WO2024064895A2 - Compositions and methods for the treatment of neuromuscular disorders - Google Patents

Compositions and methods for the treatment of neuromuscular disorders Download PDF

Info

Publication number
WO2024064895A2
WO2024064895A2 PCT/US2023/074909 US2023074909W WO2024064895A2 WO 2024064895 A2 WO2024064895 A2 WO 2024064895A2 US 2023074909 W US2023074909 W US 2023074909W WO 2024064895 A2 WO2024064895 A2 WO 2024064895A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
acid sequence
patient
promoter
seq
Prior art date
Application number
PCT/US2023/074909
Other languages
French (fr)
Other versions
WO2024064895A3 (en
Inventor
Carlos Fonck
Clarice Weili CHEN
Dwaipayan Sen
Original Assignee
Astellas Gene Therapies, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Astellas Gene Therapies, Inc. filed Critical Astellas Gene Therapies, Inc.
Publication of WO2024064895A2 publication Critical patent/WO2024064895A2/en
Publication of WO2024064895A3 publication Critical patent/WO2024064895A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/711Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination

Definitions

  • XLMTM XLMTM
  • respiratory support i.e., mechanical ventilation
  • tracheostomy a severe form of XLMTM.
  • MTM1 a gene therapy approach involving the delivery of MTM1 has been developed for the treatment of XLMTM.
  • MTM1 a gene therapy approach involving the delivery of MTM1 has been developed for the treatment of XLMTM.
  • compositions and methods useful for the treatment of neuromuscular disorders particularly X-linked myotubular myopathy (XLMTM).
  • a patient e.g., a human patient
  • XLMTM X-linked myotubular myopathy
  • a viral vector such as an adeno-associated viral (AAV) vector, having a transgene encoding myotubularin 1 (MTM1).
  • AAV adeno-associated viral
  • MTM1 myotubularin 1
  • the disclosure provides a recombinant AAV vector comprising a transgene encoding MTM1, wherein the transgene is operably linked to a promoter that is active in liver tissue.
  • the promoter is selectively active in liver tissue.
  • the vector upon separately contacting the vector with one or more liver cells and one or more non-liver cells (e.g., in vitro or in vivo) under equivalent conditions, results in a level of expression of the transgene in the one or more liver cells that is greater than the level of expression of the transgene in the one or more non-liver cells, for example, from 2-fold to 1,000-fold greater (e.g., 2- fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 200-fold, 300- fold, 400-fold, 500-fold, 600-fold, 700-fold, 800-fold, 900-fold, or 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells).
  • 2-fold to 1,000-fold greater e.g., 2- fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold,
  • the vector upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is from 10-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells.
  • the vector upon separately contacting the vector with the one or more liver cells PATENT ATTORNEY DOCKET NO.51037-075WO2 and the one or more non-liver cells, results in a level of expression of the transgene in the one or more liver cells that is from 50-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells.
  • the vector upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is from 100-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells. In some embodiments, upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is at least 2-fold, at least 5-fold, at least 10-fold, at least 50-fold, or at least 100-fold greater than the level of expression of the transgene in the one or more non-liver cells.
  • the non-liver cells are muscle cells or neural cells. In some embodiments, the non-liver cells are cardiac cells.
  • the vector upon contacting the vector with one or more liver cells (e.g., of a patient having a neuromuscular disorder), the vector results in a level of expression of the transgene in the one or more liver cells that is closer to the expression of MTM1 in wild-type/healthy liver cells, for example, the expression of the transgene may be from 0.2-fold to 10-fold (e.g., 0.2-fold, 0.3-fold, 0.4-fold.0.5-fold, 0.6-fold, 0.7-fold, 0.8-fold, 0.9-fold, 1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4- fold, 4.5-fold, 5-fold, 5.5-fold, 6-fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, or 10- fold) the expression level of M
  • the vector upon contacting the vector with one or more liver cells (e.g., of a patient having a neuromuscular disorder), results in a level of expression of the transgene in the one or more liver cells that is at least 0.2-fold, at least 0.5-fold, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold the level of wild-type MTM1 expression in a healthy liver cell.
  • the promoter comprises an LP1 promoter.
  • the LP1 promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 3: GTCCCCTAAAATGGGCAAACATTGCAAGCAGCAAACAGCAAACACACAGCCCTCCCTGCCTGCT GACCTTGGAGCTGGGGCAGAGGTCAGAGACCTCTCTGGGCCCATGCCACCTCCAACATCCACT CGACCCCTTGGAATTTCGGTGGAGAGGAGCAGAGGTTGTCCTGGCGTGGTTTAGGTAGTGTGA GAGGGGAATGACTCCTTTCGGTAAGTGCAGTGGAAGCTGTACACTGCCCAGGCAAAGCGTCCG GGCAGCGTAGGCGGGCGACTCAGATCCCAGCCAGTGcACTTAGCCCCTGTTTGCTCCTCCGATA ACTGGGGTGACCTTGGTTAATATTCACCAGCAGCCTCCCGTTGCCCCTCTGGATCCACTGCT TAAATACGGACGAGGACAGGGCCCTGTCTCCTCAGCTTCAGGCACCACCACTG
  • the LP1 promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 3 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, PATENT ATTORNEY DOCKET NO.51037-075WO2 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 3).
  • the LP1 promoter has the nucleic acid sequence of SEQ ID NO: 3.
  • the promoter comprises an apolipoprotein E (ApoE) promoter.
  • the ApoE promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 8: CCCTAAAATGGGCAAACATTGCAAGCAGCAAACAGCAAACACACAGCCCTCCCTGCCTGCTGAC CTTGGAGCTGGGGCAGAGGTCAGAGACCTCTCTGGGCCCATGCCACCTCCAACATCCACTCGA CCCCTTGGAATTTCGGTGGAGAGGAGCAGAGGTTGTCCTGGCGTGGTTTAGGTAGTGTGAGAG GGG (SEQ ID NO: 8).
  • the ApoE promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 8 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 8).
  • the ApoE promoter has the nucleic acid sequence of SEQ ID NO: 8.
  • the promoter comprises an alpha-1-antitrysin (A1AT) promoter.
  • the A1AT promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 9: AATGACTCCTTTCGGTAAGTGCAGTGGAAGCTGTACACTGCCCAGGCAAAGCGTCCGGGCAGC GTAGGCGGGCGACTCAGATCCCAGCCAGTGGACTTAGCCCCTGTTTGCTCCTCCGATAACTGGG GTGACCTTGGTTAATATTCACCAGCAGCCTCCCGTTGCCCCTCTGGATCCACTGCTTAAATAC GGACGAGGACAGGGCCCTGTCTCCTCAGCTTCAGGCACCACCACTGACCTGGGACAGTGAAT (SEQ ID NO: 9).
  • the A1AT promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 9 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 9).
  • the A1AT promoter has the nucleic acid sequence of SEQ ID NO: 9.
  • the promoter is a chimeric promoter comprising an ApoE promoter and an A1AT promoter.
  • the chimeric promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 2: CGTGATCTAGTAGGCTCAGAGGCACACAGGAGTTTCTGGGCTCACCCTGCCCCCTTCCAACCCC TCAGTTCCCATCCTCCAGCAGCTGTTTGTGTGCTGCCTCTGAAGTCCACACTGAACAAACTTCAG CCTACTCATGTCCCTAAAATGGGCAAACATTGCAAGCAGCAAACAGCAAACACACAGCCCTCCCT GCCTGCTGACCTTGGAGCTGGGGCAGAGGTCAGAGACCTCTCTGGGCCCATGCCACCTCCAAC ATCCACTCGACCCCTTGGAATTTCGGTGGAGAGGAGCAGAGGTTGTCCTGGCGTGGTTTAGGTA PATENT ATTORNEY DOCKET NO.51037-075WO2 GTGTGAGAGGGGTACCCGGGGATCTTGCTACCAGTGGAACAGCCACTAAGGATTCTGCAGTGA GAGCAGAGGGCCA
  • the chimeric promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 2 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 2).
  • the chimeric promoter has the nucleic acid sequence of SEQ ID NO: 2.
  • the promoter comprises a constitutive promoter.
  • the constitutive promoter is a phosphoglycerate kinase (PGK) promoter, an elongation factor-1 alpha (EF1alpha) promoter, a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) promoter, a cytomegalovirus (CMV) promoter, or a chicken- ⁇ -actin (CBA) promoter.
  • the AAV vector further comprises a second transgene encoding MTM1.
  • the second transgene encoding MTM1 is operably linked to a promoter that is active in muscle tissue.
  • the promoter that is active in muscle tissue is selectively active in muscle tissue.
  • the promoter that is selectively active in muscle tissue effectuates (e.g., in vitro or in vivo) a level of expression of the MTM1 transgene in muscle cells that is greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells, for example, from 2-fold to 1,000-fold greater (e.g., 2-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50- fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 200-fold, 300-fold, 400-fold, 500-fold, 600-fold, 700- fold, 800-fold, 900-fold, or 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells).
  • 2-fold to 1,000-fold greater e.g., 2-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50- fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold
  • the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 10-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. In some embodiments, the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 50-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells.
  • the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 100-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. In some embodiments, the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is at least 2-fold, at least 5-fold, at least 10-fold, at least 50-fold, or at least 100-fold greater PATENT ATTORNEY DOCKET NO.51037-075WO2 than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells.
  • the vector upon contacting the vector with one or more liver cells (e.g., of a patient having a neuromuscular disorder, results in a level of expression of the transgene in the one or more liver cells that is closer to the expression of MTM1 in wild-type/healthy liver cells, for example, the expression of the transgene may be from 0.2-fold to 10-fold (e.g., 0.2-fold, 0.3-fold, 0.4- fold.0.5-fold, 0.6-fold, 0.7-fold, 0.8-fold, 0.9-fold, 1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4- fold, 4.5-fold, 5-fold, 5.5-fold, 6-fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, or 10- fold) the expression level of MTM1 in wild-type/healthy liver cells.
  • the expression of the transgene may be from 0.2-fold to 10-fold (e.g.,
  • the vector upon contacting the vector with one or more liver cells (e.g., of a patient having a neuromuscular disorder), results in a level of expression of the transgene in the one or more liver cells that is at least 0.2-fold, at least 0.5-fold, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold the level of wild-type MTM1 expression in a healthy liver cell.
  • the second transgene encoding MTM1 is operably linked to a muscle creatine kinase (MCK) promoter or a desmin (DES) promoter.
  • each transgene encoding MTM1 independently, has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1).
  • each transgene encoding MTM1 has the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 6.
  • each transgene encoding MTM1 independently, has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 6 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 6).
  • each transgene encoding MTM1 has the nucleic acid sequence of SEQ ID NO: 6.
  • each transgene encoding MTM1 independently, has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 7. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 7 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 7).
  • each transgene encoding MTM1 has the nucleic acid sequence of SEQ ID NO: 7.
  • the MTM1 sequence is codon-optimized.
  • PATENT ATTORNEY DOCKET NO.51037-075WO2 the AAV is an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAVrh10, or AAVrh74 serotype.
  • the AAV vector is a pseudotyped AAV. In some embodiments, the pseudotyped AAV is AAV2/8 or AAV2/9.
  • the disclosure features a method of treating XLMTM in a human patient in need thereof by administering to the patient a therapeutically effective amount of the AAV vector of any of the above aspects or embodiments of the disclosure.
  • the patient is five years old or younger at the time of administration of the AAV vector.
  • the patient is four years old or younger at the time of administration of the AAV vector, optionally wherein the patient is three years old or younger, two years old or younger, one year old or younger, or six months old or younger.
  • the AAV vector is administered to the patient in an amount of less than 3 x 10 14 vg/kg.
  • the AAV vector is administered to the patient in an amount of less than 2.5 x 10 14 vg/kg, optionally wherein the AAV vector is administered to the patient in an amount of less than 2 x 10 14 vg/kg, less than 1.5 x 10 14 vg/kg, or less than 1.4 x 10 14 vg/kg.
  • the AAV vector is administered to the patient in an amount of from 3 x 10 13 vg/kg to 2.3 x 10 14 vg/kg, optionally wherein the AAV vector is administered to the patient in an amount of from 8 x 10 13 vg/kg to 1.8 x 10 14 vg/kg, from 1 x 10 14 vg/kg to 1.6 x 10 14 vg/kg, from 1.1 x 10 14 vg/kg to 1.5 x 10 14 vg/kg, or from 1.2 x 10 14 vg/kg to 1.4 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.3 x 10 14 vg/kg.
  • the AAV vector is administered to the patient by way of intravenous, intramuscular, intrahepatic, intradermal, or subcutaneous administration.
  • the patient is further administered an anti-cholestatic agent.
  • the anti-cholestatic agent is selected from the group consisting of a bile acid, a farnesoid X receptor (FXR) ligand, a fibroblast growth factor 19 (FGF-19) mimetic, a Takeda-G- protein-receptor-5 (TGR5) agonist, a peroxisome proliferator-activated receptor (PPAR) agonist, a PPAR-alpha agonist, a PPAR-delta agonist, a dual PPAR-alpha and PPAR-delta agonist, an apical sodium-dependent bile acid transporter (ASBT) inhibitor, an immunomodulatory drug, an antifibrotic therapy, and a nicotinamide adenine dinucleotide phosphat
  • the FXR ligand is obeticholic acid, cilofexor, tropifexor, tretinoin, or EDP-305;
  • the FGF-19 mimetic is aldafermin;
  • the TGR5 agonist is INT-777 or INT-767;
  • the PPAR agonist is bezafibrate, fenofibrate, seladelpar, or elafibrinor;
  • the PPAR-alpha agonist is fenofibrate;
  • the PPAR-delta agonist is seladelpar;
  • the dual PPAR-alpha and PPAR-delta agonist is elafibranor;
  • the ASBT inhibitor is odevixibat, maralixibat, or linerixibat;
  • the immunomodulatory drug is rituximab, abatacept, ustekinumab,
  • the bile acid is ursodeoxycholic acid, nor-ursodeoxycholic acid, or a pharmaceutically acceptable salt thereof.
  • the patient does not have a history of cholestasis or hyperbilirubinemia. In some embodiments, the patient does not have a history of any underlying liver disease.
  • the method comprises administering to the patient therapeutically effective amounts of: (i) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in liver tissue, and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue.
  • the promoter that is active in liver tissue comprises an LP1 promoter.
  • the LP1 promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 3.
  • the LP1 promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 3 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 3).
  • the LP1 promoter has the nucleic acid sequence of SEQ ID NO: 3.
  • the promoter that is active in liver tissue comprises an ApoE promoter.
  • the ApoE promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 8. In some embodiments, the ApoE promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 8 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 8).
  • the ApoE promoter has the nucleic acid sequence of SEQ ID NO: 8.
  • the promoter that is active in liver tissue comprises an A1AT promoter.
  • the A1AT promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 9.
  • the A1AT promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 9 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 9).
  • the A1AT promoter has the nucleic acid sequence of SEQ ID NO: 9.
  • the promoter that is active in liver tissue is a chimeric promoter comprising an ApoE promoter and an A1AT promoter.
  • the chimeric promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 2.
  • the chimeric promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 2 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 2).
  • the chimeric promoter has the nucleic acid sequence of SEQ ID NO: 2.
  • the disclosure features a method of treating XLMTM in a human patient in need thereof by administering to the patient therapeutically effective amounts of: (i) a non-viral composition comprising a nucleic acid encoding MTM1 operably linked to a promoter that is active PATENT ATTORNEY DOCKET NO.51037-075WO2 (e.g., selectively active) in liver tissue, and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue.
  • a non-viral composition comprising a nucleic acid encoding MTM1 operably linked to a promoter that is active PATENT ATTORNEY DOCKET NO.51037-075WO2 (e.g., selectively active) in liver tissue
  • an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in
  • the non-viral composition is a liposome, vesicle, synthetic vesicle, exosome, synthetic exosome, dendrimer, or nanoparticle.
  • the nanoparticle is a lipid nanoparticle, which can deliver DNA constructs similar in composition and size to those delivered via AAV capsids.
  • the promoter is a DES promoter.
  • the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1.
  • the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1.
  • the nucleic acid encoding MTM1 and the transgene encoding MTM1 each have the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 6. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 6.
  • the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 6.
  • the nucleic acid encoding MTM1 and the transgene encoding MTM1 each have the nucleic acid sequence of SEQ ID NO: 6.
  • the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 7. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 7.
  • the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 7.
  • the nucleic acid encoding MTM1 and the transgene encoding MTM1 each have the nucleic acid sequence of SEQ ID NO: 7.
  • the AAV vector is resamirigene bilparvovec.
  • the disclosure provides a kit comprising the AAV vector of any of the above aspects or embodiments of the disclosure.
  • the kit may further include a package insert instructing a user to administer the AAV vector to a patient diagnosed as having XLMTM.
  • the disclosure provides a kit comprising: (i) a non-viral composition comprising a nucleic acid encoding MTM1 (e.g., a non-viral composition described above), and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue (e.g., an AAV vector described above).
  • the kit may further include a package insert instructing a user to administer the non-viral composition and the AAV vector to a patient diagnosed as having XLMTM.
  • FIG.1 is a schematic drawing of an exemplary adeno-associated virus (AAV) vector for the expression of the human or mouse myotubularin 1 (MTM1) gene (e.g., a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8).
  • AAV adeno-associated virus
  • FIG.2 is a schematic drawing of an exemplary AAV vector for the expression of the human or mouse myotubularin 1 (MTM1) gene (e.g., a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8).
  • MTM1 human or mouse myotubularin 1
  • FIG.3 is a schematic drawing of an exemplary AAV vector for the expression of the human myotubularin 1 (hMTM1) gene (e.g., a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8).
  • hMTM1 human myotubularin 1
  • the shaded arrows and rectangles represent the nucleic acid sequences encoding a muscle-specific promoter (e.g., MCK, desmin) operatively linked to hMTM1 and a poly-adenylation signal (polyA), a transcription pause site, a liver-specific promoter (e.g., ApoE/A1AT, LP1) operatively linked to hMTM1, a poly-adenylation signal (polyA), and flanking inverted terminal repeat sequences (ITR).
  • a muscle-specific promoter e.g., MCK, desmin
  • polyA poly-adenylation signal
  • a transcription pause site e.g., a transcription pause site
  • a liver-specific promoter e.g., ApoE/A1AT, LP1
  • ITR flanking inverted terminal repeat sequences
  • FIG.4 is a schematic drawing of an exemplary AAV vector for the expression of the human myotubularin 1 (hMTM1) gene (e.g., a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8).
  • hMTM1 human myotubularin 1
  • a pseudotyped AAV vector such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8
  • the shaded arrows and rectangles represent the nucleic acid sequences encoding a ubiquitous promoter (e.g., PGK, Ef1a, GAPDH) operatively linked to hMTM1, a poly-adenylation signal (polyA), and flanking inverted terminal repeat sequences (ITR).
  • a ubiquitous promoter e.g., PGK, Ef1a, GAPDH
  • polyA poly-adenylation signal
  • ITR flanking inverted terminal repeat
  • Statistical significances HEMI PATENT ATTORNEY DOCKET NO.51037-075WO2 AAV8-Des-mMTM1 mice vs.
  • WT Vehicle mice p ⁇ 0.001 for all time points; WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP mice vs.
  • WT Vehicle mice p ⁇ 0.05 for weeks 7-16; HEMI AAV8-Des- mMTM1 + AAV8 empty capsid mice vs.
  • HEMI AAV8-Des-mMTM1 mice p ⁇ 0.05 on weeks 4 and 5; HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP mice vs. HEMI AAV8-Des-mMTM1 mice: p ⁇ 0.05 on weeks 5-6 and 8-9.
  • AFOS alkaline phosphatase
  • FIG.6A abbreviations: WT, wild type; HEMI, hemizygous; AFOS, Alkaline Phosphatase.
  • FIG.6B abbreviations: AFOS, Alkaline Phosphatase.
  • ALAT alanine aminotransferase
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des- mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8- Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP.
  • FIG.7A abbreviations: WT, wild type; HEMI, hemizygous; ALAT, Alanine Aminotransferase.
  • FIG.7B abbreviations: ALAT, Alanine Aminotransferase.
  • FIG.8A abbreviations: WT, wild type; HEMI, hemizygous; ASAT, Aspartate Aminotransferase.
  • FIG.8B abbreviations: ASAT, Aspartate Aminotransferase.
  • FIG.9A abbreviations: WT, wild type; HEMI, hemizygous.
  • GTT gamma-glutamyl transferase
  • FIG.10A abbreviations: WT, wild type; PATENT ATTORNEY DOCKET NO.51037-075WO2 HEMI, hemizygous; GGT, Gamma-Glutamyl Transferase.
  • FIG.10B abbreviations: GGT, Gamma-Glutamyl Transferase.
  • FIG.11A abbreviations: WT, wild type; HEMI, hemizygous; Prot tot, total protein.
  • FIG.11B abbreviations: Prot tot, total protein.
  • FIG.12A abbreviations: WT, wild type; HEMI, hemizygous.
  • FIG.13A abbreviations: WT, wild type; HEMI, hemizygous; LDH, Lactate Dehydrogenase.
  • FIG.13B abbreviations: LDH, Lactate Dehydrogenase.
  • Statistical differences # p ⁇ 0.05 compared to HEMI AAV8-Des-mMTM1.
  • FIG.14A abbreviations: WT, wild type; HEMI, hemizygous.
  • FIG.15 abbreviations: WT, wild type; HEMI, hemizygous; BilTot, total bilirubin.
  • FIG.16A abbreviations: WT, wild type; HEMI, hemizygous.
  • FIG.17A abbreviations: WT, wild type; HEMI, hemizygous.
  • Statistical differences *p ⁇ 0.05 compared to WT Vehicle.
  • FIG.18A abbreviations: WT, wild type; HEMI, hemizygous; CK, creatine kinase.
  • FIG.18B abbreviations: CK, creatine kinase.
  • FIG.19A abbreviations: WT, wild type; HEMI, hemizygous.
  • WBC white blood cell counts
  • HEMI AAV8-Des- mMTM1 HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP
  • HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.20A abbreviations: WT, wild type; HEMI, hemizygous; WBC, white blood cell count.
  • FIG.20B abbreviations: WBC, white blood cell count.
  • RBC red blood cell counts
  • WT Vehicle HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.21A abbreviations: WT, wild type; HEMI, hemizygous; RBC, red blood cell count.
  • FIG.21B abbreviations: RBC, red blood cell count.
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- PATENT ATTORNEY DOCKET NO.51037-075WO2 STOP.
  • FIG.22A abbreviations: WT, wild type; HEMI, hemizygous; HGB, hemoglobin.
  • FIG.22B abbreviations: HGB, hemoglobin.
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP.
  • FIG.23A abbreviations: WT, wild type; HEMI, hemizygous; HCT, hematocrit.
  • FIG.23B abbreviations: HCT, hematocrit.
  • MCV mean corpuscular volume
  • WT Vehicle HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.24B abbreviations: MCV, mean corpuscular volume.
  • WT Vehicle HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.25A abbreviations: WT, wild type; HEMI, hemizygous; MCH, mean corpuscular hemoglobin.
  • FIG.25B abbreviations: MCH, mean corpuscular hemoglobin.
  • FIG.26B abbreviations: MCHC, mean corpuscular hemoglobin concentration.
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP.
  • FIG.27A abbreviations: WT, wild type; HEMI, hemizygous; PLT, platelet or thrombocyte count.
  • FIG.27B abbreviations: PLT, platelet or thrombocyte count.
  • WT Vehicle HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.28A abbreviations: WT, wild type; HEMI, hemizygous; %NEUT, Neutrophil (%).
  • FIG.28B abbreviations: %NEUT, Neutrophil (%).
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP.
  • FIG.29A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute NEUT, absolute neutrophil count.
  • FIG.29B abbreviations: Abs/Absolute NEUT, absolute neutrophil count.
  • WT Vehicle HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.30B abbreviations: %LYM, Lymphocyte (%).
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP.
  • FIG.31A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute LYMPHS, absolute lymphocyte count.
  • FIG.31B abbreviations: Abs/Absolute LYMPHS, absolute lymphocyte count.
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP.
  • FIG.32A abbreviations: WT, wild type; HEMI, hemizygous; %MONO, Monocyte (%).
  • FIG.32B abbreviations: %MONO, Monocyte (%).
  • FIG.33A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute MONOS, absolute monocyte count.
  • FIG.33B abbreviations: Abs/Absolute MONOS, absolute monocyte count.
  • WT Vehicle HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.34B abbreviations: %EOS, Eosinophil (%).
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP.
  • FIG.35B abbreviations: Abs/Absolute EOS, absolute eosinophil count.
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP.
  • FIG.36A abbreviations: WT, wild type; HEMI, hemizygous; %BASO, Basophil (%).
  • FIG.36B PATENT ATTORNEY DOCKET NO.51037-075WO2 abbreviations: %BASO, Basophil (%).
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid
  • HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP.
  • FIG.37A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute BASOS, absolute basophil count.
  • FIG.37B abbreviations: Abs/Absolute BASOS, absolute basophil count.
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid
  • HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP and WT AAV8- Des-mMTM1 + AAV8-Des-hMTM1-STOP.
  • FIG.38A abbreviations: WT, wild type; HEMI, hemizygous; %LUC, Large unstained cells (%).
  • FIG. 38B abbreviations: %LUC, Large unstained cells (%).
  • WT Vehicle HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des- mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des- mMTM1 + AAV8-Des-hMTM1-STOP.
  • FIG.39A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute LUCS, absolute count of large unstained cells.
  • FIG.39B abbreviations: Abs/Absolute LUCS, absolute count of large unstained cells.
  • WT Vehicle HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.40A abbreviations: WT, wild type; HEMI, hemizygous; %Retic, Reticulocyte (%).
  • FIG.40B abbreviations: %Retic, Reticulocyte (%).
  • WT Vehicle HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP.
  • FIG.41A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute Retic, absolute reticulocyte cell count.
  • FIG.41B abbreviations: Abs/Absolute Retic, absolute reticulocyte cell count. Definitions As used herein, the term “about” refers to a value that is within 10% above or below the value being described. For example, “100 pounds” as used in the context of weight described herein includes quantities that are within 10% above or below 100 lbs.
  • administering refers to directly giving a patient a therapeutic agent (e.g., a pharmaceutical composition including a viral vector including a nucleic acid sequence encoding a myotubularin 1 (MTM1) gene operably linked to a promoter) by any effective route.
  • a therapeutic agent e.g., a pharmaceutical composition including a viral vector including a nucleic acid sequence encoding a myotubularin 1 (MTM1) gene operably linked to a promoter
  • routes of administration include systemic administration routes, such as intravenous injection, as well as routes of administration directly to the central nervous system of the patient, such as by way of intrathecal injection or intracerebroventricular injection, and directly to the liver of the patient, among others.
  • age-adjusted norms refers to the process of a normalization of data by age, which is a technique that is used to allow populations of subjects to be compared when the age profiles of the populations are different.
  • the term “norm” refers to data that does not undergo a normalization by age, as populations of subjects across age profiles are similar.
  • alanine aminotransferase and “ALT” refer to a protein whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring wild- type ALT protein (e.g., ALT1 and ALT2) as well as proteins whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring allelic variants of ALT (GPT or GPT2 e.g., splice variants or allelic variants).
  • GPT naturally occurring wild- type ALT protein
  • GPT2 e.g., splice variants or allelic variants
  • NCBI RefSeq Acc. No. NP_005300.1 Human GPT2 nucleic acid sequence is provided in NCBI RefSeq Acc. No. NM_001142466.2, and an exemplary wild-type ALT2 amino acid sequence is provided in NCBI RefSeq Acc. No. NP_001135938.1.
  • alkaline phosphatase and “ASP” refer to a protein whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring wild-type ASP protein as well as proteins whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring allelic variants of ASP (e.g., splice variants or allelic variants).
  • Human ASP nucleic acid sequence is provided in NCBI RefSeq Acc. No. NM_000478.5, and an exemplary wild-type ASP amino acid sequence is provided in NCBI RefSeq Acc. No. NP_000469.3.
  • anti-cholestatic agent refers to a substance, such as a small molecule that acts to increase bile formation and/or antagonize the effect of hydrophobic bile acids on biological membranes.
  • antagonistize refers to a molecule that decreases signal transduction resulting from the interaction of the protein with one or more of its binding partners. The antagonist may result in a decrease in the binding of the protein to one or more of its binding partners relative to binding of the two proteins in the absence of the antagonist.
  • AST refers to a protein whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring wild- type AST protein as well as proteins whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring allelic variants of AST (e.g., splice variants or allelic variants).
  • Human AST nucleic acid sequence is provided in NCBI RefSeq Acc. No NM_002079.2, and an exemplary wild-type ASP amino acid sequence is provided in NCBI RefSeq Acc. No. NP_002070.1.
  • bile acid test and “serum bile acid test” refer to the procedure in which a pre-prandial (i.e., before eating) blood sample is collected for a baseline, followed by a meal and followed about two hours later by the collection of a postprandial (i.e., after eating) blood sample. Both blood samples are tested for bile acid levels and the pre-prandial sample is used as a reference.
  • bile acid refers to the steroid acids found predominantly in the bile of mammals and other vertebrates.
  • CHOP INTEND refers to a validated motor outcome measure developed for the evaluation of weak infants, such as those with a disease of skeletal muscle (e.g., X-linked myotubular myopathy (XLMTM)).
  • CHOP INTEND uses a 0–64-point scale where higher scores indicate better motor function.
  • motor function score refers to a score on the 0–64-point scale of the CHOP INTEND (e.g., a scale of >45 on the CHOP INTEND).
  • cholestasis refers to a condition where bile cannot flow from the liver to the duodenum.
  • the two clinical distinctions are the “obstructive” type of cholestasis where there is a mechanical blockage in the duct system that can occur from a gallstone or malignancy, and “metabolic” types of cholestasis which are disturbances in bile formation that can occur because of genetic defects or acquired as a side effect of many medications.
  • bile refers to the digestive fluid that is secreted by the liver to aid in the digestion of fats.
  • a “combination therapy” means that two (or more) different agents or treatments are administered to a subject as part of a defined treatment regimen for a particular disease or condition (e.g., a neuromuscular disorder).
  • a “combination therapy” may include a procedure.
  • the treatment regimen defines the doses and periodicity of administration of each agent such that the effects of the separate agents on the subject overlap.
  • the delivery of the two or more agents is simultaneous or concurrent and the agents may be co-formulated.
  • the two or more agents are not co-formulated and are administered in a sequential manner as part of a prescribed regimen.
  • administration of two or more agents or treatments in combination is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other.
  • the effect of the two treatments can be partially additive, wholly additive, or greater than additive (e.g., synergistic).
  • Sequential or substantially simultaneous administration of each therapeutic agent can be affected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, intrahepatic routes, and direct absorption through mucous membrane tissues.
  • Therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination may be administered enterally.
  • an agent of the therapeutic combination may be administered by intravenous injection and a procedure (e.g., nasobiliary drainage (NBD)) of the therapeutic combination may be performed.
  • a procedure e.g., nasobiliary drainage (NBD)
  • dose refers to the quantity of a therapeutic agent, such as a viral vector described herein, that is administered to a subject at a particular instant for the treatment of a disorder, such as to treat or ameliorate one or more symptoms of a neuromuscular disorder described herein (e.g., XLMTM).
  • a therapeutic agent as described herein may be administered in a single dose or in multiple doses over the course of a treatment period, as defined herein.
  • therapeutic agent may be administered using one or more unit dosage forms of therapeutic agent, a term that refers to a one or more discrete compositions containing a therapeutic agent that collectively constitute a single dose of the agent.
  • the terms “effective amount,” “therapeutically effective amount,” and the like, when used in reference to a therapeutic composition, such as a vector construct described herein, refer to a quantity sufficient to, when administered to the subject, including a mammal, for example a human, effect beneficial or desired results, such as clinical results.
  • a composition such as a vector construct of the present disclosure, also include an amount that results in a beneficial or desired result in a subject as compared to a control.
  • gamma-glutamyl transferase and “GGT” refers to a protein whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring wild- type GGT protein as well as proteins whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring allelic variants of GGT (GGT1, GGT2, and GGT3 e.g., splice variants or allelic variants).
  • Human GGT1 nucleic acid sequence is provided in NCBI RefSeq Acc. No NM_001288833.1, and an exemplary wild-type GGT1 amino acid sequence is provided in NCBI RefSeq Acc. No. NP_001275762.1.
  • the term “hyperbilirubinemia” refers to a condition in which there is a higher- than-normal level of bilirubin in the blood.
  • the term “bilirubin” refers to a compound that occurs in the normal catabolic pathway that breaks down heme in vertebrates. This catabolism is a necessary process in the body's clearance of waste products that arise from the destruction of aged or abnormal red blood cells.
  • a “bilirubin test” refers to a measurement of the amount of bilirubin in a patient’s blood.
  • the term “level” refers to a level of a protein, as compared to a reference.
  • the reference can be any useful reference, as defined herein.
  • a “decreased level” and an “increased level” of a protein is meant a decrease or increase in protein level, as compared to a reference (e.g., a decrease or an increase by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, or more; a decrease or an increase of more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; a decrease or an increase by less than about 0.01-fold, about 0.02-fold, about 0.1-fold, about 0.3-fold, about 0.5-fold, about 0.8-fold, or less; or an increase by more than about
  • a level of a protein may be expressed in mass/vol (e.g., g/dL, mg/mL, ⁇ g/mL, or ng/mL) or percentage relative to total protein in a sample.
  • mass/vol e.g., g/dL, mg/mL, ⁇ g/mL, or ng/mL
  • percentage relative to total protein in a sample e.g., the terms “liver function test” and “LFT” refers to a hepatic panel (e.g., a group of blood tests that provide information about the state of a patient's liver).
  • a hepatic panel may include measurement of the level of gamma-glutamyl transferase, the level of alkaline phosphatase, the level of aspartate aminotransferase, the level of alanine aminotransferase, the level of albumin, the level of bilirubin, the prothrombin time, the activated partial thromboplastin time, or a combination thereof.
  • maximum inspiratory pressure and “MIP” refer to a variable in mechanical ventilation including the total airway pressure delivered, generally used to overcome both PATENT ATTORNEY DOCKET NO.51037-075WO2 respiratory system compliance as well as airway resistance.
  • the MIP includes the sum of the positive-end expiratory pressure and the “delta pressure.”
  • delta pressure refers to a variable in mechanical ventilation including the difference between the MIP and the positive-end expiratory pressure.
  • mechanical ventilatory support refers to the medical term for artificial ventilation where mechanical means are used to assist or replace spontaneous breathing.
  • invasive mechanical ventilatory support refers to the medical term for artificial ventilation where air is delivered via a tube that is inserted into a patient’s windpipe through the mouth or nose and mechanical means are used to assist or replace spontaneous breathing.
  • noninvasive mechanical ventillatory support refers to mechanical ventilatory support in which air is delivered to a patient through a sealed mask that can be placed over the mouth, nose, or the whole face.
  • operably linked refers to a first molecule joined to a second molecule, wherein the molecules are so arranged that the first molecule affects the function of the second molecule.
  • the two molecules may or may not be part of a single contiguous molecule and may or may not be adjacent.
  • a promoter is operably linked to a transcribable polynucleotide molecule if the promoter modulates transcription of the transcribable polynucleotide molecule of interest in a cell.
  • two portions of a transcription regulatory element are operably linked to one another if they are joined such that the transcription-activating functionality of one portion is not adversely affected by the presence of the other portion.
  • Two transcription regulatory elements may be operably linked to one another by way of a linker nucleic acid (e.g., an intervening non-coding nucleic acid) or may be operably linked to one another with no intervening nucleotides present.
  • a linker nucleic acid e.g., an intervening non-coding nucleic acid
  • the term “pharmaceutical composition” refers to a mixture containing a therapeutic compound to be administered to a subject, such as a mammal, e.g., a human, in order to prevent, treat or control a particular disease or condition affecting or that may affect the subject.
  • the term “pharmaceutically acceptable” refers to those compounds, materials, compositions and/or dosage forms, which are suitable for contact with the tissues of a subject, such as a mammal (e.g., a human) without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
  • the term “promoter” refers to a recognition site on DNA that is bound by an RNA polymerase. The polymerase drives transcription of the transgene.
  • promoters suitable for use with the compositions and methods described herein are described, for example, in Sandelin et al., Nature Reviews Genetics 8:424 (2007), the disclosure of which is incorporated herein by reference as it pertains to nucleic acid regulatory elements.
  • promoter may refer to a synthetic promoter, which are regulatory DNA sequences that do not occur naturally in biological systems. Synthetic promoters contain parts of naturally occurring promoters combined with polynucleotide PATENT ATTORNEY DOCKET NO.51037-075WO2 sequences that do not occur in nature and can be optimized to express recombinant DNA using a variety of transgenes, vectors, and target cell types.
  • a promoter that is “selectively active” in liver cells may achieve a level of gene expression in a liver cell that is greater than the level of gene expression achieved by the same promoter, under substantially the same conditions, in one or more non-liver cells, for example, from 2- fold to 1,000-fold greater (e.g., 2-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80- fold, 90-fold, 100-fold, 200-fold, 300-fold, 400-fold, 500-fold, 600-fold, 700-fold, 800-fold, 900-fold, or 1,000-fold greater expression).
