -0.09985 0.054741 0.139347 0.023823
0.080917 0.064236 0.171922 0.22657 0.098435 0.0564
-0.06801 -0.03697 -0.07868 0.094888 0.164296 0.056524
0.090833 0.054743 -0.01279 0.185637 0.150725 0.0514
-0.06305 -0.01414 -0.06595 0.072041 0.049724 0.088204
0.009972 0.001292 0.161378 0.150441 0.15833 0.0035
-0.26942 -0.19324 -0.05774 -0.00214 -0.07012 -0.04746
-0.05323 0.014118 0.02292 0.058138 -0.04841 -0.096
-0.04958 -0.08844 0.051942 0.069463 -0.07814 0.013415
Figure imgf000138_0001
-0.10812 0.003125 -0.08302 0.074837 0.107025 0.015
0.0782 0.035783 0.136374 0.076122 0.054898
-0.21349 -0.13665 -0.01717 0.111136 0.282699 0.12
-0.13487 0.06011 0.068638 0.161122 0.168422
-0.07201 -0.07918 0.024274 0.054882 0.090007 -0.02
0.13419 0.248364 0.382213 0.254633 0.108633
0.65393 0.820614 0.77787 0.644496 0.198681 0.233 0.414904 0.439813 0.265254 0.3062 0.160963
-0.28667 -0.07832 0.244423 -0.02565 -0.13489 -0.05
-0.22374 0.057761 -0.0943 -0.2399 -0.15198
-0.16863 -0.06526 0.234694 0.019974 -0.08284 0.015 -0.19506 -0.05054 -0.10984 0.032674 0.017637
-0.67504 -0.35187 0.080153 -0.07445 -0.09662 0.050
-0.40931 0.024598 -0.04071 -0.0839 -0.01581
0.028632 0.143322 0.24505 0.003015 -0.1461 -0.10 -0.16347 0.120638 0.086879 -0.13507 0.054289
-0.0646 0.072591 0.197804 -0.06143 -0.17216 -0.14
-0.08104 0.153672 -0.01555 -0.17279 -0.10441
Figure imgf000139_0001
-0.76058 -0.21212 -0.29913 -0.25702 -0.15356 -0.109
-0.28269 -0.28352 0.051453 -0.09703 -0.0826 -0.02111
-0.02246 -0.05432 0.25085 -0.03005 -0.05578 -0.0448
-0.02391 -0.22177 0.295238 0.097632 -0.07237 0.068719
-0.0962 0.788591 0.209246 0.016025 -0.04183 -0.045 -0.11588 -0.23567 0.19467 0.174141 -0.13979 -0.02164
-0.69953 -0.21964 -0.18242 -0.08809 0.015373 0.0696
-0.33597 -0.23693 -0.06876 -0.10432 -0.02249 0.096556
-0.14646 0.106813 -0.14788 -0.01947 -0.01272 0.03731 -0.18882 -0.09507 -0.0441 -0.08739 0.126363 0.108741
-0.23114 -0.18165 0.084896 0.067865 0.112652 0.1257
-0.14917 -0.07572 0.235185 0.060902 0.224281 0.14183
-0.66375 -0.54784 -0.46893 -0.29232 0.000395 0.2021 -0.29308 -0.2624 -0.15279 -0.1525 0.032141 0.265081
-0.08344 0.112082 -0.05714 -0.07651 -0.07703 -0.0212
-0.37831 -0.25781 -0.03259 -0.0988 0.115844 0.090822
-0.85559 -0.70297 -0.68594 -0.51458 -0.10938 0.0023
-0.51241 -0.31216 -0.25602 -0.04505 0.068215 0.120763
Figure imgf000140_0001
-0.04959 0.102175 -0.08869 0.811881 -0.01442 0.0287
-0.10069 -0.13664 -0.02819 0.245116 0.283689 0.270319
-0.04457 0.004164 0.028129 -0.01266 0.060525 0.09281
-0.31266 -0.33246 0.039967 0.076685 0.231741 0.306852
-0.87802 -0.82207 -0.51959 -0.21018 0.018467 0.06463
-0.33208 -0.23448 -0.0108 -0.01197 0.059919 0.010207
-0.10483 0.001076 -0.10551 -0.08597 0.214522 0.08093
-0.08066 -0.09026 0.013186 0.208134 0.103185 0.066696
0.047848 0.068234 -0.0931 0.048546 0.069746 0.0132
-0.08927 -0.15794 -0.11534 -0.09583 0.045844 0.003237
-0.06265 -0.02713 -0.04687 0.034852 0.104522 0.0916
0.055247 -0.11249 -0.14177 -0.09921 0.1296 0.117044
-0.04372 0.040596 0.011511 0.069762 0.285804 0.2182
-0.03271 -0.12236 -0.06379 -0.00041 0.169889 0.156067
0.134561 0.074719 -0.02595 -0.00636 0.02871 -0.0551
-0.15412 -0.18578 -0.16756 -0.08801 0.076637 0.075422
0.133573 0.17252 -0.03134 0.043672 0.05388 -0.0675
-0.21901 -0.13446 -0.04332 -0.02718 0.001963 -0.00103
-0.58206 -0.22346 -0.20434 -0.04768 0.034616 -0.0040
-0.26816 -0.19957 -0.12515 -0.10684 0.267504 0.186237
-0.21801 -0.14707 -0.11737 -0.05885 0.025946 0.0496
-0.36895 -0.26036 -0.08357 -0.05597 0.163978 0.15943
-0.06105 -0.08756 -0.18044 -0.07708 0.114815 0.0187
-0.22322 -0.11681 -0.1274 -0.08884 0.040407 0.00543
Figure imgf000141_0001
-0.41325 -0.17872 -0.1014 0.135711 0.053475 0.3278
24855 -0.01998 -0.17763 -0.09861 0.077578 0.090874
-0.42243 -0.09846 0.028985 0.147716 0.094217 0.37572
0.118924 0.191591 0.110752 0.297473 0.048007 -0.09659
-0.1111 0.003655 0.119875 0.24866 0.079254 0.3095
-0.11452 -0.00606 -0.06084 0.011539 0.121889 0.096474
-0.19812 -0.15268 -0.05819 0.142705 0.152855 0.4282
-0.17373 -0.0413 -0.0808 -0.00258 0.102667 -0.04781
0.283836 0.107959 -0.11677 0.17276 -0.08687 0.1596
-0.25985 -0.15691 -0.1801 -0.02539 0.082067 -0.05035
0.01583 0.118433 0.00833 0.132927 -0.07182 0.1909 -0.11144 -0.02544 -0.11337 0.053417 0.065341 -0.04468
-0.97407 -0.81528 -0.64702 -0.19588 -0.06344 0.21103
-0.68216 -0.25532 -0.13025 0.085791 0.062267 0.041163
-0.42066 -0.20508 0.023769 0.020343 0.015899 0.2140
-0.42102 -0.32897 -0.05719 -0.04079 0.086022 0.093541
-0.22626 -0.0583 -0.01934 -0.05696 -0.03677 0.23611
-0.20899 0.102212 -0.02849 0.015311 0.180948 -0.07971
-0.19921 0.014064 0.625155 0.066573 -0.07696 -0.1174
-0.07044 -0.17348 0.173757 0.020072 -0.2515 -0.15654
Figure imgf000142_0001
-0.74437 -0.41839 0.100401 0.009111 -0.05193 -0.067
-0.42259 -0.33806 0.026465 -0.10894 -0.04077 -0.06236
-0.54989 -0.45598 0.046517 -0.26668 -0.19605 -0.1972
-0.26988 -0.21113 0.125746 -0.15714 -0.02413 -0.07617 0.020685 0.033365 0.244032 0.116328 0.04937 -0.0096