  • 2- fold to 1,000-fold greater e.g., 2-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80- fold, 90-fold, 100-fold, 200-fold, 300-fold, 400-fold, 500-fold, 600-fold, 700-fold, 800-fold, 900-fold, or 1,000-fold greater expression.
  • a therapeutic agent is considered to be “provided” to a patient if the patient is directly administered therapeutic agent or if the patient is administered a substance that is processed or metabolized in vivo so as to yield therapeutic agent endogenously.
  • a patient such as a patient having a neuromuscular disorder described herein, may be provided a nucleic acid molecule encoding a therapeutic protein (e.g., MTM1) by direct administration of the nucleic acid molecule or by administration of a substance (e.g., viral vector or cell) that is processed in vivo so as to yield the desired nucleic acid molecule.
  • a therapeutic protein e.g., MTM1
  • the terms “patient” and “subject” refer to an organism that receives treatment for a particular disease or condition as described herein (such as a neuromuscular disorder, e.g., XLMTM). Examples of subjects and patients include mammals, such as humans, receiving treatment for a disease or condition described herein.
  • a “reference” is meant any useful reference used to compare protein levels related to cholestasis, hyperbilirubinemia, or one or more symptoms thereof.
  • the reference can be any sample, standard, standard curve, or level that is used for comparison purposes.
  • the reference can be a normal reference sample or a reference standard or level.
  • a “reference sample” can be, for example, a control, e.g., a predetermined negative control value such as a “normal control” or a prior sample taken from the same subject; a sample from a normal healthy subject, such as a normal cell or normal tissue; a sample (e.g., a cell or tissue) from a subject not having cholestasis, hyperbilirubinemia, or one or more symptoms thereof; a sample from a subject that is diagnosed with cholestasis, hyperbilirubinemia, or one or more symptoms thereof; a sample from a subject that has been treated for cholestasis, hyperbilirubinemia, or one or more symptoms thereof; or a sample of a purified protein (e.g., any described herein) at a known normal concentration.
  • a control e.g., a predetermined negative control value such as a “normal control” or a prior sample taken from the same subject
  • a sample from a normal healthy subject such as a
  • reference standard or level is meant a value or number derived from a reference sample.
  • a “normal control value” is a pre- determined value indicative of non-disease state, e.g., a value expected in a healthy control subject. Typically, a normal control value is expressed as a range (“between X and Y”), a high threshold (“no higher than X”), or a low threshold (“no lower than X”).
  • a subject having a measured value within the normal control value for a particular biomarker is typically referred to as “within normal limits” for that biomarker.
  • a normal reference standard or level can be a value or number derived from a normal subject not having cholestasis, hyperbilirubinemia, or one or more symptoms thereof.
  • the reference sample, standard, or level is matched to the sample subject sample by at least one of the following criteria: age, weight, sex, disease stage, and overall health.
  • age e.g., any described herein, within the normal reference range
  • overall health e.g., any described herein, within the normal reference range
  • the term “term age” refers to the age of a patient (e.g., a newborn) born between 37 weeks of gestational age and 42 weeks of gestational age. For example, if the patient was born at 35 weeks of gestational age, the patient is at term age at 14 days old.
  • transgene refers to a recombinant nucleic acid (e.g., DNA or cDNA) encoding a gene product (e.g., a gene product described herein).
  • the gene product may be an RNA, peptide, or protein.
  • the transgene may include or be operably linked to one or more elements to facilitate or enhance expression, such as a promoter, enhancer(s), destabilizing domain(s), response element(s), reporter element(s), insulator element(s), polyadenylation signal(s), and/or other functional elements.
  • Embodiments of the disclosure may utilize any known suitable promoter, enhancer(s), destabilizing domain(s), response element(s), reporter element(s), insulator element(s), polyadenylation signal(s), and/or other functional elements.
  • the terms “treat” and “treatment” refer to therapeutic treatment, in which the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the progression of a neuromuscular disorder, such as XLMTM, among others.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms (e.g., stiffness and/or joint contractures), diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • symptoms e.g., stiffness and/or joint contractures
  • diminishment of extent of disease e.g., stiffness and/or joint contractures
  • stabilized i.e., not worsening
  • delay or slowing of disease progression e.g., delay or slowing of disease progression
  • amelioration or palliation of the disease state e.g., palliation of the disease state
  • remission whether partial or total
  • treatment of a patient may manifest in one or more detectable changes, such as an increase in the concentration of MTM1 protein or nucleic acids (e.g., DNA or RNA, such as mRNA) encoding MTM1, or an increase in MTM1 activity (e.g., by 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, or more.
  • MTM1 protein or nucleic acids e.g., DNA or RNA, such as mRNA
  • MTM1 activity e.g., by 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%
  • the concentration of MTM1 protein may be determined using protein detection assays known in the art, including ELISA assays described herein.
  • the concentration of MTM1- encoding nucleic acids may be determined using nucleic acid detection assays (e.g., RNA Seq assays) described herein.
  • treatment of a patient suffering from a neuromuscular disorder, such as XLMTM may manifest in improvements in a patient’s muscle function (e.g., skeletal muscle function) as well as improvements in muscle coordination. For example, manifestation of an improvement may include increasing diaphragm and/or respiratory muscle progression.
  • X-linked myotubular myopathy and “XLMTM” refer to the genetically inherited neuromuscular disorder that is caused by mutations of the MTM1 gene and is characterized by symptoms including mild to profound muscle weakness, hypotonia (diminished muscle tone), feeding difficulties, and/or severe breathing complications.
  • Human MTM1 has NCBI Gene ID NO 4534.
  • An exemplary wild-type human MTM1 nucleic acid sequence is provided in NCBI PATENT ATTORNEY DOCKET NO.51037-075WO2 RefSeq Acc. No. NM_000252.3, and an exemplary wild-type myotubularin 1 amino acid sequence is provided in NCBI RefSeq Acc. No. NP_000243.1.
  • vector refers to a nucleic acid, e.g., DNA or RNA, that may function as a vehicle for the delivery of a gene of interest into a cell (e.g., a mammalian cell, such as a human cell), such as for purposes of replication and/or expression.
  • a cell e.g., a mammalian cell, such as a human cell
  • exemplary vectors useful in conjunction with the compositions and methods described herein are plasmids, DNA vectors, RNA vectors, virions, or other suitable replicon (e.g., viral vector).
  • a variety of vectors have been developed for the delivery of polynucleotides encoding exogenous proteins into a prokaryotic or eukaryotic cell.
  • Expression vectors described herein contain a polynucleotide sequence as well as, e.g., additional sequence elements used for the expression of proteins and/or the integration of these polynucleotide sequences into the genome of a mammalian cell.
  • Certain vectors that can be used for the expression of transgenes described herein include plasmids that contain regulatory sequences, such as promoter and enhancer regions, which direct gene transcription.
  • Other useful vectors for expression of transgenes contain polynucleotide sequences that enhance the rate of translation of these genes or improve the stability or nuclear export of the mRNA that results from gene transcription.
  • sequence elements include, e.g., 5’ and 3’ untranslated regions, an internal ribosomal entry site (IRES), and polyadenylation signal site in order to direct efficient transcription of the gene carried on the expression vector.
  • the expression vectors described herein may also contain a polynucleotide encoding a marker for selection of cells that contain such a vector. Examples of a suitable marker include genes that encode resistance to antibiotics, such as ampicillin, chloramphenicol, kanamycin, or nourseothricin.
  • XLMTM X-linked myotubular myopathy
  • a patient e.g., a human patient
  • a viral vector such as an adeno-associated viral (AAV) vector, that contains a transgene encoding myotubularin 1 (MTM1).
  • AAV vector may be, for example, a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8 (AAV2/8).
  • the MTM1 transgene is operably linked to a transcription regulatory element, such as a promoter that drives gene expression in a liver cell.
  • the promoter contains an LP1 promoter, apolipoprotein E (ApoE) promoter, and/or alpha-1-antitrypsin (A1AT) promoter (e.g., an LP1 promoter, ApoE promoter, and/or A1AT promoter described herein).
  • ApoE apolipoprotein E
  • A1AT alpha-1-antitrypsin
  • the MTM1 transgene is operably linked to a constitutive transcription regulatory element, such as a phosphoglycerate kinase (PGK) promoter, an elongation factor-1 alpha PATENT ATTORNEY DOCKET NO.51037-075WO2 (EF1alpha) promoter, a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) promote, a cytomegalovirus (CMV) promoter, or a chicken- ⁇ -actin (CBA) promoter.
  • PGK phosphoglycerate kinase
  • EF1alpha elongation factor-1 alpha PATENT ATTORNEY DOCKET NO.51037-075WO2
  • GPDH glyceraldehyde 3-phosphate dehydrogenase
  • CMV cytomegalovirus
  • CBA chicken- ⁇ -actin
  • the viral vector includes two MTM1 transgenes: one under the control of a promoter that is active in liver tissue, and another that is under the control of a promoter that is active in muscle tissue.
  • the nanoparticle is a lipid nanoparticle.
  • Viral vectors useful in conjunction with this aspect of the disclosure include, without limitation, resamirigene bilparvovec.
  • the disclosure describes a method of reducing stiffness and/or joint contractures in a human patient diagnosed as having XLMTM by administration of one or more compositions described herein.
  • the disclosure describes a method of increasing diaphragm and/or respiratory muscle progression in a human patient diagnosed as having XLMTM by administration of one or more compositions described herein.
  • the disclosure describes a method of preventing cholestasis or hyperbilirubinemia in a human patient diagnosed as having XLMTM by administration of one or more compositions described herein.
  • X-Linked Myotubular Myopathy XLMTM is a rare, life-threatening, congenital myopathy caused by a mutation in the MTM1 gene and is characterized in most patients by profound muscle weakness and hypotonia at birth, which results in severe respiratory insufficiency, inability to sit up, stand or walk, and early mortality.
  • the myopathy associated with XLMTM impairs the development of motor skills such as sitting, standing, and walking. Affected infants may also have difficulties with feeding due to muscle weakness.
  • XLMTM XLMTM may also have weakness in the muscles that PATENT ATTORNEY DOCKET NO.51037-075WO2 control eye movement (ophthalmoplegia), weakness in other muscles of the face, and absent reflexes (areflexia).
  • ophthalmoplegia ophthalmoplegia
  • weakness in other muscles of the face ophthalmoplegia
  • absent reflexes ophthalmoplegia
  • muscle weakness often disrupts normal bone development and can lead to fragile bones, an abnormal curvature of the spine (scoliosis), and joint deformities (contractures) of the hips and knees.
  • Patients having XLMTM may have a large head with a narrow and elongated face and a high, arched roof of the mouth (palate). Patients may also have liver disease, recurrent ear and respiratory infections, or seizures. As a consequence of their severe breathing difficulties, patients having XLMTM usually survive only into early childhood; however, some patients with this condition have lived into adulthood.
  • the compositions and methods of the disclosure provide the important medical benefit of being able to prolong the lifetimes of such patients by restoring functional MTM1 expression.
  • compositions and methods described herein can be used to improve patients’ quality of life post- treatment (e.g., reducing stiffness and/or joint contractures or increasing diaphragm and/or respiratory muscle progression), as the disclosure provides a series of guidelines that can be used to determine a patient’s eligibility for being weaned off of mechanical ventilation.
  • Methods of Treatment In some embodiments, the patient is a newborn (e.g., 0-4 months old), an infant (e.g., 0-5 months old), a toddler (e.g., 6-12 months old), a child aged 1– ⁇ 3 years old, or a child aged 3– ⁇ 5 years old at the time of administration of the viral vector.
  • the patient is a newborn (e.g., 0-4 months old) at the time of administration of the viral vector.
  • the patient is a newborn that is about 0 to about 4 months old (e.g., 0 months old to about 4 months old, 1 month old to about 4 months old, 2 months old to about 4 months old, or 3 months old to about 4 months old).
  • the patient is 0 months old.
  • the patient is 1 month old.
  • the patient is 2 months old.
  • the patient is 3 months old.
  • the patient is 4 months old.
  • the patient is a newborn (e.g., less than about 4 months old) at the time of administration of the viral vector.
  • the patient is a newborn that is less than about 4 months old.
  • the patient is less than about 4 months old.
  • the patient is less than about 3 months old.
  • the patient is less than about 2 months old.
  • the patient is less than about 1 month old.
  • the patient is an infant (e.g., 0-5 months old) at the time of administration of the viral vector. For example.
  • the patient is an infant that is about 0 months old to about 5 months old (e.g., 0 months old to about 5 months old, 1 month old to about 5 months old, 2 months old to about 5 months old, 3 months old to about 5 months old, or 4 months old to about 5 months old).
  • the patient is 0 months old.
  • the patient is 1 month old.
  • the patient is 2 months old.
  • the patient is 3 months old.
  • the patient is 4 months old.
  • the patient is 3 months old.
  • the patient is 5 months old.
  • the patient is an infant (e.g., less than about 5 months old) at the time of administration of the viral vector.
  • the patient is an infant that is less than about 5 months old.
  • the patient is less than about 5 months old.
  • the patient is less than about 4 months old.
  • the patient is less than about 3 months old.
  • the patient is less than about 2 months old.
  • the patient is less than about 1 month old.
  • the patient is a toddler (e.g., 6-12 months old) at the time of administration of the viral vector.
  • the patient is an infant that is about 6 months old to about 12 months old (e.g., 6 months old to about 12 months old, 7 months old to about 12 months old, 8 months old to about 12 months old, 9 months old to about 12 months old, 10 months old to about 12 months old, or 11 months old to about 12 months old).
  • the patient is 6 months old.
  • the patient is 7 months old.
  • the patient is 8 months old.
  • the patient is 9 months old.
  • the patient is 10 months old.
  • the patient is 11 months old.
  • the patient is 12 months old.
  • the patient is a toddler (e.g., less than about 12 months old) at the time of administration of the viral vector.
  • the patient is a toddler that is less than about 12 months old.
  • the patient is less than about 12 months old.
  • the patient is less than about 11 months old.
  • the patient is less than about 10 months old.
  • the patient is less than about 9 months old.
  • the patient is less than about 8 months old.
  • the patient is less than about 7 months old.
  • the patient is less than about 6 months old.
  • the patient is less than about 5 months old. In some embodiments, the patient is less than about 4 months old.
  • the patient is less than about 3 months old. In some embodiments, the patient is less than about 2 months old. In some embodiments, the patient is less than about 1 month old. In some embodiments, the patient is a child aged 1- ⁇ 3 years old at the time of administration of the viral vector. For example, in some embodiments, the patient is a child that is about 1 year old to about 3 years old (e.g., 1 year old to about 3 years old or 2 years old to about 3 years old). In some embodiments, the patient is 1 year old. In some embodiments, the patient is 2 years old. In some embodiments, the patient is 3 years old. In some embodiments, the patient is a child (e.g., less than about 3 years old) at the time of administration of the viral vector.
  • the patient is a child that is less than about 3 years old. In some embodiments, the patient is less than about 3 years old. In some embodiments, the patient is less than about 2 years old. In some embodiments, the patient is less than about 1 year old. In some embodiments, the patient is less than about 12 months old. In some embodiments, the patient is less than about 11 months old. In some embodiments, the patient is less than about 10 months old. In some embodiments, the patient is less than about 9 months old. In PATENT ATTORNEY DOCKET NO.51037-075WO2 some embodiments, the patient is less than about 8 months old. In some embodiments, the patient is less than about 7 months old. In some embodiments, the patient is less than about 6 months old.
  • the patient is less than about 5 months old. In some embodiments, the patient is less than about 4 months old. In some embodiments, the patient is less than about 3 months old. In some embodiments, the patient is less than about 2 months old. In some embodiments, the patient is less than about 1 month old. In some embodiments, the patient is a child aged 3-5 years old at the time of administration of the viral vector. For example, in some embodiments, the patient is a child that is about 3 years old to about 5 years old (e.g., 3 years old to about 5 years old or 4 years old to about 5 years old). In some embodiments, the patient is 3 years old. In some embodiments, the patient is 4 years old. In some embodiments, the patient is 5 years old.
  • the patient is a child (e.g., less than about 5 years old) at the time of administration of the viral vector.
  • the patient is a child that is less than about 5 years old.
  • the patient is less than about 5 years old.
  • the patient is less than about 4 years old.
  • the patient is less than about 3 years old.
  • the patient is less than about 2 years old.
  • the patient is less than about 1 year old.
  • the patient is less than about 12 months old.
  • the patient is less than about 11 months old.
  • the patient is less than about 10 months old.
  • the patient is less than about 9 months old.
  • the patient is less than about 8 months old. In some embodiments, the patient is less than about 7 months old. In some embodiments, the patient is less than about 6 months old. In some embodiments, the patient is less than about 5 months old. In some embodiments, the patient is less than about 4 months old. In some embodiments, the patient is less than about 3 months old. In some embodiments, the patient is less than about 2 months old. In some embodiments, the patient is less than about 1 month old.
  • the patient is from about 1 month old to about 5 years old (e.g., about 1 month old to about 5 years old, about 2 months old to about 5 years old, about 3 months old to about 5 years old, about 4 months old to about 5 years old, about 5 months old to about 5 years old, about 6 months old to about 5 years old, about 1 year old to about 5 years old, about 2 years old to about 5 years old, about 3 years old to about 5 years old, or about 4 years old to about 5 years old) at the time of administration of the viral vector.
  • the patient was born at greater than or equal to 35 weeks of gestational age (e.g., 35 weeks of gestational age, 36 weeks of gestational age, 37 weeks of gestational age, 38 weeks of gestational age, 39 weeks of gestational age, 40 weeks of gestational age, 41 weeks of gestational age, and 42 weeks of gestational age) and is between adjusted term age (e.g., 37 weeks of gestational age or greater) to about 5 years old at the time of administration of the viral vector. For example, if the patient was born at 35 weeks of gestational age, the patient is at term age at 14 days old.
  • adjusted term age e.g., 37 weeks of gestational age or greater
  • the patient was born at 35 weeks of gestational age and is between adjusted term age to about 5 years old (e.g., 14 days old to about 5 years old, 15 days old to about 5 PATENT ATTORNEY DOCKET NO.51037-075WO2 years old, 16 days old to about 5 years old, 17 days old to about 5 years old, 18 days old to about 5 years old, 19 days old to about 5 years old, 20 days old to about 5 years old, 25 days old to about 5 years old, one month old to about 5 years old, two months old to about 5 years old, 3 months old to about 5 years old, 4 months old to about 5 years old, 5 months old to about 5 years old, 6 months old to about 5 years old, 1 year old to about 5 years old, 2 years old to about 5 years old, 3 years old to about 5 years old, and 4 years old to about 5 years old) at the time of administration of the viral vector.
  • adjusted term age to about 5 years old e.g., 14 days old to about 5 years old, 15 days old to about 5 PAT
  • the patient was born at 36 weeks of gestational age and is between adjusted term age to about 5 years old (e.g., 7 days old to about 5 years old, 8 days old to about 5 years old, 9 days old to about 5 years old, 10 days old to about 5 years old, 11 days old to about 5 years old, 12 days old to about 5 years old, 13 days old to about 5 years old, 14 days old to about 5 years old, 15 days old to about 5 years old, 16 days old to about 5 years old, 17 days old to about 5 years old, 18 days old to about 5 years old, 19 days old to about 5 years old, 20 days old to about 5 years old, 25 days old to about 5 years old, one month old to about 5 years old, two months old to about 5 years old, 3 months old to about 5 years old, 4 months old to about 5 years old, 5 months old to about 5 years old, 6 months old to about 5 years old, 1 year old to about 5 years old, 2 years old to about 5 years old, 3 years old to about 5 years old, and 4 years old to
  • the patient was born at 37 weeks of gestational age and is between adjusted term age to about 5 years old (e.g., 1 day old to about 5 years old, 2 days old to about 5 years old, 3 days old to about 5 years old, 4 days old to about 5 years old, 5 days old to about 5 years old, 6 days old to about 5 years old, 7 days old to about 5 years old, 8 days old to about 5 years old, 9 days old to about 5 years old, 10 days old to about 5 years old, 11 days old to about 5 years old, 12 days old to about 5 years old, 13 days old to about 5 years old, 14 days old to about 5 years old, 15 days old to about 5 years old, 16 days old to about 5 years old, 17 days old to about 5 years old, 18 days old to about 5 years old, 19 days old to about 5 years old, 20 days old to about 5 years old, 25 days old to about 5 years old, one month old to about 5 years old, two months old to about 5 years old, 3 months old to about 5 years old, 4 months old to about 5 years old,
  • the patient is male. In some embodiments, the patient is female.
  • Cholestasis and Hyperbilirubinemia Cholestasis is any condition in which the flow of bile acid from the liver is slowed or blocked, while hyperbilirubinemia is a condition in which there is an accumulation of bilirubin in the blood and serum bile acids appear to remain normal.
  • cholestatic syndromes are characterized by marked bile acidemia with normal to slightly elevated bilirubin levels.
  • PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, the patient is monitored for the development of cholestasis. In some embodiments, the patient is monitored for the development of hyperbilirubinemia.
  • the patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof.
  • the patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof by evaluating a parameter in a blood sample obtained from the patient, wherein a finding that the parameter is above a reference level identifies the patient as having cholestasis, hyperbilirubinemia, or one or more symptoms thereof.
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof when the patient exhibits one or more parameters (e.g., total bile acids level, gamma-glutamyl transferase (GGT) level, alkaline phosphatase (ASP) level, aspartate aminotransferase (AST) level, and/or alanine aminotransferase (ALT) level), as measured in a serum bile acid test and/or blood test (e.g., a liver function test (LFT)), that is greater than or less than the age-adjusted norm.
  • GTT total bile acids level
  • ASP alkaline phosphatase
  • AST aspartate aminotransferase
  • ALT alanine aminotransferase
  • a patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof when the patient exhibits a bilirubin level, as measured in a blood test (e.g., a bilirubin test), that is greater than the norm.
  • a blood test e.g., a bilirubin test
  • the patient does not have a history of cholestasis or hyperbilirubinemia.
  • the patient does not have a history of any underlying liver disease.
  • Vectors for Delivery of Exogenous Nucleic Acids to Target Cells Viral Vectors for Nucleic Acid Delivery
  • a gene of interest e.g., a transgene encoding MTM1
  • a target cell e.g., a mammalian cell, such as a human cell
  • Recombinant viral genomes are particularly useful vectors for gene delivery because they deliver the gene of interest to the nucleus of the target cells.
  • polynucleotides contained within such genomes may be incorporated into the genome of a target cell by generalized or specialized transduction.
  • recombinant viral vectors used to deliver genes of interest include AAV, adenovirus (e.g., Ad5, Ad26, Ad34, Ad35, and Ad48), parvovirus (e.g., adeno-associated viruses), coronavirus, negative strand RNA viruses such as orthomyxovirus (e.g., influenza virus), rhabdovirus (e.g., rabies and vesicular stomatitis virus), paramyxovirus (e.g., measles and Sendai), positive strand RNA viruses, such as picornavirus and alphavirus, and double stranded DNA viruses including adenovirus, herpesvirus (e.g., Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomegalovirus), and poxvirus (e.g., vaccinia, modified vaccinia Ankara (MV
  • viruses useful for delivering polynucleotides encoding antibody light and heavy chains or antibody fragments of the invention include Norwalk virus, togavirus, flavivirus, reoviruses, papovavirus, hepadnavirus, and hepatitis virus, PATENT ATTORNEY DOCKET NO.51037-075WO2 for example.
  • retroviruses include: avian leukosis- sarcoma, mammalian C-type, B-type viruses, D-type viruses, HTLV-BLV group, lentivirus, spumavirus (Coffin, J. M., Retroviridae: The viruses and their replication, In Fundamental Virology, Third Edition, B. N.
  • murine leukemia viruses include murine leukemia viruses, murine sarcoma viruses, mouse mammary tumor virus, bovine leukemia virus, feline leukemia virus, feline sarcoma virus, avian leukemia virus, human T-cell leukemia virus, baboon endogenous virus, Gibbon ape leukemia virus, Mason Pfizer monkey virus, simian immunodeficiency virus, simian sarcoma virus, Rous sarcoma virus and lentiviruses.
  • nucleic acids of the compositions and methods described herein are incorporated into recombinant AAV (rAAV) vectors and/or virions in order to facilitate their introduction into a cell.
  • rAAV vectors useful in the invention are recombinant nucleic acid constructs that include (1) a transgene to be expressed (e.g., a polynucleotide encoding a MTM1 protein) and (2) viral nucleic acids that facilitate integration and expression of the heterologous genes.
  • the viral nucleic acids may include those sequences of AAV that are required in cis for replication and packaging (e.g., functional inverted terminal repeats (ITRs)) of the DNA into a virion.
  • ITRs functional inverted terminal repeats
  • the transgene encodes MTM1, which is useful for correcting a MTM1 mutation in patients suffering from neuromuscular disorders, such as XLMTM.
  • Such rAAV vectors may also contain marker or reporter genes.
  • Useful rAAV vectors have one or more of the AAV wild type genes deleted in whole or in part but retain functional flanking ITR sequences.
  • the AAV ITRs may be of any serotype (e.g., derived from serotype 2) suitable for a particular application.
  • rAAV vectors Methods for using rAAV vectors are described, for example, in Tal et al., J. Biomed. Sci.7:279-291 (2000), and Monahan and Samulski, Gene Delivery 7:24-30 (2000), the disclosures of each of which are incorporated herein by reference as they pertain to AAV vectors for gene delivery.
  • the nucleic acids and vectors described herein can be incorporated into a rAAV virion in order to facilitate introduction of the nucleic acid or vector into a cell.
  • the capsid proteins of AAV compose the exterior, non-nucleic acid portion of the virion and are encoded by the AAV cap gene.
  • the cap gene encodes three viral coat proteins, VP1, VP2 and VP3, which are required for virion assembly.
  • rAAV virions useful in conjunction with the compositions and methods described herein include PATENT ATTORNEY DOCKET NO.51037-075WO2 those derived from a variety of AAV serotypes including AAV 1, 2, 3, 4, 5, 6, 7, 8 and 9.
  • rAAV virions that include at least one serotype 1 capsid protein may be particularly useful.
  • rAAV virions that include at least one serotype 6 capsid protein may also be particularly useful, as serotype 6 capsid proteins are structurally similar to serotype 1 capsid proteins, and thus are expected to also result in high expression of MTM1 in muscle cells.
  • rAAV serotype 9 has also been found to be an efficient transducer of muscle cells.
  • AAV vectors and AAV proteins of different serotypes are described, for example, in Chao et al., Mol. Ther.2:619-623 (2000); Davidson et al., Proc. Natl. Acad. Sci. USA 97:3428-3432 (2000); Xiao et al., J. Virol.72:2224-2232 (1998); Halbert et al., J. Virol.74:1524-1532 (2000); Halbert et al., J. Virol.75:6615-6624 (2001); and Auricchio et al., Hum. Molec.
  • Pseudotyped vectors include AAV vectors of a given serotype (e.g., AAV9) pseudotyped with a capsid gene derived from a serotype other than the given serotype (e.g., AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, etc.).
  • a representative pseudotyped vector is an AAV8 vector encoding a therapeutic protein pseudotyped with a capsid gene derived from AAV serotype 2.
  • Techniques involving the construction and use of pseudotyped rAAV virions are known in the art and are described, for example, in Duan et al., J. Virol.75:7662-7671 (2001); Halbert et al., J. Virol. 74:1524-1532 (2000); Zolotukhin et al., Methods, 28:158-167 (2002); and Auricchio et al., Hum. Molec. Genet., 10:3075-3081 (2001).
  • AAV virions that have mutations within the virion capsid may be used to infect particular cell types more effectively than non-mutated capsid virions.
  • suitable AAV mutants may have ligand insertion mutations for the facilitation of targeting AAV to specific cell types.
  • the construction and characterization of AAV capsid mutants including insertion mutants, alanine screening mutants, and epitope tag mutants are described in Wu et al., J. Virol.74:8635-45 (2000).
  • Other rAAV virions that can be used in methods of the invention include those capsid hybrids that are generated by molecular breeding of viruses as well as by exon shuffling. See, e.g., Soong et al., Nat.
  • Resamirigene Bilparvovec As described herein, a pseudotyped AAV vector including a nucleic acid sequence encoding a MTM1 gene operably linked to a desmin promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8) as well as the other genetic components listed in Table 1, refers to the compound known by the international proprietary name (INN) of resamirigene bilparvovec.
  • INN international proprietary name
  • Resamirigene bilparvovec is a non-replicating recombinant AAV8 vector expressing a non- codon-optimized human MTM1 cDNA under the control of the muscle-specific human desmin PATENT ATTORNEY DOCKET NO.51037-075WO2 promoter.
  • the MTM1 expression cassette was built by cloning a synthetic DNA sequence complementary to the coding portion (nucleotides 43-1864) of the wild-type human MTM1 transcript (NCBI Ref. Seq NM_000252.3) downstream of the 1.05-kb human desmin enhancer/promoter region.
  • the second intron and polyadenylation sequence of the human ⁇ -globin gene were inserted upstream and downstream respectively of the MTM1 synthetic cDNA to mediate RNA processing.
  • the expression cassette was flanked by AAV serotype-2 (AAV2) inverted terminal repeats (ITRs).
  • AAV2 AAV2
  • ITRs inverted terminal repeats
  • a method of treating a disorder (e.g., XLMTM) or alleviating one or more symptoms (e.g., stiffness and/or join contractures or need for diaphragm and/or respiratory muscle progression) of a disorder (e.g., XLMTM) in a human patient in need thereof includes administering to the patient a therapeutically effective amount of resamirigene bilparvovec during a treatment period.
  • a method of weaning a human patient off of mechanical ventilation includes a patient that has previously been administered a therapeutically effective amount of resamirigene bilparvovec.
  • Table 1 The components of resamirigene bilparvovec are shown in Table 1, below:
  • Resamirigene Bilparvovec nucleic acid sequence (SEQ ID NO: 1) As described herein, resamirigene bilparvovec refers to the AAV vector having the nucleic acid sequence of SEQ ID NO: 1, shown below: TCGCGCGTTT CGGTGATGAC GGTGAAAACC TCTGACACAT GCAGCTCCCG 50 GACGTCATTG TCGATCCTGC AGGCGTACGG CATAGCTAAC 100 AAGGTGTGGA AAAAGAATTA GTGGTTAGAG AGTGAGCTAT TCGTTGAAAC 150 AATTGCGTTC TTGAAACAAT TCTTGCTGGT AAAATGTCAC ATTTTATGTG 200 ACTACAGGTG GAGGATTGGC ACATAACCTA ACCAGTGGGG GAAACAATTG 250 ACCTCTGGAT TTGTCCAAGT GTATAGTAGC ATTTGCCCAA TCGAATGGTC 300 CTGGTAAGGT GTTAATGT
  • transcription regulatory elements described herein may contain an ApoE and/or A1AT promoter, such as the chimeric promoter set forth in SEQ ID NO: 2, or a functional portion thereof.
  • Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequence.
  • transcription regulatory elements described herein may contain an LP1 promoter, or functional portion thereof.
  • the regulatory element may contain a PATENT ATTORNEY DOCKET NO.51037-075WO2 LP1 promoter as set forth in SEQ ID NO: 3, or a functional portion thereof.
  • Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequences.
  • Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein include promoters that stimulate expression of a transgene operably linked to a liver-specific promoter.
  • promoters are the ApoE/A1At and LP1 promoter, or a variant thereof (e.g., a variant having at least 85% sequence identity (for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity) to the nucleic acid sequence of the wild-type promoter locus and that is able to stimulate transcription of a transgene operably linked thereto upon introduction into a cell) or functional portion thereof.
  • Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein include promoters that stimulate expression of a transgene operably linked to a muscle-specific promoter.
  • promoters are the desmin or MCK promoters, or a variant thereof (e.g., a variant having at least 85% sequence identity (for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity) to the nucleic acid sequence of the wild-type promoter locus and that is able to stimulate transcription of a transgene operably linked thereto upon introduction into a cell) or functional portion thereof.
  • Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein include promoters that stimulate expression of a transgene operably linked to a ubiquitous promoter.
  • promoters are PGK, Ef1a, and GAPDH or a variant thereof (e.g., a variant having at least 85% sequence identity (for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity) to the nucleic acid sequence of the wild-type promoter locus and that is able to stimulate transcription of a transgene operably linked thereto upon introduction into a cell) or functional portion thereof.
  • Transcription regulatory elements described herein may contain a SV40 enhancer or a functional portion thereof.
  • the regulatory element may contain a SV40 enhancer as set forth in SEQ ID NO: 4, or a functional portion thereof.
  • Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequences.
  • Transcription regulatory elements described herein may contain a ⁇ -globin enhancer or a functional portion thereof.
  • the regulatory element may contain a ⁇ -globin enhancer as set forth in SEQ ID NO: 5, or a functional portion thereof.
  • Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, PATENT ATTORNEY DOCKET NO.51037-075WO2 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequences.
  • Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein include enhancers that enhance expression of a transgene.
  • promoters examples include PGK, Ef1alpha, and GAPDH, or a variant thereof (e.g., a variant having at least 85% sequence identity (for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity) to the nucleic acid sequence of the wild- type promoter locus and that is able to stimulate transcription of a transgene operably linked thereto upon introduction into a cell) or functional portion thereof.
  • sequence identity for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity
  • nucleic acid regulatory elements PATENT ATTORNEY DOCKET NO.51037-075WO2
  • Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the nucleic acid sequences set forth in Table 2.
  • Transgenes Pseudotyped AAV vectors described herein may include a nucleic acid sequence encoding a MTM1 gene operably linked to a promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8).
  • the nucleic acid sequence encoding a MTM1 gene encodes a human MTM1 gene.
  • the nucleic acid sequence encoding a MTM1 gene encodes a mouse MTM1 gene.
  • the MTM1 gene is codon-optimized.
  • the promoter is a liver-specific promoter. In some embodiments, the promoter is a muscle-specific promoter.
  • the promoter is a ubiquitously expressed promoter.
  • two MTM1 genes may be expressed simultaneously using different promoters on the same AAV vector.
  • Transgenes that may be used in conjunction with the compositions and methods described herein may contain a human MTM1 transgene, as set forth in SEQ ID NO: 6, or a functional portion thereof. Additional transgenes useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequence.
  • PATENT ATTORNEY DOCKET NO.51037-075WO2 Transgenes that may be used in conjunction with the compositions and methods described herein may contain a mouse MTM1 transgene, as set forth in SEQ ID NO: 7, or a functional portion thereof. Additional transgenes useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequence.
  • sequence identity e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity
  • a method of treating a disorder (e.g., XLMTM) or alleviating one or more symptoms (e.g., stiffness and/or join contractures or need for diaphragm and/or respiratory muscle progression) of a disorder (e.g., XLMTM) in a human patient in need thereof includes administering to the patient a therapeutically effective amount of a pseudotyped AAV vector including a nucleic acid sequence encoding a MTM1 gene operably linked to a liver-specific or muscle specific promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8) as well as the other genetic components listed in Table 2 during a treatment period.
  • a pseudotyped AAV vector including a nucleic acid sequence encoding a MTM1 gene operably linked to a liver-specific or muscle specific promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8) as well as the other genetic components listed in Table 2 during a treatment period
  • a method of weaning a human patient off of mechanical ventilation includes a patient that has previously been administered a therapeutically effective amount of a pseudotyped AAV vector including a nucleic acid sequence encoding a MTM1 gene operably linked to a liver-specific or muscle specific promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8) as well as the other genetic components listed in Table 2.
  • a pseudotyped AAV vector including a nucleic acid sequence encoding a MTM1 gene operably linked to a liver-specific or muscle specific promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8) as well as the other genetic components listed in Table 2.
  • transgenes PATENT ATTORNEY DOCKET NO.51037-075WO2
  • Additional transgenes useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the nucleic acid sequences set forth in Table 3.
  • a transgene such as a MTM1 transgene described herein
  • electroporation can be used to permeabilize mammalian cells (e.g., human target cells) by the application of an electrostatic potential to the cell of interest.
  • Mammalian cells, such as human cells, subjected to an external electric field in this manner are subsequently predisposed to the uptake of exogenous nucleic acids (e.g., nucleic acids capable of expression in e.g., neurons, glial cells, or non-neural cells, such as colon and kidney cells).
  • exogenous nucleic acids e.g., nucleic acids capable of expression in e.g., neurons, glial cells, or non-neural cells, such as colon and kidney cells.
  • NUCLEOFECTIONTM utilizes an applied electric field in order to stimulate the uptake of exogenous polynucleotides into the nucleus of a eukaryotic cell.
  • NUCLEOFECTIONTM and protocols useful for performing this technique are described in detail, e.g., in Distler et al., Experimental Dermatology 14:315 (2005), as well as in US 2010/0317114, the disclosures of each of which are incorporated herein by reference.
  • An additional technique useful for the transfection of target cells is the squeeze-poration methodology.
  • This technique induces the rapid mechanical deformation of cells in order to stimulate the uptake of exogenous DNA through membranous pores that form in response to the applied stress.
  • This technology is advantageous in that a vector is not required for delivery of nucleic acids into a cell, such as a human target cell.