0.059398 -0.02703 0.214368 -0.03109 -0.02121 0.024638
Figure imgf000143_0001
-0.32257 -0.12703 0.169066 -0.01736 -0.15525 -0.1523
-U H -0.15824 -0.24433 0.227852 -0.04323 0.115277 0.080266
-0.10035 -0.09845 0.248169 0.043588 -0.1236 -0.0602
-0.2601 -0.22831 0.120281 -0.10858 0.093309 -0.12665 -0.5206 -0.51731 0.132098 -0.46216 -0.07551 -0.1172
-0.26567 -0.33853 0.326514 -0.11652 -0.03106 -0.05012
-0.31799 0.03268 0.700648 0.274722 -0.10511 -0.1311
-0.25176 0.038027 0.858522 0.388847 0.188277 -0.01143
-0.11458 -0.08803 0.169573 0.422768 -0.21245 -0.2242
-0.28033 -0.32057 0.232238 0.024718 -0.18128 -0.06701 -0.46725 -0.0797 0.488593 -0.11626 -0.07 0.03703
-0.28478 -0.21956 -0.28569 -0.24606 -0.16902 -0.04347
Figure imgf000143_0002
-0.03893 0.135863 -0.01836 -0.09981 -0.02931 0.0548
-0.01262 0.00778 -0.23324 -0.16801 -0.0238 -0.03749
-0.27397 -0.0419 -0.03065 -0.01841 0.058196 0.08805
-0.37289 -0.29705 -0.11541 -0.10416 0.106979 0.077404
-0.20381 0.192525 0.156466 0.080356 0.077815 0.2736
-0.15765 -0.0001 -0.09062 -0.08702 0.258223 0.29017
-0.03479 0.082644 -0.05691 -0.06002 -0.09205 0.0729
-0.22131 -0.08792 -0.17823 -0.15019 0.162681 0.180298
-0.22617 -0.06029 -0.02503 -0.04484 -0.00471 0.08931
-0.42418 -0.18142 -0.211 -0.31742 0.027319 -0.05788
-0.53042 -0.40387 -0.30434 -0.22009 -0.05717 -0.0262
-0.27272 -0.20468 0.029944 -0.02922 -0.05535 -0.03979
-0.08446 0.037338 -0.02381 0.374119 -0.10528 -0.0285
-0.22456 -0.18626 0.305427 -0.24734 0.016383 -0.02436
-0.09806 -0.03658 -0.05458 0.367031 -0.17043 -0.1296
-0.1392 -0.13534 -0.24914 0.029543 -0.0433 -0.18148
0.492521 0.316872 0.075794 0.024288 0.045591 0.00348
0.084957 0.079978 0.538134 -0.15279 -0.13073 -0.10986
Figure imgf000144_0001
-0.37649 0.632329 -0.18793 -0.04769 0.014928 0.0310
-0.38412 -0.14949 -0.10558 -0.09525 0.028574 -0.03464
°·°74776 °·180163 °·°8931 -0.02345 0.088315 0.2219
-0.02531 -0.05932 0.0522
^07402 °·170025 °·1046
Figure imgf000145_0001
^j ^cl -0.12335 -0.08151 0.044627 0.0600
°·0135 -0.04645 0.031867 -0.12019 0.04866 0.028511 °·120114 0.145443
-0.12022 -0.12833 -0.1125 -0.23369 0.03033 0.034191 ^00588 °·025087 "^02349 -0·00182 -°·°7538 -0·0598 0.046349 0.031565 -0.07161 -0.024 -0.03177 -0.05084 ^02456 0'613352 -°·°6661 °·475794 ^16097 "0-1 137
-0.0655 -0.06378 -0.05174 0.223537 -0.23737 -0.1173 -0.25868 0.01214 -0.13637 0.244112 -0.09286 -0.05672
0.048145 0.272817 -0.08232 0.630831 -0.13844 -0.1271 -™350200959 0 075732 °·7»2787 -°·03737 -°·0505
Figure imgf000145_0002
-0.15932 -0.1004 0.190709 0.37466 -0.00253 0.0209
-0.21623 0.032285 0.213771 0.505233 0.092351 0.057181
0.007087 0.035479 -0.04895 0.159095 0.20071 0.1485
0.067328 0.226559 -0.09211 0.29265 0.158809 0.0905
-0.60889 -0.43691 -0.2175 -0.06604 -0.01981 -0.0034
-0.13567 -0.01472 -0.03387 0.11985 0.05066 0.08584
-0.10343 -0.15378 -0.06828 0.060007 0.002953 0.00773
-0.17798 0.127215 -0.12308 0.156744 0.097585 0.222053
-0.04251 -0.02534 -0.09397 -0.11937 -0.01966 -0.0799
-0.09282 0.011484 -0.09429 0.035935 0.03833 0.083936
-0.38263 -0.27247 -0.17555 -0.02637 -0.26056 -0.1759
-0.09631 0.017419 -0.23012 -0.13721 0.048553 0.05467
-0.16941 -0.17397 -0.13737 0.001928 -0.21997 -0.1518
-0.36998 -0.14699 -0.09676 0.214356 -0.06589 -0.05564
-0.67155 -0.49001 -0.17811 -0.14266 -0.24382 -0.1557
-0.26129 -0.41316 0.050639 -0.08742 -0.22041 -0.06072
-0.44108 -0.16794 0.130143 0.025731 -0.14436 -0.0470
-0.39406 -0.32943 0.334811 0.050891 -0.09877 0.010128
-0.40261 -0.26611 -0.02616 -0.05693 -0.0996 0.0234
-0.15121 -0.24954 0.262262 0.036462 -0.15158 -0.19457
-0.47476 -0.335 0.154507 0.124102 -0.02919 0.0754
-0.184 -0.30618 0.26304 0.028289 -0.07984 -0.03323
Figure imgf000146_0001
-0.76529 -0.82778 -0.50782 -0.42068 -0.15434 -0.0505
0.208544 -0.13802 0.299236 -0.13446 -0.03964 0.030752
-0.17205 -0.01497 0.124156 0.016836 -0.06275 0.01301
0.231706 -0.16632 0.482484 0.152623 -0.1638 -0.07067
-0.07326 -0.02505 0.115487 0.003792 -0.02327 0.1058
0.239851 -0.16031 0.372282 -0.01338 -0.11882 -0.03474
-0.05859 -0.02695 0.172476 -0.04648 -0.06967 0.12331
0.078276 -0.21651 0.440789 0.046581 -0.04272 0.003071
-0.11564 -0.06607 0.095363 0.439771 -0.17363 0.0410
0.003215 -0.28859 0.451101 0.199815 -0.16331 -0.11554
-0.16114 -0.0939 -0.03481 0.008423 -0.14272 -0.0171
-0.14278 -0.07909 0.01498 -0.09733 -0.07688 -0.06746
-0.13903 -0.15525 -0.03652 -0.02435 0.02077 0.0766
-0.1688 -0.03447 0.055323 -0.14544 -0.01338 0.031021
-0.39231 -0.39598 -0.2656 -0.22733 -0.07654 0.0638
-0.80472 -0.31004 -0.10014 -0.19646 -0.01854 -0.12741
-0.65557 -0.49133 -0.01618 -0.06112 0.007377 0.1194
-0.19437 -0.10511 -0.18533 -0.24823 0.051262 -0.08852
Figure imgf000147_0001
-0.43164 0.433277 -0.11882 -0.06939 -0.04686 0.0508
-0.25938 -0.18203 -0.07084 -0.21066 -0.02073 0.22256 -0.28005 -0.2055 -0.1049 -0.08261 0.002443 0.1664
-0.807 -0.54297 -0.06955 -0.18512 -0.17031 -0.02866
-0.17515 0.087039 -0.0409 -0.00848 0.067902 0.21132