  • Squeeze-poration is described in detail, e.g., in Sharei et al., J. Vis. Exp. 81:e50980 (2013), the disclosure of which is incorporated herein by reference.
  • Lipofection represents another technique useful for transfection of target cells.
  • This method involves the loading of nucleic acids into a liposome, which often presents cationic functional groups, PATENT ATTORNEY DOCKET NO.51037-075WO2 such as quaternary or protonated amines, towards the liposome exterior.
  • PATENT ATTORNEY DOCKET NO.51037-075WO2 such as quaternary or protonated amines
  • This promotes electrostatic interactions between the liposome and a cell due to the anionic nature of the cell membrane, which ultimately leads to uptake of the exogenous nucleic acids, for example, by direct fusion of the liposome with the cell membrane or by endocytosis of the complex.
  • Lipofection is described in detail, for example, in US 7,442,386, the disclosure of which is incorporated herein by reference.
  • cationic molecules that associate with polynucleotides so as to impart a positive charge favorable for interaction with the cell membrane are activated dendrimers (described, e.g., in Dennig, Top Curr Chem.228:227 (2003), the disclosure of which is incorporated herein by reference) polyethylenimine, and DEAE-dextran, the use of which as a transfection agent is described in detail, for example, in Gulick et al., Curr Protoc Mol Biol.40:1:9.2:9.2.1 (1997), the disclosure of which is incorporated herein by reference.
  • laserfection also called optical transfection
  • Impalefection is another technique that can be used to deliver genetic material to target cells. It relies on the use of nanomaterials, such as carbon nanofibers, carbon nanotubes, and nanowires. Needle-like nanostructures are synthesized perpendicular to the surface of a substrate. DNA containing the gene, intended for intracellular delivery, is attached to the nanostructure surface.
  • MAGNETOFECTIONTM can also be used to deliver nucleic acids to target cells.
  • the principle of MAGNETOFECTIONTM is to associate nucleic acids with cationic magnetic nanoparticles.
  • the magnetic nanoparticles are made of iron oxide, which is fully biodegradable, and coated with specific cationic proprietary molecules varying upon the applications.
  • sonoporation a technique that involves the use of sound (typically ultrasonic frequencies) for modifying the permeability of the cell plasma membrane permeabilize the cells and allow PATENT ATTORNEY DOCKET NO.51037-075WO2 polynucleotides to penetrate the cell membrane.
  • sound typically ultrasonic frequencies
  • PATENT ATTORNEY DOCKET NO.51037-075WO2 polynucleotides to penetrate the cell membrane.
  • This technique is described in detail, e.g., in Rhodes et al., Methods Cell Biol.82:309 (2007), the disclosure of which is incorporated herein by reference.
  • Microvesicles represent another potential vehicle that can be used to modify the genome of a target cell according to the methods described herein.
  • microvesicles that have been induced by the co-overexpression of the glycoprotein VSV-G with, e.g., a genome-modifying protein, such as a nuclease, can be used to efficiently deliver proteins into a cell that subsequently catalyze the site-specific cleavage of an endogenous polynucleotide sequence so as to prepare the genome of the cell for the covalent incorporation of a polynucleotide of interest, such as a gene or regulatory sequence.
  • a genome-modifying protein such as a nuclease
  • vesicles also referred to as Gesicles
  • Methylation changes in early embryonic genes in cancer [abstract] in: Proceedings of the 18th Annual Meeting of the American Society of Gene and Cell Therapy; 2015 May 13, Abstract No.122.
  • Incorporation of Target Genes by Gene Editing Techniques In addition to the above, a variety of tools have been developed that can be used for the incorporation of a gene of interest into a target cell, such as a human cell.
  • Transposons are polynucleotides that encode transposase enzymes and contain a polynucleotide sequence or gene of interest flanked by 5’ and 3’ excision sites. Once a transposon has been delivered into a cell, expression of the transposase gene commences and results in active enzymes that cleave the gene of interest from the transposon. This activity is mediated by the site- specific recognition of transposon excision sites by the transposase. In some instances, these excision sites may be terminal repeats or inverted terminal repeats.
  • the gene of interest can be integrated into the genome of a mammalian cell by transposase-catalyzed cleavage of similar excision sites that exist within the nuclear genome of the cell. This allows the gene of interest to be inserted into the cleaved nuclear DNA at the complementary excision sites, and subsequent covalent ligation of the phosphodiester bonds that join the gene of interest to the DNA of the mammalian cell genome completes the incorporation process.
  • the transposon may be a retrotransposon, such that the gene encoding the target gene is first transcribed to an RNA product and then reverse-transcribed to DNA before incorporation in the mammalian cell genome.
  • transposon systems are the piggybac transposon (described in detail in, e.g., WO 2010/085699) and the sleeping beauty transposon (described in detail in, e.g., US 2005/0112764), the disclosures of each of which are incorporated herein by reference as they pertain to transposons for use in gene delivery to a cell of interest.
  • Another tool for the integration of target genes into the genome of a target cell is the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas system, a system that originally evolved as an adaptive defense mechanism in bacteria and archaea against viral infection.
  • CRISPR clustered regularly interspaced short palindromic repeats
  • the CRISPR/Cas system includes palindromic repeat sequences within plasmid DNA and an associated Cas9 nuclease. This ensemble of DNA and protein directs site specific DNA cleavage of a target PATENT ATTORNEY DOCKET NO.51037-075WO2 sequence by first incorporating foreign DNA into CRISPR loci. Polynucleotides containing these foreign sequences and the repeat-spacer elements of the CRISPR locus are in turn transcribed in a host cell to create a guide RNA, which can subsequently anneal to a target sequence and localize the Cas9 nuclease to this site.
  • CRISPR/Cas to modulate gene expression has been described in, for example, US Patent No.8,697,359, the disclosure of which is incorporated herein by reference as it pertains to the use of the CRISPR/Cas system for genome editing.
  • Alternative methods for site-specifically cleaving genomic DNA prior to the incorporation of a gene of interest in a target cell include the use of zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs).
  • ZFNs zinc finger nucleases
  • TALENs transcription activator-like effector nucleases
  • these enzymes do not contain a guiding polynucleotide to localize to a specific target sequence. Target specificity is instead controlled by DNA binding domains within these enzymes.
  • ZFNs and TALENs in genome editing applications is described, e.g., in Urnov et al., Nat. Rev. Genet.11:636 (2010); and in Joung et al., Nat. Rev. Mol. Cell Biol.14:49 (2013), the disclosure of each of which are incorporated herein by reference as they pertain to compositions and methods for genome editing.
  • Additional genome editing techniques that can be used to incorporate polynucleotides encoding target genes into the genome of a target cell include the use of ARCUSTM meganucleases that can be rationally designed so as to site-specifically cleave genomic DNA.
  • Single chain meganucleases can be modified at certain amino acid positions in order to create nucleases that selectively cleave DNA at desired locations, enabling the site-specific incorporation of a target gene into the nuclear DNA of a target cell.
  • These single-chain nucleases have been described extensively in, for example, US Patent Nos.8,021,867 and US 8,445,251, the disclosures of each of which are incorporated herein by reference as they pertain to compositions and methods for genome editing.
  • compositions and Routes of Administration may contain a transgene, such as a transgene encoding MTM1 and may be incorporated into a vehicle for administration into a patient, such as a human patient suffering from a neuromuscular disorder (for example, XLMTM).
  • a transgene such as a transgene encoding MTM1
  • a vehicle for administration into a patient such as a human patient suffering from a neuromuscular disorder (for example, XLMTM).
  • Pharmaceutical compositions containing vectors, such as viral vectors, that contain the transcription regulatory elements (e.g., a ApoE/A1AT promoter) described herein operably linked to a therapeutic transgene can be prepared using methods known in the art.
  • compositions can be prepared using, e.g., physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical PATENT ATTORNEY DOCKET NO.51037-075WO2 Sciences 16th edition, Osol, A. Ed. (1980); incorporated herein by reference), and in a desired form, e.g., in the form of lyophilized formulations or aqueous solutions.
  • Viral vectors such as AAV vectors and others described herein, containing the transcription regulatory element operably linked to a therapeutic transgene may be administered to a patient (e.g., a human patient) by a variety of routes of administration.
  • the route of administration may vary, for example, with the onset and severity of disease, and may include, e.g., intradermal, transdermal, parenteral, intravenous, intramuscular, intrahepatic, intranasal, subcutaneous, percutaneous, intratracheal, intraperitoneal, intraarterial, intravascular, inhalation, perfusion, lavage, and oral administration.
  • Intravascular administration includes delivery into the vasculature of a patient.
  • the administration is into a vessel considered to be a vein (intravenous), and in some administration, the administration is into a vessel considered to be an artery (intraarterial).
  • Veins include, but are not limited to, the internal jugular vein, a peripheral vein, a coronary vein, a hepatic vein, the portal vein, great saphenous vein, the pulmonary vein, superior vena cava, inferior vena cava, a gastric vein, a splenic vein, inferior mesenteric vein, superior mesenteric vein, cephalic vein, and/or femoral vein.
  • Arteries include, but are not limited to, coronary artery, pulmonary artery, hepatic artery, brachial artery, internal carotid artery, aortic arch, femoral artery, peripheral artery, and/or ciliary artery.
  • delivery may be through or to an arteriole or capillary.
  • Mixtures of the nucleic acids and viral vectors described herein may be prepared in water suitably mixed with one or more excipients, carriers, or diluents.
  • Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (described in US 5,466,468, the disclosure of which is incorporated herein by reference).
  • the formulation may be sterile and may be fluid to the extent that easy syringability exists. Formulations may be stable under the conditions of manufacture and storage and may be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • polyol e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • a solution containing a pharmaceutical composition described herein may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, PATENT ATTORNEY DOCKET NO.51037-075WO2 intrahepatic, subcutaneous, and intraperitoneal administration.
  • sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in 1 mL of isotonic NaCl solution and either added to 1000 mL of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • kits for human administration, preparations may meet sterility, pyrogenicity, general safety, and purity standards as required by FDA Office of Biologics standards.
  • Kits The compositions described herein can be provided in a kit for use in treating a neuromuscular disorder (e.g., XLMTM).
  • the kit may include one or more viral vectors as described herein.
  • the kit can include a package insert that instructs a user of the kit, such as a physician of skill in the art, to perform any one of the methods described herein.
  • the kit may optionally include a syringe or other device for administering the composition.
  • the kit may include one or more additional therapeutic agents.
  • the kit can include a package insert that instructs a user of the kit, such as a physician of skill in the art, to perform any one of the methods described herein.
  • the kit may optionally include a syringe or other device for administering the composition.
  • the kit may include one or more additional therapeutic agents.
  • Dosing Regimens Dosing Regimens Involving AAV-MTM1 Vectors Using the compositions and methods of the disclosure, a patient having a neuromuscular disorder (e.g., XLMTM) may be administered an AAV vector containing a transgene encoding MTM1 in an amount of about 1.3 x 10 14 vg/kg.
  • a neuromuscular disorder e.g., XLMTM
  • the AAV vector is administered to the patient in an amount of less than about 3 x 10 14 vg/kg (e.g., in an amount of less than about 3 x 10 14 vg/kg, 2.9 x 10 14 vg/kg, 2.8 x 10 14 vg/kg, 2.7 x 10 14 vg/kg, 2.6 x 10 14 vg/kg, 2.5 x 10 14 vg/kg, 2.4 x 10 14 vg/kg, 2.3 x 10 14 vg/kg, 2.2 x 10 14 vg/kg, 2.1 x 10 14 vg/kg, 2 x 10 14 vg/kg, 1.9 x 10 14 vg/kg, 1.8 x 10 14 vg/kg, 1.7 x
  • the AAV vector may be administered to the patient in an amount of about 3 x 10 14 vg/kg, 2.9 x 10 14 vg/kg, 2.8 x 10 14 vg/kg, 2.7 x 10 14 vg/kg, 2.6 x 10 14 vg/kg, 2.5 x 10 14 vg/kg, 2.4 x 10 14 vg/kg, 2.3 x 10 14 vg/kg, 2.2 x 10 14 vg/kg, 2.1 x 10 14 vg/kg, 2 x 10 14 vg/kg, 1.9 x 10 14 vg/kg, 1.8 x 10 14 vg/kg, 1.7 x 10 14 vg/kg, 1.6 x 10 14 vg/kg, 1.5 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1
  • the AAV vector is administered to the patient in an amount of less than about 2.5 x 10 14 vg/kg (e.g., in an amount of less than about 2.5 x 10 14 vg/kg, 2.4 x 10 14 vg/kg, 2.3 x 10 14 vg/kg, 2.2 x 10 14 vg/kg, 2.1 x 10 14 vg/kg, 2 x 10 14 vg/kg, 1.9 x 10 14 vg/kg, 1.8 x 10 14 vg/kg, 1.7 x 10 14 vg/kg, 1.6 x 10 14 vg/kg, 1.5 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10
  • the AAV vector may be administered to the patient in an amount of about 2.5 x 10 14 vg/kg, 2.4 x 10 14 vg/kg, 2.3 x 10 14 vg/kg, 2.2 x 10 14 vg/kg, 2.1 x 10 14 vg/kg, 2 x 10 14 vg/kg, 1.9 x 10 14 vg/kg, 1.8 x 10 14 vg/kg, 1.7 x 10 14 vg/kg, 1.6 x 10 14 vg/kg, 1.5 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 13 vg/kg, 1 x 10 12 vg/kg, 1 x 10 11 vg/kg, 1 x 10 10 10
  • the AAV vector is administered to the patient in an amount of less than about 2 x 10 14 vg/kg (e.g., in an amount of less than about 2 x 10 14 vg/kg, 1.9 x 10 14 vg/kg, 1.8 x 10 14 vg/kg, 1.7 x 10 14 vg/kg, 1.6 x 10 14 vg/kg, 1.5 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 13 vg/kg, 1 x 10 12 vg/kg, 1 x 10 11 vg/kg, 1 x 10 10 vg/kg, 1 x 10 9 vg/kg, 1 x 10 8 vg/kg, or less).
  • the AAV vector may be administered to the patient in an amount of about 2 x 10 14 vg/kg, 1.9 x 10 14 vg/kg, 1.8 x 10 14 vg/kg, 1.7 x 10 14 vg/kg, 1.6 x 10 14 vg/kg, 1.5 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 13 vg/kg, 1 x 10 12 vg/kg, 1 x 10 11 vg/kg, 1 x 10 10 vg/kg, 1 x 10 9 vg/kg, 1 x 10 8 vg/kg.
  • the AAV vector is administered to the patient in an amount of less than about 1.5 x 10 14 vg/kg (e.g., less than about 1.5 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 13 vg/kg, 1 x 10 12 vg/kg, 1 x 10 11 vg/kg, 1 x 10 10 vg/kg, 1 x 10 9 vg/kg, 1 x 10 8 vg/kg, or less).
  • 1.5 x 10 14 vg/kg e.g., less than about 1.5 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.2 x 10 14 vg/kg,
  • the AAV vector may be administered to the patient in an amount of about 1.5 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 13 vg/kg, 1 x 10 12 vg/kg, 1 x 10 11 vg/kg, 1 x 10 10 vg/kg, 1 x 10 9 vg/kg, 1 x 10 8 vg/kg.
  • the AAV vector is administered to the patient in an amount of less than about 1 x 10 14 vg/kg (e.g., less than about 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 13 vg/kg, 1 x 10 12 vg/kg, 1 x 10 11 vg/kg, 1 x 10 10 vg/kg, 1 x 10 9 vg/kg, 1 x 10 8 vg/kg, or less).
  • 1 x 10 14 vg/kg e.g., less than about 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 13 vg/kg, 1 x 10 12 vg/kg, 1 x 10 11 vg/kg, 1 x 10 10 vg/kg, 1 x 10 9 vg/kg, 1 x 10 8 vg/kg, or less.
  • the AAV vector may be administered to the patient in an amount of about 1 x 10 14 vg/kg, 1 x 10 14 vg/kg, 1 x 10 13 vg/kg, 1 x 10 12 vg/kg, 1 x 10 11 vg/kg, 1 x 10 10 vg/kg, 1 x 10 9 vg/kg, 1 x 10 8 vg/kg.
  • the AAV vector is administered to the patient in an amount of from about 3 x 10 13 vg/kg to about 2.3 x 10 14 vg/kg (e.g., in an amount of from about 3 x 10 13 vg/kg to about 2.3 x 10 14 vg/kg).
  • the AAV vector may be administered to the patient in an amount of about 3 x 10 13 vg/kg, 3.1 x 10 13 vg/kg, 3.2 x 10 13 vg/kg, 3.3 x 10 13 vg/kg, 3.4 x 10 13 vg/kg, 3.5 x 10 13 vg/kg, 3.6 x 10 13 vg/kg, 3.7 x 10 13 vg/kg, 3.8 x 10 13 vg/kg, 3.9 x 10 13 vg/kg, 4 x 10 13 vg/kg, 4.1 x 10 13 vg/kg, 4.2 x 10 13 vg/kg, 4.3 x 10 13 vg/kg, 4.4 x 10 13 vg/kg, 4.5 x 10 13 vg/kg, 4.6 x 10 13 vg/kg, 4.7 x 10 13 vg/kg, 4.8 x 10 13 vg/kg, 4.9 x 10 13 vg/kg,
  • the AAV vector is administered to the patient in an amount of from about 4 x 10 13 vg/kg to about 2.3 x 10 14 vg/kg, such as an in amount of about 4 x 10 13 vg/kg, 4.1 x 10 13 vg/kg, 4.2 x 10 13 vg/kg, 4.3 x 10 13 vg/kg, 4.4 x 10 13 vg/kg, 4.5 x 10 13 vg/kg, 4.6 x 10 13 vg/kg, 4.7 x 10 13 vg/kg, 4.8 x 10 13 vg/kg, 4.9 x 10 13 vg/kg, 5 x 10 13 vg/kg, 5.1 x 10 13 vg/kg, 5.2 x 10 13 vg/kg, 5.3 x 10 13 vg/kg, 5.4 x 10 13 vg/kg, 5.5 x 10 13 vg/kg, 5.6 x 10 13 vg/kg, 5.7
  • the AAV vector is administered to the patient in an amount of from about 5 x 10 13 vg/kg to about 2.3 x 10 14 vg/kg, such as an in amount of about 5 x 10 13 vg/kg, 5.1 x 10 13 vg/kg, 5.2 x 10 13 vg/kg, 5.3 x 10 13 vg/kg, 5.4 x 10 13 vg/kg, 5.5 x 10 13 vg/kg, 5.6 x 10 13 vg/kg, 5.7 x 10 13 vg/kg, 5.8 x 10 13 vg/kg, 5.9 x 10 13 vg/kg, 6 x 10 13 vg/kg, 6.1 x 10 13 vg/kg, 6.2 x 10 13 vg/kg, 6.3 x 10 13 vg/kg, 6.4 x 10 13 vg/kg, 6.5 x 10 13 vg/kg, 6.6 x 10 13 vg/kg, 6.7
  • the AAV vector is administered to the patient in an amount of from about 6 x 10 13 vg/kg to about 2.3 x 10 14 vg/kg, such as an in amount of about 6 x 10 13 vg/kg, 6.1 x 10 13 vg/kg, 6.2 x 10 13 vg/kg, 6.3 x 10 13 vg/kg, 6.4 x 10 13 vg/kg, 6.5 x 10 13 vg/kg, 6.6 x 10 13 vg/kg, 6.7 x PATENT ATTORNEY DOCKET NO.51037-075WO2 10 13 vg/kg, 6.8 x 10 13 vg/kg, 6.9 x 10 13 vg/kg, 7 x 10 13 vg/kg, 7.1 x 10 13 vg/kg, 7.2 x 10 13 vg/kg, 7.3 x 10 13 vg/kg, 7.4 x 10 13 vg/kg, 7.5 x 10 13 vg/kg
  • the AAV vector is administered to the patient in an amount of from about 7 x 10 13 vg/kg to about 2.3 x 10 14 vg/kg, such as an in amount of about 7 x 10 13 vg/kg, 7.1 x 10 13 vg/kg, 7.2 x 10 13 vg/kg, 7.3 x 10 13 vg/kg, 7.4 x 10 13 vg/kg, 7.5 x 10 13 vg/kg, 7.6 x 10 13 vg/kg, 7.7 x 10 13 vg/kg, 7.8 x 10 13 vg/kg, 7.9 x 10 13 vg/kg, 8 x 10 13 vg/kg, 8.1 x 10 13 vg/kg, 8.2 x 10 13 vg/kg, 8.3 x 10 13 vg/kg, 8.4 x 10 13 vg/kg, 8.5 x 10 13 vg/kg, 8.6 x 10 13 vg/kg, 8.7
  • the AAV vector is administered to the patient in an amount of from about 8 x 10 13 vg/kg to about 2.3 x 10 14 vg/kg, such as an in amount of about 8 x 10 13 vg/kg, 8.1 x 10 13 vg/kg, 8.2 x 10 13 vg/kg, 8.3 x 10 13 vg/kg, 8.4 x 10 13 vg/kg, 8.5 x 10 13 vg/kg, 8.6 x 10 13 vg/kg, 8.7 x 10 13 vg/kg, 8.8 x 10 13 vg/kg, 8.9 x 10 13 vg/kg, 9 x 10 13 vg/kg, 9.1 x 10 13 vg/kg, 9.2 x 10 13 vg/kg, 9.3 x 10 13 vg/kg, 9.4 x 10 13 vg/kg, 9.5 x 10 13 vg/kg, 9.6 x 10 13 vg/kg, 9.7
  • the AAV vector is administered to the patient in an amount of from about 9 x 10 13 vg/kg to about 2.3 x 10 14 vg/kg, such as an in amount of 9 x 10 13 vg/kg, 9.1 x 10 13 vg/kg, 9.2 x 10 13 vg/kg, 9.3 x 10 13 vg/kg, 9.4 x 10 13 vg/kg, 9.5 x 10 13 vg/kg, 9.6 x 10 13 vg/kg, 9.7 x 10 13 vg/kg, 9.8 x 10 13 vg/kg, 9.9 x 10 13 vg/kg, 1 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.5 x 10 14 vg/kg, 1.6 x 10 14 vg/kg, 1.7 x
  • the AAV vector is administered to the patient in an amount of from about 1 x 10 14 vg/kg to about 2.3 x 10 14 vg/kg, such as an in amount 1 x 10 14 vg/kg, 1.1 x 10 14 vg/kg, 1.2 x 10 14 vg/kg, 1.3 x 10 14 vg/kg, 1.4 x 10 14 vg/kg, 1.5 x 10 14 vg/kg, 1.6 x 10 14 vg/kg, 1.7 x 10 14 vg/kg, 1.8 x 10 14 vg/kg, 1.9 x 10 14 vg/kg, 2 x 10 14 vg/kg, 2.1 x 10 14 vg/kg, 2.2 x 10 14 vg/kg, or 2.3 x 10 14 vg/kg.
  • the AAV vector is administered to the patient in an amount of about 3 x 10 13 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 4 x 10 13 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 5 x 10 13 vg/kg. In some embodiments, the AAV vector is administered to the patient PATENT ATTORNEY DOCKET NO.51037-075WO2 in an amount of about 6 x 10 13 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 7 x 10 13 vg/kg.
  • the AAV vector is administered to the patient in an amount of about 8 x 10 13 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 9 x 10 13 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.1 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.2 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.3 x 10 14 vg/kg.
  • the AAV vector is administered to the patient in an amount of about 1.4 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.5 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.6 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.7 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.8 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.9 x 10 14 vg/kg.
  • the AAV vector is administered to the patient in an amount of about 2 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.1 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.2 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.3 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.4 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.5 x 10 14 vg/kg.
  • the AAV vector is administered to the patient in an amount of about 2.6 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.7 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.8 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.9 x 10 14 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 3 x 10 14 vg/kg.
  • the AAV vector is administered to the patient in a single dose comprising the amount (e.g., less than about 3 x 10 14 vg/kg). In some embodiments, the AAV vector is administered to the patient in two or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) doses that, together, comprise the amount (e.g., less than about 3 x 10 14 vg/kg). In some embodiments, the AAV vector is administered to the patient in two or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) doses that each, individually, comprise the amount (e.g., less than about 3 x 10 14 vg/kg).
  • the two or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) doses are separated from one another by one year or more (e.g., one year, one year and one day, one year and one month, one year and six months, two years, three years, four years, or five years).
  • the two or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) doses are administered to the patient within about 12 months (e.g., about 12 months, about 11 months, about 10 months, about 9 months, about 8 months, about 7 months, about 6 months, about 5 months, about 4 months, about 3 months, about 2 months, or about 1 month) of one another
  • An AAV vector containing a transgene encoding MTM1 described herein can be administered in combination with a one or more additional AAV vectors containing a transgene encoding MTM1 described herein or in combination with one or more therapeutic procedures (e.g., nasobiliary drainage (NBD)) and/or agents (e.g., an anti-cholestatic agent) for the treatment of a neuromuscular disorder (e.g., XLMTM).
  • NBD nasobiliary drainage
  • agents e.g., an anti-cholestatic agent
  • NBD is a therapeutic procedure that is performed to help drain bile (e.g., when the bile duct is blocked, a biliary drain may help bile to flow from the liver into the intestine).
  • NBD is performed with a biliary drain (also known as a biliary stent), which is a thin, hollow, flexible tube with several small holes along the sides. A biliary drain may be inserted in a patient’s bile duct to allow it to drain.
  • a biliary drain also known as a biliary stent
  • the one or more additional therapeutic agents is an anti-cholestatic agent (e.g., a bile acid, a farnesoid X receptor (FXR) ligand, a fibroblast growth factor 19 (FGF-19) mimetic, a Takeda-G-protein-receptor-5 (TGR5) agonist, a peroxisome proliferator-activated receptor (PPAR) agonist, a PPAR-alpha agonist, a PPAR-delta agonist, a dual PPAR-alpha and PPAR-delta agonist, an apical sodium-dependent bile acid transporter (ASBT) inhibitor, an immunomodulatory drug, an antifibrotic therapy, and a nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitor) or a combination thereof.
  • an anti-cholestatic agent e.g., a bile acid, a farnesoid X receptor (FXR)
  • the anti-cholestatic agent is administered to the patient in one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy) doses that commence within about six weeks before or after (e.g., about six weeks before or after, about five weeks before or after, about four weeks before or after, about three weeks before or after, about two weeks before or after, or about one week before or after) administration of the viral vector to the patient.
  • one or more e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy
  • doses that commence within about six weeks before or after (e.g., about six weeks before or after, about five weeks before or after, about four weeks before or after, about three weeks before or after, about two weeks before or after, or about one week before or after) administration of the viral vector to the patient.
  • the anti-cholestatic agent is administered to the patient in one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy) doses that commence within about five weeks before or after (e.g., about five weeks before or after, about four weeks before or after, about three weeks before or after, about two weeks before or after, or about one week before or after) administration of the viral vector to the patient.
  • one or more e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy
  • doses that commence within about five weeks before or after (e.g., about five weeks before or after, about four weeks before or after, about three weeks before or after, about two weeks before or after, or about one week before or after) administration of the viral vector to the patient.
  • the anti-cholestatic agent is administered to the patient in one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy) doses that commence within about one week before or after (e.g., about one week before or PATENT ATTORNEY DOCKET NO.51037-075WO2 after, about six days before or after, about five days before or after, about four days before or after, about three days before or after, about two days before or after, or about one day before or after) administration of the viral vector to the patient.
  • one week before or after e.g., about one week before or PATENT ATTORNEY DOCKET NO.51037-075WO2 after, about six days before or after, about five days before or after, about four days before or after, about three days before or after, about two days before or after, or about one day before or after
  • the anti-cholestatic agent is administered to the patient in one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy) doses that commence on the same day (e.g., 24 th hour, on the 23 rd hour, on the 22 nd hour, on the 21 st hour, on the 20 th hour, on the 19 th hour, on the 18 th hour, on the 17 th hour, on the 16 th hour, on the 15 th hour, on the 14 th hour, on the 13 th hour, on the 12 th hour, on the 11 th hour, on the 10 th hour, on the 9 th hour, on the 8 th hour, on the 7 th hour, on the 6 th hour, on the 5 th hour, on the 4 th hour, on the 3 rd hour, on the 2 nd hour, on the 1 st hour, on the 60 th minute
  • the anti-cholestatic agent is a bile acid.
  • the bile acid is ursodeoxycholic acid or a derivative thereof or nor-ursodeoxycholic acid.
  • the bile acid is ursodiol.
  • the anti-cholestatic agent is an FXR ligand.
  • the FXR ligand is obeticholic acid, cilofexor, tropifexor, tretinoin, or EDP-305.
  • the one or more anti-cholestatic agent is an FGF-19 mimetic. In some embodiment, the FGF-19 mimetic is aldafermin.
  • the anti-cholestatic agent is a TGR5 agonist. In some embodiments, the TGR5 agonist is INT-777 or INT-767. In some embodiments, the anti-cholestatic agent is a PPAR agonist. In some embodiments, the PPAR agonist is bezafibrate, seladelpar, or elafibrinor. In some embodiments, the anti-cholestatic agent is a PPAR-alpha agonist. In some embodiments, the PPAR-alpha agonist is fenofibrate. In some embodiments, the anti-cholestatic agent is a PPAR-delta agonist. In some embodiments, the PPAR-delta agonist is seladelpar.
  • the anti-cholestatic agent is a dual PPAR-alpha and PPAR-delta agonist. In some embodiments, the dual PPAR-alpha -delta agonist is elafibranor. In some embodiments, the one or more anti-cholestatic agent is an ASBT inhibitor. In some embodiments, the ASBT inhibitor is odevixibat, maralixibat, or linerixibat. In some embodiments, the anti-cholestatic agent is an immunomodulatory drug. In some embodiments, the immunomodulatory drug is rituximab, abatacept, ustekinumab, infliximab, baricitinib, or FFP104.
  • the anti-cholestatic agent is an antifibrotic therapy.
  • the antifibrotic therapy is a vitamin D receptor (VDR) agonist or pumpuzumab.
  • the anti-cholestatic agent is a NOX inhibitor.
  • the NOX inhibitor is setanaxib. PATENT ATTORNEY DOCKET NO.51037-075WO2 Recommended Clinical Parameters for Monitoring a Patient for Development of Cholestasis or Hyperbilirubinemia
  • a patient is monitored for the development of cholestasis by a serum bile acid test and/or blood test (e.g., an LFT), as described herein.
  • a patient is monitored for the development of hyperbilirubinemia by a blood test (e.g., bilirubin test), as described herein.
  • a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti- cholestatic agent.
  • a patient is monitored for the development of hyperbilirubinemia, and if the patient exhibits hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by a blood test (e.g., a serum acid bile test or a liver function test).
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by a blood test (e.g., a serum acid bile test or a liver function test), and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • the patient is determined to exhibit cholestasis, hyperbilirubinemia, or one or more symptoms thereof by a finding that the patient exhibits a parameter (e.g., a serum bile acid level) in blood test (e.g., a serum acid bile test) that is increased relative to a reference level.
  • a parameter e.g., a serum bile acid level
  • blood test e.g., a serum acid bile test
  • the patient is determined to exhibit cholestasis, hyperbilirubinemia, or one or more symptoms thereof by a finding that the patient exhibits a serum bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) level in blood test (e.g., a serum acid bile test) that is increased relative to a reference level.
  • a serum bile acid e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid
  • the blood test is a liver function test.
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by a liver function test, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • the patient is determined to exhibit cholestasis, hyperbilirubinemia, or one or more symptoms thereof by a finding that the patient exhibits a parameter (e.g., aspartate aminotransferase level or alanine aminotransferase level) in liver function test that is increased or decreased relative to a reference level.
  • a parameter e.g., aspartate aminotransferase level or alanine aminotransferase level
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia with a serum bile acid test. In some embodiments, a patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is PATENT ATTORNEY DOCKET NO.51037-075WO2 administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia with a serum bile acid test, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for cholestasis or hyperbilirubinemia by the patient’s bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) levels, as measured with a serum bile acid test.
  • bile acid e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more of the patient’s bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) levels, as measured with a serum bile acid test, is greater than the norm.
  • bile acid e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s cholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL).
  • 5 nmol/mL e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL,
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s cholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 5 nmol/mL e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/m
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s chenodeoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL).
  • 6 nmol/mL e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s chenodeoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL).
  • 6 nmol/mL e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s deoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 6 nmol/mL e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s deoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 PATENT ATTORNEY DOCKET NO.51037-075WO2 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 6 nmol/mL e.g., 6 PATENT ATTORNEY DOCKET NO.51037-075WO2 nmol/m
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s ursodeoxycholic acid level, as measured with a serum bile acid test, is greater than 2 nmol/mL (e.g., 2 nmol/mL, 3 nmol/mL, 4 nmol/mL, 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 2 nmol/mL e.g., 2 nmol/mL, 3 nmol/mL, 4 n
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ursodeoxycholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 5 nmol/mL e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia with an LFT.
  • a patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia with an LFT, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when a parameter (e.g., ASP level or AST level) of the patient’s LFT is greater than the age-adjusted norm, as described herein.
  • a parameter e.g., ASP level or AST level
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s AST level in an LFT.
  • a patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s AST level in an LFT, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s AST level in an LFT and it is determined that a patient PATENT ATTORNEY DOCKET NO.51037-075WO2 exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than the norm.
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53 U/L,
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L,
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s ALT level in an LFT.
  • a patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s ALT level in an LFT, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s ALT level in an LFT and it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than the norm.
  • a patient exhibits cholestasis or one or more symptoms thereof when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53 U/
  • a patient is monitored for the development of cholestasis. In some embodiments, a patient is monitored for the development of cholestasis with a serum bile acid test. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis with a serum bile acid test, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bile acids level, as measured with a serum bile acid test, is greater than the norm.
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bile acids level, as measured with a serum bile acid test, is greater than 14 ⁇ mol/L (e.g., 15 ⁇ mol/L, 16 ⁇ mol/L, 17 ⁇ mol/L, 18 ⁇ mol/L, 19 ⁇ mol/L, 20 ⁇ mol/L, 21 ⁇ mol/L, 22 ⁇ mol/L, 23 ⁇ mol/L, 24 ⁇ mol/L, 25 ⁇ mol/L, 26 ⁇ mol/L, 27 ⁇ mol/L, 28 ⁇ mol/L, 29 ⁇ mol/L, 30 ⁇ mol/L, 31 ⁇ mol/L, 32 ⁇ mol/L, 33 ⁇ mol/L, 34 ⁇ mol/L, 35 ⁇ mol/L, 36 ⁇ mol/L, 37 ⁇ mol/L, 38 ⁇ mol/L, 39 ⁇ mol/L, 40 ⁇ mol/L,
  • a patient is monitored for the development of cholestasis with a blood test (e.g., LFT or a bilirubin test).
  • a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis with an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more parameters (e.g., GGT level, ASP level, AST level, ALT level, and bilirubin level) of the patient’s blood test (e.g., a LFT or a bilirubin test) is greater than the age-adjusted norm, as described herein.
  • one or more parameters e.g., GGT level, ASP level, AST level, ALT level, and bilirubin level
  • a LFT or a bilirubin test e.g., a LFT or a bilirubin test
  • a patient is monitored for the development of cholestasis with an LFT.
  • a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis with an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more parameters (e.g., GGT level, ASP level, AST level, and ALT level) of the patient’s LFT is greater than the age-adjusted norm, as described herein.
  • one or more parameters e.g., GGT level, ASP level, AST level, and ALT level
  • GGT level e.g., GGT level, ASP level, AST level, and ALT level
  • a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis by measuring the patient’s GGT level in an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • the patient is a newborn (e.g., 0-6 months old), a toddler (e.g., 6-12 months old), or a child aged 1- ⁇ 5 years old. In some embodiments, the patient is a newborn of the age from 0-6 months old. In some embodiments, the patient is a toddler of the age from 6-12 months old. In some embodiments, the patient is a child of the age from 1-5 years old.
  • a patient is a newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 12-122 U/L (e.g., 12-122 U/L, 13-122 U/L, 14-122 U/L,15-122 U/L, 16-122 U/L, 17-122 U/L, 18-122 U/L, 19-122 U/L, 20-122 U/L, 25-122 U/L, 30-122 U/L, 40-122 U/L, 50-122 U/L, 60-122 U/L, 70-122 U/L, 80-122 U/L, 90-122 U/L, 100-122 U/L, 110-122 U/L, 120-122 U/L, or 121-122 U/L).
  • 12-122 U/L e.g., 12-122 U/L, 13-122 U/L, 14-122
  • the patient is a male newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than 12 U/L (e.g., 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L).
  • 12 U/L e.g., 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L.
  • the patient is a male newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 122 U/L (e.g., 123 U/L, 124 U/L, PATENT ATTORNEY DOCKET NO.51037-075WO2 125 U/L, 126 U/L, 127 U/L, 128 U/L, 129 U/L, 130 U/L, 135 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • 122 U/L e.g., 123 U/L, 124 U/L, PATENT ATTORNEY DOCKET NO.51037-075WO2 125 U/L, 126 U/L, 127 U/L, 128 U/L, 129 U
  • the patient is a male toddler (e.g., 6-12 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 1-39 U/L (e.g., 2-39 U/L, 3-39 U/L, 4-39 U/L, 5-39 U/L, 6-39 U/L, 7-39 U/L, 8-39 U/L, 9-39 U/L, 10-39 U/L, 11-39 U/L, 12-39 U/L, 13-39 U/L, 14-39 U/L, 15-39 U/L, 16-39 U/L, 17-39 U/L, 18-39 U/L, 19-39 U/L, 20-39 U/L, 21-39 U/L, 22-39 U/L, 23-39 U/L, 24-39 U/L, 25-39 U/L, 26-39 U/L, 27-39 U/L
  • the patient is a male toddler (e.g., 6-12 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 39 U/L (e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/L, 48 U/L, 49 U/L, 50 U/L, 55 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • U/L e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/
  • the patient is a male child aged 1- ⁇ 5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 3-22 U/L (e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22 U/L, 13- 22 U/L, 14-22 U/L, 15-22 U/L, 16-22 U/L, 17-22 U/L, 18-22 U/L, 19-22 U/L, 20-22 U/L, and 21-22 U/L).
  • 3-22 U/L e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22
  • the patient is a male child aged 1- ⁇ 5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient is GGT level is less than 3 U/L (e.g., 2 U/L and 1 U/L).
  • the patient is a male child aged 1- ⁇ 5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 22 U/L (e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40 U/L, 50 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • 22 U/L e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35
  • the patient is a female newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 15-132 U/L (e.g., 15-132 U/L, 16-132 U/L, 17-132 U/L, 18-132 U/L, 19-132 U/L, 20-132 U/L, 25-132 U/L, 30- 132 U/L, 40-132 U/L, 50-132 U/L, 60-132 U/L, 70-132 U/L, 80-132 U/L, 90-132 U/L, 100-132 U/L, 110-132 U/L, 120-132 U/L, 130-132 U/L, and 131-132 U/L).
  • 15-132 U/L e.g., 15-132 U/L, 16-132 U/L, 17-132 U/L, 18-132 U/
  • the patient is a female newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than 15 U/L (e.g., 14 U/L, 13 U/L, 12 U/L, 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L).
  • 15 U/L e.g., 14 U/L, 13 U/L, 12 U/L, 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L.
  • the patient is a female newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 132 U/L (e.g., 133 U/L, 134 U/L, 135 U/L, 136 U/L, 137 U/L, 138 U/L, 139 U/L, 140 U/L, 145 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • 132 U/L e.g., 133 U/L, 134 U/L, 135 U/L, 136 U/L, 137 U/L, 138 U/L, 139 U/L, 140 U/L, 145 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200
  • the patient is a female toddler (e.g., 6-12 months old) it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is outside of the normal range of about 1-39 U/L (e.g., 2-39 U/L, 3-39 U/L, 4-39 U/L, 5-39 U/L, 6-39 U/L, 7-39 U/L, 8-39 U/L, 9-39 U/L, 10-39 U/L, 11-39 U/L, 12-39 U/L, 13-39 U/L, 14-39 U/L, 15-39 U/L, 16-39 U/L, 17-39 U/L, 18-39 U/L, 19-39 U/L, 20-39 U/L, 21-39 U/L, 22-39 U/L, 23-39 U/L, 24-39 U/L, 25-39 U/L, 26-39 U/L, 27-39 U/L
  • the patient is a female toddler (e.g., 6-12 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 39 U/L (e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/L, 48 U/L, 49 U/L, 50 U/L, 55 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • U/L e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/
  • the patient is a female child aged 1- ⁇ 5 years old it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 3-22 U/L (e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22 U/L, 13- 22 U/L, 14-22 U/L, 15-22 U/L, 16-22 U/L, 17-22 U/L, 18-22 U/L, 19-22 U/L, 20-22 U/L, and 21-22 U/L).
  • 3-22 U/L e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U
  • the patient is a female child aged 1- ⁇ 5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is less than 3 U/L (e.g., 2 U/L and 1 U/L).
  • the patient is a female child aged 1- ⁇ 5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is greater than 22 U/L (e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40 U/L, 50 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • 22 U/L e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L,
  • a patient is monitored for the development of cholestasis by measuring the patient’s ASP level in an LFT.
  • a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis by measuring the patient’s ASP level in an LFT, and if the patient PATENT ATTORNEY DOCKET NO.51037-075WO2 exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis by measuring the patient’s ASP level in an LFT and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level that is greater than the norm.
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is outside of the normal range of about 50 to 300 U/L e.g., about 51 to 300 U/L, about 52 to U/L, about 53 to 300 U/L, about 54 to 300 U/L, about 55 to 300 U/L, about 56 to 300 U/L, about 57 to 300 U/L, about 58 to 300 U/L, about 59 to 300 U/L, about 60 to 300 U/L, about 65 to 300 U/L, about 70 to 300 U/L, about 80 to 300 U/L, about 90 to 300 U/L, about 100 to 300 U/L, about 125 to 300 U/L, about 150 to 300 U/L, about 175 to 300 U/L, about 200 to 300 U/L, about 225 to 300 U/L, about 250 to 300 U/L, or about 275 to 300 U/L).
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is less than 50 U/L (e.g., 50 U/L, 49 U/L, 48 U/L, 47 U/L, 46 U/L, 45 U/L, 44 U/L, 43 U/L, 42 U/L, 41 U/L, 40 U/L, 39 U/L, 38 U/L, 37 U/L, 36 U/L, 35 U/L, 34 U/L, 33 U/L, 32 U/L, 31 U/L, 30 U/L, 29 U/L, 28 U/L, 27 U/L, 26 U/L, 25 U/L, 24 U/L, 23 U/L, 22 U/L, 21 U/L, 20 U/L, 19 U/L, 18 U/L, 17 U/L, 16 U/L, 15 U/L, 14 U/L, 13 U/L, 12 U/L, 11 U/L
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is greater than 300 U/L (e.g., 300 U/L, 301 U/L, 302 U/L, 303 U/L, 304 U/L, 305 U/L, 306 U/L, 307 U/L, 308 U/L, 309 U/L, 310 U/L, 311 U/L, 312 U/L, 313 U/L, 314 U/L, 315 U/L, 316 U/L, 317 U/L, 318 U/L, 319 U/L, 320 U/L, 321 U/L, 322 U/L, 323 U/L, 324 U/L, 325 U/L, 330 U/L, 340 U/L, 350 U/L, 400 U/L, and 500 U/L).
  • 300 U/L e.g., 300 U/L, 301 U/L,
  • a patient is monitored for the development of cholestasis by measuring the patient’s AST level in an LFT. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s AST level in an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis by measuring the patient’s AST level in an LFT and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than the norm.
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L,
  • a patient is monitored for the development of cholestasis by measuring the patient’s ALT level in an LFT. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s ALT level in an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of cholestasis by measuring the patient’s ALT level in an LFT and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than the norm.
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53 U/
  • a patient is monitored for the development of hyperbilirubinemia.
  • a patient is monitored for the development of hyperbilirubinemia with a bilirubin test.
  • a patient is monitored for the development of hyperbilirubinemia, and if the patient exhibits hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient is monitored for the development of hyperbilirubinemia with a bilirubin test, and if the patient exhibits hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent.
  • a patient exhibits hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a bilirubin level that is greater than the norm.
  • PATENT ATTORNEY DOCKET NO.51037-075WO2 it is determined that a patient exhibits hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bilirubin level is greater than 1.2 mg/dL (e.g., 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL,
  • mg/dL 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, and 100 mg/dL).
  • a patient exhibits hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s direct bilirubin level is greater than 0.2 mg/dL (e.g., 0.2 mg/dL, 0.3 mg/dL, 0.4 mg/dL, 0.5 mg/dL, 0.6 mg/dL, 0.7 mg/dL, 0.8 mg/dL, 0.9 mg/dL, 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL,
  • mg/dL 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, and 100 mg/dL).
  • the patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a bilirubin level that is greater than 1 mg/dL (e.g., greater than 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3.
  • 1 mg/dL e.g., greater than 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3
  • mg/dL 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, or 100 mg/dL) in a bilirubin test.
  • a Patient Exhibits Cholestasis or Hyperbilirubinemia or a Symptom Thereof
  • it is determined that the patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof by determining that one or more parameters (e.g., total bile acids level, GGT level, ASP level, AST level, and ALT level) of the patient’s serum bile acid test and/or blood test (e.g., an LFT) is greater than or less than the age-adjusted norm, as described herein, and the patient is administered an anti-cholestatic agent.
  • one or more parameters e.g., total bile acids level, GGT level, ASP level, AST level, and ALT level
  • serum bile acid test and/or blood test e.g., an LFT
  • one or more parameters e.g., bilirubin level
  • blood test e.g., bilirubin test
  • PATENT ATTORNEY DOCKET NO.51037-075WO2 it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more of the patient’s bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) levels, as measured with a serum bile acid test, is greater than the norm.
  • bile acid e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when a parameter (e.g., ASP level or AST level) of the patient’s LFT is greater than the age-adjusted norm, as described herein.
  • a parameter e.g., ASP level or AST level
  • Serum Bile Acid Test it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more of the patient’s bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) levels, as measured with a serum bile acid test, is greater than the norm.
  • bile acid e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s cholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL).
  • 5 nmol/mL e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL,
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s cholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 5 nmol/mL e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/m
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s chenodeoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL).
  • 6 nmol/mL e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s chenodeoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL).
  • 6 nmol/mL e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s deoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 6 nmol/mL e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s deoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 6 nmol/mL e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s ursodeoxycholic acid level, as measured with a serum bile acid test, is greater than 2 nmol/mL (e.g., 2 nmol/mL, 3 nmol/mL, 4 nmol/mL, 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 2 nmol/mL e.g., 2 nmol/mL, 3 nmol/mL, 4 n
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ursodeoxycholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 2 nmol/mL, 3 nmol/mL, 4 nmol/mL, 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL.
  • 5 nmol/mL e.g., 2 nmol/mL, 3
  • Liver Function Test it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when a parameter (e.g., ASP level or AST level) of the patient’s LFT is greater than the age-adjusted norm, as described herein. IIa.
  • a parameter e.g., ASP level or AST level
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s AST level in an LFT and it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than the norm.
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53 U/L,
  • a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L,
  • a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s ALT level in an LFT and it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than the norm.
  • a patient exhibits cholestasis or one or more symptoms thereof when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/
  • a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53 U/
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits an acid bile level, as measured with a serum bile acid test, that is greater than the norm.
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bile acids level, as measured determined with a serum bile acid test, is greater than 14 ⁇ mol/L (e.g., 15 ⁇ mol/L, 16 ⁇ mol/L, 17 ⁇ mol/L, 18 ⁇ mol/L, 19 ⁇ mol/L, 20 ⁇ mol/L, 21 ⁇ mol/L, 22 ⁇ mol/L, 23 ⁇ mol/L, 24 ⁇ mol/L, 25 ⁇ mol/L, 26 ⁇ mol/L, 27 ⁇ mol/L, 28 ⁇ mol/L, 29 ⁇ mol/L, 30 ⁇ mol/L, 31 ⁇ mol/L, 32 ⁇ mol/L, 33 ⁇ mol/L, 34 ⁇ mol/L, 35 ⁇ mol/L, 36 ⁇ mol/L, 37 ⁇ mol/L, 38 ⁇ mol/L, 39 ⁇ mol/L, 40 ⁇ mol/L
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more parameters (e.g., PATENT ATTORNEY DOCKET NO.51037-075WO2 GGT level, ASP level, AST level, ALT level, and bilirubin level) of the patient’s blood test (e.g., a LFT or a bilirubin test) is greater than the age-adjusted norm, as described herein. IIa.
  • one or more parameters e.g., PATENT ATTORNEY DOCKET NO.51037-075WO2 GGT level, ASP level, AST level, ALT level, and bilirubin level
  • a LFT or a bilirubin test e.g., a LFT or a bilirubin test
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more parameters (e.g., GGT level, ASP level, AST level, and ALT level) of the patient’s LFT is greater than the age-adjusted norm, as described herein.
  • one or more parameters e.g., GGT level, ASP level, AST level, and ALT level
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a GGT level, as measured in a LFT, that is greater than the age-adjusted norm.
  • the patient is a newborn (e.g., 0-6 months old), a toddler (e.g., 6-12 months old), or a child aged 1- ⁇ 5 years old. In some embodiments, the patient is a newborn of the age from 0-6 months old. In some embodiments, the patient is a toddler of the age from 6-12 months old. In some embodiments, the patient is a child of the age from 1-5 years old.
  • a patient is a newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 12-122 U/L (e.g., 12-122 U/L, 13-122 U/L, 14-122 U/L,15-122 U/L, 16-122 U/L, 17-122 U/L, 18-122 U/L, 19-122 U/L, 20-122 U/L, 25-122 U/L, 30-122 U/L, 40-122 U/L, 50-122 U/L, 60-122 U/L, 70-122 U/L, 80-122 U/L, 90-122 U/L, 100-122 U/L, 110-122 U/L, 120-122 U/L, or 121-122 U/L).
  • 12-122 U/L e.g., 12-122 U/L, 13-122 U/L, 14-122 U/L,15
  • the patient is a male newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than 12 U/L (e.g., 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L).
  • 12 U/L e.g., 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L.
  • the patient is a male newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 122 U/L (e.g., 123 U/L, 124 U/L, 125 U/L, 126 U/L, 127 U/L, 128 U/L, 129 U/L, 130 U/L, 135 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • 122 U/L e.g., 123 U/L, 124 U/L, 125 U/L, 126 U/L, 127 U/L, 128 U/L, 129 U/L, 130 U/L, 135 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U
  • the patient is a male toddler (e.g., 6-12 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 1-39 U/L (e.g., 2-39 U/L, 3-39 U/L, 4-39 U/L, 5-39 U/L, 6-39 U/L, 7-39 U/L, 8-39 U/L, 9-39 U/L, 10-39 U/L, 11-39 U/L, 12-39 U/L, 13- 39 U/L, 14-39 U/L, 15-39 U/L, 16-39 U/L, 17-39 U/L, 18-39 U/L, 19-39 U/L, 20-39 U/L, 21-39 U/L, 22- 39 U/L, 23-39 U/L, 24-39 U/L, 25-39 U/L, 26-39 U/L, 27-39 U/L, 28-39 U
  • the patient is a male toddler (e.g., 6-12 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 39 U/L (e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/L, 48 U/L, 49 U/L, 50 U/L, 55 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • U/L e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/
  • the patient is a male child aged 1- ⁇ 5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 3-22 U/L (e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22 U/L, 13-22 U/L, 14-22 U/L, 15-22 U/L, 16-22 U/L, 17-22 U/L, 18-22 U/L, 19-22 U/L, 20-22 U/L, and 21-22 U/L).
  • 3-22 U/L e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22
  • the patient is a male child aged 1- ⁇ 5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than about 3 U/L (e.g., 2 U/L and 1 U/L).
  • the patient is a male child aged 1- ⁇ 5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 22 U/L (e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40 U/L, 50 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • 22 U/L e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40
  • the patient is a female newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is outside of the normal range of about 15-132 U/L (e.g., 15-132 U/L, 16-132 U/L, 17-132 U/L, 18-132 U/L, 19-132 U/L, 20-132 U/L, 25-132 U/L, 30-132 U/L, 40-132 U/L, 50-132 U/L, 60-132 U/L, 70-132 U/L, 80-132 U/L, 90-132 U/L, 100-132 U/L, 110-132 U/L, 120-132 U/L, 130-132 U/L, and 131-132 U/L).
  • 15-132 U/L e.g., 15-132 U/L, 16-132 U/L, 17-132 U/L, 18-132 U/L, 19-132
  • the patient is a female newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is less than about 15 U/L (e.g., 14 U/L, 13 U/L, 12 U/L, 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L).
  • 15 U/L e.g., 14 U/L, 13 U/L, 12 U/L, 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L.
  • the patient is a female newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is greater than 132 U/L (e.g., 133 U/L, 134 U/L, 135 U/L, 136 U/L, 137 U/L, 138 U/L, 139 U/L, 140 U/L, 145 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • 132 U/L e.g., 133 U/L, 134 U/L, 135 U/L, 136 U/L, 137 U/L, 138 U/L, 139 U/L, 140 U/L, 145 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L.
  • the patient is a female toddler (e.g., 6-12 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is outside of the normal range of about 1-39 U/L (e.g., 2-39 U/L, 3-39 U/L, 4-39 U/L, 5-39 U/L, 6-39 U/L, 7-39 U/L, 8-39 U/L, 9-39 U/L, 10-39 U/L, 11-39 U/L, 12-39 U/L, 13-39 U/L, 14-39 U/L, 15-39 U/L, 16-39 U/L, 17-39 U/L, 18-39 U/L, 19-39 U/L, 20-39 U/L, 21-39 U/L, 22-39 U/L, 23-39 U/L, 24-39 U/L, 25-39 U/L, 26-39 U/L, 27-39 U/L, 28-39
  • the patient is a female toddler (e.g., 6-12 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is greater than 39 U/L (e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/L, 48 U/L, 49 U/L, 50 U/L, 55 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • U/L e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U
  • the patient is a female child aged 1- ⁇ 5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 3-22 U/L (e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22 U/L, 13-22 U/L, 14-22 U/L, 15-22 U/L, 16-22 U/L, 17-22 U/L, 18-22 U/L, 19-22 U/L, 20-22 U/L, and 21-22 U/L).
  • 3-22 U/L e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22
  • the patient is a female child aged 1- ⁇ 5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than about 3 U/L (e.g., 2 U/L and 1 U/L).
  • the patient is a female child aged 1- ⁇ 5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 22 U/L (e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40 U/L, 50 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L).
  • 22 U/L e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits an ASP level, as measured in a LFT, that is greater than the norm.
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is outside of the normal range of about 50 to 300 U/L (e.g., about 51 to 300 U/L, about 52 to U/L, about 53 to 300 U/L, about 54 to 300 U/L, about 55 to 300 U/L, about 56 to 300 U/L, about 57 to 300 U/L, about 58 to 300 U/L, about 59 to 300 U/L, about 60 to 300 U/L, about 65 to 300 U/L, about 70 to 300 U/L, about 80 to 300 U/L, about 90 to 300 U/L, about 100 to 300 U/L, about 125 to 300 U/L, about 150 to 300 U/L, about 175 to 300 U/L, about 200 to 300 U/L, about 225 to 300 U/L, about 250 to 300 U/L, or about 275 to 300 U/L).
  • about 50 to 300 U/L e.g.
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is less than about 50 U/L (e.g., 50 U/L, 49 U/L, 48 U/L, 47 U/L, 46 U/L, 45 U/L, 44 U/L, 43 U/L, 42 U/L, 41 U/L, 40 U/L, 39 U/L, 38 U/L, 37 U/L, 36 U/L, 35 U/L, 34 U/L, 33 U/L, 32 U/L, 31 U/L, 30 U/L, 29 U/L, 28 U/L, 27 U/L, 26 U/L, 25 U/L, 24 U/L, 23 U/L, 22 U/L, 21 U/L, 20 U/L, 19 U/L, 18 U/L, 17 U/L, 16 PATENT ATTORNEY DOCKET NO.51037-075WO2 U/L, 15 U/L,
  • a is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is greater than 300 U/L (e.g., 300 U/L, 301 U/L, 302 U/L, 303 U/L, 304 U/L, 305 U/L, 306 U/L, 307 U/L, 308 U/L, 309 U/L, 310 U/L, 311 U/L, 312 U/L, 313 U/L, 314 U/L, 315 U/L, 316 U/L, 317 U/L, 318 U/L, 319 U/L, 320 U/L, 321 U/L, 322 U/L, 323 U/L, 324 U/L, 325 U/L, 330 U/L, 340 U/L, 350 U/L, 400 U/L, and 500 U/L).
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits an AST level, as measured in a LFT, that is greater than the norm.
  • a is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53 U
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits an ALT level, as measured in a LFT, that is greater than the norm.
  • a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L).
  • 50 U/L e.g., 51 U/L, 52 U/L, 53
  • a patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a bilirubin level, as measured in a blood test (e.g., a bilirubin test), that is greater than the norm.
  • a blood test e.g., a bilirubin test
  • a patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bilirubin level is greater than 1.2 mg/dL (e.g., 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3.
  • 1.2 mg/dL e.g., 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/
  • a patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s direct bilirubin level is greater than 0.2 mg/dL (e.g., 0.2 mg/dL, 0.3 mg/dL, 0.4 mg/dL, 0.5 mg/dL, 0.6 mg/dL, 0.7 mg/dL, 0.8 mg/dL, 0.9 mg/dL, 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL,
  • mg/dL 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, and 100 mg/dL).
  • the patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a bilirubin level that is greater than 1 mg/dL (e.g., greater than 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 ]mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3.
  • 1 mg/dL e.g., greater than 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL
  • mg/dL 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, or 100 mg/dL) in a bilirubin test.
  • Example 1 Evaluating safety and efficacy of a pseudotyped AAV8 vector including a nucleic acid sequence encoding a myotubularin 1 gene in a murine model of X-Linked myotubular myopathy Objective and Study Methodology The objective of this study was to perform an efficacy and sampling study using myotubularin 1 (MTM1) hemizygous (“HEMI”) knock out (KO) mice and determine the possible effects of liver- specific MTM1 expression in mice.
  • MTM1 myotubularin 1
  • HEMI myotubularin 1
  • mice were dosed with different test articles intravenously at ⁇ 4 weeks of age. Mice were sampled at ⁇ 16 weeks of age ( ⁇ 12 weeks post-dosing). Tissue samples were used for myotubularin and bile salt transporter expression, bile acid level measurements, and histopathology. Whole blood samples were used for hematology analysis and serum samples were PATENT ATTORNEY DOCKET NO.51037-075WO2 used for clinical chemistry analysis. In addition, a group of na ⁇ ve MTM1 KO mice were sampled at ⁇ 5 weeks of age, and tissues were used for histopathology. This study was performed according to appropriate methodologies and standard operating procedures (SOPs) at the testing facility or test sites.
  • SOPs standard operating procedures
  • IVC Individually ventilated caging systems
  • polycarbonate Type II Long cages were used to house the mice.
  • KO mice were randomized into groups so that whole litters of mice did not end up in a single testing group to avoid a “litter effect”. Mice were housed in groups of up to 4-5. Each cage included 3-4 KO mice, and 1-2 WT mice for socializing purposes. None of the KO mice were housed individually.
  • Dose levels were selected based on previous studies with mice. The high dose was a multiple of the previous high dose in mice, as the previous high dose showed significant pharmacological effect without toxicity. Animals were dosed by slow intravenous (IV) bolus injection into a tail vein. Dose volumes (total 16.33 mL/kg) were administered as a split IV bolus dose with 2 to 3 hours between administrations. The dose site, end time, and dose volume were documented. Animals were anesthetized with isoflurane, if necessary. This administration route is consistent with the proposed route of administration in humans and is expected to provide appropriate systemic exposure for investigation and associated pharmacological activity. The administration frequency was one dose on day 1. Dose levels were anticipated to identify achievable associated pharmacological activity.
  • Test article volumes used prior to dosing are summarized in Table 5, below. Table 5.
  • Test article volumes prior to dosing PATENT ATTORNEY DOCKET NO.51037-075WO2
  • the following humane endpoint criteria are applicable for the study. If the mice met the predefined humane endpoint criteria, they underwent a planned euthanasia for welfare reasons. If any of the mice needed to be euthanized for welfare reasons, they were opened, and macroscopic observations were recorded. In case the general health status of an animal deteriorated significantly, a shortened tissue isolation protocol was applied, if possible.
  • the shortened tissue isolation protocol consisted of collecting a skeletal muscle (quadriceps) and 1 liver lobe in liquid nitrogen and 1 liver lobe and heart (dorsal portion) tissue in formalin.
  • the skeletal muscle and liver lobe were stored at - 80oC while the liver lobe and heart (dorsal portion) in formalin were stored at room temperature. Due to a limited staff capacity during evenings and weekends to perform sampling on acute cases needing immediate action, mice were euthanized by an overdose of CO2 and decapitated. In this case, no sampling was performed. Body weight was measured three times a week and mortality were recorded.
  • mice were euthanized by deep anesthetization with sodium pentobarbital (180 mg/kg). As much whole blood was collected as possible via cardiac puncture. Blood was first collected for hematology (100 ⁇ L of EDTA whole blood), and then for clinical chemistry (200 ⁇ L of whole blood to separate minimum 80 ⁇ L of serum). Any remaining serum was collected. Then, the mice were transcardially perfused with heparinized saline to remove blood from the tissues. The group of 10 na ⁇ ve MTM1 KO mice were sampled at ⁇ 5 wks of age.
  • Tissue samples were used for myotubularin and bile salt transporter expression, bile acid level measurements, and histopathology. In-life and endpoint whole blood samples were used for hematology analysis and serum samples for clinical chemistry analysis. The same portion of the sample from each mouse was used for each analysis. The heart was collected with the left and right atria, then halved in the frontal plane. Tissue collection and analysis is summarized in Table 6, below. Table 6.
  • a clinical biochemistry panel of markers was analyzed in unhemolysed serum for the following parameters: alanine aminotransferase (ALAT), aspartate aminotransferase (ASAT), alkaline phosphatase (AFOS), gamma-glutamyl transferase (GGT), lactate dehydrogenase (LDH), creatine kinase (CK), albumin, total bilirubin, total protein, creatinine, bile acids, calculated globulin, calculated albumin/globulin ratio, and urea nitrogen. If the blood sample was limited, analysis priority was given in the following order: 1. Alanine aminotransferase (ALAT) 2.
  • ASAT aspartate aminotransferase
  • AFOS alkaline phosphatase
  • GTT gamma-glutamyl transferase
  • LDH lactate dehydrogenase
  • CK creatine kinase
  • albumin total bilirubin
  • total protein
  • Bile acids 4. Aspartate aminotransferase (ASAT) 5. Alkaline phosphatase (AFOS) 6. Gamma-glutamyl transferase (GGT) 7. Lactate dehydrogenase (LDH) 8. Creatine kinase (CK) 9. Albumin 10. Total protein 11. Creatinine 12. Urea Nitrogen Only ALAT was analyzed from in-life samples due to limited serum sample.
  • ASAT Aspartate aminotransferase
  • AFOS Alkaline phosphatase
  • GTT Gamma-glutamyl transferase
  • LDH Lactate dehydrogenase
  • CK Creatine kinase
  • Albumin 10. Total protein 11. Creatinine 12. Urea Nitrogen Only ALAT was analyzed from in-life samples due to limited serum sample.
  • Hematology Panel At in-life sampling and terminal necropsy, a hematology panel of markers was analyzed in EDTA whole blood for the following parameters: hemoglobin, red blood cell counts, white blood cell counts including relative and absolute differential counts, absolute reticulocyte counts, reticulocyte%, and thrombocyte counts. Additional parameters, if assay quantity of whole blood allowed, included: hematocrit, mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular PATENT ATTORNEY DOCKET NO.51037-075WO2 hemoglobin concentration. Genotyping Samples A tail sample was taken at the endpoint sampling for possible regenotyping with regular PCR.
  • Body weight was significantly less in WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP mice compared to WT Vehicle mice (Two-Way-ANOVA: Group: p ⁇ 0.05) on weeks 7-16 (Fisher’s LSD multiple comparison: p ⁇ 0.05, for all) (FIG.5). Body weight was significantly altered among HEMI mice (Two-Way-ANOVA: Group effect: NS, Group x Time interaction effect: p ⁇ 0.0001).
  • ALAT Alanine Aminotransferase
  • ALAT levels from na ⁇ ve mice at 5 weeks of age are shown in FIG.7B.
  • Aspartate Aminotransferase (ASAT) ASAT levels were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test: p ⁇ 0.01) (FIG.8A).
  • FIG.8B ASAT levels from na ⁇ ve mice at 5 weeks of age are shown in FIG.8B.
  • Albumin There were no statistically significant differences between the groups in albumin levels at 16 weeks of age (FIG.9A).
  • Albumin levels from na ⁇ ve mice at 5 weeks of age are shown in FIG.9B.
  • GGT Gamma-Glutamyl Transferase
  • Creatine Kinase CK levels were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test: p ⁇ 0.05) (FIG.18A). CK levels from na ⁇ ve mice at 5 weeks of age are shown in FIG.18B. Creatinine There were no statistically significant differences between the groups in creatinine levels at 16 weeks of age (FIG.19A). Creatinine levels from na ⁇ ve mice at 5 weeks of age are shown in FIG.19B.
  • WBC White Blood Cell Count
  • FIG.20B White blood cell counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.20B.
  • Red Blood Cell Count (RBC) There were no statistically significant differences between the groups in red blood cell counts (FIG.21A). Red blood cell counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.21B.
  • Hemoglobin (HGB) There were no statistically significant differences between the groups in hemoglobin levels (FIG.22A). Hemoglobin levels from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.22B.
  • HCT Hematocrit
  • MCV Mean Corpuscular Volume
  • MCHC Mean Corpuscular Hemoglobin Concentration
  • FIG.26B MCHC levels from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.26B.
  • Thrombocyte Count (PLT) Thrombocyte or platelet counts were significantly decreased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 6 weeks of age (Two-Way ANOVA: Group effect: p ⁇ 0.05, Dunnett’s post-hoc comparison: p ⁇ 0.01) (FIG.27A).
  • FIG. 27B Thrombocyte or platelet counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG. 27B.
  • Neutrophil Count (Relative) Neutrophil Count (Relative) (Neut(%)
  • FIG.28B Relative neutrophil counts from na ⁇ ve at 4 and 5 weeks of age mice are shown in FIG.28B.
  • Neutrophil Count Absolute
  • Neut Neut
  • FIG.29A Absolute neutrophil counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.29B.
  • Lymphocyte Count Relative lymphocyte counts were significantly decreased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice at 5 and 6 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: NS, Group x Time interaction effect: p ⁇ 0.05, Dunnett’s post-hoc comparison: p ⁇ 0.05, for all) (FIG.30A). Relative lymphocyte counts from na ⁇ ve mice are shown in FIG.30B.
  • Lymphocyte Count Absolute lymphocyte counts were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test, p ⁇ 0.01) (FIG.31A). Absolute lymphocyte counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.31B.
  • Monocyte Count (Relative) (Mono(%)) Relative monocyte counts were significantly increased in WT AAV8-Des-mMTM1 + AAV8- PATENT ATTORNEY DOCKET NO.51037-075WO2 Des-hMTM1-STOP mice compared to WT Vehicle mice at 16 weeks of age (t-test, p ⁇ 0.05) (FIG. 32A). Relative monocyte counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.32B.
  • Monocyte Count Absolute monocyte counts were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test, p ⁇ 0.01) (FIG.33A). Absolute monocyte counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.33B.
  • Eosinophil Count (Relative) (Eos(%)) Eosinophil counts were significantly increased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice at 4 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: p ⁇ 0.05, Dunnett’s post-hoc comparison: p ⁇ 0.01) (FIG.34A). Eosinophil counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.34B.
  • Basophil Count Relative basophil counts significantly decreased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice and in HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1- STOP mice at 5 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: NS, Group x Time interaction effect: p ⁇ 0.01, Dunnett’s post-hoc comparison: p ⁇ 0.05) (FIG. 36A).
  • FIG.36B Relative basophil counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.36B.
  • Basophil Count Absolute basophil counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.37B.
  • FIG.39B Absolute count of large unstained cells from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.39B.
  • Reticulocyte Count (Relative) (Retic(%)) There were no statistically significant differences between the groups in relative reticulocyte counts (FIG.40A).
  • Relative reticulocyte counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG.40B.
  • Reticulocyte Cell Count (Absolute) (Retic) There were no statistically significant differences between the groups in absolute reticulocyte cell counts (FIG.41A). Absolute reticulocyte cell counts from na ⁇ ve mice at 4 and 5 weeks of age are shown in FIG. 41B.
  • a patient having XLMTM may be concurrently administered a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter (FIG.1 and FIG.2) and a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec), each in a dose of less than about 3 x 10 14 vg/kg (e.g., in an
  • the patient Upon concurrently administering the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter (FIG.1 and FIG.2) and the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient, the patient displays a change from baseline in maximal inspiratory pressure.
  • a desmin promoter e.g., resamirigene bilparvovec
  • the patient displays the change from baseline in maximal inspiratory pressure by about 24 weeks (e.g., by about 20 weeks, 16 weeks, 12 weeks, 8 weeks, or 4 weeks) after concurrent administration of the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a liver- specific promoter (FIG.1 and FIG.2) and the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient.
  • a desmin promoter e.g., resamirigene bilparvovec
  • a patient having XLMTM may be administered a pseudotyped AAV2/8 vector including both a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter and a nucleic acid sequence encoding an MTM1 gene operably linked to a muscle-specific promoter (FIG.3) in a dose of less than about 3 x 10 14 vg/kg (e.g., in an amount of less than about 3 x 10 14 vg/kg, 2.9 x 10 14 vg/kg, 2.8 x 10 14 vg/kg, 2.7 x 10 14 vg
  • the patient Upon administering the AAV2/8 vector including both a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter and a nucleic acid sequence encoding an MTM1 gene operably linked to a muscle-specific promoter (FIG.3) to the patient, the patient exhibits a change from baseline in hours of mechanical ventilation support over time.
  • the patient exhibits the change from baseline in hours of mechanical ventilation support over time by about 24 weeks (e.g., by about 20 weeks, 16 weeks, 12 weeks, 8 weeks, or 4 weeks) after administration of the AAV2/8 vector including both a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter and a nucleic acid sequence encoding an MTM1 gene operably linked to a muscle-specific promoter (FIG.3) to the patient.
  • a patient having XLMTM may be administered a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a ubiquitous promoter (FIG.4) in a dose of less than about 3 x 10 14 vg/kg (e.g., in an amount of less than about 3 x 10 14 vg/kg, 2.9 x 10 14 vg/kg, 2.8 x 10 14 vg/kg, 2.7 x 10 14 vg/kg, 2.6 x 10 14 vg/kg, 2.5 x 10 14 vg/kg, 2.4 x 10 14 vg/kg, 2.3 x 10 14 vg/kg, 2.2 x 10 14 vg/kg,
  • the patient Upon administering the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a ubiquitous promoter (FIG.4) to the patient, the patient achieves functionally independent sitting for at least 30 seconds. For example, the patient achieves the functionally independent sitting by about 24 weeks (e.g., by about 20 weeks, 16 weeks, 12 weeks, 8 weeks, or 4 weeks) after administration of the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a ubiquitous promoter (FIG.4) to the patient.
  • Example 5 Example 5
  • a patient having XLMTM may be administered a non-viral vector (e.g.
  • lipid nanoparticle including a liver-expressing construct in combination with a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec), with the AAV2/8 vector administered in a dose of less than about 3 x 10 14 vg/kg (e.g., in an amount of less than about 3 x 10 14 vg/kg, 2.9 x 10 14 vg/kg, 2.8 x 10 14 vg/kg, 2.7 x 10 14 vg/kg, 2.6 x 10 14 vg/kg, 2.5 x 10 14 vg/kg, 2.4 x 10 14 vg/kg, 2.3 x 10 14 vg/kg, 2.2 x 10 14 vg/kg, 2.1 x 10 14 vg/kg, 2 x 10 14 vg/kg, 1.9 x 10 14 vg/kg,
  • non-viral vector e.g. a lipid nanoparticle
  • the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec)
  • the patient Upon administering the non-viral vector (e.g. a lipid nanoparticle) including a liver-expressing construct in combination with the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient, the patient achieves functionally independent sitting for at least 30 seconds.
  • a desmin promoter e.g., resamirigene bilparvovec
  • the patient achieves functionally independent sitting for at least 30 seconds by about 24 weeks (e.g., by about 20 weeks, 16 weeks, 12 weeks, 8 weeks, or 4 weeks) after administration of the non-viral vector (e.g., a lipid nanoparticle) including a liver-expressing construct in combination with the PATENT ATTORNEY DOCKET NO.51037-075WO2 pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient.
  • the non-viral vector e.g., a lipid nanoparticle
  • the PATENT ATTORNEY DOCKET NO.51037-075WO2 pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present disclosure relates to adeno-associated virus (AAV)-mediated delivery of nucleic acids for treating neuromuscular disorders, e.g., X-linked myotubular myopathy (XLMTM), in patients in need thereof. The AAV vectors of the disclosure may include, e.g., a transgene encoding a myotubularin protein operably linked to one or more transcription regulatory elements. In some embodiments, the one or more transcription regulatory elements are specifically active in muscle and/or liver tissue.