-0.50848 -0.31844 -0.06437 -0.27222 -0.05419 -0.06598
0.068022 0.138879 -0.09102 -0.07378 0.087503 0.2617
-0.14404 -0.16313 -0.10163 -0.10631 -0.05991 0.135113
-0.16722 -0.07784 -0.06877 0.398536 -0.02563 0.10863
0.117864 -0.22087 0.110761 0.007084 -0.16903 -0.01141
-0.25891 0.680714 -0.10897 -0.04166 -0.00739 -0.062 -0.54016 -0.36931 -0.16817 -0.09532 -0.0743 -0.06415
-0.29339 -0.24189 0.089865 0.044516 0.06141 0.0624 -0.58921 -0.33057 -0.24176 -0.20121 0.031738 0.012035
0.037476 0.070056 0.05082 0.131647 0.170683 0.0683
-0.03236 0.007978 -0.06524 -0.13777 0.105794 -0.01316
Figure imgf000148_0001
-0.79542 -0.79747 -0.38589 -0.23595 0.002645 -0.0217
-0.1921 -0.19378 -0.35848 -0.25502 0.129504 0.069213
-0.46616 -0.30837 -0.20535 -0.10195 0.009306 -0.0355
-0.31324 -0.26222 -0.44339 -0.42239 -0.01127 0.111865
-0.12946 -0.16661 -0.06749 -0.02764 0.050825 0.0323
-0.07634 -0.1393 -0.24829 -0.25468 0.016504 -0.09293
0.026753 0.025944 0.122911 0.16412 0.222536 0.15942
0.175649 -0.14293 0.303709 0.084449 0.198894 0.161688
-0.82567 -0.82768 -0.43631 -0.19335 0.15918 0.1702
-0.37053 -0.30668 -0.28824 -0.2641 0.054979 0.133901
-0.95114 -0.94412 -0.81115 -0.82504 -0.44546 -0.2195
-0.61611 -0.54331 -0.50779 -0.52012 -0.23505 -0.14067
-0.62751 -0.36487 -0.15749 0.272197 -0.09115 -0.1564
-0.28069 -0.12305 -0.05957 0.251367 -0.12469 -0.05394
-0.11165 0.052667 0.078208 0.272065 0.154978 -0.1200
-0.28885 -0.14442 -0.05189 0.199586 0.060071 0.070496
Figure imgf000149_0001
-0.11953 -0.1257 0.002758 0.306791 -0.06684 -0.2006
-0.06764 -0.05843 -0.16665 0.147588 -0.07498 -0.03601 -0.24698 -0.15335 -0.01305 0.280534 0.003197 -0.0838
-0.08699 -0.17626 -0.15503 0.152008 -0.01856 0.060121
-0.27161 -0.20826 -0.031 0.295491 -0.12124 -0.2127
-0.20634 -0.05892 -0.26379 0.044863 -0.0336 0.012695
-0.44485 -0.30888 -0.07468 0.038724 -0.07096 -0.2303
-0.29757 -0.11275 -0.31244 -0.25074 -0.12352 -0.08286
-0.3491 -0.21671 -0.06567 0.170298 0.035372 -0.0455
-0.17747 -0.17067 -0.31069 -0.09823 -0.01328 0.049078
-0.05546 -0.08958 -0.03674 0.129874 0.068852 -0.0843
-0.24068 -0.24192 -0.12288 -0.02123 0.024993 0.109064
0.116571 0.242268 0.125519 0.224732 -0.04939 -0.190
-0.10731 0.059746 0.169313 0.139503 -0.16139 -0.05273
0.194099 0.101568 0.050277 0.049819 0.055504 0.0766
0.147617 -0.1243 0.076768 -0.11318 -0.07437 -0.10104
-0.02989 -0.05956 0.083398 0.042438 -0.05498 -0.0081
0.102061 -0.18227 0.052838 -0.20674 -0.05977 -0.04135
-0.33783 -0.2863 0.066634 0.091249 0.181302 0.2482
-0.14399 -0.27981 0.127648 0.014718 -0.04907 -0.10179
Figure imgf000150_0001
-0.13522 -0.10895 0.048377 0.05318 0.197922 0.2561
0.009496 -0.12025 0.134491 -0.02208 -0.01895 0.008769
-0.57245 -0.48402 0.073235 0.035764 0.023039 0.0771
-0.41493 -0.41098 0.151031 -0.09703 -0.09775 -0.15406
0.088375 0.071297 0.114077 0.066133 0.018295 0.06548
0.036114 -0.1578 0.110443 -0.19931 -0.1068 -0.05289
-0.35325 -0.16952 -0.06595 -0.04422 0.027822 -0.0261
-0.37223 -0.36256 0.105686 -0.20886 -0.08264 -0.02905
-0.73838 -0.48286 -0.19484 -0.18426 -0.004 0.06263
-0.50502 -0.3617 -0.16795 -0.24429 -0.17637 -0.16258
0.107427 0.046604 -0.08672 0.358137 0.10445 0.0923
0.143636 -0.1092 0.117312 0.438014 -0.16267 -0.0989
-0.51981 -0.30724 -0.20684 -0.1204 0.134217 0.0973
-0.27922 -0.27925 -0.15162 -0.13289 -0.12861 -0.07388
-0.38236 -0.12559 -0.09387 -0.08497 0.045054 0.17882
-0.07473 -0.12475 -0.16204 -0.163 0.020952 0.015347
Figure imgf000151_0001
0.061797 0.124656 -0.03865 -0.03678 0.186132 0.2814
-0.07382 -0.09607 -0.08097 -0.09401 -0.05118 -0.00414
-0.77623 -0.60158 -0.3483 -0.26663 0.195791 0.40541
-0.81956 -0.51961 -0.31472 -0.15017 0.121456 0.108646
-0.34188 -0.09766 -0.17441 -0.13432 0.082496 0.1889
-0.89481 -0.57808 -0.28915 -0.38973 -0.24233 -0.14468
Figure imgf000152_0001
-0.41302 -0.19978 -0.10704 -0.08939 0.075721 0.1622
-0.4694 -0.20236 0.453591 -0.30877 0.017633 -0.09246
-0.27686 -0.08666 0.247787 -0.05605 0.056465 0.1360
-0.21559 -0.10854 -0.12921 -0.28226 -0.01629 0.046633
-0.45861 -0.03424 0.180663 -0.14881 0.192984 0.22103
-0.28555 -0.20885 -0.33998 -0.48398 -0.13132 -0.09222
0.225677 0.313656 -0.02658 0.300119 0.100519 0.2054
-0.05938 -0.05439 -0.19977 0.458591 -0.03363 -0.03535
0.240422 0.187406 -0.02723 0.020319 0.241791 0.1136
-0.03791 0.013527 -0.12823 -0.14146 -0.08287 -0.16031
-0.69145 -0.57753 -0.37416 -0.21279 0.019636 -0.0010
-0.2531 -0.25133 -0.41956 -0.38682 -0.04637 -0.02692
-0.0403 -0.01304 -0.09863 -0.03377 0.351326 0.2330
-0.20525 -0.14382 -0.26947 -0.25815 -0.06852 -0.05036
Figure imgf000152_0002
-0.24089 -0.17085 -0.16441 -0.03545 0.351744 0.2495
-0.43887 -0.33039 -0.37631 -0.23673 0.058231 -0.08689
0.064016 -0.00332 -0.00668 -0.02152 0.178442 0.09518
-0.25508 -0.22495 -0.30506 -0.2961 -0.16141 -0.17671
-0.11174 0.104536 0.023406 0.108521 0.192868 0.1396
-0.61974 -0.38561 -0.08176 -0.10732 0.025184 -0.051
0.267411 0.238943 -0.06144 -0.00204 0.111411 0.0919
-0.15386 -0.09047 -0.38895 -0.30079 0.003075 -0.04997
0.229911 0.17612 -0.04523 -0.00341 0.222202 0.15183
-0.22717 -0.2121 -0.48988 -0.36651 -0.14321 -0.12286
0.340359 0.24425 -0.0251 0.024793 0.086705 0.02951
-0.02043 -0.08708 -0.38531 -0.29038 -0.04983 -0.04763
0.020667 0.11712 -0.03494 0.124522 0.003171 0.0102
-0.04283 0.010223 0.007066 0.092996 -0.05703 0.018789
Figure imgf000153_0001
0.003802 0.087984 0.03423 0.291773 0.00045 0.03411
-0.16898 -0.08414 -0.07246 0.237282 -0.0666 0.008299
0.005609 0.050688 0.063879 0.277051 0.165736 0.0583
-0.26863 -0.19261 -0.03505 0.403648 -0.07574 0.050707
-0.40373 -0.13192 0.177795 0.392803 0.304589 0.1680
-0.17048 0.036129 -0.04805 0.271155 -0.04251 0.076333
Figure imgf000153_0002
-0.01274 -0.02608 -0.04499 0.210222 0.04793 0.0134
-0.18058 -0.1345 -0.26537 0.146551 -0.10254 -0.059
0.00476 0.011297 0.010094 0.185718 -0.001 -0.0470
-0.17339 -0.13773 -0.38124 0.029898 -0.11635 0.02668
0.023125 -0.00635 -0.04019 0.246271 0.00831 0.0191
-0.20673 -0.03158 -0.33379 0.250724 -0.12822 0.008395
-0.04551 0.00725 -0.13228 0.077233 0.032829 0.1750
-0.17263 -0.0629 -0.23549 0.12954 -0.13536 -0.0643
0.102625 0.124135 0.075543 0.229275 0.020047 0.2034
-0.28553 0.013402 -0.33338 0.075789 -0.00615 -0.00427
0.222035 0.265221 0.459894 0.262607 -0.02657 0.1419
0.554403 0.208938 0.322626 0.026635 0.136895 0.026316
-0.17975 -0.06232 0.223885 0.037432 0.065887 0.1571
0.166574 -0.09547 -0.08198 -0.36828 0.060289 -0.06339
-0.32647 -0.21747 0.216156 0.098201 -0.02918 0.1220
0.071395 -0.11434 0.126336 -0.27929 0.047018 0.003263
-0.06928 -0.00605 0.312033 0.14099 0.100447 0.2077
0.289426 -0.05273 0.090221 -0.26257 0.086246 -0.03334
Figure imgf000154_0001
-0.50755 -0.306 0.261087 0.133802 0.033667 0.1065
-0.19705 -0.2548 0.122226 -0.34201 0.153377 0.022982 -0.53691 -0.39748 0.245686 0.02884 0.057487 0.1617
0.00319 -0.17005 0.135243 -0.28015 0.03293 -0.07662
-0.49723 -0.20857 0.145217 0.020358 -0.00937 0.0442
-0.23895 -0.246 0.004591 -0.31196 -0.0268 -0.06418
-0.50174 -0.27755 0.229584 0.015675 0.03672 0.1271
-0.01124 -0.16569 -0.02935 -0.33122 -0.0136 -0.11918
-0.10634 -0.0389 0.095348 0.067418 0.049147 -0.0071
0.04819 -0.15744 -0.21904 -0.31746 0.025895 -0.13937
-0.82337 -0.75975 -0.26451 -0.13441 0.167633 0.1451
-0.25851 -0.22852 -0.61886 -0.40514 -0.11017 0.01557
0.021788 0.125797 0.020008 -0.01616 0.177233 0.2793
-0.17625 -0.17687 -0.37438 -0.33175 -0.13449 0.037702
-0.21889 -0.1459 -0.12775 -0.14166 0.07424 -0.0182
-0.02855 -0.03881 -0.16169 -0.30592 0.059009 -0.02306
-0.07647 0.110987 0.100147 0.08324 0.243433 0.3788
0.006174 -0.08999 -0.37458 -0.21876 0.01886 -0.07126
-0.17855 -0.0349 0.19218 0.056899 0.091447 0.1419
-0.12559 -0.1124 0.066268 0.019802 -0.05775 0.047939
Figure imgf000155_0001
■0.60439 -0.39708 0.017872 -0.01196 0.166093 0.2273
-0.17851 -0.1985 -0.2817 -0.34455 -0.0655 0.014667
0.01132 0.109831 0.048862 0.013354 0.24172 0.27253
-0.11025 -0.06953 -0.22964 -0.21199 -0.02066 -0.04389
■0.46977 -0.30197 -0.21647 -0.0815 0.13222 0.3542
-0.34317 -0.31633 -0.59354 -0.40877 -0.1313 -0.15478
■0.18923 -0.00439 0.025631 0.020822 -0.08406 0.169
-0.40303 -0.25265 -0.46151 -0.21382 -0.06941 -0.05659
■0.98717 -0.98592 -0.98627 -0.67759 0.032027 -0.1627
-0.92242 -0.90969 -1.89572 -0.92663 -0.08032 -0.07384
■0.19332 -0.15695 -0.28367 -0.39401 0.0859 0.0610
-0.31288 -0.31801 -0.58522 -0.49297 -0.08698 -0.08969
-0.0832 -0.04064 -0.13382 -0.0938 0.056747 0.0108
-0.36883 -0.25425 -0.61619 -0.50103 0.076149 0.049281
Figure imgf000156_0001
■0.35342 -0.22043 -0.36026 -0.2683 0.05336 0.0355
-0.06918 -0.23202 -0.3378 -0.52229 -0.08303 -0.22046
■0.85034 -0.80632 -0.78601 -0.62293 -0.07718 -0.1077
-0.20354 -0.21189 -0.44881 -0.43588 0.023158 -0.08055
■0.47973 -0.3332 -0.34207 -0.26729 -0.0048 -0.0631
0.150969 -0.05031 0.096611 -0.31965 -0.09119 -0.16614
■0.24224 -0.11213 -0.2855 -0.24679 -0.01961 -0.0567
0.137845 0.050275 1.020312 0.097239 -0.09355 -0.10093
■0.81764 -0.69142 -0.68102 -0.40402 0.32546 0.1751
-0.64808 -0.50406 -1.08063 -0.86881 -0.11223 -0.11102
■0.98639 -0.98524 -0.91304 -0.47102 0.011347 0.0228
-0.91907 -0.91782 -1.85987 -0.4387 -0.02655 -0.04962
■0.65715 -0.54906 -0.50574 -0.42782 -0.00733 0.0194
-0.65003 -0.31871 -0.90956 -0.52345 -0.20559 -0.27929
Figure imgf000157_0001
-0.98055 -0.98159 -0.41209 -0.12955 0.076073 0.05198
-0.78681 -0.348 -0.28377 0.028242 -0.08557 -0.02882
-0.758 -0.69316 -0.43202 -0.15371 0.120407 0.0725
-0.50245 -0.33461 -0.62505 -0.32003 0.051807 0.096167
Figure imgf000158_0001
In view of the many possible embodiments to which the principles of the disclosed invention may be applied, it should be recognized that the illustrated embodiments are only preferred examples of the invention and should not be taken as limiting the scope of the invention. Rather, the scope of the invention is defined by the following claims. We therefore claim as our invention all that comes within the scope and spirit of these claims.