Description

PATENT ATTORNEY DOCKET NO.51037-075WO2 COMPOSITIONS AND METHODS FOR THE TREATMENT OF NEUROMUSCULAR DISORDERS Sequence Listing The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on September 14, 2023, is named “51037-075WO2_Sequence_Listing_9_14_23” and is 27,146 bytes in size. Background of the Invention X-linked myotubular myopathy (XLMTM) is a fatal monogenic disease of skeletal muscle, resulting from mutations in the myotubularin 1 (MTM1) gene. Approximately one in every 50,000 newborn boys has XLMTM, which typically displays as marked hypotonia and respiratory failure. In extremely rare cases, females can develop a severe form of XLMTM. Survival beyond the postnatal period requires intensive support, including respiratory support (i.e., mechanical ventilation) at birth in 85-90% of patients, ongoing 24-hour ventilator dependence in nearly 50% of patients, and tracheostomy in ~60% of patients. Until recently, only supportive treatment options, such as ventilator use or a feeding tube, were available. Recently, a gene therapy approach involving the delivery of MTM1 has been developed for the treatment of XLMTM. However, there is a need in the art for improved methods of administering gene therapy to patients having XLMTM. Summary of the Invention The present disclosure features compositions and methods useful for the treatment of neuromuscular disorders, particularly X-linked myotubular myopathy (XLMTM). Using the compositions and methods described herein, a patient (e.g., a human patient) having XLMTM may be administered a viral vector, such as an adeno-associated viral (AAV) vector, having a transgene encoding myotubularin 1 (MTM1). Exemplary composition and methods of the disclosure are described below. In a first aspect, the disclosure provides a recombinant AAV vector comprising a transgene encoding MTM1, wherein the transgene is operably linked to a promoter that is active in liver tissue. In some embodiments, the promoter is selectively active in liver tissue. In some embodiments, upon separately contacting the vector with one or more liver cells and one or more non-liver cells (e.g., in vitro or in vivo) under equivalent conditions, the vector results in a level of expression of the transgene in the one or more liver cells that is greater than the level of expression of the transgene in the one or more non-liver cells, for example, from 2-fold to 1,000-fold greater (e.g., 2- fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 200-fold, 300- fold, 400-fold, 500-fold, 600-fold, 700-fold, 800-fold, 900-fold, or 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells). In some embodiments, upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is from 10-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells. In some embodiments, upon separately contacting the vector with the one or more liver cells PATENT ATTORNEY DOCKET NO.51037-075WO2 and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is from 50-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells. In some embodiments, upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is from 100-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells. In some embodiments, upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is at least 2-fold, at least 5-fold, at least 10-fold, at least 50-fold, or at least 100-fold greater than the level of expression of the transgene in the one or more non-liver cells. In some embodiments, the non-liver cells are muscle cells or neural cells. In some embodiments, the non-liver cells are cardiac cells. In some embodiments, upon contacting the vector with one or more liver cells (e.g., of a patient having a neuromuscular disorder), the vector results in a level of expression of the transgene in the one or more liver cells that is closer to the expression of MTM1 in wild-type/healthy liver cells, for example, the expression of the transgene may be from 0.2-fold to 10-fold (e.g., 0.2-fold, 0.3-fold, 0.4-fold.0.5-fold, 0.6-fold, 0.7-fold, 0.8-fold, 0.9-fold, 1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4- fold, 4.5-fold, 5-fold, 5.5-fold, 6-fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, or 10- fold) the expression level of MTM1 in wild-type/healthy liver cells. In some embodiments, upon contacting the vector with one or more liver cells (e.g., of a patient having a neuromuscular disorder), the vector results in a level of expression of the transgene in the one or more liver cells that is at least 0.2-fold, at least 0.5-fold, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold the level of wild-type MTM1 expression in a healthy liver cell. In some embodiments, the promoter comprises an LP1 promoter. In some embodiments, the LP1 promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 3: GTCCCCTAAAATGGGCAAACATTGCAAGCAGCAAACAGCAAACACACAGCCCTCCCTGCCTGCT GACCTTGGAGCTGGGGCAGAGGTCAGAGACCTCTCTGGGCCCATGCCACCTCCAACATCCACT CGACCCCTTGGAATTTCGGTGGAGAGGAGCAGAGGTTGTCCTGGCGTGGTTTAGGTAGTGTGA GAGGGGAATGACTCCTTTCGGTAAGTGCAGTGGAAGCTGTACACTGCCCAGGCAAAGCGTCCG GGCAGCGTAGGCGGGCGACTCAGATCCCAGCCAGTGcACTTAGCCCCTGTTTGCTCCTCCGATA ACTGGGGTGACCTTGGTTAATATTCACCAGCAGCCTCCCCCGTTGCCCCTCTGGATCCACTGCT TAAATACGGACGAGGACAGGGCCCTGTCTCCTCAGCTTCAGGCACCACCACTGACCTGGGACA GTGAA (SEQ ID NO: 3). In some embodiments, the LP1 promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 3 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, PATENT ATTORNEY DOCKET NO.51037-075WO2 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 3). In some embodiments, the LP1 promoter has the nucleic acid sequence of SEQ ID NO: 3. In some embodiments, the promoter comprises an apolipoprotein E (ApoE) promoter. In some embodiments, the ApoE promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 8: CCCTAAAATGGGCAAACATTGCAAGCAGCAAACAGCAAACACACAGCCCTCCCTGCCTGCTGAC CTTGGAGCTGGGGCAGAGGTCAGAGACCTCTCTGGGCCCATGCCACCTCCAACATCCACTCGA CCCCTTGGAATTTCGGTGGAGAGGAGCAGAGGTTGTCCTGGCGTGGTTTAGGTAGTGTGAGAG GGG (SEQ ID NO: 8). In some embodiments, the ApoE promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 8 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 8). In some embodiments, the ApoE promoter has the nucleic acid sequence of SEQ ID NO: 8. In some embodiments, the promoter comprises an alpha-1-antitrysin (A1AT) promoter. In some embodiments, the A1AT promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 9: AATGACTCCTTTCGGTAAGTGCAGTGGAAGCTGTACACTGCCCAGGCAAAGCGTCCGGGCAGC GTAGGCGGGCGACTCAGATCCCAGCCAGTGGACTTAGCCCCTGTTTGCTCCTCCGATAACTGGG GTGACCTTGGTTAATATTCACCAGCAGCCTCCCCCGTTGCCCCTCTGGATCCACTGCTTAAATAC GGACGAGGACAGGGCCCTGTCTCCTCAGCTTCAGGCACCACCACTGACCTGGGACAGTGAAT (SEQ ID NO: 9). In some embodiments, the A1AT promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 9 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 9). In some embodiments, the A1AT promoter has the nucleic acid sequence of SEQ ID NO: 9. In some embodiments, the promoter is a chimeric promoter comprising an ApoE promoter and an A1AT promoter. In some embodiments, the chimeric promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 2: CGTGATCTAGTAGGCTCAGAGGCACACAGGAGTTTCTGGGCTCACCCTGCCCCCTTCCAACCCC TCAGTTCCCATCCTCCAGCAGCTGTTTGTGTGCTGCCTCTGAAGTCCACACTGAACAAACTTCAG CCTACTCATGTCCCTAAAATGGGCAAACATTGCAAGCAGCAAACAGCAAACACACAGCCCTCCCT GCCTGCTGACCTTGGAGCTGGGGCAGAGGTCAGAGACCTCTCTGGGCCCATGCCACCTCCAAC ATCCACTCGACCCCTTGGAATTTCGGTGGAGAGGAGCAGAGGTTGTCCTGGCGTGGTTTAGGTA PATENT ATTORNEY DOCKET NO.51037-075WO2 GTGTGAGAGGGGTACCCGGGGATCTTGCTACCAGTGGAACAGCCACTAAGGATTCTGCAGTGA GAGCAGAGGGCCAGCTAAGTGGTACTCTCCCAGAGACTGTCTGACTCACGCCACCCCCTCCAC CTTGGACACAGGACGCTGTGGTTTCTGAGCCAGGTACAATGACTCCTTTCGGTAAGTGCAGTGG AAGCTGTACACTGCCCAGGCAAAGCGTCCGGGCAGCGTAGGCGGGCGACTCAGATCCCAGCCA GTGGACTTAGCCCCTGTTTGCTCCTCCGATAACTGGGGTGACCTTGGTTAATATTCACCAGCAGC CTCCCCCGTTGCCCCTCTGGATCCACTGCTTAAATACGGACGAGGACAGGGCCCTGTCTCCTCA GCTTCAGGCACCACCACTGACCTGGGACAGTGAATGATCCCCCTGATCTGCGGCCTCGACGGTA TCGATAAG (SEQ ID NO: 2). In some embodiments, the chimeric promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 2 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 2). In some embodiments, the chimeric promoter has the nucleic acid sequence of SEQ ID NO: 2. In some embodiments, the promoter comprises a constitutive promoter. In some embodiments, the constitutive promoter is a phosphoglycerate kinase (PGK) promoter, an elongation factor-1 alpha (EF1alpha) promoter, a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) promoter, a cytomegalovirus (CMV) promoter, or a chicken-β-actin (CBA) promoter. In some embodiments, the AAV vector further comprises a second transgene encoding MTM1. In some embodiments, the second transgene encoding MTM1 is operably linked to a promoter that is active in muscle tissue. In some embodiments, the promoter that is active in muscle tissue is selectively active in muscle tissue. In some embodiments, the promoter that is selectively active in muscle tissue effectuates (e.g., in vitro or in vivo) a level of expression of the MTM1 transgene in muscle cells that is greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells, for example, from 2-fold to 1,000-fold greater (e.g., 2-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50- fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 200-fold, 300-fold, 400-fold, 500-fold, 600-fold, 700- fold, 800-fold, 900-fold, or 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells). In some embodiments, the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 10-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. In some embodiments, the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 50-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. In some embodiments, the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 100-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. In some embodiments, the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is at least 2-fold, at least 5-fold, at least 10-fold, at least 50-fold, or at least 100-fold greater PATENT ATTORNEY DOCKET NO.51037-075WO2 than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. In some embodiments, upon contacting the vector with one or more liver cells (e.g., of a patient having a neuromuscular disorder, the vector results in a level of expression of the transgene in the one or more liver cells that is closer to the expression of MTM1 in wild-type/healthy liver cells, for example, the expression of the transgene may be from 0.2-fold to 10-fold (e.g., 0.2-fold, 0.3-fold, 0.4- fold.0.5-fold, 0.6-fold, 0.7-fold, 0.8-fold, 0.9-fold, 1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4- fold, 4.5-fold, 5-fold, 5.5-fold, 6-fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, or 10- fold) the expression level of MTM1 in wild-type/healthy liver cells. In some embodiments, upon contacting the vector with one or more liver cells (e.g., of a patient having a neuromuscular disorder), the vector results in a level of expression of the transgene in the one or more liver cells that is at least 0.2-fold, at least 0.5-fold, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold the level of wild-type MTM1 expression in a healthy liver cell. In some embodiments, the second transgene encoding MTM1 is operably linked to a muscle creatine kinase (MCK) promoter or a desmin (DES) promoter. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1). In some embodiments, each transgene encoding MTM1 has the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 6. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 6 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 6). In some embodiments, each transgene encoding MTM1 has the nucleic acid sequence of SEQ ID NO: 6. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 7. In some embodiments, each transgene encoding MTM1, independently, has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 7 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 7). In some embodiments, each transgene encoding MTM1 has the nucleic acid sequence of SEQ ID NO: 7. In some embodiments, the MTM1 sequence is codon-optimized. PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, the AAV is an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAVrh10, or AAVrh74 serotype. In some embodiments, the AAV vector is a pseudotyped AAV. In some embodiments, the pseudotyped AAV is AAV2/8 or AAV2/9. In another aspect, the disclosure features a method of treating XLMTM in a human patient in need thereof by administering to the patient a therapeutically effective amount of the AAV vector of any of the above aspects or embodiments of the disclosure. In some embodiments, the patient is five years old or younger at the time of administration of the AAV vector. In some embodiments, the patient is four years old or younger at the time of administration of the AAV vector, optionally wherein the patient is three years old or younger, two years old or younger, one year old or younger, or six months old or younger. In some embodiments, the AAV vector is administered to the patient in an amount of less than 3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of less than 2.5 x 1014 vg/kg, optionally wherein the AAV vector is administered to the patient in an amount of less than 2 x 1014 vg/kg, less than 1.5 x 1014 vg/kg, or less than 1.4 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from 3 x 1013 vg/kg to 2.3 x 1014 vg/kg, optionally wherein the AAV vector is administered to the patient in an amount of from 8 x 1013 vg/kg to 1.8 x 1014 vg/kg, from 1 x 1014 vg/kg to 1.6 x 1014 vg/kg, from 1.1 x 1014 vg/kg to 1.5 x 1014 vg/kg, or from 1.2 x 1014 vg/kg to 1.4 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient by way of intravenous, intramuscular, intrahepatic, intradermal, or subcutaneous administration. In some embodiments, the patient is further administered an anti-cholestatic agent. In some embodiments, the anti-cholestatic agent is selected from the group consisting of a bile acid, a farnesoid X receptor (FXR) ligand, a fibroblast growth factor 19 (FGF-19) mimetic, a Takeda-G- protein-receptor-5 (TGR5) agonist, a peroxisome proliferator-activated receptor (PPAR) agonist, a PPAR-alpha agonist, a PPAR-delta agonist, a dual PPAR-alpha and PPAR-delta agonist, an apical sodium-dependent bile acid transporter (ASBT) inhibitor, an immunomodulatory drug, an antifibrotic therapy, and a nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitor. In some embodiments: (i) the FXR ligand is obeticholic acid, cilofexor, tropifexor, tretinoin, or EDP-305; (ii) the FGF-19 mimetic is aldafermin; (iii) the TGR5 agonist is INT-777 or INT-767; (iv) the PPAR agonist is bezafibrate, fenofibrate, seladelpar, or elafibrinor; (v) the PPAR-alpha agonist is fenofibrate; (vi) the PPAR-delta agonist is seladelpar; (vii) the dual PPAR-alpha and PPAR-delta agonist is elafibranor; (viii) the ASBT inhibitor is odevixibat, maralixibat, or linerixibat; (ix) the immunomodulatory drug is rituximab, abatacept, ustekinumab, infliximab, baricitinib, or FFP-104; (x) the antifibrotic therapy is a vitamin D receptor agonist or simtuzumab; and/or (xi) the NOX inhibitor is setanaxib. In some embodiments, the bile acid is ursodeoxycholic acid, nor-ursodeoxycholic acid, or a pharmaceutically acceptable salt thereof. In some embodiments, the patient does not have a history of cholestasis or hyperbilirubinemia. In some embodiments, the patient does not have a history of any underlying liver disease. PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, the method comprises administering to the patient therapeutically effective amounts of: (i) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in liver tissue, and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue. In some embodiments, the promoter that is active in liver tissue comprises an LP1 promoter. In some embodiments, the LP1 promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 3. In some embodiments, the LP1 promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 3 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 3). In some embodiments, the LP1 promoter has the nucleic acid sequence of SEQ ID NO: 3. In some embodiments, the promoter that is active in liver tissue comprises an ApoE promoter. In some embodiments, the ApoE promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 8. In some embodiments, the ApoE promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 8 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 8). In some embodiments, the ApoE promoter has the nucleic acid sequence of SEQ ID NO: 8. In some embodiments, the promoter that is active in liver tissue comprises an A1AT promoter. In some embodiments, the A1AT promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 9. In some embodiments, the A1AT promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 9 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 9). In some embodiments, the A1AT promoter has the nucleic acid sequence of SEQ ID NO: 9. In some embodiments, the promoter that is active in liver tissue is a chimeric promoter comprising an ApoE promoter and an A1AT promoter. In some embodiments, the chimeric promoter has a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 2. In some embodiments, the chimeric promoter has a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 2 (e.g., a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 2). In some embodiments, the chimeric promoter has the nucleic acid sequence of SEQ ID NO: 2. In a further aspect, the disclosure features a method of treating XLMTM in a human patient in need thereof by administering to the patient therapeutically effective amounts of: (i) a non-viral composition comprising a nucleic acid encoding MTM1 operably linked to a promoter that is active PATENT ATTORNEY DOCKET NO.51037-075WO2 (e.g., selectively active) in liver tissue, and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue. In some embodiments, the non-viral composition is a liposome, vesicle, synthetic vesicle, exosome, synthetic exosome, dendrimer, or nanoparticle. In some embodiments, the nanoparticle is a lipid nanoparticle, which can deliver DNA constructs similar in composition and size to those delivered via AAV capsids. In some embodiments, the promoter is a DES promoter. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each have the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 6. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 6. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 6. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each have the nucleic acid sequence of SEQ ID NO: 6. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 7. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 7. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, have a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 7. In some embodiments, the nucleic acid encoding MTM1 and the transgene encoding MTM1 each have the nucleic acid sequence of SEQ ID NO: 7. In some embodiments, the AAV vector is resamirigene bilparvovec. PATENT ATTORNEY DOCKET NO.51037-075WO2 In a further aspect, the disclosure provides a kit comprising the AAV vector of any of the above aspects or embodiments of the disclosure. The kit may further include a package insert instructing a user to administer the AAV vector to a patient diagnosed as having XLMTM. In another aspect, the disclosure provides a kit comprising: (i) a non-viral composition comprising a nucleic acid encoding MTM1 (e.g., a non-viral composition described above), and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue (e.g., an AAV vector described above). The kit may further include a package insert instructing a user to administer the non-viral composition and the AAV vector to a patient diagnosed as having XLMTM. Brief Description of the Drawings FIG.1 is a schematic drawing of an exemplary adeno-associated virus (AAV) vector for the expression of the human or mouse myotubularin 1 (MTM1) gene (e.g., a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8). From left to right, the shaded arrows and rectangles represents the nucleic acid sequences encoding an ApoE/A1AT liver-specific promoter (ApoE/A1AT) operatively linked to a Beta- globin Intron (b-globin intron), a human or mouse MTM1 gene, an SV40 poly-adenylation signal (SV40-polyA), and flanking inverted terminal repeat sequences (ITR). FIG.2 is a schematic drawing of an exemplary AAV vector for the expression of the human or mouse myotubularin 1 (MTM1) gene (e.g., a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8). From left to right, the shaded arrows and rectangles represent the nucleic acid sequences encoding a LP1 liver- specific promoter (LP1 promoter) operatively linked to an SV40 Intron, a human or mouse MTM1 gene, an SV40 poly-adenylation signal (SV40-polyA), and flanking inverted terminal repeat sequences (ITR). FIG.3 is a schematic drawing of an exemplary AAV vector for the expression of the human myotubularin 1 (hMTM1) gene (e.g., a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8). From left to right, the shaded arrows and rectangles represent the nucleic acid sequences encoding a muscle-specific promoter (e.g., MCK, desmin) operatively linked to hMTM1 and a poly-adenylation signal (polyA), a transcription pause site, a liver-specific promoter (e.g., ApoE/A1AT, LP1) operatively linked to hMTM1, a poly-adenylation signal (polyA), and flanking inverted terminal repeat sequences (ITR). FIG.4 is a schematic drawing of an exemplary AAV vector for the expression of the human myotubularin 1 (hMTM1) gene (e.g., a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8). From left to right, the shaded arrows and rectangles represent the nucleic acid sequences encoding a ubiquitous promoter (e.g., PGK, Ef1a, GAPDH) operatively linked to hMTM1, a poly-adenylation signal (polyA), and flanking inverted terminal repeat sequences (ITR). FIG.5 is a graph showing body weight (g) over a period (age in weeks), mean + SEM (n= 10- 14, for all), measured in the manner described in Example 1, below. Statistical significances: HEMI PATENT ATTORNEY DOCKET NO.51037-075WO2 AAV8-Des-mMTM1 mice vs. WT Vehicle mice: p < 0.001 for all time points; WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP mice vs. WT Vehicle mice: p < 0.05 for weeks 7-16; HEMI AAV8-Des- mMTM1 + AAV8 empty capsid mice vs. HEMI AAV8-Des-mMTM1 mice: p < 0.05 on weeks 4 and 5; HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP mice vs. HEMI AAV8-Des-mMTM1 mice: p < 0.05 on weeks 5-6 and 8-9. FIG.5 abbreviations: WT, wild type; HEMI, hemizygous. FIG.6A is a graph showing alkaline phosphatase (AFOS) (U/L) levels, mean + SEM (n=10- 14, for all), measured in the manner described in Example 1, below. No statistical differences were observed. FIG.6A abbreviations: WT, wild type; HEMI, hemizygous; AFOS, Alkaline Phosphatase. FIG.6B is a graph showing alkaline phosphatase (AFOS) (U/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.6B abbreviations: AFOS, Alkaline Phosphatase. FIG.7A is a graph showing alanine aminotransferase (ALAT) (U/L) levels, mean + SEM (in- life samples (weeks 4-10): n=0-7; terminal samples (week 16): n=10-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des- mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8- Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP. Data are not shown for groups that fell below the limit of detection. Statistical differences: *p<0.05 compared to WT Vehicle, #p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.7A abbreviations: WT, wild type; HEMI, hemizygous; ALAT, Alanine Aminotransferase. FIG.7B is a graph showing alanine aminotransferase (ALAT) (U/L) levels in naïve mice, mean + SEM (n=5-11), measured in the manner described in Example 1, below. FIG.7B abbreviations: ALAT, Alanine Aminotransferase. FIG.8A is a graph showing aspartate aminotransferase (ASAT) (U/L) levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. Statistical differences: **p<0.01 compared to WT Vehicle. FIG.8A abbreviations: WT, wild type; HEMI, hemizygous; ASAT, Aspartate Aminotransferase. FIG.8B is a graph showing aspartate aminotransferase (ASAT) (U/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.8B abbreviations: ASAT, Aspartate Aminotransferase. FIG.9A is a graph showing albumin (g/L) levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. No statistical differences were observed. FIG.9A abbreviations: WT, wild type; HEMI, hemizygous. FIG.9B is a graph showing albumin (g/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.10A is a graph showing gamma-glutamyl transferase (GGT) (U/L) levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. Statistical differences: #p<0.05, ##p<0.01 compared to HEMI AAV8-Des-mMTM1. FIG.10A abbreviations: WT, wild type; PATENT ATTORNEY DOCKET NO.51037-075WO2 HEMI, hemizygous; GGT, Gamma-Glutamyl Transferase. FIG.10B is a graph showing gamma-glutamyl transferase (GGT) (U/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.10B abbreviations: GGT, Gamma-Glutamyl Transferase. FIG.11A is a graph showing total protein (g/L) levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. No statistical differences were observed. FIG.11A abbreviations: WT, wild type; HEMI, hemizygous; Prot tot, total protein. FIG.11B is a graph showing total protein (g/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.11B abbreviations: Prot tot, total protein. FIG.12A is a graph showing urea (mmol/L) levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. No statistical differences were observed. FIG.12A abbreviations: WT, wild type; HEMI, hemizygous. FIG.12B is a graph showing urea (mmol/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.13A is a graph showing lactate dehydrogenase (LDH) (U/I) levels, mean + SEM (n=10- 14, for all), measured in the manner described in Example 1, below. No statistical differences were observed. FIG.13A abbreviations: WT, wild type; HEMI, hemizygous; LDH, Lactate Dehydrogenase. FIG.13B is a graph showing lactate dehydrogenase (LDH) (U/I) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.13B abbreviations: LDH, Lactate Dehydrogenase. FIG.14A is a graph showing levels of bile acids (µmol/L), mean + SEM (n=0-3, for all), measured in the manner described in Example 1, below. Data are not shown for groups that fell below the limit of detection. Statistical differences: #p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.14A abbreviations: WT, wild type; HEMI, hemizygous. FIG.14B is a graph showing levels of bile acids (µmol/L) in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.15 is a graph showing total bilirubin (µmol/L) levels, mean + SEM (n=0-8, for all), measured in the manner described in Example 1, below. Data are not shown for groups that fell below the limit of detection. No statistical differences were observed. FIG.15 abbreviations: WT, wild type; HEMI, hemizygous; BilTot, total bilirubin. FIG.16A is a graph showing calculated globulin (g/L) levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. Statistical differences: *p<0.05 compared to WT Vehicle. FIG.16A abbreviations: WT, wild type; HEMI, hemizygous. FIG.16B is a graph showing calculated globulin (g/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.17A is a graph showing calculated albumin/globulin ratio levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. No statistical differences were observed. FIG.17A abbreviations: WT, wild type; HEMI, hemizygous. FIG.17B is a graph showing calculated albumin/globulin ratio levels in naïve mice, mean + PATENT ATTORNEY DOCKET NO.51037-075WO2 SEM (n=11), measured in the manner described in Example 1, below. FIG.18A is a graph showing creatine kinase (U/L) levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. Statistical differences: *p<0.05 compared to WT Vehicle. FIG.18A abbreviations: WT, wild type; HEMI, hemizygous; CK, creatine kinase. FIG.18B is a graph showing creatine kinase (U/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.18B abbreviations: CK, creatine kinase. FIG.19A is a graph showing creatinine (umol/L) levels, mean + SEM (n=10-14, for all), measured in the manner described in Example 1, below. No statistical differences were observed. FIG.19A abbreviations: WT, wild type; HEMI, hemizygous. FIG.19B is a graph showing creatinine (umol/L) levels in naïve mice, mean + SEM (n=11), measured in the manner described in Example 1, below. FIG.20A is a graph showing white blood cell counts (WBC) (x109cells/L), mean + SEM (n=5- 14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. Statistical differences: *p<0.05 compared to WT Vehicle, #p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.20A abbreviations: WT, wild type; HEMI, hemizygous; WBC, white blood cell count. FIG.20B is a graph showing white blood cell counts (WBC) (x109cells/L) levels in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.20B abbreviations: WBC, white blood cell count. FIG.21A is a graph showing red blood cell counts (RBC) (x1012cells/L), mean + SEM (n=5- 14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. No statistical differences were observed. FIG.21A abbreviations: WT, wild type; HEMI, hemizygous; RBC, red blood cell count. FIG.21B is a graph showing red blood cell counts (RBC) (x1012cells/L) levels in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.21B abbreviations: RBC, red blood cell count. FIG.22A is a graph showing hemoglobin (g/L) levels, mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- PATENT ATTORNEY DOCKET NO.51037-075WO2 STOP. No statistical differences were observed. FIG.22A abbreviations: WT, wild type; HEMI, hemizygous; HGB, hemoglobin. FIG.22B is a graph showing hemoglobin (g/L) levels in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.22B abbreviations: HGB, hemoglobin. FIG.23A is a graph showing hematocrit (HCT) (%) levels, mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. Statistical differences: #p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.23A abbreviations: WT, wild type; HEMI, hemizygous; HCT, hematocrit. FIG.23B is a graph showing hematocrit (HCT) (%) levels in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.23B abbreviations: HCT, hematocrit. FIG.24A is a graph showing mean corpuscular volume (MCV) (fL) levels, mean + SEM (n=5- 14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. Statistical differences: *p<0.05 compared to WT Vehicle, #p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.24A abbreviations: WT, wild type; HEMI, hemizygous; MCV, mean corpuscular volume. FIG.24B is a graph showing mean corpuscular volume (MCV) (fL) levels in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.24B abbreviations: MCV, mean corpuscular volume. FIG.25A is a graph showing mean corpuscular hemoglobin (MCH) (pg) levels, mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. No statistical differences were observed. FIG.25A abbreviations: WT, wild type; HEMI, hemizygous; MCH, mean corpuscular hemoglobin. FIG.25B is a graph showing mean corpuscular hemoglobin (MCH) (pg) levels in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.25B abbreviations: MCH, mean corpuscular hemoglobin. FIG.26A is a graph showing mean corpuscular hemoglobin concentration (MCHC) (g/L) levels, mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At PATENT ATTORNEY DOCKET NO.51037-075WO2 each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8- Des-mMTM1 + AAV8-Des-hMTM1-STOP. Statistical differences: #p<0.05 compared to HEMI AAV8- Des-mMTM1. FIG.26A abbreviations: WT, wild type; HEMI, hemizygous; MCHC, mean corpuscular hemoglobin concentration. FIG.26B is a graph showing mean corpuscular hemoglobin concentration (MCHC) (g/L) levels in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.26B abbreviations: MCHC, mean corpuscular hemoglobin concentration. FIG.27A is a graph showing thrombocyte counts (x109cells/L), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. Statistical differences: *p<0.05 compared to WT Vehicle. FIG.27A abbreviations: WT, wild type; HEMI, hemizygous; PLT, platelet or thrombocyte count. FIG.27B is a graph showing thrombocyte counts (x109cells/L) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.27B abbreviations: PLT, platelet or thrombocyte count. FIG.28A is a graph showing relative neutrophil counts (neutrophil (%)), mean + SEM (n=5- 14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. No statistical differences were observed. FIG.28A abbreviations: WT, wild type; HEMI, hemizygous; %NEUT, Neutrophil (%). FIG.28B is a graph showing relative neutrophil counts (neutrophil (%)) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.28B abbreviations: %NEUT, Neutrophil (%). FIG.29A is a graph showing absolute neutrophil counts (x109cells/L), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. No statistical differences were observed. FIG.29A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute NEUT, absolute neutrophil count. PATENT ATTORNEY DOCKET NO.51037-075WO2 FIG.29B is a graph showing absolute neutrophil counts (x109cells/L) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.29B abbreviations: Abs/Absolute NEUT, absolute neutrophil count. FIG.30A is a graph showing relative lymphocyte counts (Lymphocyte (%)), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. Statistical differences: #p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.30A abbreviations: WT, wild type; HEMI, hemizygous; %LYM, Lymphocyte (%). FIG.30B is a graph showing relative lymphocyte counts (Lymphocyte (%)) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.30B abbreviations: %LYM, Lymphocyte (%). FIG.31A is a graph showing absolute lymphocyte counts (x109cells/L), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. Statistical differences: *p<0.05 compared to WT Vehicle. FIG.31A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute LYMPHS, absolute lymphocyte count. FIG.31B is a graph showing absolute lymphocyte counts (x109cells/L) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.31B abbreviations: Abs/Absolute LYMPHS, absolute lymphocyte count. FIG.32A is a graph showing relative monocyte counts (Monocyte (%)), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. Statistical differences: *p<0.05 compared to WT Vehicle. FIG.32A abbreviations: WT, wild type; HEMI, hemizygous; %MONO, Monocyte (%). FIG.32B is a graph showing relative monocyte counts (Monocyte (%)) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.32B abbreviations: %MONO, Monocyte (%). FIG.33A is a graph showing absolute monocyte counts (x109cells/L), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty PATENT ATTORNEY DOCKET NO.51037-075WO2 capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. Statistical differences: *p<0.05 compared to WT Vehicle. FIG.33A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute MONOS, absolute monocyte count. FIG.33B is a graph showing absolute monocyte counts (x109cells/L) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.33B abbreviations: Abs/Absolute MONOS, absolute monocyte count. FIG.34A is a graph showing relative eosinophil counts (Eosinophil (%)), mean + SEM (n=5- 14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. Statistical differences: ##p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.34A abbreviations: WT, wild type; HEMI, hemizygous; %EOS, Eosinophil (%). FIG.34B is a graph showing relative eosinophil counts (Eosinophil (%)) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.34B abbreviations: %EOS, Eosinophil (%). FIG.35A is a graph showing absolute eosinophil counts (x109cells/L), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. Statistical differences: #p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.35A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute EOS, absolute eosinophil count. FIG.35B is a graph showing absolute eosinophil counts (x109cells/L) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.35B abbreviations: Abs/Absolute EOS, absolute eosinophil count. FIG.36A is a graph showing relative basophil counts (Basophil (%)), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. Data are not shown for groups that fell below the limit of detection. Statistical differences: #p<0.05 compared to HEMI AAV8-Des-mMTM1. FIG.36A abbreviations: WT, wild type; HEMI, hemizygous; %BASO, Basophil (%). FIG.36B is a graph showing relative basophil counts (Basophil (%)) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.36B PATENT ATTORNEY DOCKET NO.51037-075WO2 abbreviations: %BASO, Basophil (%). FIG.37A is a graph showing absolute basophil counts (x109cells/L), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8- APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1- STOP. No statistical differences were observed. Data are not shown for groups that fell below the limit of detection. FIG.37A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute BASOS, absolute basophil count. FIG.37B is a graph showing absolute basophil counts (x109cells/L) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. Data are not shown for groups that fell below the limit of detection. FIG.37B abbreviations: Abs/Absolute BASOS, absolute basophil count. FIG.38A is a graph showing the relative count of large unstained cells (Large unstained cells (%)), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8- Des-mMTM1 + AAV8-Des-hMTM1-STOP. Statistical differences: **p<0.01 compared to WT Vehicle. FIG.38A abbreviations: WT, wild type; HEMI, hemizygous; %LUC, Large unstained cells (%). FIG.38B is a graph showing the relative count of large unstained cells (Large unstained cells (%)), naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG. 38B abbreviations: %LUC, Large unstained cells (%). FIG.39A is a graph showing the absolute count of large unstained cells (x109cells/L), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des- mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des- mMTM1 + AAV8-Des-hMTM1-STOP. Data are not shown for groups that fell below the limit of detection. Statistical differences: #p<0.05, ##p<0.01 compared to HEMI AAV8-Des-mMTM1. FIG.39A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute LUCS, absolute count of large unstained cells. FIG.39B is a graph showing the absolute count of large unstained