Claims

We claim:
1. A method for treating a subject with muscular dystrophy, comprising: administering an effective amount of an α7β1 integrin modulatory agent to the subject with muscular dystrophy, wherein the α7β1 integrin modulatory agent comprises ciclopirox ethanolamine, deferoxamine, 2,2-dipyridyl; 5a- cholestan-3p-ol-6-one, N032-0003, N066-0070, N069-0071, N069-0075, N064-0028, N066-0053, N069- 0073, 1080-0573, an agent having a formula selected from
Figure imgf000160_0001
B
I I I
\C wherein each R1 and R2 independently is selected from Ci-ioalkyl, substituted Ci-ioalkyl, Ci-ioalkoxy, substituted Ci-ioalkoxy, acyl, acylamino, acyloxy, acylCi-ioalkyloxy, amino, substituted amino, aminoacyl, aminocarbonylCi-ioalkyl, aminocarbonylamino, aminodicarbonylamino, aminocarbonyloxy, aminosulfonyl, C6-i5aryl, substituted C6-isaryl, C6-i5aryloxy, substituted C6-i5aryloxy, C6-i5arylthio, substituted C6-isarylthio, carboxyl, carboxyester, (carboxyester)amino, (carboxyester)oxy, cyano, C3-8cycloalkyl, substituted C3- 8cycloalkyl, (C3-8cycloalkyl)oxy, substituted (C3-scycloalkyl)oxy, (C3-8cycloalkyl)thio, substituted (C3- 8cycloalkyl)thio, halo, hydroxyl, Ci-ioheteroaryl, substituted Ci-ioheteroaryl, Ci-ioheteroaryloxy, substituted Ci-ioheteroaryloxy, Ci-ioheteroarylthio, substituted Ci-ioheteroarylthio, C2-ioheterocyclyl, C2-iosubstituted heterocyclyl, C2-ioheterocyclyloxy, substituted C2-ioheterocyclyloxy, C2-ioheterocyclylthio, substituted C2- loheterocyclylthio, imino, oxo, sulfonyl, sulfonylamino, thiol, Ci-ioalkylthio, substituted Ci-ioalkythio, or thiocarbonyl; or
two R1 substituents, together with the atom to which each is bound, may form ring selected from a C6-i5aryl, substituted C6-isaryl, C3-8cycloalkyl, substituted C3-8cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C2-iosubstituted heterocyclyl, or C2-ioheterocyclyloxy, substituted;
two R2 substituents, together with the atom to which each is bound, may form ring selected from a C6-i5aryl, substituted C6-isaryl, C3-8cycloalkyl, substituted C3-8cycloalkyl, Ci-ioheteroaryl, substituted Ci. loheteroaryl, C2-iosubstituted heterocyclyl, or C2-ioheterocyclyloxy, substituted;
each of A, B, C, D, E, and F independently may be selected from carbon, nitrogen, oxygen, or sulfur; and
n may be zero, 1, 2, 3, 4, or 5; or a combination of any of these compounds, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby treating the subject with muscular dystrophy.
2. The method of claim 1 , wherein the α7β1 integrin modulatory agent is selected from compound #1001 , compound #1002, compound #1003,
Figure imgf000161_0001
3. The method of claim 1 wherein the α7β1 integrin modulatory agent has a formula selected from
Figure imgf000161_0002
wherein Y1 may be selected from oxygen, sulfur, or NRb, wherein Rb is selected from hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclyl, or substituted heterocyclyl; and
R6 is selected from Ci-ioalkyl, substituted Ci-ioalkyl, Ci-ioalkoxy, substituted Ci-ioalkoxy, acyl, acylamino, acyloxy, acylCi-ioalkyloxy, amino, substituted amino, aminoacyl, aminocarbonylCi-ioalkyl, aminocarbonylamino, aminodicarbonylamino, aminocarbonyloxy, aminosulfonyl, C6-isaryl, substituted Ce- lsaryl, C6-i5aryloxy, substituted C6-isaryloxy, C6-i5arylthio, substituted C6-i5arylthio, carboxyl, carboxyester, (carboxyester)amino, (carboxyester)oxy, cyano, C3-scycloalkyl, substituted C3-scycloalkyl, (C3- 8cycloalkyl)oxy, substituted (C3-8cycloalkyl)oxy, (C3-scycloalkyl)thio, substituted (C3-8cycloalkyl)thio, halo, hydroxyl, Ci-ioheteroaryl, substituted Ci-ioheteroaryl, Ci-ioheteroaryloxy, substituted Ci-ioheteroaryloxy, G- loheteroarylthio, substituted Ci-ioheteroarylthio, C2-ioheterocyclyl, C2-iosubstituted heterocyclyl, C2- loheterocyclyloxy, substituted C2-ioheterocyclyloxy, C2-ioheterocyclylthio, substituted C2-ioheterocyclylthio, imino, oxo, sulfonyl, sulfonylamino, thiol, Ci-ioalkylthio, substituted Ci-ioalkythio, or thiocarbonyl.
4. The method of claim 1 wherein the α7 1 integrin modulatory agent has a formula
Figure imgf000162_0001
wherein:
R1 is selected from C(0)NH2, C(0)N[(CH2)o-5CH3]2, C(0)OH, C(0)OP(0)(OH)2,
C(0)OC(0)C(CH3)3, C(0)OC(0)NMe2, or C(0)0(CH2)o-5CH3;
R1' is selected from NH2 or N[(CH2)o-5CH3]2;
R2 is selected from N[(CH2)o-5CH3]2; and
Y1 is selected from oxygen, sulfur, NH, N(CH2)o-5CH3, and NCH2OP(0)(OH)2.
5. The method of claim 4 whe integrin modulatory agent is
Figure imgf000162_0002
6. The method of claim 1 wherein the α7 1 integrin modulatory agent has a formula
Figure imgf000162_0003
wherein:
R1 is selected from C(0)NH(Ph)/?CH2NHC(0)Oi-Bu,
C(0)N[CH2OP(0)(OH)2](Ph)^CH2NHC(0)Oi-Bu,
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[CH2OP(0)(OH)2]C(0)Oi-Bu,
C(0)NH(Ph)^CH2N[CH2OP(0)(OH)2]C(0)Oi-Bu, C(0)NH(Ph)/?CH2NHC(0)OH,
C(0)N[CH2OP(0)(OH)2](Ph)^CH2NHC(0)OH, C(0)NH(Ph)/?CH2NHC(0)OMe,
C(0)N[CH2OP(0)(OH)2](Ph)^CH2NHC(0)OMe,
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[CH2OP(0)(OH)2]C(0)OMe, C(0)N[(CH2)o- 5CH3](Ph)/?CH2NHC(0)Oi-Bu, C(0)N[(CH2)o-5CH3](Ph)^CH2N[CH20P(0)(OH)2]C(0)Oi-Bu, C(0)N[(CH2)o-5CH3](Ph)^CH2NHC(0)OH, C(0)N[(CH2)o-5CH3](Ph)^CH2N[CH20P(0)(OH)2]C(0)OH, C(0)N[(CH2)o-5CH3](Ph)^CH2NHC(0)OMe, C(0)N[(CH2)o-5CH3](Ph)^CH2N[CH2OP(0)(OH)2]C(0)OMe,
Figure imgf000163_0001
C(0)N[CH2OP(0)(OH)2](Ph)pCH2NH2, C(0)NH(Ph)/?CH2OH,
C(0)N[CH2OP(0)(OH)2](Ph)^CH2OH, C(0)NH(Ph)/?CH2N[(CH2)o-5CH3]2,
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[(CH2)o-5CH3]2, C(0)NH(Ph)/?CH2OMe,
Figure imgf000163_0002
C(0)NH(Ph)/?CH2SMe, C(0)N[CH2OP(0)(OH)2](Ph)^CH2SMe, C(0)N[(CH2)o-5CH3](Ph)/?CH2NH2,
Figure imgf000163_0003
C(0)N[CH20P(0)(OH)2](Ph)^CH2OH, C(0)N[(CH2)o-5CH3](Ph)^CH2N[(CH2)o-5CH3]2,
C(0)N[CH2OP(0)(OH)2](Ph)^CH2N[(CH2)o-5CH3]2, C(0)N[(CH2)o-5CH3](Ph)^CH2OMe,
Figure imgf000163_0004
C(0)N[CH2OP(0)(OH)2] (Ph)/?CH2SH, C(0)N[(CH2)o-5CH3] (Ph)/?CH2SMe, or
C(0)N[CH2OP(0)(OH)2](Ph)^CH2SMe;
R2 is selected from (CEbVsCEb; and
Y1 is selected from oxygen, sulfur, NH, N(CH2)o-5CH3, or NCH2OP(0)(OH)2.
7. The method of claim 6 wherein the α7β1 integrin modulatory agent is
Figure imgf000163_0005
8. The method of claim 1 wherein the α7 1 integrin modulatory agent has a formula
Figure imgf000163_0006
wherein:
Rl is selected from hydrogen, C(0)-Ph-/?-Br, C(0)-Ph-/?-Cl, C(0)-Ph-/?-F, C(0)-Ph-/?-I, C(0)-Ph-/ CF3, C(0)-Ph-m-Br, C(0)-Ph-m-Cl, C(0)-Ph-m-F, C(0)-Ph-m-I, C(0)-Ph-m-CF3, C(0)-Ph-o-Br, C(0)-Ph- o-Cl, C(0)-Ph-o-F, C(0)-Ph-o-I, C(0)-Ph-o-CF3, or (CH2)o-5CH3; and
R6 is selected from N[CH2OP(0)(OH)2]C(0)-3-bromo-5-pyridyl, NHC(0)-3-bromo-5-pyridyl, N[(CH2)o-5CH3]C(0)-3-bromo-5-pyridyl, N[(CH2)o-5CH3]C(0)-3-fiuoro-5-pyridyl,
N[CH2OP(0)(OH)2]C(0)-3-fiuoro-5-pyridyl, or NHC(0)-3-fiuoro-5-pyridyl.
9. The method of claim 8 wherein the α7β1 integrin modulatory agent is
Figure imgf000164_0001
The method of c nt has a formula
Figure imgf000164_0002
R1 is selected from hydrogen and (CJ hVsCEb;
R2 is selected from OMe, SH, SMe, NH2, N[(CH2)o-5CH3]2, OH, OP(0)(OH)2, OC(0)C(CH3)3, or OC(0)NMe2;
R2' is N02; and
optional ring A, if present, is phenyl.
11. The method of claim 10 wherein the α7β1 integrin modulatory agent is
Figure imgf000164_0003
The method of claim 1 wherein the α7β1 integrin modulatory agent has a formula
Figure imgf000164_0004
wherein: R1 is selected from 0(CH2)o-5CH3, OH, OP(0)(OH)2, OC(0)C(CH3)3, OC(0)NMe2, S(CH2)0. SH, NH2, N[(CH2)o-5CH3]2, OCF3, or CF3;
Y1 is selected from NH, O, NCH2OP(0)(OH)2, or S; and
Y2 is selected from NH, O, NCH2OP(0)(OH)2, or S.
The method of claim 12 wherein the α7β1 integrin modulatory agent is
Figure imgf000165_0001
14. The method of claim 1, wherein the α7β1 integrin modulatory agent is selected from one or more of compounds 1-269.
The method of claim 1, wherein the α7β1 integrin modulatory agent is selected from
Figure imgf000165_0002
Figure imgf000166_0001
165
Figure imgf000167_0001
Figure imgf000167_0002
166
Figure imgf000168_0001
16. The method of claim 1, wherein the muscular dystrophy is merosin deficient congenital muscular dystrophy Type 1A (MDCIA), merosin deficient congenital muscular dystrophy Type ID (MDCID), limb-girdle muscular dystrophy (LGMD), Duchenne muscular dystrophy (DMD), Fukuyama congenital muscular dystrophy (FCMD) or Facioscapulohumeral muscular dystrophy (FHMD).
17. The method of claim 16, wherein the muscular dystrophy is DMD, MDCIA or FCMD.
18. The method of claim 16, wherein the muscular dystrophy is DMD.
19. The method of claim 1, wherein the α7β1 integrin modulatory agent is administered with an additional therapeutic agent.
20. The method of claim 19, wherein the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional α7β1 integrin modulatory agent.
21. The method of claim 20, wherein the additional α7β1 integrin modulatory agent is laminin- 111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
22. The method of claim 1, further comprising selecting a subject with muscular dystrophy.
23. The method of claim 22, wherein selecting a subject with muscular dystrophy comprises diagnosing the subject with muscular dystrophy prior to administering an effective amount of the α7β1 integrin modulatory agent to the subject.
24. A method of enhancing muscle regeneration, repair, or maintenance in a subject, comprising:
administering an effective amount of an α7β1 integrin modulatory agent to the subject in need of muscle regeneration, repair, or maintenance, wherein the α7β1 integrin modulatory agent is as provided by any one of claims 1-15, or combinations thereof, and
wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby enhancing muscle regeneration, repair or maintenance in a subject.
25. The method of claim 24, wherein the α7β1 modulatory agent is administered prior to the subject experiencing muscle damage or disease.
26. The method of claim 24, wherein the method is a method of enhancing muscle maintenance in a subject.
27. The method of claim 26, wherein the α7β1 integrin modulatory agent is administered to the subject prior to the subject exercising.
28. The method of claim 26, wherein the α7β1 integrin modulatory agent is administered to a subject at risk of acquiring a muscle disease or damage.
29. The method of claim 24, further comprising selecting a subject in need of enhancing muscle regeneration, repair, or maintenance.
30. The method of claim 29, wherein selecting a subject in need of enhancing muscle regeneration, repair, or maintenance comprises diagnosing the subject with a condition characterized by impaired muscle regeneration prior to administering an effective amount of the α7β1 integrin modulatory agent to the subject.
31. The method of claim 29, wherein selecting a subject in need of enhancing muscle regeneration, repair, or maintenance comprises diagnosing the subject with a condition characterized by impaired production of a component of α7β1 integrin prior to administering an effective amount of the α7β1 integrin modulatory agent to the subject.
32. The method of claim 24, wherein the α7β1 integrin modulatory agent is administered with an additional therapeutic agent.
33. The method of claim 32, wherein the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional α7β1 integrin modulatory agent.
34. The method of claim 33, wherein the additional α7β1 integrin modulatory agent is laminin- 111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
35. A method of prospectively preventing or reducing muscle injury or damage in a subject, comprising:
administering an effective amount of an α7β1 integrin modulatory agent as provided by any one of claims 1-15, or a combination thereof, wherein the α7β1 integrin modulatory agent increases α7β1 integrin expression or activity as compared to α7β1 integrin expression or activity prior to treatment, thereby prospectively preventing or reducing muscle injury or damage in the subject.
36. The method of claim 35, wherein the subject is at risk of developing a muscle injury or damage.
37. The method of claim 35, wherein the α7β1 integrin modulatory agent is administered with an additional therapeutic agent.
38. The method of claim 36, wherein the additional therapeutic agent is a costameric protein, a growth factor, satellite cells, stem cells, myocytes or an additional α7β1 integrin modulatory agent.
39. The method of claim 37, wherein the additional α7β1 integrin modulatory agent is laminin- 111, a laminin-111 fragment, valproic acid, or a valproic acid analog.
40. A method of enhancing α7β1 integrin expression, comprising:
contacting a cell with an effective amount of an α7β1 integrin modulatory agent, wherein the α7β1 integrin modulatory agent is as provided by any one of claims 1-15, or
a combination thereof, and increases α7β1 integrin expression in the treated cell relative to α7β1 integrin expression in an untreated cell, thereby enhancing α7β1 integrin expression.
41. The method of claim 30, wherein the cell is a muscle cell.
42. The method of claim 41, wherein the muscle cell is present in a mammal, and wherein contacting the cell with an agent comprises administering the agent to the mammal.
PCT/US2013/059074 2012-09-10 2013-09-10 Methods of treating muscular dystrophy WO2014040077A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
EP13835809.8A EP2892525B1 (en) 2012-09-10 2013-09-10 Methods of treating muscular dystrophy
DK13835809.8T DK2892525T3 (en) 2012-09-10 2013-09-10 METHODS OF TREATING MUSCLE DYROPHY
CA2919516A CA2919516C (en) 2012-09-10 2013-09-10 Use of .alpha.7.beta.1 integrin modulators in the treatment of muscular dystrophy
US14/426,928 US10028992B2 (en) 2012-09-10 2013-09-10 Methods of treating muscular dystrophy
PL13835809T PL2892525T3 (en) 2012-09-10 2013-09-10 Methods of treating muscular dystrophy
JP2015531326A JP2015527409A (en) 2012-09-10 2013-09-10 How to treat muscular dystrophy
EP18173735.4A EP3406246A3 (en) 2012-09-10 2013-09-10 Alpha7 beta 1 integrin modulators for treating muscular dystrophy
ES13835809T ES2688023T3 (en) 2012-09-10 2013-09-10 Methods of treatment of muscular dystrophy
US16/019,194 US10398749B2 (en) 2012-09-10 2018-06-26 Methods of treating muscular dystrophy