cells (x109cells/L), naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. Data are not shown for groups that fell below the limit of detection. FIG.39B abbreviations: Abs/Absolute LUCS, absolute count of large unstained cells. FIG.40A is a graph showing relative reticulocyte counts (Reticulocyte (%)), mean + SEM PATENT ATTORNEY DOCKET NO.51037-075WO2 (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. No statistical differences were observed. FIG.40A abbreviations: WT, wild type; HEMI, hemizygous; %Retic, Reticulocyte (%). FIG.40B is a graph showing relative reticulocyte counts (Reticulocyte (%)) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.40B abbreviations: %Retic, Reticulocyte (%). FIG.41A is a graph showing absolute reticulocyte cell counts (x109cells/L), mean + SEM (n=5-14, for all), measured in the manner described in Example 1, below. At each time point, 4, 5, 6, 7, 10, and 16 weeks, the following groups were tested from left to right: WT Vehicle, HEMI AAV8-Des- mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP, HEMI AAV8-Des-mMTM1 + AAV8 empty capsid, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1, HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP, and WT AAV8-Des-mMTM1 + AAV8-Des- hMTM1-STOP. No statistical differences were observed. FIG.41A abbreviations: WT, wild type; HEMI, hemizygous; Abs/Absolute Retic, absolute reticulocyte cell count. FIG.41B is a graph showing absolute reticulocyte cell counts (x109cells/L) in naïve mice, mean + SEM (n=3/9), measured in the manner described in Example 1, below. FIG.41B abbreviations: Abs/Absolute Retic, absolute reticulocyte cell count. Definitions As used herein, the term “about” refers to a value that is within 10% above or below the value being described. For example, “100 pounds” as used in the context of weight described herein includes quantities that are within 10% above or below 100 lbs. Additionally, when used in the context of a list of numerical quantities, it is to be understood that the term “about,” when preceding a list of numerical quantities, applies to each individual quantity recited in the list. As used herein, the terms “administering,” “administration,” and the like refer to directly giving a patient a therapeutic agent (e.g., a pharmaceutical composition including a viral vector including a nucleic acid sequence encoding a myotubularin 1 (MTM1) gene operably linked to a promoter) by any effective route. Exemplary routes of administration are described herein and include systemic administration routes, such as intravenous injection, as well as routes of administration directly to the central nervous system of the patient, such as by way of intrathecal injection or intracerebroventricular injection, and directly to the liver of the patient, among others. As used herein, the term “age-adjusted norms” refers to the process of a normalization of data by age, which is a technique that is used to allow populations of subjects to be compared when the age profiles of the populations are different. As used herein, the term “norm” refers to data that does not undergo a normalization by age, as populations of subjects across age profiles are similar. PATENT ATTORNEY DOCKET NO.51037-075WO2 As used herein, the terms “alanine aminotransferase” and “ALT” refer to a protein whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring wild- type ALT protein (e.g., ALT1 and ALT2) as well as proteins whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring allelic variants of ALT (GPT or GPT2 e.g., splice variants or allelic variants). Human GPT nucleic acid sequence is provided in NCBI RefSeq Acc. No. NM_005309.2, and an exemplary wild-type ALT1 amino acid sequence is provided in NCBI RefSeq Acc. No. NP_005300.1. Human GPT2 nucleic acid sequence is provided in NCBI RefSeq Acc. No. NM_001142466.2, and an exemplary wild-type ALT2 amino acid sequence is provided in NCBI RefSeq Acc. No. NP_001135938.1. As used herein, the terms “alkaline phosphatase” and “ASP” refer to a protein whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring wild-type ASP protein as well as proteins whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring allelic variants of ASP (e.g., splice variants or allelic variants). Human ASP nucleic acid sequence is provided in NCBI RefSeq Acc. No. NM_000478.5, and an exemplary wild-type ASP amino acid sequence is provided in NCBI RefSeq Acc. No. NP_000469.3. As used herein, the term “anti-cholestatic agent” refers to a substance, such as a small molecule that acts to increase bile formation and/or antagonize the effect of hydrophobic bile acids on biological membranes. The term “antagonize,” as used herein with regard to a protein, refers to a molecule that decreases signal transduction resulting from the interaction of the protein with one or more of its binding partners. The antagonist may result in a decrease in the binding of the protein to one or more of its binding partners relative to binding of the two proteins in the absence of the antagonist. As used herein, the terms “aspartate aminotransferase” and “AST” refer to a protein whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring wild- type AST protein as well as proteins whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring allelic variants of AST (e.g., splice variants or allelic variants). Human AST nucleic acid sequence is provided in NCBI RefSeq Acc. No NM_002079.2, and an exemplary wild-type ASP amino acid sequence is provided in NCBI RefSeq Acc. No. NP_002070.1. As used herein, the terms “bile acid test” and “serum bile acid test” refer to the procedure in which a pre-prandial (i.e., before eating) blood sample is collected for a baseline, followed by a meal and followed about two hours later by the collection of a postprandial (i.e., after eating) blood sample. Both blood samples are tested for bile acid levels and the pre-prandial sample is used as a reference. As used herein, “bile acid” refers to the steroid acids found predominantly in the bile of mammals and other vertebrates. As used herein, the terms “Children’s Hospital of Philadelphia Infant Test of Neuromuscular Disorders” and “CHOP INTEND” refer to a validated motor outcome measure developed for the evaluation of weak infants, such as those with a disease of skeletal muscle (e.g., X-linked myotubular myopathy (XLMTM)). CHOP INTEND uses a 0–64-point scale where higher scores indicate better motor function. As used herein, the term “motor function score” refers to a score on the 0–64-point scale of the CHOP INTEND (e.g., a scale of >45 on the CHOP INTEND). PATENT ATTORNEY DOCKET NO.51037-075WO2 As used herein, the term “cholestasis” refers to a condition where bile cannot flow from the liver to the duodenum. The two clinical distinctions are the “obstructive” type of cholestasis where there is a mechanical blockage in the duct system that can occur from a gallstone or malignancy, and “metabolic” types of cholestasis which are disturbances in bile formation that can occur because of genetic defects or acquired as a side effect of many medications. As used herein, the term “bile” refers to the digestive fluid that is secreted by the liver to aid in the digestion of fats. As used herein, a “combination therapy” means that two (or more) different agents or treatments are administered to a subject as part of a defined treatment regimen for a particular disease or condition (e.g., a neuromuscular disorder). In some embodiments, a “combination therapy” may include a procedure. The treatment regimen defines the doses and periodicity of administration of each agent such that the effects of the separate agents on the subject overlap. In some embodiments, the delivery of the two or more agents is simultaneous or concurrent and the agents may be co-formulated. In other embodiments, the two or more agents are not co-formulated and are administered in a sequential manner as part of a prescribed regimen. In some embodiments, administration of two or more agents or treatments in combination is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive (e.g., synergistic). Sequential or substantially simultaneous administration of each therapeutic agent can be affected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, intrahepatic routes, and direct absorption through mucous membrane tissues. Therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination may be administered enterally. In another example, an agent of the therapeutic combination may be administered by intravenous injection and a procedure (e.g., nasobiliary drainage (NBD)) of the therapeutic combination may be performed. As used herein, the term “dose” refers to the quantity of a therapeutic agent, such as a viral vector described herein, that is administered to a subject at a particular instant for the treatment of a disorder, such as to treat or ameliorate one or more symptoms of a neuromuscular disorder described herein (e.g., XLMTM). A therapeutic agent as described herein may be administered in a single dose or in multiple doses over the course of a treatment period, as defined herein. In each case, therapeutic agent may be administered using one or more unit dosage forms of therapeutic agent, a term that refers to a one or more discrete compositions containing a therapeutic agent that collectively constitute a single dose of the agent. As used herein, the terms “effective amount,” “therapeutically effective amount,” and the like, when used in reference to a therapeutic composition, such as a vector construct described herein, refer to a quantity sufficient to, when administered to the subject, including a mammal, for example a human, effect beneficial or desired results, such as clinical results. For example, in the context of treating neuromuscular disorders, such as XLMTM, these terms refer to an amount of the composition PATENT ATTORNEY DOCKET NO.51037-075WO2 sufficient to achieve a treatment response as compared to the response obtained without administration of the composition of interest. An “effective amount,” "therapeutically effective amount,” and the like, of a composition, such as a vector construct of the present disclosure, also include an amount that results in a beneficial or desired result in a subject as compared to a control. As used herein, the terms “gamma-glutamyl transferase” and “GGT” refers to a protein whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring wild- type GGT protein as well as proteins whose amino acid sequence comprises or consists of an amino acid sequence of a naturally occurring allelic variants of GGT (GGT1, GGT2, and GGT3 e.g., splice variants or allelic variants). Human GGT1 nucleic acid sequence is provided in NCBI RefSeq Acc. No NM_001288833.1, and an exemplary wild-type GGT1 amino acid sequence is provided in NCBI RefSeq Acc. No. NP_001275762.1. As used herein, the term “hyperbilirubinemia” refers to a condition in which there is a higher- than-normal level of bilirubin in the blood. As used herein, the term “bilirubin” refers to a compound that occurs in the normal catabolic pathway that breaks down heme in vertebrates. This catabolism is a necessary process in the body's clearance of waste products that arise from the destruction of aged or abnormal red blood cells. As used herein, a “bilirubin test” refers to a measurement of the amount of bilirubin in a patient’s blood. As used herein, the term “level” refers to a level of a protein, as compared to a reference. The reference can be any useful reference, as defined herein. By a “decreased level” and an “increased level” of a protein is meant a decrease or increase in protein level, as compared to a reference (e.g., a decrease or an increase by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, or more; a decrease or an increase of more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; a decrease or an increase by less than about 0.01-fold, about 0.02-fold, about 0.1-fold, about 0.3-fold, about 0.5-fold, about 0.8-fold, or less; or an increase by more than about 1.2-fold, about 1.4-fold, about 1.5-fold, about 1.8-fold, about 2.0-fold, about 3.0-fold, about 3.5- fold, about 4.5-fold, about 5.0-fold, about 10-fold, about 15-fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold, about 1000-fold, or more). A level of a protein may be expressed in mass/vol (e.g., g/dL, mg/mL, μg/mL, or ng/mL) or percentage relative to total protein in a sample. As used herein, the terms “liver function test” and “LFT” refers to a hepatic panel (e.g., a group of blood tests that provide information about the state of a patient's liver). A hepatic panel may include measurement of the level of gamma-glutamyl transferase, the level of alkaline phosphatase, the level of aspartate aminotransferase, the level of alanine aminotransferase, the level of albumin, the level of bilirubin, the prothrombin time, the activated partial thromboplastin time, or a combination thereof. As used herein, the terms “maximal inspiratory pressure” and “MIP” refer to a variable in mechanical ventilation including the total airway pressure delivered, generally used to overcome both PATENT ATTORNEY DOCKET NO.51037-075WO2 respiratory system compliance as well as airway resistance. In as pressure-controlled mode, the MIP includes the sum of the positive-end expiratory pressure and the “delta pressure.” As used herein, the term “delta pressure” refers to a variable in mechanical ventilation including the difference between the MIP and the positive-end expiratory pressure. As used herein, the term “mechanical ventilatory support” refers to the medical term for artificial ventilation where mechanical means are used to assist or replace spontaneous breathing. As used herein, the term “invasive mechanical ventilatory support” refers to the medical term for artificial ventilation where air is delivered via a tube that is inserted into a patient’s windpipe through the mouth or nose and mechanical means are used to assist or replace spontaneous breathing. As used herein, the term “noninvasive mechanical ventillatory support” refers to mechanical ventilatory support in which air is delivered to a patient through a sealed mask that can be placed over the mouth, nose, or the whole face. As used herein, the term “operably linked” refers to a first molecule joined to a second molecule, wherein the molecules are so arranged that the first molecule affects the function of the second molecule. The two molecules may or may not be part of a single contiguous molecule and may or may not be adjacent. For example, a promoter is operably linked to a transcribable polynucleotide molecule if the promoter modulates transcription of the transcribable polynucleotide molecule of interest in a cell. Additionally, two portions of a transcription regulatory element are operably linked to one another if they are joined such that the transcription-activating functionality of one portion is not adversely affected by the presence of the other portion. Two transcription regulatory elements may be operably linked to one another by way of a linker nucleic acid (e.g., an intervening non-coding nucleic acid) or may be operably linked to one another with no intervening nucleotides present. As used herein, the term “pharmaceutical composition” refers to a mixture containing a therapeutic compound to be administered to a subject, such as a mammal, e.g., a human, in order to prevent, treat or control a particular disease or condition affecting or that may affect the subject. As used herein, the term “pharmaceutically acceptable” refers to those compounds, materials, compositions and/or dosage forms, which are suitable for contact with the tissues of a subject, such as a mammal (e.g., a human) without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio. As used herein, the term “promoter” refers to a recognition site on DNA that is bound by an RNA polymerase. The polymerase drives transcription of the transgene. Exemplary promoters suitable for use with the compositions and methods described herein are described, for example, in Sandelin et al., Nature Reviews Genetics 8:424 (2007), the disclosure of which is incorporated herein by reference as it pertains to nucleic acid regulatory elements. Additionally, the term “promoter” may refer to a synthetic promoter, which are regulatory DNA sequences that do not occur naturally in biological systems. Synthetic promoters contain parts of naturally occurring promoters combined with polynucleotide PATENT ATTORNEY DOCKET NO.51037-075WO2 sequences that do not occur in nature and can be optimized to express recombinant DNA using a variety of transgenes, vectors, and target cell types. As used herein in the context of a promoter, the term “selectively active” refers to a promoter that preferentially drives gene expression in one cell or tissue type as compared to another cell or tissue type. For example, a promoter that is “selectively active” in liver cells may achieve a level of gene expression in a liver cell that is greater than the level of gene expression achieved by the same promoter, under substantially the same conditions, in one or more non-liver cells, for example, from 2- fold to 1,000-fold greater (e.g., 2-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80- fold, 90-fold, 100-fold, 200-fold, 300-fold, 400-fold, 500-fold, 600-fold, 700-fold, 800-fold, 900-fold, or 1,000-fold greater expression). Exemplary methods for measuring gene expression are known in the art and described herein. As used herein, a therapeutic agent is considered to be “provided” to a patient if the patient is directly administered therapeutic agent or if the patient is administered a substance that is processed or metabolized in vivo so as to yield therapeutic agent endogenously. For example, a patient, such as a patient having a neuromuscular disorder described herein, may be provided a nucleic acid molecule encoding a therapeutic protein (e.g., MTM1) by direct administration of the nucleic acid molecule or by administration of a substance (e.g., viral vector or cell) that is processed in vivo so as to yield the desired nucleic acid molecule. As used herein, the terms “patient” and “subject” refer to an organism that receives treatment for a particular disease or condition as described herein (such as a neuromuscular disorder, e.g., XLMTM). Examples of subjects and patients include mammals, such as humans, receiving treatment for a disease or condition described herein. By a “reference” is meant any useful reference used to compare protein levels related to cholestasis, hyperbilirubinemia, or one or more symptoms thereof. The reference can be any sample, standard, standard curve, or level that is used for comparison purposes. The reference can be a normal reference sample or a reference standard or level. A “reference sample” can be, for example, a control, e.g., a predetermined negative control value such as a “normal control” or a prior sample taken from the same subject; a sample from a normal healthy subject, such as a normal cell or normal tissue; a sample (e.g., a cell or tissue) from a subject not having cholestasis, hyperbilirubinemia, or one or more symptoms thereof; a sample from a subject that is diagnosed with cholestasis, hyperbilirubinemia, or one or more symptoms thereof; a sample from a subject that has been treated for cholestasis, hyperbilirubinemia, or one or more symptoms thereof; or a sample of a purified protein (e.g., any described herein) at a known normal concentration. By “reference standard or level” is meant a value or number derived from a reference sample. A “normal control value” is a pre- determined value indicative of non-disease state, e.g., a value expected in a healthy control subject. Typically, a normal control value is expressed as a range (“between X and Y”), a high threshold (“no higher than X”), or a low threshold (“no lower than X”). A subject having a measured value within the normal control value for a particular biomarker is typically referred to as “within normal limits” for that biomarker. A normal reference standard or level can be a value or number derived from a normal subject not having cholestasis, hyperbilirubinemia, or one or more symptoms thereof. In preferred PATENT ATTORNEY DOCKET NO.51037-075WO2 embodiments, the reference sample, standard, or level is matched to the sample subject sample by at least one of the following criteria: age, weight, sex, disease stage, and overall health. A standard curve of levels of a purified protein, e.g., any described herein, within the normal reference range can also be used as a reference. As used herein, the term “term age” refers to the age of a patient (e.g., a newborn) born between 37 weeks of gestational age and 42 weeks of gestational age. For example, if the patient was born at 35 weeks of gestational age, the patient is at term age at 14 days old. As used herein, the term "transgene" refers to a recombinant nucleic acid (e.g., DNA or cDNA) encoding a gene product (e.g., a gene product described herein). The gene product may be an RNA, peptide, or protein. In addition to the coding region for the gene product, the transgene may include or be operably linked to one or more elements to facilitate or enhance expression, such as a promoter, enhancer(s), destabilizing domain(s), response element(s), reporter element(s), insulator element(s), polyadenylation signal(s), and/or other functional elements. Embodiments of the disclosure may utilize any known suitable promoter, enhancer(s), destabilizing domain(s), response element(s), reporter element(s), insulator element(s), polyadenylation signal(s), and/or other functional elements. As used herein, the terms “treat” and “treatment” refer to therapeutic treatment, in which the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the progression of a neuromuscular disorder, such as XLMTM, among others. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms (e.g., stiffness and/or joint contractures), diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. In the context of neuromuscular disorders, such as XLMTM, treatment of a patient may manifest in one or more detectable changes, such as an increase in the concentration of MTM1 protein or nucleic acids (e.g., DNA or RNA, such as mRNA) encoding MTM1, or an increase in MTM1 activity (e.g., by 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, or more. The concentration of MTM1 protein may be determined using protein detection assays known in the art, including ELISA assays described herein. The concentration of MTM1- encoding nucleic acids may be determined using nucleic acid detection assays (e.g., RNA Seq assays) described herein. Additionally, treatment of a patient suffering from a neuromuscular disorder, such as XLMTM, may manifest in improvements in a patient’s muscle function (e.g., skeletal muscle function) as well as improvements in muscle coordination. For example, manifestation of an improvement may include increasing diaphragm and/or respiratory muscle progression. As used herein, the terms “X-linked myotubular myopathy” and “XLMTM” refer to the genetically inherited neuromuscular disorder that is caused by mutations of the MTM1 gene and is characterized by symptoms including mild to profound muscle weakness, hypotonia (diminished muscle tone), feeding difficulties, and/or severe breathing complications. Human MTM1 has NCBI Gene ID NO 4534. An exemplary wild-type human MTM1 nucleic acid sequence is provided in NCBI PATENT ATTORNEY DOCKET NO.51037-075WO2 RefSeq Acc. No. NM_000252.3, and an exemplary wild-type myotubularin 1 amino acid sequence is provided in NCBI RefSeq Acc. No. NP_000243.1. As used herein, the term “vector” refers to a nucleic acid, e.g., DNA or RNA, that may function as a vehicle for the delivery of a gene of interest into a cell (e.g., a mammalian cell, such as a human cell), such as for purposes of replication and/or expression. Exemplary vectors useful in conjunction with the compositions and methods described herein are plasmids, DNA vectors, RNA vectors, virions, or other suitable replicon (e.g., viral vector). A variety of vectors have been developed for the delivery of polynucleotides encoding exogenous proteins into a prokaryotic or eukaryotic cell. Examples of such expression vectors are disclosed in, e.g., WO 1994/11026, the disclosure of which is incorporated herein by reference. Expression vectors described herein contain a polynucleotide sequence as well as, e.g., additional sequence elements used for the expression of proteins and/or the integration of these polynucleotide sequences into the genome of a mammalian cell. Certain vectors that can be used for the expression of transgenes described herein include plasmids that contain regulatory sequences, such as promoter and enhancer regions, which direct gene transcription. Other useful vectors for expression of transgenes contain polynucleotide sequences that enhance the rate of translation of these genes or improve the stability or nuclear export of the mRNA that results from gene transcription. These sequence elements include, e.g., 5’ and 3’ untranslated regions, an internal ribosomal entry site (IRES), and polyadenylation signal site in order to direct efficient transcription of the gene carried on the expression vector. The expression vectors described herein may also contain a polynucleotide encoding a marker for selection of cells that contain such a vector. Examples of a suitable marker include genes that encode resistance to antibiotics, such as ampicillin, chloramphenicol, kanamycin, or nourseothricin. Detailed Description The present disclosure provides compositions and methods that can be used for treating neuromuscular disorders, particularly X-linked myotubular myopathy (XLMTM). In accordance with the compositions and methods described herein, a patient (e.g., a human patient) having XLMTM may be administered a viral vector, such as an adeno-associated viral (AAV) vector, that contains a transgene encoding myotubularin 1 (MTM1). The AAV vector may be, for example, a pseudotyped AAV vector, such as an AAV vector containing AAV2 inverted terminal repeats packaged within capsid proteins from AAV8 (AAV2/8). In some embodiments, the MTM1 transgene is operably linked to a transcription regulatory element, such as a promoter that drives gene expression in a liver cell. This aspect of the disclosure is based, at least in part, on the discovery that expressing an MTM1 transgene in liver tissue in XLMTM patients may improve the safety of MTM1 gene therapy. In some embodiments, the promoter contains an LP1 promoter, apolipoprotein E (ApoE) promoter, and/or alpha-1-antitrypsin (A1AT) promoter (e.g., an LP1 promoter, ApoE promoter, and/or A1AT promoter described herein). In some embodiments, the MTM1 transgene is operably linked to a constitutive transcription regulatory element, such as a phosphoglycerate kinase (PGK) promoter, an elongation factor-1 alpha PATENT ATTORNEY DOCKET NO.51037-075WO2 (EF1alpha) promoter, a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) promote, a cytomegalovirus (CMV) promoter, or a chicken-β-actin (CBA) promoter. In some embodiments, the viral vector includes two MTM1 transgenes: one under the control of a promoter that is active in liver tissue, and another that is under the control of a promoter that is active in muscle tissue. These features of the disclosure are based, at least in part, on the discovery that simultaneously expressing an MTM1 transgene in a plurality of tissues (e.g., liver tissue and muscle tissue) may improve the safety of MTM1 gene therapy. Also described herein are methods of treating XLMTM in a patient in need thereof by administering to the patient two compositions: (i) a non-viral composition containing a nucleic acid encoding MTM1 operably linked to a promoter that is active (e.g., selectively active) in liver tissue, and (ii) a viral vector containing an MTM1 transgene operably linked to a muscle-specific promoter. Non-viral compositions useful in conjunction with this aspect of the disclosure include, without limitation, a liposome, vesicle, synthetic vesicle, exosome, synthetic exosome, dendrimer, and nanoparticle. In some embodiments, the nanoparticle is a lipid nanoparticle. Viral vectors useful in conjunction with this aspect of the disclosure include, without limitation, resamirigene bilparvovec. In some embodiments, the disclosure describes a method of reducing stiffness and/or joint contractures in a human patient diagnosed as having XLMTM by administration of one or more compositions described herein. In some embodiments, the disclosure describes a method of increasing diaphragm and/or respiratory muscle progression in a human patient diagnosed as having XLMTM by administration of one or more compositions described herein. In some embodiments, the disclosure describes a method of preventing cholestasis or hyperbilirubinemia in a human patient diagnosed as having XLMTM by administration of one or more compositions described herein. The sections that follow provide a description of therapeutic agents and parameters for assessing cholestasis, hyperbilirubinemia, or one or more symptoms thereof. The following sections also describe various transduction agents that may be used in conjunction with the compositions and methods of the disclosure. X-Linked Myotubular Myopathy XLMTM is a rare, life-threatening, congenital myopathy caused by a mutation in the MTM1 gene and is characterized in most patients by profound muscle weakness and hypotonia at birth, which results in severe respiratory insufficiency, inability to sit up, stand or walk, and early mortality. The myopathy associated with XLMTM impairs the development of motor skills such as sitting, standing, and walking. Affected infants may also have difficulties with feeding due to muscle weakness. Individuals with this condition often do not have the muscle strength to breathe on their own and must be supported with mechanical ventilation. Some affected individuals require mechanical ventilation only periodically, such as during sleep, while others require mechanical ventilation continuously. Patients having XLMTM may also have weakness in the muscles that PATENT ATTORNEY DOCKET NO.51037-075WO2 control eye movement (ophthalmoplegia), weakness in other muscles of the face, and absent reflexes (areflexia). In XLMTM, muscle weakness often disrupts normal bone development and can lead to fragile bones, an abnormal curvature of the spine (scoliosis), and joint deformities (contractures) of the hips and knees. Patients having XLMTM may have a large head with a narrow and elongated face and a high, arched roof of the mouth (palate). Patients may also have liver disease, recurrent ear and respiratory infections, or seizures. As a consequence of their severe breathing difficulties, patients having XLMTM usually survive only into early childhood; however, some patients with this condition have lived into adulthood. The compositions and methods of the disclosure provide the important medical benefit of being able to prolong the lifetimes of such patients by restoring functional MTM1 expression. Moreover, the compositions and methods described herein can be used to improve patients’ quality of life post- treatment (e.g., reducing stiffness and/or joint contractures or increasing diaphragm and/or respiratory muscle progression), as the disclosure provides a series of guidelines that can be used to determine a patient’s eligibility for being weaned off of mechanical ventilation. Methods of Treatment In some embodiments, the patient is a newborn (e.g., 0-4 months old), an infant (e.g., 0-5 months old), a toddler (e.g., 6-12 months old), a child aged 1–^3 years old, or a child aged 3–^5 years old at the time of administration of the viral vector. In some embodiments, the patient is a newborn (e.g., 0-4 months old) at the time of administration of the viral vector. For example, in some embodiments, the patient is a newborn that is about 0 to about 4 months old (e.g., 0 months old to about 4 months old, 1 month old to about 4 months old, 2 months old to about 4 months old, or 3 months old to about 4 months old). In some embodiments, the patient is 0 months old. In some embodiments, the patient is 1 month old. In some embodiments, the patient is 2 months old. In some embodiments, the patient is 3 months old. In some embodiments, the patient is 4 months old. In some embodiments, the patient is a newborn (e.g., less than about 4 months old) at the time of administration of the viral vector. For example, in some embodiments, the patient is a newborn that is less than about 4 months old. In some embodiments, the patient is less than about 4 months old. In some embodiments, the patient is less than about 3 months old. In some embodiments, the patient is less than about 2 months old. In some embodiments, the patient is less than about 1 month old. In some embodiments, the patient is an infant (e.g., 0-5 months old) at the time of administration of the viral vector. For example. in some embodiments, the patient is an infant that is about 0 months old to about 5 months old (e.g., 0 months old to about 5 months old, 1 month old to about 5 months old, 2 months old to about 5 months old, 3 months old to about 5 months old, or 4 months old to about 5 months old). In some embodiments, the patient is 0 months old. In some embodiments, the patient is 1 month old. In some embodiments, the patient is 2 months old. In some PATENT ATTORNEY DOCKET NO.51037-075WO2 embodiments, the patient is 3 months old. In some embodiments, the patient is 4 months old. In some embodiments, the patient is 3 months old. In some embodiments, the patient is 5 months old. In some embodiments, the patient is an infant (e.g., less than about 5 months old) at the time of administration of the viral vector. For example, in some embodiments, the patient is an infant that is less than about 5 months old. In some embodiments, the patient is less than about 5 months old. In some embodiments, the patient is less than about 4 months old. In some embodiments, the patient is less than about 3 months old. In some embodiments, the patient is less than about 2 months old. In some embodiments, the patient is less than about 1 month old. In some embodiments, the patient is a toddler (e.g., 6-12 months old) at the time of administration of the viral vector. For example, in some embodiments, the patient is an infant that is about 6 months old to about 12 months old (e.g., 6 months old to about 12 months old, 7 months old to about 12 months old, 8 months old to about 12 months old, 9 months old to about 12 months old, 10 months old to about 12 months old, or 11 months old to about 12 months old). In some embodiments, the patient is 6 months old. In some embodiments, the patient is 7 months old. In some embodiments, the patient is 8 months old. In some embodiments, the patient is 9 months old. In some embodiments, the patient is 10 months old. In some embodiments, the patient is 11 months old. In some embodiments, the patient is 12 months old. In some embodiments, the patient is a toddler (e.g., less than about 12 months old) at the time of administration of the viral vector. For example, in some embodiments, the patient is a toddler that is less than about 12 months old. In some embodiments, the patient is less than about 12 months old. In some embodiments, the patient is less than about 11 months old. In some embodiments, the patient is less than about 10 months old. In some embodiments, the patient is less than about 9 months old. In some embodiments, the patient is less than about 8 months old. In some embodiments, the patient is less than about 7 months old. In some embodiments, the patient is less than about 6 months old. In some embodiments, the patient is less than about 5 months old. In some embodiments, the patient is less than about 4 months old. In some embodiments, the patient is less than about 3 months old. In some embodiments, the patient is less than about 2 months old. In some embodiments, the patient is less than about 1 month old. In some embodiments, the patient is a child aged 1- ^3 years old at the time of administration of the viral vector. For example, in some embodiments, the patient is a child that is about 1 year old to about 3 years old (e.g., 1 year old to about 3 years old or 2 years old to about 3 years old). In some embodiments, the patient is 1 year old. In some embodiments, the patient is 2 years old. In some embodiments, the patient is 3 years old. In some embodiments, the patient is a child (e.g., less than about 3 years old) at the time of administration of the viral vector. For example, in some embodiments, the patient is a child that is less than about 3 years old. In some embodiments, the patient is less than about 3 years old. In some embodiments, the patient is less than about 2 years old. In some embodiments, the patient is less than about 1 year old. In some embodiments, the patient is less than about 12 months old. In some embodiments, the patient is less than about 11 months old. In some embodiments, the patient is less than about 10 months old. In some embodiments, the patient is less than about 9 months old. In PATENT ATTORNEY DOCKET NO.51037-075WO2 some embodiments, the patient is less than about 8 months old. In some embodiments, the patient is less than about 7 months old. In some embodiments, the patient is less than about 6 months old. In some embodiments, the patient is less than about 5 months old. In some embodiments, the patient is less than about 4 months old. In some embodiments, the patient is less than about 3 months old. In some embodiments, the patient is less than about 2 months old. In some embodiments, the patient is less than about 1 month old. In some embodiments, the patient is a child aged 3-5 years old at the time of administration of the viral vector. For example, in some embodiments, the patient is a child that is about 3 years old to about 5 years old (e.g., 3 years old to about 5 years old or 4 years old to about 5 years old). In some embodiments, the patient is 3 years old. In some