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201261699189P 2012-09-10 2012-09-10
US61/699,189 2012-09-10
US201361798479P 2013-03-15 2013-03-15
US61/798,479 2013-03-15
US13/842,781 US9566310B2 (en) 2012-09-10 2013-03-15 Methods of treating muscular dystrophy
US13/842,781 2013-03-15

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/842,781 Continuation-In-Part US9566310B2 (en) 2012-09-10 2013-03-15 Methods of treating muscular dystrophy

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/426,928 A-371-Of-International US10028992B2 (en) 2012-09-10 2013-09-10 Methods of treating muscular dystrophy
US16/019,194 Continuation US10398749B2 (en) 2012-09-10 2018-06-26 Methods of treating muscular dystrophy

Publications (1)

Publication Number Publication Date
WO2014040077A1 true WO2014040077A1 (en) 2014-03-13

Family

ID=50233488

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/059074 WO2014040077A1 (en) 2012-09-10 2013-09-10 Methods of treating muscular dystrophy

Country Status (8)

Country Link
US (4) US9566310B2 (en)
EP (2) EP2892525B1 (en)
JP (2) JP2015527409A (en)
CA (1) CA2919516C (en)
DK (1) DK2892525T3 (en)
ES (1) ES2688023T3 (en)
PL (1) PL2892525T3 (en)
WO (1) WO2014040077A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016090313A1 (en) * 2014-12-05 2016-06-09 Aquinnah Pharmaceuticals, Inc. Compounds, compositions and methods of use
EP2968273A4 (en) * 2013-03-15 2016-08-31 Univ Nevada Methods of treating muscular dystrophy
WO2017009644A1 (en) * 2015-07-15 2017-01-19 King's College London Kinase inhibitors for use in the treatment of fascioscapulohumeral dystrophy
US9670163B2 (en) 2005-12-28 2017-06-06 Vertex Pharmaceuticals Incorporated Solid forms of N-[2,4-bis(1,1-dimethylethyl)-5-hydroxyphenyl]-1,4-dihydro-4-oxoquinoline-3-carboxamide
US9701639B2 (en) 2014-10-07 2017-07-11 Vertex Pharmaceuticals Incorporated Co-crystals of modulators of cystic fibrosis transmembrane conductance regulator
WO2017210527A1 (en) * 2016-06-03 2017-12-07 University Of Tennessee Research Foundation Autotaxin inhibitors
US10272069B2 (en) 2012-09-10 2019-04-30 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
US10588894B2 (en) 2017-06-21 2020-03-17 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US10870657B2 (en) 2015-12-22 2020-12-22 SHY Therapeutics LLC Compounds for the treatment of cancer and inflammatory disease
CN113527172A (en) * 2020-04-21 2021-10-22 上海交通大学医学院附属仁济医院 M2 acetylcholine receptor antagonists and uses thereof
US11299467B2 (en) 2017-07-21 2022-04-12 Antabio Sas Chemical compounds
WO2023215367A1 (en) * 2022-05-04 2023-11-09 Cytokinetics, Inc. Bicyclic piperazinones and therapeutic uses thereof
US11963964B2 (en) 2011-06-16 2024-04-23 President And Fellows Of Harvard College Small molecules for mouse satellite cell proliferation

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10398749B2 (en) * 2012-09-10 2019-09-03 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
JP6755778B2 (en) * 2016-11-11 2020-09-16 花王株式会社 PPARα activator
TW202028456A (en) * 2018-09-28 2020-08-01 日商日產化學股份有限公司 Medium additive for promoting generation of paracrine factor
EA202191082A1 (en) * 2018-11-06 2021-09-10 Эджвайз Терапьютикс, Инк. COMPOUNDS OF PYRIDAZINONES AND THEIR APPLICATIONS
CN113272291A (en) 2018-11-06 2021-08-17 艾知怀斯治疗学公司 Pyridazinone compound and use thereof
WO2020097258A1 (en) 2018-11-06 2020-05-14 Edgewise Therapeutics, Inc. Pyridazinone compounds and uses thereof
EP4149465A1 (en) * 2020-05-13 2023-03-22 Edgewise Therapeutics, Inc. Substituted pyridazinone for use in the treatment of neuromuscular diseases
WO2021262878A1 (en) * 2020-06-24 2021-12-30 Oregon Health & Science University Novel molecule for modulation of innate immune responses controlled by sting protein
EP4192503A2 (en) * 2020-08-05 2023-06-14 Ohio State Innovation Foundation Adapter polypeptides and methods of using the same
WO2023164118A1 (en) * 2022-02-24 2023-08-31 The Research Institute At Nationwide Children's Hospital Bacterial biofilm inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050165039A1 (en) * 2002-03-13 2005-07-28 Meissner Robert S. Fluorinated 4-azasteroid derivatives as androgen receptor modulators
US20110224128A1 (en) * 2009-04-13 2011-09-15 Anne Whalen Methods and compositions for treatment of muscular dystrophy
US20120207720A1 (en) * 2007-10-09 2012-08-16 Nevada, Reno Laminins, derivatives, and compositions including same and methods for their therapeutic use

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0514481B1 (en) 1990-01-30 2000-05-03 La Jolla Cancer Research Foundation Merosin, nucleic acids encoding, fragments and uses thereof
US6283761B1 (en) 1992-09-08 2001-09-04 Raymond Anthony Joao Apparatus and method for processing and/or for providing healthcare information and/or healthcare-related information
US5962427A (en) 1994-02-18 1999-10-05 The Regent Of The University Of Michigan In vivo gene transfer methods for wound healing
DE69738331T2 (en) 1996-10-08 2008-10-16 University Of Washington, Seattle THERAPEUTIC APPLICATIONS OF LAMININ AND PROTEIN FRAGMENTS DERIVED FROM LAMININ
US6933280B2 (en) 1996-10-08 2005-08-23 Gerardo Castillo Peptides for the treatment of Alzheimer's disease and other beta-amyloid protein fibrillogenesis disorders
US6682911B1 (en) 1997-10-10 2004-01-27 The General Hospital Corporation Laminins and uses thereof
US6294356B1 (en) 1998-01-16 2001-09-25 Northwestern University Methods and materials for making and using laminin-5
US6693169B1 (en) 1998-10-13 2004-02-17 The General Hospital Corporation Laminin 5, 13 and 14 and uses thereof
WO2000055181A1 (en) 1999-03-15 2000-09-21 The General Hospital Corporation Methods of modulating cell attachment and migration
AU4678400A (en) 1999-04-30 2000-11-17 Biostratum, Inc. Laminin 8 and methods for its use
WO2000066730A2 (en) 1999-04-30 2000-11-09 University Of Medicine And Dentistry Of New Jersey Robert Wood Johnson Medical School Laminin 2 and methods for its use
US6703363B1 (en) 1999-04-30 2004-03-09 Biostratum, Inc. Recombinant laminin 5
US20050069985A1 (en) 2001-02-20 2005-03-31 The Board Of Trustees Of The University Of Illinois Assay methods and amelioration of muscular dystrophy symptoms
WO2002066989A2 (en) 2001-02-20 2002-08-29 The Board Of Trustees Of The University Of Illinois Diagnostics, assay methods and amelioration of muscular dystrophy symptoms
EP1497410A4 (en) 2002-04-01 2005-05-25 Peter K Law Cellular transplantation for heart regeneration
US7078379B2 (en) 2002-06-03 2006-07-18 Santhera Pharmaceuticals (Schweiz) Gmbh Treatment of congenital muscular dystrophies
US20060223888A1 (en) 2002-12-16 2006-10-05 Abbott Frank S Valproic acid analogues and pharmaceutical composition thereof
US7727550B2 (en) 2003-02-21 2010-06-01 The Uab Research Foundation Biologically active native biomatrix composition
US20050058687A1 (en) 2003-09-12 2005-03-17 Becton, Dickinson And Company Covalently attached collagen VI for cell attachment and proliferation
JP2005194198A (en) 2003-12-26 2005-07-21 Takeda Chem Ind Ltd Thienopyridine compound
US20070219234A1 (en) * 2004-04-12 2007-09-20 Kiyoshi Oizumi Thienopyridine Derivatives
PL1758986T3 (en) 2004-06-09 2016-03-31 Univ Court Univ Of Edinburgh Neural stem cells
US7749754B2 (en) 2004-06-09 2010-07-06 The Board Of Trustees Of The Leland Stanford Junior University Isolation and characterization of muscle regenerating cells
US7531355B2 (en) 2005-07-29 2009-05-12 The Regents Of The University Of California Methods and compositions for smooth muscle reconstruction
WO2007075911A2 (en) 2005-12-22 2007-07-05 Yale University Inhibition of glycogen synthase kinase and methods of treating autoimmune or immune inflammatory disease
US20080044390A1 (en) 2006-08-11 2008-02-21 Xiaowei Jin Methods and compositions for the treatment of neurodegenerative disorders
WO2010080581A2 (en) 2008-12-19 2010-07-15 The Board Of Regents Of The Nevada System Of Higher Education, On Behalf Of The University Of Nevada, Reno Valproic acid, derivatives, analogues, and compositions including same and methods for their therapeutic use
WO2012174126A1 (en) 2011-06-13 2012-12-20 Universyty Of Medicine And Dentistry Of New Jesey METHOD OF INHIBITING NONSENSE-MEDIATED mRNA DECAY
US9782417B2 (en) 2011-06-16 2017-10-10 Presidents And Fellows Of Harvard College Methods of increasing satellite cell proliferation with kinase inhibitors
WO2012174537A2 (en) 2011-06-16 2012-12-20 President And Fellows Of Harvard College Small molecule screening for mouse satellite cell proliferation
WO2013138623A1 (en) 2012-03-14 2013-09-19 Children's Medical Center Corporation High-throughput image-based chemical screening in zebrafish blastomere cell culture
US9566310B2 (en) 2012-09-10 2017-02-14 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
US10398749B2 (en) * 2012-09-10 2019-09-03 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
JP2016516051A (en) * 2013-03-15 2016-06-02 ボード オブ リージェンツ オブ ザ ネヴァダ システム オブ ハイヤー エデュケーション オン ビハーフ オブ ザ ユニヴァーシティー オブ ネヴァダ リノ How to treat muscular dystrophy