embodiments, the patient is 4 years old. In some embodiments, the patient is 5 years old. In some embodiments, the patient is a child (e.g., less than about 5 years old) at the time of administration of the viral vector. For example, in some embodiments, the patient is a child that is less than about 5 years old. In some embodiments, the patient is less than about 5 years old. In some embodiments, the patient is less than about 4 years old. In some embodiments, the patient is less than about 3 years old. In some embodiments, the patient is less than about 2 years old. In some embodiments, the patient is less than about 1 year old. In some embodiments, the patient is less than about 12 months old. In some embodiments, the patient is less than about 11 months old. In some embodiments, the patient is less than about 10 months old. In some embodiments, the patient is less than about 9 months old. In some embodiments, the patient is less than about 8 months old. In some embodiments, the patient is less than about 7 months old. In some embodiments, the patient is less than about 6 months old. In some embodiments, the patient is less than about 5 months old. In some embodiments, the patient is less than about 4 months old. In some embodiments, the patient is less than about 3 months old. In some embodiments, the patient is less than about 2 months old. In some embodiments, the patient is less than about 1 month old. In some embodiments, the patient is from about 1 month old to about 5 years old (e.g., about 1 month old to about 5 years old, about 2 months old to about 5 years old, about 3 months old to about 5 years old, about 4 months old to about 5 years old, about 5 months old to about 5 years old, about 6 months old to about 5 years old, about 1 year old to about 5 years old, about 2 years old to about 5 years old, about 3 years old to about 5 years old, or about 4 years old to about 5 years old) at the time of administration of the viral vector. In some embodiments, the patient was born at greater than or equal to 35 weeks of gestational age (e.g., 35 weeks of gestational age, 36 weeks of gestational age, 37 weeks of gestational age, 38 weeks of gestational age, 39 weeks of gestational age, 40 weeks of gestational age, 41 weeks of gestational age, and 42 weeks of gestational age) and is between adjusted term age (e.g., 37 weeks of gestational age or greater) to about 5 years old at the time of administration of the viral vector. For example, if the patient was born at 35 weeks of gestational age, the patient is at term age at 14 days old. In some embodiments, the patient was born at 35 weeks of gestational age and is between adjusted term age to about 5 years old (e.g., 14 days old to about 5 years old, 15 days old to about 5 PATENT ATTORNEY DOCKET NO.51037-075WO2 years old, 16 days old to about 5 years old, 17 days old to about 5 years old, 18 days old to about 5 years old, 19 days old to about 5 years old, 20 days old to about 5 years old, 25 days old to about 5 years old, one month old to about 5 years old, two months old to about 5 years old, 3 months old to about 5 years old, 4 months old to about 5 years old, 5 months old to about 5 years old, 6 months old to about 5 years old, 1 year old to about 5 years old, 2 years old to about 5 years old, 3 years old to about 5 years old, and 4 years old to about 5 years old) at the time of administration of the viral vector. In some embodiments, the patient was born at 36 weeks of gestational age and is between adjusted term age to about 5 years old (e.g., 7 days old to about 5 years old, 8 days old to about 5 years old, 9 days old to about 5 years old, 10 days old to about 5 years old, 11 days old to about 5 years old, 12 days old to about 5 years old, 13 days old to about 5 years old, 14 days old to about 5 years old, 15 days old to about 5 years old, 16 days old to about 5 years old, 17 days old to about 5 years old, 18 days old to about 5 years old, 19 days old to about 5 years old, 20 days old to about 5 years old, 25 days old to about 5 years old, one month old to about 5 years old, two months old to about 5 years old, 3 months old to about 5 years old, 4 months old to about 5 years old, 5 months old to about 5 years old, 6 months old to about 5 years old, 1 year old to about 5 years old, 2 years old to about 5 years old, 3 years old to about 5 years old, and 4 years old to about 5 years old) at the time of administration of the viral vector. In some embodiments, the patient was born at 37 weeks of gestational age and is between adjusted term age to about 5 years old (e.g., 1 day old to about 5 years old, 2 days old to about 5 years old, 3 days old to about 5 years old, 4 days old to about 5 years old, 5 days old to about 5 years old, 6 days old to about 5 years old, 7 days old to about 5 years old, 8 days old to about 5 years old, 9 days old to about 5 years old, 10 days old to about 5 years old, 11 days old to about 5 years old, 12 days old to about 5 years old, 13 days old to about 5 years old, 14 days old to about 5 years old, 15 days old to about 5 years old, 16 days old to about 5 years old, 17 days old to about 5 years old, 18 days old to about 5 years old, 19 days old to about 5 years old, 20 days old to about 5 years old, 25 days old to about 5 years old, one month old to about 5 years old, two months old to about 5 years old, 3 months old to about 5 years old, 4 months old to about 5 years old, 5 months old to about 5 years old, 6 months old to about 5 years old, 1 year old to about 5 years old, 2 years old to about 5 years old, 3 years old to about 5 years old, and 4 years old to about 5 years old) at the time of administration of the viral vector. In some embodiments, the patient is male. In some embodiments, the patient is female. Cholestasis and Hyperbilirubinemia Cholestasis is any condition in which the flow of bile acid from the liver is slowed or blocked, while hyperbilirubinemia is a condition in which there is an accumulation of bilirubin in the blood and serum bile acids appear to remain normal. By contrast, cholestatic syndromes are characterized by marked bile acidemia with normal to slightly elevated bilirubin levels. PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, the patient is monitored for the development of cholestasis. In some embodiments, the patient is monitored for the development of hyperbilirubinemia. In some embodiments, the patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof. In some embodiments, the patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof by evaluating a parameter in a blood sample obtained from the patient, wherein a finding that the parameter is above a reference level identifies the patient as having cholestasis, hyperbilirubinemia, or one or more symptoms thereof. In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof when the patient exhibits one or more parameters (e.g., total bile acids level, gamma-glutamyl transferase (GGT) level, alkaline phosphatase (ASP) level, aspartate aminotransferase (AST) level, and/or alanine aminotransferase (ALT) level), as measured in a serum bile acid test and/or blood test (e.g., a liver function test (LFT)), that is greater than or less than the age-adjusted norm. In some embodiments, a patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof when the patient exhibits a bilirubin level, as measured in a blood test (e.g., a bilirubin test), that is greater than the norm. In some embodiments, the patient does not have a history of cholestasis or hyperbilirubinemia. In some embodiments, the patient does not have a history of any underlying liver disease. Vectors for Delivery of Exogenous Nucleic Acids to Target Cells Viral Vectors for Nucleic Acid Delivery Recombinant viral genomes provide a rich source of vectors that can be used for the efficient delivery of a gene of interest (e.g., a transgene encoding MTM1) into the genome of a target cell (e.g., a mammalian cell, such as a human cell). Recombinant viral genomes are particularly useful vectors for gene delivery because they deliver the gene of interest to the nucleus of the target cells. For select viruses, polynucleotides contained within such genomes may be incorporated into the genome of a target cell by generalized or specialized transduction. These processes occur as part of the natural viral replication cycle, and do not require added proteins or reagents. Examples of recombinant viral vectors used to deliver genes of interest include AAV, adenovirus (e.g., Ad5, Ad26, Ad34, Ad35, and Ad48), parvovirus (e.g., adeno-associated viruses), coronavirus, negative strand RNA viruses such as orthomyxovirus (e.g., influenza virus), rhabdovirus (e.g., rabies and vesicular stomatitis virus), paramyxovirus (e.g., measles and Sendai), positive strand RNA viruses, such as picornavirus and alphavirus, and double stranded DNA viruses including adenovirus, herpesvirus (e.g., Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomegalovirus), and poxvirus (e.g., vaccinia, modified vaccinia Ankara (MVA), fowlpox and canarypox). Other viruses useful for delivering polynucleotides encoding antibody light and heavy chains or antibody fragments of the invention include Norwalk virus, togavirus, flavivirus, reoviruses, papovavirus, hepadnavirus, and hepatitis virus, PATENT ATTORNEY DOCKET NO.51037-075WO2 for example. Examples of retroviruses include: avian leukosis- sarcoma, mammalian C-type, B-type viruses, D-type viruses, HTLV-BLV group, lentivirus, spumavirus (Coffin, J. M., Retroviridae: The viruses and their replication, In Fundamental Virology, Third Edition, B. N. Fields, et al., Eds., Lippincott-Raven Publishers, Philadelphia, 1996). Other examples include murine leukemia viruses, murine sarcoma viruses, mouse mammary tumor virus, bovine leukemia virus, feline leukemia virus, feline sarcoma virus, avian leukemia virus, human T-cell leukemia virus, baboon endogenous virus, Gibbon ape leukemia virus, Mason Pfizer monkey virus, simian immunodeficiency virus, simian sarcoma virus, Rous sarcoma virus and lentiviruses. Other examples of vectors are described, for example, in US Patent No.5,801,030, the disclosure of which is incorporated herein by reference as it pertains to viral vectors for use in gene therapy. AAV Vectors for Nucleic Acid Delivery In some embodiments, nucleic acids of the compositions and methods described herein are incorporated into recombinant AAV (rAAV) vectors and/or virions in order to facilitate their introduction into a cell. rAAV vectors useful in the invention are recombinant nucleic acid constructs that include (1) a transgene to be expressed (e.g., a polynucleotide encoding a MTM1 protein) and (2) viral nucleic acids that facilitate integration and expression of the heterologous genes. The viral nucleic acids may include those sequences of AAV that are required in cis for replication and packaging (e.g., functional inverted terminal repeats (ITRs)) of the DNA into a virion. In typical applications, the transgene encodes MTM1, which is useful for correcting a MTM1 mutation in patients suffering from neuromuscular disorders, such as XLMTM. Such rAAV vectors may also contain marker or reporter genes. Useful rAAV vectors have one or more of the AAV wild type genes deleted in whole or in part but retain functional flanking ITR sequences. The AAV ITRs may be of any serotype (e.g., derived from serotype 2) suitable for a particular application. Methods for using rAAV vectors are described, for example, in Tal et al., J. Biomed. Sci.7:279-291 (2000), and Monahan and Samulski, Gene Delivery 7:24-30 (2000), the disclosures of each of which are incorporated herein by reference as they pertain to AAV vectors for gene delivery. The nucleic acids and vectors described herein can be incorporated into a rAAV virion in order to facilitate introduction of the nucleic acid or vector into a cell. The capsid proteins of AAV compose the exterior, non-nucleic acid portion of the virion and are encoded by the AAV cap gene. The cap gene encodes three viral coat proteins, VP1, VP2 and VP3, which are required for virion assembly. The construction of rAAV virions has been described, for example, in US Patent Nos.5,173,414; 5,139,941; 5,863,541; 5,869,305; 6,057,152; and 6,376,237; as well as in Rabinowitz et al., J. Virol. 76:791-801 (2002) and Bowles et al., J. Virol.77:423-432 (2003), the disclosures of each of which are incorporated herein by reference as they pertain to AAV vectors for gene delivery. rAAV virions useful in conjunction with the compositions and methods described herein include PATENT ATTORNEY DOCKET NO.51037-075WO2 those derived from a variety of AAV serotypes including AAV 1, 2, 3, 4, 5, 6, 7, 8 and 9. For targeting muscle cells, rAAV virions that include at least one serotype 1 capsid protein may be particularly useful. rAAV virions that include at least one serotype 6 capsid protein may also be particularly useful, as serotype 6 capsid proteins are structurally similar to serotype 1 capsid proteins, and thus are expected to also result in high expression of MTM1 in muscle cells. rAAV serotype 9 has also been found to be an efficient transducer of muscle cells. Construction and use of AAV vectors and AAV proteins of different serotypes are described, for example, in Chao et al., Mol. Ther.2:619-623 (2000); Davidson et al., Proc. Natl. Acad. Sci. USA 97:3428-3432 (2000); Xiao et al., J. Virol.72:2224-2232 (1998); Halbert et al., J. Virol.74:1524-1532 (2000); Halbert et al., J. Virol.75:6615-6624 (2001); and Auricchio et al., Hum. Molec. Genet.10:3075-3081 (2001), the disclosures of each of which are incorporated herein by reference as they pertain to AAV vectors for gene delivery. Also useful in conjunction with the compositions and methods described herein are pseudotyped rAAV vectors. Pseudotyped vectors include AAV vectors of a given serotype (e.g., AAV9) pseudotyped with a capsid gene derived from a serotype other than the given serotype (e.g., AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, etc.). For example, a representative pseudotyped vector is an AAV8 vector encoding a therapeutic protein pseudotyped with a capsid gene derived from AAV serotype 2. Techniques involving the construction and use of pseudotyped rAAV virions are known in the art and are described, for example, in Duan et al., J. Virol.75:7662-7671 (2001); Halbert et al., J. Virol. 74:1524-1532 (2000); Zolotukhin et al., Methods, 28:158-167 (2002); and Auricchio et al., Hum. Molec. Genet., 10:3075-3081 (2001). AAV virions that have mutations within the virion capsid may be used to infect particular cell types more effectively than non-mutated capsid virions. For example, suitable AAV mutants may have ligand insertion mutations for the facilitation of targeting AAV to specific cell types. The construction and characterization of AAV capsid mutants including insertion mutants, alanine screening mutants, and epitope tag mutants are described in Wu et al., J. Virol.74:8635-45 (2000). Other rAAV virions that can be used in methods of the invention include those capsid hybrids that are generated by molecular breeding of viruses as well as by exon shuffling. See, e.g., Soong et al., Nat. Genet., 25:436-439 (2000) and Kolman and Stemmer, Nat. Biotechnol.19:423-428 (2001). Resamirigene Bilparvovec As described herein, a pseudotyped AAV vector including a nucleic acid sequence encoding a MTM1 gene operably linked to a desmin promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8) as well as the other genetic components listed in Table 1, refers to the compound known by the international proprietary name (INN) of resamirigene bilparvovec. Resamirigene bilparvovec is a non-replicating recombinant AAV8 vector expressing a non- codon-optimized human MTM1 cDNA under the control of the muscle-specific human desmin PATENT ATTORNEY DOCKET NO.51037-075WO2 promoter. The MTM1 expression cassette was built by cloning a synthetic DNA sequence complementary to the coding portion (nucleotides 43-1864) of the wild-type human MTM1 transcript (NCBI Ref. Seq NM_000252.3) downstream of the 1.05-kb human desmin enhancer/promoter region. The second intron and polyadenylation sequence of the human β-globin gene (HBB) were inserted upstream and downstream respectively of the MTM1 synthetic cDNA to mediate RNA processing. The expression cassette was flanked by AAV serotype-2 (AAV2) inverted terminal repeats (ITRs). The vector was produced in an AAV8 capsid by two-plasmid transfection in HEK293 cells in suspension culture in bioreactors with a full GMP process. In some embodiments, a method of treating a disorder (e.g., XLMTM) or alleviating one or more symptoms (e.g., stiffness and/or join contractures or need for diaphragm and/or respiratory muscle progression) of a disorder (e.g., XLMTM) in a human patient in need thereof, includes administering to the patient a therapeutically effective amount of resamirigene bilparvovec during a treatment period. In some embodiments, a method of weaning a human patient off of mechanical ventilation, includes a patient that has previously been administered a therapeutically effective amount of resamirigene bilparvovec. The components of resamirigene bilparvovec are shown in Table 1, below:
PATENT ATTORNEY DOCKET NO.51037-075WO2 Table 1. Resamirigene Bilparvovec nucleic acid sequence (SEQ ID NO: 1)
Figure imgf000036_0003
As described herein, resamirigene bilparvovec refers to the AAV vector having the nucleic acid sequence of SEQ ID NO: 1, shown below: TCGCGCGTTT CGGTGATGAC GGTGAAAACC TCTGACACAT GCAGCTCCCG 50 GACGTCATTG TCGATCCTGC AGGCGTACGG
Figure imgf000036_0001
CATAGCTAAC 100 AAGGTGTGGA AAAAGAATTA GTGGTTAGAG AGTGAGCTAT TCGTTGAAAC 150 AATTGCGTTC TTGAAACAAT TCTTGCTGGT AAAATGTCAC ATTTTATGTG 200 ACTACAGGTG GAGGATTGGC ACATAACCTA ACCAGTGGGG GAAACAATTG 250 ACCTCTGGAT TTGTCCAAGT GTATAGTAGC ATTTGCCCAA TCGAATGGTC 300 CTGGTAAGGT GTTAATGTTG ACTAGAACCA AAGGTGGAAG TTGCAGGGAA 350 ACTGGTTTAG TACAAGGGTG GACACCAGGC AGTCATCCAG AGGCCCATTA 400 AAGGCCTTGG AATGTTTTTC CGAAGGAGAA TCACTCCCTC TTCTCTCGCT 450 TAAAGTTTTA GGGGATTCAT GAACAGCTGC TGTGGGATAG TTTCATGTCC 500 CTAGCAATTG TAAAGCAACT GAGGGTGGCT TAAACCAGTT TTAGCTTTAG 550 GGTTAGGGTT ACTGGACTAA AATTTGAGAA ATTCATAAAT CTTAAGGAAA 600 TCCATTGTGA GTTTTCATTA TGAGTGCATC CAATGTATAA TTTCCATGAC 650 CCTCCCATGC AAGTGAGCAT GTGAATCAGG AAACGTTACA AGAACCCAAC 700 AAACTCAACC ACTACTAGAC AGGCGATCAC TTCCAGTTAG TATGCAACTT 750 TCTGTGTAAT TTTAGTTACC
Figure imgf000036_0002
GGATGACCTT AGTGTAAGGA 800 PATENT ATTORNEY DOCKET NO.51037-075WO2 AAAAATACCT TGAATAGTGT TAAAGATGTA CACTTGGTGT CAGGCATTGT 850 AACATTGATA AATCTGTGTA AGGTGCTTTT TGAAAACTTC AAAGCTGCAT 900 CAAGTCAAGT ACAAGAAAGG CCATGGCTGC TAAAGCTGTT GAAGATGTGG 950 GATGGAACTG GGTCACATTG GTGTTAACAG CGTTGTGCAG AGCCGGCAGG 1000 ATCTTGGTGT GAGCGAACAT TAGTCTATTT AATAAAGCTG TGTGAATGTT 1050 GTAGAGGTGA GGATGCTCAC TTGAAAACTC ACTGAAGAAC ACTTGGCCCC 1100 TTGAACTAAA GTGCTTCTAT CAAGTTCAGT GAGAAATTCC GAATTACAAG 1150 CATAGGTACT AGAAAAGTTT TGAAAAGCAG TATAGAGCAA CATAAGCACA 1200
Figure imgf000037_0001
CTTGTTGCAT AGGTTGGTCT CAAACTCCTG GCCTCAAGTG ATTCTCCTGC 1650 CTCTGCCTCC CAAAGTGCTG AGATTACAGG TGTGAGGCAC CATGCCAGGT 1700 CTCTTACTGT TTGTAATTAA ATACATACAC ATTTTGTGTG TTTGTGTGCA 1750 CCTTTATAAA GTCAAAGGTG ATAGTAACCC ATTTAAGTTC CTACTCAATT 1800 TTACTTTCCA GGGATAACTA ACTACTTTTT CTTTTTGAGA TGGAGTCTCG 1850 CTGTGTAGCC CAGGCTGGAG TGCAGTGGCA CCATCTCGGC TCACTGCAAG 1900 CTCCTCCTCC CTGGTTCACG CTATTCTCCT GCCTCAGCCT CCCCAACAAC 1950 TAGGACTACA GGCTCACCTC GCCATACCTG GCTAATTTTT TGTATTTTTA 2000 GTAGAGACAG GGTTTCACTG TGTTAGCCAG GATGGTCTCG ATCTCCTGAC 2050 CTTGTGATCC GCCTGCCTCT GCCTCCCAAA GTGCTGGGAT TACAGGCATG 2100 AGCAACCTCA CCCAGCTGGG ATAACTACTT TTTACAGGTT GATATTCTTT 2150 TGGACTTTTC CCCTGTGTAA AAATATACTA TATTTGTTAT GTACATATTA 2200 TGTACATACA GACACAAATT GGACCATTCT CAGTATAATG ATTCTCAGGT 2250 TTTTTGAGGT GGGGAACTAG ATAATTATGG ACATCTTTCC 2300 ATACTAGCAT ATCAATATCT ACCTCATTCT TTTTAATATT TTTGCTAGTA 2350 TTCCATTGTA TGAATGTCCT ATGATTTACT TAACCTGTCC ATCAATATTT 2400 GTTTCCAGGT TTTTGCTATT ATAATGCTGC TGCAAAGTAC ATCCTCACAC 2450 ATCTTTATTT TGTCTATTCA TATTTCTGTA AGATAGGTTA CTAAAGTTGG 2500 AACTGCCAAA TTAACACTAT CATACTATTT TGTTTTTTAA TTTTAATTTT 2550 TAAAATGTGC AATTTCAAGA GGAGAAACTT GAACACAAGG 2600 AGCAAAATCT ATTTTTATAA CATCCTATTA AAAGCTTGCT TTACATAAAG 2650 ATTTTGAAAG AATAGCATAA ATACAAGATT TCTATTTTAA TTGGATTCTT 2700 AGGGCTAATA AAATAATCAG CCTTAGCACT TATTTATTTA TTTTTTTTGA 2750 GAGGGAGTCT CGCTCTGTTG TCCATGCTGG AGTGCAGTGG CGTGATCTCG 2800 GCTCACTGCA AGCTCCACCT CATGAGTTCA CACCATTCTC CTGCCTCAGT 2850 PATENT ATTORNEY DOCKET NO.51037-075WO2 CTCCCGAGTA GCTGGGACTC CAGGCGCCCT CTACAAAGCC CGTCTAATTT 2900 TTTTTGTATT TTTAGTAGAG ACAGGGTTTC ACTGTGTTAG CCAGGATGGT 2950 CTTGATCTCC TGACCTTGTG ATCTGCCCGC CTCGGCCTCC CAAAGTGCTG 3000 GGATTATAGG CTTGAGCCAC TGCTCCCGGC CAGCACTTAT TTTTATAATT 3050 CTTCATGATT ACTGTGTTAC TGTCCCATGG GCCGCCAGGG CCAGCTAGGT 3100 TGGCCACTCC CTCTCTGCGC GCTCGCTCGC TCACTGAGGC CGGGCGACCA 3150 AAGGTCGCCC GACGCCCGGG CTTTGCCCGG GCGGCCTCAG TGAGCGAGCG 3200 AGCGCGCAGA GAGGGAGTGG CCAACTCCAT CACTAGGGGT TCCTCCTAGC 3250 ACGCGCTACC CCCTGCCCCC CACAGCTCCT CTCCTGTGCC TTGTTTCCCA 3300 GCCATGCGTT CTCCTCTATA AATACCCGCT CTGGTATTTG GGGTTGGCAG 3350 CTGTTGCTGC CAGGGAGATG GTTGGGTTGA CATGCGGCTC CTGACAAAAC 3400 ACAAACCCCT GGTGTGTGTG GGCGTGGGTG GTGTGAGTAG GGGGATGAAT 3450 CAGGGAGGGG GCGGGGGACC CAGGGGGCAG GAGCCACACA AAGTCTGTGC 3500 GGGGGTGGGA GCGCACATAG CAATTGGAAA CTGAAAGCTT ATCAGACCCT 3550 TTCTGGAAAT CAGCCCACTG TTTATAAACT TGAGGCCCCA CCCTCGACAG 3600 TACCGGGGAG GAAGAGGGCC TGCACTAGTC CAGAGGGAAA CTGAGGCTCA 3650 GGGCCAGCTC GCCCATAGAC ATACATGGCA GGCAGGCTTT GGCCAGGATC 3700 CCTCCGCCTG CCAGGCGTCT CCCTGCCCTC CCTTCCTGCC TAGAGACCCC 3750 CACCCTCAAG CCTGGCTGGT CTTTGCCTGA GACCCAAACC TCTTCGACTT 3800 CAAGAGAATA TTTAGGAACA AGGTGGTTTA GGGCCTTTCC TGGGAACAGG 3850 CCTTGACCCT TTAAGAAATG ACCCAAAGTC TCTCCTTGAC CAAAAAGGGG 3900 ACCCTCAAAC TAAAGGGAAG CCTCTCTTCT GCTGTCTCCC CTGACCCCAC 3950 TCCCCCCCAC CCCAGGACGA GGAGATAACC AGGGCTGAAA GAGGCCCGCC 4000 TGGGGGCTGC AGACATGCTT GCTGCCTGCC CTGGCGAAGG ATTGGTAGGC 4050 TTGCCCGTCA CAGGACCCCC GCTGGCTGAC TCAGGGGCGC AGGCCTCTTG 4100 CGGGGGAGCT GGCCTCCCCG CCCCCACGGC CACGGGCCGC CCTTTCCTGG 4150 CAGGACAGCG GGATCTTGCA GCTGTCAGGG GAGGGGAGGC GGGGGCTGAT 4200 GTCAGGAGGG ATACAAATAG TGCCGACGGC TGGGGGCCCT GTCTCCCCTC 4250 GCCGCATCCA CTCTCCGGCC GGCCGCCTGC CCGCCGCCTC CTCCGTGCGC 4300 CCGCCAGCCT CGCCCGGACT CTAGAGGATC CAGATCTAAG CTTCTCTGGT 4350 CACCGATCCT GAGAACTTCA GGGTGAGTCT ATGGGACCCT TGATGTTTTC 4400 TTTCCCCTTC TTTTCTATGG TTAAGTTCAT GTCATAGGAA GGGGAGAAGT 4450 AACAGGGTAC ACATATTGAC CAAATCAGGG TAATTTTGCA TTTGTAATTT 4500 TAAAAAATGC TTTCTTCTTT TAATATACTT TTTTGTTTAT CTTATTTCTA 4550 ATACTTTCCC TAATCTCTTT CTTTCAGGGC AATAATGATA CAATGTATCA 4600 TGCCTCTTTG CACCATTCTA AAGAATAACA GTGATAATTT CTGGGTTAAG 4650 GCAATAGCAA TATTTCTGCA TATAAATATT TCTGCATATA AATTGTAACT 4700 GATGTAAGAG GTTTCATATT GCTAATAGCA GCTACAATCC AGCTACCATT 4750 CTGCTTTTAT TTTATGGTTG GGATAAGGCT GGATTATTCT GAGTCCAAGC 4800 TAGGCCCTTT TGCTAATCAT GTTCATACCT CTTATCTTCC TCCCACAGCT 4850 CCTGGGCAAC GTGCTGGTCT GTGTGCTGGC CCATCACTTT GGCAAAGAAT 4900 PATENT ATTORNEY DOCKET NO.51037-075WO2 TCCGCGGGCG GCCGCAAGTT TCCAGGATGG CTTCTGCATC AACTTCTAAA 4950 TATAATTCAC ACTCCTTGGA GAATGAGTCT ATTAAGAGGA CGTCTCGAGA 5000 TGGAGTCAAT CGAGATCTCA CTGAGGCTGT TCCTCGACTT CCAGGAGAAA 5050 CACTAATCAC TGACAAAGAA GTTATTTACA TATGTCCTTT CAATGGCCCC 5100 ATTAAGGGAA GAGTTTACAT CACAAATTAT CGTCTTTATT TAAGAAGTTT 5150 GGAAACGGAT TCTTCTCTAA TACTTGATGT TCCTCTGGGT GTGATCTCGA 5200 GAATTGAAAA AATGGGAGGC GCGACAAGTA GAGGAGAAAA TTCCTATGGT 5250 CTAGATATTA CTTGTAAAGA CATGAGAAAC CTGAGGTTCG CTTTGAAACA 5300 GGAAGGCCAC AGCAGAAGAG ATATGTTTGA GATCCTCACG AGATACGCGT 5350 TTCCCCTGGC TCACAGTCTG CCATTATTTG CATTTTTAAA TGAAGAAAAG 5400 TTTAACGTGG ATGGATGGAC AGTTTACAAT CCAGTGGAAG AATACAGGAG 5450 GCAGGGCTTG CCCAATCACC ATTGGAGAAT AACTTTTATT AATAAGTGCT 5500 ATGAGCTCTG TGACACTTAC CCTGCTCTTT TGGTGGTTCC GTATCGTGCC 5550 TCAGATGATG ACCTCCGGAG AGTTGCAACT TTTAGGTCCC GAAATCGAAT 5600 TCCAGTGCTG TCATGGATTC ATCCAGAAAA TAAGACGGTC ATTGTGCGTT 5650 GCAGTCAGCC TCTTGTCGGT ATGAGTGGGA AACGAAATAA AGATGATGAG 5700 AAATATCTCG ATGTTATCAG GGAGACTAAT AAACAAATTT CTAAACTCAC 5750 CATTTATGAT GCAAGACCCA GCGTAAATGC AGTGGCCAAC AAGGCAACAG 5800 GAGGAGGATA TGAAAGTGAT GATGCATATC ATAACGCCGA ACTTTTCTTC 5850 TTAGACATTC ATAATATTCA TGTTATGCGG GAATCTTTAA AAAAAGTGAA 5900 GGACATTGTT TATCCTAATG TAGAAGAATC TCATTGGTTG TCCAGTTTGG 5950 AGTCTACTCA TTGGTTAGAA CATATCAAGC TCGTTTTGAC AGGAGCCATT 6000 CAAGTAGCAG ACAAAGTTTC TTCAGGGAAG AGTTCAGTGC TTGTGCATTG 6050 CAGTGACGGA TGGGACAGGA CTGCTCAGCT GACATCCTTG GCCATGCTGA 6100 TGTTGGATAG CTTCTATAGG AGCATTGAAG GGTTCGAAAT ACTGGTACAA 6150 AAAGAATGGA TAAGTTTTGG ACATAAATTT GCATCTCGAA TAGGTCATGG 6200 TGATAAAAAC CACACCGATG CTGACCGTTC TCCTATTTTT CTCCAGTTTA 6250 TTGATTGTGT GTGGCAAATG TCAAAACAGT TCCCTACAGC TTTTGAATTC 6300 AATGAACAAT TTTTGATTAT AATTTTGGAT CATCTGTATA GTTGCCGATT 6350 TGGTACTTTC TTATTCAACT GTGAATCTGC TCGAGAAAGA CAGAAGGTTA 6400 CAGAAAGGAC TGTTTCTTTA TGGTCACTGA TAAACAGTAA TAAAGAAAAA 6450 TTCAAAAACC CCTTCTATAC TAAAGAAATC AATCGAGTTT TATATCCAGT 6500 TGCCAGTATG CGTCACTTGG AACTCTGGGT GAATTACTAC ATTAGATGGA 6550 ACCCCAGGAT CAAGCAACAA CAGCCGAATC CAGTGGAGCA GCGTTACATG 6600 GAGCTCTTAG CCTTACGCGA CGAATACATA AAGCGGCTTG AGGAACTGCA 6650 GCTCGCCAAC TCTGCCAAGC TTTCTGATCC CCCAACTTCA CCTTCCAGTC 6700 CTTCGCAAAT GATGCCCCAT GTGCAAACTC ACTTCTGACC GGTCCGAGGG 6750 CCCAGATCTA ATTCACCCCA CCAGTGCAGG CTGCCTATCA GAAAGTGGTG 6800 GCTGGTGTGG CTAATGCCCT GGCCCACAAG TATCACTAAG CTCGCTTTCT 6850 TGCTGTCCAA TTTCTATTAA AGGTTCCTTT GTTCCCTAAG TCCAACTACT 6900 AAACTGGGGG ATATTATGAA GGGCCTTGAG CATCTGGATT CTGCCTAATA 6950 PATENT ATTORNEY DOCKET NO.51037-075WO2 AAAAACATTT ATTTTCATTG CAATGATGTA TTTAAATTAT TTCTGAATAT 7000 AGGGAATGTG GGAGGTCAGT GCATTTAAAA CATAAAGAAA 7050 TGAAGAGCTA GTTCAAACCT TGGGAAAATA CACTATATCT TAAACTCCAT 7100 GAAAGAAGGT GAGGCTGCAA ACAGCTAATG CACATTGGCA ACAGCCCCTG 7150 ATGCCTATGC CTTATTCATC CCTCAGAAAA GGATTCAAGT AGAGGCTTGA 7200 TTTGGAGGTT AAAGTTTTGC TATGCTGTAT TTTACATTAC TTATTGTTTT 7250 AGCTGTCCTC ATGAATGTCT TTTCACTACC CATTTGCTTA TCCTGCATCT 7300 CTCAGCCTTG ACTCCACTCA GTTCTCTTGC TTAGAGATAC CACCTTTCCC 7350 CTGAAGTGTT CCTTCCATGT TTTACGGCGA GATGGTTTCT CCTCGCCTGG 7400 CCACTCAGCC TTAGTTGTCT CTGTTGTCTT ATAGAGGTCT ACTTGAAGAA 7450 GGAAAAACAG GGGGCATGGT TTGACTGTCC TGTGAGCCCT TCTTCCCTGC 7500 CTCCCCCACT CACAGTGACC GGCCGCTCTA GGAGGAACCC CTAGTGATGG 7550 AGTTGGCCAC TCCCTCTCTG CGCGCTCGCT CGCTCACTGA GGCCGGGCGA 7600 CCAAAGGTCG CCCGACGCCC GGGCTTTGCC CGGGCGGCCT CAGTGAGCGA 7650 GCGAGCGCGC AGAGAGGGAG TGGCCAACCT AGAGGCCGCC AGGGCCATAT 7700 TTCTCAATTT TTAAATTTTT
Figure imgf000040_0001
AATCCTTAAT GTGCATATTT 7750 TTGAATTGTT AATATAACTT TTTGAGGTGA TGTCTTCATG TGTTTCAACT 7800 ACTTAAAAAC TTTTAAACAG TATATAATAA
Figure imgf000040_0002
AGGCCACTCA 7850 CACCTGTAAT CCCAGCACTT TGGGAGGCTG AGGTGGGCAG ATCACCTGAG 7900 GGCAGGAGTT CGAGACCAGC CTGGCCAATA TATATATATT CATATATTCA 7950 TATATATATA TATATTCATA TATTCATATA TATATATTCA TATATTCATA 8000 TATATATATA TATATATATA TAGCAAAACC TCATCTCTAA TAAAATACAA 8050 AAATTAGCTG AGCGTGGTGA TGGATGCCTG TAGTCCCAGC TACTCGGGAG 8100 GCTGAGGCAG GAGAATCTCT TGAACCTGGG AGGTGGAGGT TGCAGTGAGC 8150 TGAGATGGTG CCACTGCCCT CCAGCCTGAG TGACAGAGCG AGACTCGGTC 8200
Figure imgf000040_0003
AAATCTTCCA TCCTTGTCTC CCATCCACCC 8250 CTTCCCCCCA GCATGTACTT GCAGACTTTA TGCATATACA GTGAGTACTG 8300 TATATACACA AATAATAAAA AAATCATATA TATAATATAT GTAATTCCCC 8350 TTTACATGAA AGGTAGCACA CTGGTCTGTA CAGTCTGTCT GCACTGTGCT 8400 ATTTCACTTT ATATTTTTAT AGTTTGACAG AGTTCTAACA TTTCTTTTTT 8450 TTTTTTTTTA ACAGAGTCTT GTTCCTGATT GTTAAATTTT AAAGCATCCT 8500 AAAGTTTGGT TTCACACTTG AATGAATACC ATGTAAGGAT TCACTTACAT 8550 AGATGTGGTT GCCTGAATCT TAAGAATAAA ATAACATTGT TTGTATTTAT 8600 TTAAATTAGT GTTCCTTTTA TGGTTTGCCT GAAAGCACAA
Figure imgf000040_0004
8650 ACCAAGATAT TACAATTATG ACTCCCATAC AGGTAAACTG TTTAGAGATT 8700 GGCAAGCACC TTTTAATGAA AGGAGTCAGC CAGCTTAGTG TGCAGTATTT 8750 ATTTCTGCCG GAAGAGGGAG CTTCAGGGAC AGACTTTGGT TTAGTCATGA 8800 AGCCTCCAGC ACTCCCAAGC GGTTGTGGTT GACCAAGCAA TTTATGCTTT 8850 TACCTTTCTA CTTCCAGAGG CTTGTTTACT TATCAGTAAG CATTAATTTA 8900 GTGTCCCCTC AGATGCCTTT TACTTTCTTC TTTTCTGCCT AGAATAAGCT 8950 GCTCTTCCAA TTTTGCAGCT ACATGTTTCC ACCCCAGTTG GAATTTCTCC 9000 PATENT ATTORNEY DOCKET NO.51037-075WO2 ATAACATCCA TTGTAGCTAT CCTTCAATCT ACAGCCTCTA TTTCCTGTTA 9050 TAGCTGGTCA GGTCTAATCC CTCAAAATAC TCTGTCCCCT GCTTCCCTTA 9100 TCTGCTGGCC ACCTTTTTCC CCCACATACA CACTGCCATG TCCCACCCTT 9150 CACTCAAGTT GTTCCCTGCC ACCTCAACAA ATTTAAGTCC ATAAAATAGA 9200 GTAAGTGTTC CTGACTGTTA
Figure imgf000041_0001
CATCCCAAAG TCTGATTTCA 9250 CACTCGAATG AATACTATGT ACGGATTCAT TTACATAGAT GCGGTTGCAT 9300 GAGTCTTAAC
Figure imgf000041_0002
CATTATTTGT ATTTATTCAA AGTACTGTCA 9350 AGATATAATG TCAAGACCTA ATTCAAAGGT TCCACAAAGC CTTCCTTGAC 9400 TGCCCCCAAC GAAGATTATC CATTTTCCCT GAAATCCCAT TGACTTTTCT 9450 ATTTTGTAAG GAGGCTCGTG AGACTCTGTC
Figure imgf000041_0003
9500 AAGAAACAAT CAAACGGCTT GCTTCTGTTC TTTGATCTGC TAGTAAGCAA 9550 AAATTACACA TGGTGACAGG AGCTATGTGA GGCTGTCAGG TTGAATGGGA 9600 GGAGTTTGGG ATCCTGCTTG TGGATGGTTG GAAGAGGCTT TCGGGAAAGA 9650 CAGTATTTAT GTGAGACCTG GAAGATGGGC CTTAGCTTTG CAGAAGGTGG 9700 AGAGGCAGGA AATAGCACGG GGGCCCTGGG GCTGGAAGAC TTGGGCATAT 9750 TTGAGGAACA GAAAGGAGAC CAGCATAACT GAGGTGGGAA AAGCATGTGA 9800 AGAGATGGGG CTGGAGGAGG CCGGGAGTGG TGGCTCACGC CTGTAATCCC 9850 AGCACTTTGG GAGGCCAAGG CAGGCGGATC ATGAGCTCAG GAGATTGAGA 9900
Figure imgf000041_0004
TTGGGGTTAG AACACAAGTT TTGATGGGAA ACAGGTTAGA ACACATTCAT 10600 CTCTTCCCAT AGCGATGGTC ATAGAAAAAC GGGGCATATT TATAAACTCT 10650 CAGTTGATCT TAAAATGTGC AAAAGCTGCC GAACTCCTGG GAGTGAGCTC 10700 GAGCCCTGCA GGATCATTGT CACATGTGAG CAAAAGGCCA GCAAAAGGCC 10750 AGGAACCGTA AAAAGGCCGC GTTGCTGGCG TTTTTCCATA GGCTCCGCCC 10800 CCCTGACGAG CATCACAAAA ATCGACGCTC AAGTCAGAGG TGGCGAAACC 10850 CGACAGGACT ATAAAGATAC CAGGCGTTTC CCCCTGGAAG CTCCCTCGTG 10900 CGCTCTCCTG TTCCGACCCT GCCGCTTACC GGATACCTGT CCGCCTTTCT 10950 CCCTTCGGGA AGCGTGGCGC TTTCTCATAG CTCACGCTGT AGGTATCTCA 11000 GTTCGGTGTA GGTCGTTCGC TCCAAGCTGG GCTGTGTGCA CGAACCCCCC 11050 PATENT ATTORNEY DOCKET NO.51037-075WO2 GTTCAGCCCG ACCGCTGCGC CTTATCCGGT AACTATCGTC TTGAGTCCAA 11100 CCCGGTAAGA CACGACTTAT CGCCACTGGC AGCAGCCACT GGTAACAGGA 11150 TTAGCAGAGC GAGGTATGTA GGCGGTGCTA CAGAGTTCTT GAAGTGGTGG 11200 CCTAACTACG GCTACACTAG AAGAACAGTA TTTGGTATCT GCGCTCTGCT 11250 GAAGCCAGTT ACCTTCGGAA AAAGAGTTGG TAGCTCTTGA TCCGGCAAAC 11300 AAACCACCGC TGGTAGCGGT GGTTTTTTTG TTTGCAAGCA GCAGATTACG 11350 CGCAGAAAAA AAGGATCTCA AGAAGATCCT TTGATCTTTT CTACGGGGTC 11400 TGACGCTCAG TGGAACGAAA ACTCACGTTA AGGGATTTTG GTCATGAGAT 11450 TATCAAAAAG GATCTTCACC TAGATCCTTT TAAATTAAAA ATGAAGTTTT 11500 AAATCAAGCC CAATCTGAAT AATGTTACAA CCAATTAACC AATTCTGATT 11550 AGAAAAACTC ATCGAGCATC AAATGAAACT GCAATTTATT CATATCAGGA 11600 TTATCAATAC CATATTTTTG AAAAAGCCGT TTCTGTAATG AAGGAGAAAA 11650 CTCACCGAGG CAGTTCCATA GGATGGCAAG ATCCTGGTAT CGGTCTGCGA 11700 TTCCGACTCG TCCAACATCA ATACAACCTA TTAATTTCCC
Figure imgf000042_0001
11750 ATAAGGTTAT CAAGTGAGAA ATCACCATGA GTGACGACTG AATCCGGTGA 11800 GAATGGCAAA AGTTTATGCA TTTCTTTCCA GACTTGTTCA ACAGGCCAGC 11850 CATTACGCTC GTCATCAAAA TCACTCGCAT CAACCAAACC GTTATTCATT 11900 CGTGATTGCG CCTGAGCGAG ACGAAATACG CGATCGCTGT TAAAAGGACA 11950 ATTACAAACA GGAATCGAAT GCAACCGGCG CAGGAACACT GCCAGCGCAT 12000 CAACAATATT TTCACCTGAA TCAGGATATT CTTCTAATAC CTGGAATGCT 12050 GTTTTTCCGG GGATCGCAGT GGTGAGTAAC CATGCATCAT CAGGAGTACG 12100 GATAAAATGC TTGATGGTCG GAAGAGGCAT AAATTCCGTC AGCCAGTTTA 12150 GTCTGACCAT CTCATCTGTA ACATCATTGG CAACGCTACC TTTGCCATGT 12200 TTCAGAAACA ACTCTGGCGC ATCGGGCTTC CCATACAAGC GATAGATTGT 12250 CGCACCTGAT TGCCCGACAT TATCGCGAGC CCATTTATAC CCATATAAAT 12300 CAGCATCCAT GTTGGAATTT AATCGCGGCC TCGACGTTTC CCGTTGAATA 12350 TGGCTCATAA CACCCCTTGT ATTACTGTTT ATGTAAGCAG ACAGTTTTAT 12400 TGTTCATGAT GATATATTTT TATCTTGTGC AATGTAACAT CAGAGATTTT 12450 GAGACACGGG CCAGAGCTGC A 12500 Transcription Regulatory Elements Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein may contain various portions operably linked to one another. For example, transcription regulatory elements described herein may contain an ApoE and/or A1AT promoter, such as the chimeric promoter set forth in SEQ ID NO: 2, or a functional portion thereof. Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequence. Additionally or alternatively, transcription regulatory elements described herein may contain an LP1 promoter, or functional portion thereof. For example, the regulatory element may contain a PATENT ATTORNEY DOCKET NO.51037-075WO2 LP1 promoter as set forth in SEQ ID NO: 3, or a functional portion thereof. Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequences. Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein include promoters that stimulate expression of a transgene operably linked to a liver-specific promoter. Examples of such promoters are the ApoE/A1At and LP1 promoter, or a variant thereof (e.g., a variant having at least 85% sequence identity (for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity) to the nucleic acid sequence of the wild-type promoter locus and that is able to stimulate transcription of a transgene operably linked thereto upon introduction into a cell) or functional portion thereof. Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein include promoters that stimulate expression of a transgene operably linked to a muscle-specific promoter. Examples of such promoters are the desmin or MCK promoters, or a variant thereof (e.g., a variant having at least 85% sequence identity (for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity) to the nucleic acid sequence of the wild-type promoter locus and that is able to stimulate transcription of a transgene operably linked thereto upon introduction into a cell) or functional portion thereof. Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein include promoters that stimulate expression of a transgene operably linked to a ubiquitous promoter. Examples of such promoters are PGK, Ef1a, and GAPDH or a variant thereof (e.g., a variant having at least 85% sequence identity (for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity) to the nucleic acid sequence of the wild-type promoter locus and that is able to stimulate transcription of a transgene operably linked thereto upon introduction into a cell) or functional portion thereof. Transcription regulatory elements described herein may contain a SV40 enhancer or a functional portion thereof. For example, the regulatory element may contain a SV40 enhancer as set forth in SEQ ID NO: 4, or a functional portion thereof. Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequences. Transcription regulatory elements described herein may contain a β-globin enhancer or a functional portion thereof. For example, the regulatory element may contain a β-globin enhancer as set forth in SEQ ID NO: 5, or a functional portion thereof. Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, PATENT ATTORNEY DOCKET NO.51037-075WO2 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequences. Transcription regulatory elements that may be used in conjunction with the compositions and methods described herein include enhancers that enhance expression of a transgene. Examples of such promoters are PGK, Ef1alpha, and GAPDH, or a variant thereof (e.g., a variant having at least 85% sequence identity (for example, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or more, sequence identity) to the nucleic acid sequence of the wild- type promoter locus and that is able to stimulate transcription of a transgene operably linked thereto upon introduction into a cell) or functional portion thereof. The foregoing nucleic acid regulatory elements are summarized in Table 2, below. Table 2. Exemplary nucleic acid regulatory elements
Figure imgf000044_0001
PATENT ATTORNEY DOCKET NO.51037-075WO2
Figure imgf000045_0001
Additional nucleic acid regulatory elements useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the nucleic acid sequences set forth in Table 2. Transgenes Pseudotyped AAV vectors described herein may include a nucleic acid sequence encoding a MTM1 gene operably linked to a promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8). In some embodiments, the nucleic acid sequence encoding a MTM1 gene encodes a human MTM1 gene. In some embodiments, the nucleic acid sequence encoding a MTM1 gene encodes a mouse MTM1 gene. In some embodiments, the MTM1 gene is codon-optimized. In some embodiments, the promoter is a liver-specific promoter. In some embodiments, the promoter is a muscle-specific promoter. In some embodiments, the promoter is a ubiquitously expressed promoter. In some embodiments two MTM1 genes may be expressed simultaneously using different promoters on the same AAV vector. Transgenes that may be used in conjunction with the compositions and methods described herein may contain a human MTM1 transgene, as set forth in SEQ ID NO: 6, or a functional portion thereof. Additional transgenes useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequence. PATENT ATTORNEY DOCKET NO.51037-075WO2 Transgenes that may be used in conjunction with the compositions and methods described herein may contain a mouse MTM1 transgene, as set forth in SEQ ID NO: 7, or a functional portion thereof. Additional transgenes useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the above nucleic acid sequence. In some embodiments, a method of treating a disorder (e.g., XLMTM) or alleviating one or more symptoms (e.g., stiffness and/or join contractures or need for diaphragm and/or respiratory muscle progression) of a disorder (e.g., XLMTM) in a human patient in need thereof, includes administering to the patient a therapeutically effective amount of a pseudotyped AAV vector including a nucleic acid sequence encoding a MTM1 gene operably linked to a liver-specific or muscle specific promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8) as well as the other genetic components listed in Table 2 during a treatment period. In some embodiments, a method of weaning a human patient off of mechanical ventilation, includes a patient that has previously been administered a therapeutically effective amount of a pseudotyped AAV vector including a nucleic acid sequence encoding a MTM1 gene operably linked to a liver-specific or muscle specific promoter flanked by AAV2 ITR and packaged within capsid proteins from AAV8 (AAV2/8) as well as the other genetic components listed in Table 2. The foregoing transgenes are summarized in Table 3, below. Table 3. Exemplary transgenes
Figure imgf000046_0001