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050165039A1 (en) * 2002-03-13 2005-07-28 Meissner Robert S. Fluorinated 4-azasteroid derivatives as androgen receptor modulators
US20120207720A1 (en) * 2007-10-09 2012-08-16 Nevada, Reno Laminins, derivatives, and compositions including same and methods for their therapeutic use
US20110224128A1 (en) * 2009-04-13 2011-09-15 Anne Whalen Methods and compositions for treatment of muscular dystrophy

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LIU JIANMING ET AL.: "Beta1D chain increases alha7beta1 integrin and laminin and protects against sarcolemmal damage in mdx mice", HUMAN MOLECULAR GENETICS, vol. 21, no. 7, 2012, pages 1592 - 1603, XP055236033 *
See also references of EP2892525A4 *
WAGNER KATHRYN R. ET AL.: "Current treatment of adult Duchenne muscular dystrophy.", BIOCHIMICA ET BIOPHYSYCA ACTA, vol. 1772, 2007, pages 229 - 237, XP005856208 *

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9670163B2 (en) 2005-12-28 2017-06-06 Vertex Pharmaceuticals Incorporated Solid forms of N-[2,4-bis(1,1-dimethylethyl)-5-hydroxyphenyl]-1,4-dihydro-4-oxoquinoline-3-carboxamide
US11963964B2 (en) 2011-06-16 2024-04-23 President And Fellows Of Harvard College Small molecules for mouse satellite cell proliferation
US10272069B2 (en) 2012-09-10 2019-04-30 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
US10398680B2 (en) 2012-09-10 2019-09-03 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
EP2968273A4 (en) * 2013-03-15 2016-08-31 Univ Nevada Methods of treating muscular dystrophy
US9707210B2 (en) 2013-03-15 2017-07-18 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
US9980943B2 (en) 2013-03-15 2018-05-29 Board Of Regents Of The Nevada Systems Of Higher Education On Behalf Of The Nevada, Reno Methods of treating muscular dystrophy
US10206903B2 (en) 2013-03-15 2019-02-19 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
US10537553B2 (en) 2013-03-15 2020-01-21 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Reno Methods of treating muscular dystrophy
US9701639B2 (en) 2014-10-07 2017-07-11 Vertex Pharmaceuticals Incorporated Co-crystals of modulators of cystic fibrosis transmembrane conductance regulator
WO2016090313A1 (en) * 2014-12-05 2016-06-09 Aquinnah Pharmaceuticals, Inc. Compounds, compositions and methods of use
WO2017009644A1 (en) * 2015-07-15 2017-01-19 King's College London Kinase inhibitors for use in the treatment of fascioscapulohumeral dystrophy
US11560390B2 (en) 2015-12-22 2023-01-24 SHY Therapeutics LLC Compounds for the treatment of cancer and inflammatory disease
US10870657B2 (en) 2015-12-22 2020-12-22 SHY Therapeutics LLC Compounds for the treatment of cancer and inflammatory disease
US11124490B2 (en) 2016-06-03 2021-09-21 University Of Tennessee Research Foundation Autotaxin inhibitors
WO2017210527A1 (en) * 2016-06-03 2017-12-07 University Of Tennessee Research Foundation Autotaxin inhibitors
US10940139B2 (en) 2017-06-21 2021-03-09 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11000515B2 (en) 2017-06-21 2021-05-11 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11026930B1 (en) 2017-06-21 2021-06-08 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US10933054B2 (en) 2017-06-21 2021-03-02 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11213515B1 (en) 2017-06-21 2022-01-04 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11541041B1 (en) 2017-06-21 2023-01-03 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, Rasopathies, and fibrotic disease
US10588894B2 (en) 2017-06-21 2020-03-17 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11299467B2 (en) 2017-07-21 2022-04-12 Antabio Sas Chemical compounds
CN113527172A (en) * 2020-04-21 2021-10-22 上海交通大学医学院附属仁济医院 M2 acetylcholine receptor antagonists and uses thereof
WO2023215367A1 (en) * 2022-05-04 2023-11-09 Cytokinetics, Inc. Bicyclic piperazinones and therapeutic uses thereof

Also Published As

Publication number Publication date
US20140072536A1 (en) 2014-03-13
EP2892525A1 (en) 2015-07-15
DK2892525T3 (en) 2018-09-24
US20150290276A1 (en) 2015-10-15
US10272069B2 (en) 2019-04-30
US10028992B2 (en) 2018-07-24
CA2919516A1 (en) 2014-03-13
EP2892525A4 (en) 2016-08-03
US20180263959A1 (en) 2018-09-20
EP2892525B1 (en) 2018-06-13
US20170224657A1 (en) 2017-08-10
EP3406246A2 (en) 2018-11-28
JP2018197252A (en) 2018-12-13
CA2919516C (en) 2021-07-27
EP3406246A3 (en) 2019-03-27
US10398680B2 (en) 2019-09-03
PL2892525T3 (en) 2018-11-30
ES2688023T3 (en) 2018-10-30
JP2015527409A (en) 2015-09-17
US9566310B2 (en) 2017-02-14

Similar Documents

Publication Publication Date Title
US10398680B2 (en) Methods of treating muscular dystrophy
DK2902035T3 (en) METHODS FOR TREATING MUSCLE DYROPHY
US10537553B2 (en) Methods of treating muscular dystrophy
EP2211851B1 (en) Laminin-1 for use in enhancing muscle regeneration after injury or for improving wound healing if administered systemically
US20170258873A1 (en) Methods for diagnosing, prognosing and treating muscular dystrophy
JP2009534415A (en) Pharmacological control of the action of AMPA receptor modulators on neurotrophin expression
US10398749B2 (en) Methods of treating muscular dystrophy
US20160101086A1 (en) Methods of treating muscular dystrophy
Baudy Efficacy of glucocorticoids in muscular dystrophy: Signaling, hormonal activities, and muscle inflammation

Legal Events

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

Ref document number: 13835809

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14426928

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2015531326

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2013835809

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2919516

Country of ref document: CA