PATENT ATTORNEY DOCKET NO.51037-075WO2
Figure imgf000047_0001
PATENT ATTORNEY DOCKET NO.51037-075WO2
Figure imgf000048_0001
Additional transgenes useful in conjunction with the compositions and methods described herein include nucleic acid molecules that have at least 85% sequence identity (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99,9%, or greater, sequence identity) with respect to the nucleic acid sequences set forth in Table 3. Methods for the Delivery of Exogenous Nucleic Acids to Target Cells Transfection Techniques Techniques that can be used to introduce a transgene, such as a MTM1 transgene described herein, into a target cell are known in the art. For example, electroporation can be used to permeabilize mammalian cells (e.g., human target cells) by the application of an electrostatic potential to the cell of interest. Mammalian cells, such as human cells, subjected to an external electric field in this manner are subsequently predisposed to the uptake of exogenous nucleic acids (e.g., nucleic acids capable of expression in e.g., neurons, glial cells, or non-neural cells, such as colon and kidney cells). Electroporation of mammalian cells is described in detail, e.g., in Chu et al., Nucleic Acids Research 15:1311 (1987), the disclosure of which is incorporated herein by reference. A similar technique, NUCLEOFECTION™, utilizes an applied electric field in order to stimulate the uptake of exogenous polynucleotides into the nucleus of a eukaryotic cell. NUCLEOFECTION™ and protocols useful for performing this technique are described in detail, e.g., in Distler et al., Experimental Dermatology 14:315 (2005), as well as in US 2010/0317114, the disclosures of each of which are incorporated herein by reference. An additional technique useful for the transfection of target cells is the squeeze-poration methodology. This technique induces the rapid mechanical deformation of cells in order to stimulate the uptake of exogenous DNA through membranous pores that form in response to the applied stress. This technology is advantageous in that a vector is not required for delivery of nucleic acids into a cell, such as a human target cell. Squeeze-poration is described in detail, e.g., in Sharei et al., J. Vis. Exp. 81:e50980 (2013), the disclosure of which is incorporated herein by reference. Lipofection represents another technique useful for transfection of target cells. This method involves the loading of nucleic acids into a liposome, which often presents cationic functional groups, PATENT ATTORNEY DOCKET NO.51037-075WO2 such as quaternary or protonated amines, towards the liposome exterior. This promotes electrostatic interactions between the liposome and a cell due to the anionic nature of the cell membrane, which ultimately leads to uptake of the exogenous nucleic acids, for example, by direct fusion of the liposome with the cell membrane or by endocytosis of the complex. Lipofection is described in detail, for example, in US 7,442,386, the disclosure of which is incorporated herein by reference. Similar techniques that exploit ionic interactions with the cell membrane to provoke the uptake of foreign nucleic acids are contacting a cell with a cationic polymer-nucleic acid complex. Exemplary cationic molecules that associate with polynucleotides so as to impart a positive charge favorable for interaction with the cell membrane are activated dendrimers (described, e.g., in Dennig, Top Curr Chem.228:227 (2003), the disclosure of which is incorporated herein by reference) polyethylenimine, and DEAE-dextran, the use of which as a transfection agent is described in detail, for example, in Gulick et al., Curr Protoc Mol Biol.40:1:9.2:9.2.1 (1997), the disclosure of which is incorporated herein by reference. Another useful tool for inducing the uptake of exogenous nucleic acids by target cells is laserfection, also called optical transfection, a technique that involves exposing a cell to electromagnetic radiation of a particular wavelength in order to gently permeabilize the cells and allow polynucleotides to penetrate the cell membrane. The bioactivity of this technique is similar to, and in some cases found superior to, electroporation. Impalefection is another technique that can be used to deliver genetic material to target cells. It relies on the use of nanomaterials, such as carbon nanofibers, carbon nanotubes, and nanowires. Needle-like nanostructures are synthesized perpendicular to the surface of a substrate. DNA containing the gene, intended for intracellular delivery, is attached to the nanostructure surface. A chip with arrays of these needles is then pressed against cells or tissue. Cells that are impaled by nanostructures can express the delivered gene(s). An example of this technique is described in Shalek et al., PNAS 107:251870 (2010), the disclosure of which is incorporated herein by reference. MAGNETOFECTION™ can also be used to deliver nucleic acids to target cells. The principle of MAGNETOFECTION™ is to associate nucleic acids with cationic magnetic nanoparticles. The magnetic nanoparticles are made of iron oxide, which is fully biodegradable, and coated with specific cationic proprietary molecules varying upon the applications. Their association with the gene vectors (DNA, siRNA, viral vector, etc.) is achieved by salt-induced colloidal aggregation and electrostatic interaction. The magnetic particles are then concentrated on the target cells by the influence of an external magnetic field generated by magnets. This technique is described in detail in Scherer et al., Gene Ther.9:102 (2002), the disclosure of which is incorporated herein by reference. Magnetic beads are another tool that can be used to transfect target cells in a mild and efficient manner, as this methodology utilizes an applied magnetic field in order to direct the uptake of nucleic acids. This technology is described in detail, for example, in US2010/0227406, the disclosure of which is incorporated herein by reference. Another useful tool for inducing the uptake of exogenous nucleic acids by target cells is sonoporation, a technique that involves the use of sound (typically ultrasonic frequencies) for modifying the permeability of the cell plasma membrane permeabilize the cells and allow PATENT ATTORNEY DOCKET NO.51037-075WO2 polynucleotides to penetrate the cell membrane. This technique is described in detail, e.g., in Rhodes et al., Methods Cell Biol.82:309 (2007), the disclosure of which is incorporated herein by reference. Microvesicles represent another potential vehicle that can be used to modify the genome of a target cell according to the methods described herein. For example, microvesicles that have been induced by the co-overexpression of the glycoprotein VSV-G with, e.g., a genome-modifying protein, such as a nuclease, can be used to efficiently deliver proteins into a cell that subsequently catalyze the site-specific cleavage of an endogenous polynucleotide sequence so as to prepare the genome of the cell for the covalent incorporation of a polynucleotide of interest, such as a gene or regulatory sequence. The use of such vesicles, also referred to as Gesicles, for the genetic modification of eukaryotic cells is described in detail, e.g., in Quinn et al., Genetic Modification of Target Cells by Direct Delivery of Active Protein [abstract]. In: Methylation changes in early embryonic genes in cancer [abstract], in: Proceedings of the 18th Annual Meeting of the American Society of Gene and Cell Therapy; 2015 May 13, Abstract No.122. Incorporation of Target Genes by Gene Editing Techniques In addition to the above, a variety of tools have been developed that can be used for the incorporation of a gene of interest into a target cell, such as a human cell. One such method that can be used for incorporating polynucleotides encoding target genes into target cells involves the use of transposons. Transposons are polynucleotides that encode transposase enzymes and contain a polynucleotide sequence or gene of interest flanked by 5’ and 3’ excision sites. Once a transposon has been delivered into a cell, expression of the transposase gene commences and results in active enzymes that cleave the gene of interest from the transposon. This activity is mediated by the site- specific recognition of transposon excision sites by the transposase. In some instances, these excision sites may be terminal repeats or inverted terminal repeats. Once excised from the transposon, the gene of interest can be integrated into the genome of a mammalian cell by transposase-catalyzed cleavage of similar excision sites that exist within the nuclear genome of the cell. This allows the gene of interest to be inserted into the cleaved nuclear DNA at the complementary excision sites, and subsequent covalent ligation of the phosphodiester bonds that join the gene of interest to the DNA of the mammalian cell genome completes the incorporation process. In certain cases, the transposon may be a retrotransposon, such that the gene encoding the target gene is first transcribed to an RNA product and then reverse-transcribed to DNA before incorporation in the mammalian cell genome. Exemplary transposon systems are the piggybac transposon (described in detail in, e.g., WO 2010/085699) and the sleeping beauty transposon (described in detail in, e.g., US 2005/0112764), the disclosures of each of which are incorporated herein by reference as they pertain to transposons for use in gene delivery to a cell of interest. Another tool for the integration of target genes into the genome of a target cell is the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas system, a system that originally evolved as an adaptive defense mechanism in bacteria and archaea against viral infection. The CRISPR/Cas system includes palindromic repeat sequences within plasmid DNA and an associated Cas9 nuclease. This ensemble of DNA and protein directs site specific DNA cleavage of a target PATENT ATTORNEY DOCKET NO.51037-075WO2 sequence by first incorporating foreign DNA into CRISPR loci. Polynucleotides containing these foreign sequences and the repeat-spacer elements of the CRISPR locus are in turn transcribed in a host cell to create a guide RNA, which can subsequently anneal to a target sequence and localize the Cas9 nuclease to this site. In this manner, highly site-specific cas9-mediated DNA cleavage can be engendered in a foreign polynucleotide because the interaction that brings cas9 within close proximity of the target DNA molecule is governed by RNA:DNA hybridization. As a result, one can design a CRISPR/Cas system to cleave any target DNA molecule of interest. This technique has been exploited in order to edit eukaryotic genomes (Hwang et al., Nature Biotechnology 31:227 (2013)) and can be used as an efficient means of site-specifically editing target cell genomes in order to cleave DNA prior to the incorporation of a gene encoding a target gene. The use of CRISPR/Cas to modulate gene expression has been described in, for example, US Patent No.8,697,359, the disclosure of which is incorporated herein by reference as it pertains to the use of the CRISPR/Cas system for genome editing. Alternative methods for site-specifically cleaving genomic DNA prior to the incorporation of a gene of interest in a target cell include the use of zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs). Unlike the CRISPR/Cas system, these enzymes do not contain a guiding polynucleotide to localize to a specific target sequence. Target specificity is instead controlled by DNA binding domains within these enzymes. The use of ZFNs and TALENs in genome editing applications is described, e.g., in Urnov et al., Nat. Rev. Genet.11:636 (2010); and in Joung et al., Nat. Rev. Mol. Cell Biol.14:49 (2013), the disclosure of each of which are incorporated herein by reference as they pertain to compositions and methods for genome editing. Additional genome editing techniques that can be used to incorporate polynucleotides encoding target genes into the genome of a target cell include the use of ARCUSTM meganucleases that can be rationally designed so as to site-specifically cleave genomic DNA. The use of these enzymes for the incorporation of genes encoding target genes into the genome of a mammalian cell is advantageous in view of the defined structure-activity relationships that have been established for such enzymes. Single chain meganucleases can be modified at certain amino acid positions in order to create nucleases that selectively cleave DNA at desired locations, enabling the site-specific incorporation of a target gene into the nuclear DNA of a target cell. These single-chain nucleases have been described extensively in, for example, US Patent Nos.8,021,867 and US 8,445,251, the disclosures of each of which are incorporated herein by reference as they pertain to compositions and methods for genome editing. Pharmaceutical Compositions and Routes of Administration The gene therapy agents described herein may contain a transgene, such as a transgene encoding MTM1 and may be incorporated into a vehicle for administration into a patient, such as a human patient suffering from a neuromuscular disorder (for example, XLMTM). Pharmaceutical compositions containing vectors, such as viral vectors, that contain the transcription regulatory elements (e.g., a ApoE/A1AT promoter) described herein operably linked to a therapeutic transgene can be prepared using methods known in the art. For example, such compositions can be prepared using, e.g., physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical PATENT ATTORNEY DOCKET NO.51037-075WO2 Sciences 16th edition, Osol, A. Ed. (1980); incorporated herein by reference), and in a desired form, e.g., in the form of lyophilized formulations or aqueous solutions. Viral vectors, such as AAV vectors and others described herein, containing the transcription regulatory element operably linked to a therapeutic transgene may be administered to a patient (e.g., a human patient) by a variety of routes of administration. The route of administration may vary, for example, with the onset and severity of disease, and may include, e.g., intradermal, transdermal, parenteral, intravenous, intramuscular, intrahepatic, intranasal, subcutaneous, percutaneous, intratracheal, intraperitoneal, intraarterial, intravascular, inhalation, perfusion, lavage, and oral administration. Intravascular administration includes delivery into the vasculature of a patient. In some embodiments, the administration is into a vessel considered to be a vein (intravenous), and in some administration, the administration is into a vessel considered to be an artery (intraarterial). Veins include, but are not limited to, the internal jugular vein, a peripheral vein, a coronary vein, a hepatic vein, the portal vein, great saphenous vein, the pulmonary vein, superior vena cava, inferior vena cava, a gastric vein, a splenic vein, inferior mesenteric vein, superior mesenteric vein, cephalic vein, and/or femoral vein. Arteries include, but are not limited to, coronary artery, pulmonary artery, hepatic artery, brachial artery, internal carotid artery, aortic arch, femoral artery, peripheral artery, and/or ciliary artery. It is contemplated that delivery may be through or to an arteriole or capillary. Mixtures of the nucleic acids and viral vectors described herein may be prepared in water suitably mixed with one or more excipients, carriers, or diluents. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (described in US 5,466,468, the disclosure of which is incorporated herein by reference). In any case the formulation may be sterile and may be fluid to the extent that easy syringability exists. Formulations may be stable under the conditions of manufacture and storage and may be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. For example, a solution containing a pharmaceutical composition described herein may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, PATENT ATTORNEY DOCKET NO.51037-075WO2 intrahepatic, subcutaneous, and intraperitoneal administration. In this connection, sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in 1 mL of isotonic NaCl solution and either added to 1000 mL of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations may meet sterility, pyrogenicity, general safety, and purity standards as required by FDA Office of Biologics standards. Kits The compositions described herein can be provided in a kit for use in treating a neuromuscular disorder (e.g., XLMTM). In some embodiments, the kit may include one or more viral vectors as described herein. The kit can include a package insert that instructs a user of the kit, such as a physician of skill in the art, to perform any one of the methods described herein. The kit may optionally include a syringe or other device for administering the composition. In some embodiments, the kit may include one or more additional therapeutic agents. The kit can include a package insert that instructs a user of the kit, such as a physician of skill in the art, to perform any one of the methods described herein. The kit may optionally include a syringe or other device for administering the composition. In some embodiments, the kit may include one or more additional therapeutic agents. Dosing Regimens Dosing Regimens Involving AAV-MTM1 Vectors Using the compositions and methods of the disclosure, a patient having a neuromuscular disorder (e.g., XLMTM) may be administered an AAV vector containing a transgene encoding MTM1 in an amount of about 1.3 x 1014 vg/kg. Administration of the vector to the patient in such a quantity can achieve the beneficial effect of augmenting MTM1 expression in the patient, e.g., to within 50% or 200% of wild-type levels, without inducing toxic side effects. In some embodiments, the AAV vector is administered to the patient in an amount of less than about 3 x 1014 vg/kg (e.g., in an amount of less than about 3 x 1014 vg/kg, 2.9 x 1014 vg/kg, 2.8 x 1014 vg/kg, 2.7 x 1014 vg/kg, 2.6 x 1014 vg/kg, 2.5 x 1014 vg/kg, 2.4 x 1014 vg/kg, 2.3 x 1014 vg/kg, 2.2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). For example, the AAV vector may be administered to the patient in an amount of about 3 x 1014 vg/kg, 2.9 x 1014 vg/kg, 2.8 x 1014 vg/kg, 2.7 x 1014 vg/kg, 2.6 x 1014 vg/kg, 2.5 x 1014 vg/kg, 2.4 x 1014 vg/kg, 2.3 x 1014 vg/kg, 2.2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg. PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, the AAV vector is administered to the patient in an amount of less than about 2.5 x 1014 vg/kg (e.g., in an amount of less than about 2.5 x 1014 vg/kg, 2.4 x 1014 vg/kg, 2.3 x 1014 vg/kg, 2.2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). For example, the AAV vector may be administered to the patient in an amount of about 2.5 x 1014 vg/kg, 2.4 x 1014 vg/kg, 2.3 x 1014 vg/kg, 2.2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of less than about 2 x 1014 vg/kg (e.g., in an amount of less than about 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). For example, the AAV vector may be administered to the patient in an amount of about 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of less than about 1.5 x 1014 vg/kg (e.g., less than about 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). For example, the AAV vector may be administered to the patient in an amount of about 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of less than about 1 x 1014 vg/kg (e.g., less than about 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). For example, the AAV vector may be administered to the patient in an amount of about 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from about 3 x 1013 vg/kg to about 2.3 x 1014 vg/kg (e.g., in an amount of from about 3 x 1013 vg/kg to about 2.3 x 1014 vg/kg). For example, the AAV vector may be administered to the patient in an amount of about 3 x 1013 vg/kg, 3.1 x 1013 vg/kg, 3.2 x 1013 vg/kg, 3.3 x 1013 vg/kg, 3.4 x 1013 vg/kg, 3.5 x 1013 vg/kg, 3.6 x 1013 vg/kg, 3.7 x 1013 vg/kg, 3.8 x 1013 vg/kg, 3.9 x 1013 vg/kg, 4 x 1013 vg/kg, 4.1 x 1013 vg/kg, 4.2 x 1013 vg/kg, 4.3 x 1013 vg/kg, 4.4 x 1013 vg/kg, 4.5 x 1013 vg/kg, 4.6 x 1013 vg/kg, 4.7 x 1013 vg/kg, 4.8 x 1013 vg/kg, 4.9 x 1013 vg/kg, 5 x 1013 vg/kg, 5.1 x 1013 vg/kg, 5.2 x 1013 vg/kg, 5.3 x 1013 vg/kg, 5.4 x 1013 vg/kg, 5.5 x 1013 vg/kg, 5.6 x 1013 vg/kg, 5.7 x 1013 vg/kg, 5.8 x 1013 vg/kg, 5.9 x 1013 vg/kg, 6 x 1013 vg/kg, 6.1 x 1013 vg/kg, 6.2 x 1013 vg/kg, 6.3 x 1013 vg/kg, 6.4 x 1013 vg/kg, 6.5 x 1013 PATENT ATTORNEY DOCKET NO.51037-075WO2 vg/kg, 6.6 x 1013 vg/kg, 6.7 x 1013 vg/kg, 6.8 x 1013 vg/kg, 6.9 x 1013 vg/kg, 7 x 1013 vg/kg, 7.1 x 1013 vg/kg, 7.2 x 1013 vg/kg, 7.3 x 1013 vg/kg, 7.4 x 1013 vg/kg, 7.5 x 1013 vg/kg, 7.6 x 1013 vg/kg, 7.7 x 1013 vg/kg, 7.8 x 1013 vg/kg, 7.9 x 1013 vg/kg, 8 x 1013 vg/kg, 8.1 x 1013 vg/kg, 8.2 x 1013 vg/kg, 8.3 x 1013 vg/kg, 8.4 x 1013 vg/kg, 8.5 x 1013 vg/kg, 8.6 x 1013 vg/kg, 8.7 x 1013 vg/kg, 8.8 x 1013 vg/kg, 8.9 x 1013 vg/kg, 9 x 1013 vg/kg, 9.1 x 1013 vg/kg, 9.2 x 1013 vg/kg, 9.3 x 1013 vg/kg, 9.4 x 1013 vg/kg, 9.5 x 1013 vg/kg, 9.6 x 1013 vg/kg, 9.7 x 1013 vg/kg, 9.8 x 1013 vg/kg, 9.9 x 1013 vg/kg, 1 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.9 x 1014 vg/kg, 2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2.2 x 1014 vg/kg, or 2.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from about 4 x 1013 vg/kg to about 2.3 x 1014 vg/kg, such as an in amount of about 4 x 1013 vg/kg, 4.1 x 1013 vg/kg, 4.2 x 1013 vg/kg, 4.3 x 1013 vg/kg, 4.4 x 1013 vg/kg, 4.5 x 1013 vg/kg, 4.6 x 1013 vg/kg, 4.7 x 1013 vg/kg, 4.8 x 1013 vg/kg, 4.9 x 1013 vg/kg, 5 x 1013 vg/kg, 5.1 x 1013 vg/kg, 5.2 x 1013 vg/kg, 5.3 x 1013 vg/kg, 5.4 x 1013 vg/kg, 5.5 x 1013 vg/kg, 5.6 x 1013 vg/kg, 5.7 x 1013 vg/kg, 5.8 x 1013 vg/kg, 5.9 x 1013 vg/kg, 6 x 1013 vg/kg, 6.1 x 1013 vg/kg, 6.2 x 1013 vg/kg, 6.3 x 1013 vg/kg, 6.4 x 1013 vg/kg, 6.5 x 1013 vg/kg, 6.6 x 1013 vg/kg, 6.7 x 1013 vg/kg, 6.8 x 1013 vg/kg, 6.9 x 1013 vg/kg, 7 x 1013 vg/kg, 7.1 x 1013 vg/kg, 7.2 x 1013 vg/kg, 7.3 x 1013 vg/kg, 7.4 x 1013 vg/kg, 7.5 x 1013 vg/kg, 7.6 x 1013 vg/kg, 7.7 x 1013 vg/kg, 7.8 x 1013 vg/kg, 7.9 x 1013 vg/kg, 8 x 1013 vg/kg, 8.1 x 1013 vg/kg, 8.2 x 1013 vg/kg, 8.3 x 1013 vg/kg, 8.4 x 1013 vg/kg, 8.5 x 1013 vg/kg, 8.6 x 1013 vg/kg, 8.7 x 1013 vg/kg, 8.8 x 1013 vg/kg, 8.9 x 1013 vg/kg, 9 x 1013 vg/kg, 9.1 x 1013 vg/kg, 9.2 x 1013 vg/kg, 9.3 x 1013 vg/kg, 9.4 x 1013 vg/kg, 9.5 x 1013 vg/kg, 9.6 x 1013 vg/kg, 9.7 x 1013 vg/kg, 9.8 x 1013 vg/kg, 9.9 x 1013 vg/kg, 1 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.9 x 1014 vg/kg, 2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2.2 x 1014 vg/kg, or 2.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from about 5 x 1013 vg/kg to about 2.3 x 1014 vg/kg, such as an in amount of about 5 x 1013 vg/kg, 5.1 x 1013 vg/kg, 5.2 x 1013 vg/kg, 5.3 x 1013 vg/kg, 5.4 x 1013 vg/kg, 5.5 x 1013 vg/kg, 5.6 x 1013 vg/kg, 5.7 x 1013 vg/kg, 5.8 x 1013 vg/kg, 5.9 x 1013 vg/kg, 6 x 1013 vg/kg, 6.1 x 1013 vg/kg, 6.2 x 1013 vg/kg, 6.3 x 1013 vg/kg, 6.4 x 1013 vg/kg, 6.5 x 1013 vg/kg, 6.6 x 1013 vg/kg, 6.7 x 1013 vg/kg, 6.8 x 1013 vg/kg, 6.9 x 1013 vg/kg, 7 x 1013 vg/kg, 7.1 x 1013 vg/kg, 7.2 x 1013 vg/kg, 7.3 x 1013 vg/kg, 7.4 x 1013 vg/kg, 7.5 x 1013 vg/kg, 7.6 x 1013 vg/kg, 7.7 x 1013 vg/kg, 7.8 x 1013 vg/kg, 7.9 x 1013 vg/kg, 8 x 1013 vg/kg, 8.1 x 1013 vg/kg, 8.2 x 1013 vg/kg, 8.3 x 1013 vg/kg, 8.4 x 1013 vg/kg, 8.5 x 1013 vg/kg, 8.6 x 1013 vg/kg, 8.7 x 1013 vg/kg, 8.8 x 1013 vg/kg, 8.9 x 1013 vg/kg, 9 x 1013 vg/kg, 9.1 x 1013 vg/kg, 9.2 x 1013 vg/kg, 9.3 x 1013 vg/kg, 9.4 x 1013 vg/kg, 9.5 x 1013 vg/kg, 9.6 x 1013 vg/kg, 9.7 x 1013 vg/kg, 9.8 x 1013 vg/kg, 9.9 x 1013 vg/kg, 1 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.9 x 1014 vg/kg, 2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2.2 x 1014 vg/kg, or 2.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from about 6 x 1013 vg/kg to about 2.3 x 1014 vg/kg, such as an in amount of about 6 x 1013 vg/kg, 6.1 x 1013 vg/kg, 6.2 x 1013 vg/kg, 6.3 x 1013 vg/kg, 6.4 x 1013 vg/kg, 6.5 x 1013 vg/kg, 6.6 x 1013 vg/kg, 6.7 x PATENT ATTORNEY DOCKET NO.51037-075WO2 1013 vg/kg, 6.8 x 1013 vg/kg, 6.9 x 1013 vg/kg, 7 x 1013 vg/kg, 7.1 x 1013 vg/kg, 7.2 x 1013 vg/kg, 7.3 x 1013 vg/kg, 7.4 x 1013 vg/kg, 7.5 x 1013 vg/kg, 7.6 x 1013 vg/kg, 7.7 x 1013 vg/kg, 7.8 x 1013 vg/kg, 7.9 x 1013 vg/kg, 8 x 1013 vg/kg, 8.1 x 1013 vg/kg, 8.2 x 1013 vg/kg, 8.3 x 1013 vg/kg, 8.4 x 1013 vg/kg, 8.5 x 1013 vg/kg, 8.6 x 1013 vg/kg, 8.7 x 1013 vg/kg, 8.8 x 1013 vg/kg, 8.9 x 1013 vg/kg, 9 x 1013 vg/kg, 9.1 x 1013 vg/kg, 9.2 x 1013 vg/kg, 9.3 x 1013 vg/kg, 9.4 x 1013 vg/kg, 9.5 x 1013 vg/kg, 9.6 x 1013 vg/kg, 9.7 x 1013 vg/kg, 9.8 x 1013 vg/kg, 9.9 x 1013 vg/kg, 1 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.9 x 1014 vg/kg, or 2 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from about 7 x 1013 vg/kg to about 2.3 x 1014 vg/kg, such as an in amount of about 7 x 1013 vg/kg, 7.1 x 1013 vg/kg, 7.2 x 1013 vg/kg, 7.3 x 1013 vg/kg, 7.4 x 1013 vg/kg, 7.5 x 1013 vg/kg, 7.6 x 1013 vg/kg, 7.7 x 1013 vg/kg, 7.8 x 1013 vg/kg, 7.9 x 1013 vg/kg, 8 x 1013 vg/kg, 8.1 x 1013 vg/kg, 8.2 x 1013 vg/kg, 8.3 x 1013 vg/kg, 8.4 x 1013 vg/kg, 8.5 x 1013 vg/kg, 8.6 x 1013 vg/kg, 8.7 x 1013 vg/kg, 8.8 x 1013 vg/kg, 8.9 x 1013 vg/kg, 9 x 1013 vg/kg, 9.1 x 1013 vg/kg, 9.2 x 1013 vg/kg, 9.3 x 1013 vg/kg, 9.4 x 1013 vg/kg, 9.5 x 1013 vg/kg, 9.6 x 1013 vg/kg, 9.7 x 1013 vg/kg, 9.8 x 1013 vg/kg, 9.9 x 1013 vg/kg, 1 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.9 x 1014 vg/kg, 2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2.2 x 1014 vg/kg, or 2.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from about 8 x 1013 vg/kg to about 2.3 x 1014 vg/kg, such as an in amount of about 8 x 1013 vg/kg, 8.1 x 1013 vg/kg, 8.2 x 1013 vg/kg, 8.3 x 1013 vg/kg, 8.4 x 1013 vg/kg, 8.5 x 1013 vg/kg, 8.6 x 1013 vg/kg, 8.7 x 1013 vg/kg, 8.8 x 1013 vg/kg, 8.9 x 1013 vg/kg, 9 x 1013 vg/kg, 9.1 x 1013 vg/kg, 9.2 x 1013 vg/kg, 9.3 x 1013 vg/kg, 9.4 x 1013 vg/kg, 9.5 x 1013 vg/kg, 9.6 x 1013 vg/kg, 9.7 x 1013 vg/kg, 9.8 x 1013 vg/kg, 9.9 x 1013 vg/kg, 1 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.9 x 1014 vg/kg, 2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2.2 x 1014 vg/kg, or 2.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from about 9 x 1013 vg/kg to about 2.3 x 1014 vg/kg, such as an in amount of 9 x 1013 vg/kg, 9.1 x 1013 vg/kg, 9.2 x 1013 vg/kg, 9.3 x 1013 vg/kg, 9.4 x 1013 vg/kg, 9.5 x 1013 vg/kg, 9.6 x 1013 vg/kg, 9.7 x 1013 vg/kg, 9.8 x 1013 vg/kg, 9.9 x 1013 vg/kg, 1 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.9 x 1014 vg/kg, 2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2.2 x 1014 vg/kg, or 2.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of from about 1 x 1014 vg/kg to about 2.3 x 1014 vg/kg, such as an in amount 1 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.9 x 1014 vg/kg, 2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2.2 x 1014 vg/kg, or 2.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 3 x 1013 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 4 x 1013 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 5 x 1013 vg/kg. In some embodiments, the AAV vector is administered to the patient PATENT ATTORNEY DOCKET NO.51037-075WO2 in an amount of about 6 x 1013 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 7 x 1013 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 8 x 1013 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 9 x 1013 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.1 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.2 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.4 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.5 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.6 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.7 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.8 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 1.9 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.1 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.2 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.4 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.5 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.6 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.7 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.8 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 2.9 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in an amount of about 3 x 1014 vg/kg. In some embodiments, the AAV vector is administered to the patient in a single dose comprising the amount (e.g., less than about 3 x 1014 vg/kg). In some embodiments, the AAV vector is administered to the patient in two or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) doses that, together, comprise the amount (e.g., less than about 3 x 1014 vg/kg). In some embodiments, the AAV vector is administered to the patient in two or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) doses that each, individually, comprise the amount (e.g., less than about 3 x 1014 vg/kg). In some embodiments, the two or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) doses are separated from one another by one year or more (e.g., one year, one year and one day, one year and one month, one year and six months, two years, three years, four years, or five years). PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, the two or more (e.g., two, three, four, five, six, seven, eight, nine, or ten) doses are administered to the patient within about 12 months (e.g., about 12 months, about 11 months, about 10 months, about 9 months, about 8 months, about 7 months, about 6 months, about 5 months, about 4 months, about 3 months, about 2 months, or about 1 month) of one another Combination Therapies An AAV vector containing a transgene encoding MTM1 described herein can be administered in combination with a one or more additional AAV vectors containing a transgene encoding MTM1 described herein or in combination with one or more therapeutic procedures (e.g., nasobiliary drainage (NBD)) and/or agents (e.g., an anti-cholestatic agent) for the treatment of a neuromuscular disorder (e.g., XLMTM). Therapeutic Procedure In some embodiments, the one or more additional therapeutic procedures is NBD. NBD is a therapeutic procedure that is performed to help drain bile (e.g., when the bile duct is blocked, a biliary drain may help bile to flow from the liver into the intestine). In some embodiments, NBD is performed with a biliary drain (also known as a biliary stent), which is a thin, hollow, flexible tube with several small holes along the sides. A biliary drain may be inserted in a patient’s bile duct to allow it to drain. Therapeutic Agent In some embodiments, the one or more additional therapeutic agents is an anti-cholestatic agent (e.g., a bile acid, a farnesoid X receptor (FXR) ligand, a fibroblast growth factor 19 (FGF-19) mimetic, a Takeda-G-protein-receptor-5 (TGR5) agonist, a peroxisome proliferator-activated receptor (PPAR) agonist, a PPAR-alpha agonist, a PPAR-delta agonist, a dual PPAR-alpha and PPAR-delta agonist, an apical sodium-dependent bile acid transporter (ASBT) inhibitor, an immunomodulatory drug, an antifibrotic therapy, and a nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitor) or a combination thereof. In some embodiments, the anti-cholestatic agent is administered to the patient in one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy) doses that commence within about six weeks before or after (e.g., about six weeks before or after, about five weeks before or after, about four weeks before or after, about three weeks before or after, about two weeks before or after, or about one week before or after) administration of the viral vector to the patient. In some embodiments, the anti-cholestatic agent is administered to the patient in one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy) doses that commence within about five weeks before or after (e.g., about five weeks before or after, about four weeks before or after, about three weeks before or after, about two weeks before or after, or about one week before or after) administration of the viral vector to the patient. In some embodiments, the anti-cholestatic agent is administered to the patient in one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy) doses that commence within about one week before or after (e.g., about one week before or PATENT ATTORNEY DOCKET NO.51037-075WO2 after, about six days before or after, about five days before or after, about four days before or after, about three days before or after, about two days before or after, or about one day before or after) administration of the viral vector to the patient. In some embodiments, the anti-cholestatic agent is administered to the patient in one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, fifteen, twenty, thirty, forty, fifty, sixty, and seventy) doses that commence on the same day (e.g., 24th hour, on the 23rd hour, on the 22nd hour, on the 21st hour, on the 20th hour, on the 19th hour, on the 18th hour, on the 17th hour, on the 16th hour, on the 15th hour, on the 14th hour, on the 13th hour, on the 12th hour, on the 11th hour, on the 10th hour, on the 9th hour, on the 8th hour, on the 7th hour, on the 6th hour, on the 5th hour, on the 4th hour, on the 3rd hour, on the 2nd hour, on the 1st hour, on the 60th minute, on the 59th minute, on the 58th minute, on the 57th minute, on the 56th minute, on the 55th minute, on the 50th minute, on the 40th minute, on the 30th minute, on the 20th minute, on the 10th minute, or on the same minute) of administration of the viral vector to the patient. In some embodiments, the anti-cholestatic agent is a bile acid. In some embodiments, the bile acid is ursodeoxycholic acid or a derivative thereof or nor-ursodeoxycholic acid. In some embodiments, the bile acid is ursodiol. In some embodiments, the anti-cholestatic agent is an FXR ligand. In some embodiments, the FXR ligand is obeticholic acid, cilofexor, tropifexor, tretinoin, or EDP-305. In some embodiments, the one or more anti-cholestatic agent is an FGF-19 mimetic. In some embodiment, the FGF-19 mimetic is aldafermin. In some embodiments, the anti-cholestatic agent is a TGR5 agonist. In some embodiments, the TGR5 agonist is INT-777 or INT-767. In some embodiments, the anti-cholestatic agent is a PPAR agonist. In some embodiments, the PPAR agonist is bezafibrate, seladelpar, or elafibrinor. In some embodiments, the anti-cholestatic agent is a PPAR-alpha agonist. In some embodiments, the PPAR-alpha agonist is fenofibrate. In some embodiments, the anti-cholestatic agent is a PPAR-delta agonist. In some embodiments, the PPAR-delta agonist is seladelpar. In some embodiments, the anti-cholestatic agent is a dual PPAR-alpha and PPAR-delta agonist. In some embodiments, the dual PPAR-alpha -delta agonist is elafibranor. In some embodiments, the one or more anti-cholestatic agent is an ASBT inhibitor. In some embodiments, the ASBT inhibitor is odevixibat, maralixibat, or linerixibat. In some embodiments, the anti-cholestatic agent is an immunomodulatory drug. In some embodiments, the immunomodulatory drug is rituximab, abatacept, ustekinumab, infliximab, baricitinib, or FFP104. In some embodiments, the anti-cholestatic agent is an antifibrotic therapy. In some embodiments, the antifibrotic therapy is a vitamin D receptor (VDR) agonist or simtuzumab. In some embodiments, the anti-cholestatic agent is a NOX inhibitor. In some embodiments, the NOX inhibitor is setanaxib. PATENT ATTORNEY DOCKET NO.51037-075WO2 Recommended Clinical Parameters for Monitoring a Patient for Development of Cholestasis or Hyperbilirubinemia In some embodiments, a patient is monitored for the development of cholestasis by a serum bile acid test and/or blood test (e.g., an LFT), as described herein. In some embodiments, a patient is monitored for the development of hyperbilirubinemia by a blood test (e.g., bilirubin test), as described herein. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti- cholestatic agent. In some embodiments, a patient is monitored for the development of hyperbilirubinemia, and if the patient exhibits hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by a blood test (e.g., a serum acid bile test or a liver function test). In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by a blood test (e.g., a serum acid bile test or a liver function test), and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, the patient is determined to exhibit cholestasis, hyperbilirubinemia, or one or more symptoms thereof by a finding that the patient exhibits a parameter (e.g., a serum bile acid level) in blood test (e.g., a serum acid bile test) that is increased relative to a reference level. In some embodiments, the patient is determined to exhibit cholestasis, hyperbilirubinemia, or one or more symptoms thereof by a finding that the patient exhibits a serum bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) level in blood test (e.g., a serum acid bile test) that is increased relative to a reference level. In some embodiments, the blood test is a liver function test. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by a liver function test, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, the patient is determined to exhibit cholestasis, hyperbilirubinemia, or one or more symptoms thereof by a finding that the patient exhibits a parameter (e.g., aspartate aminotransferase level or alanine aminotransferase level) in liver function test that is increased or decreased relative to a reference level. I. Serum Bile Acid Test In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia with a serum bile acid test. In some embodiments, a patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is PATENT ATTORNEY DOCKET NO.51037-075WO2 administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia with a serum bile acid test, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for cholestasis or hyperbilirubinemia by the patient’s bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) levels, as measured with a serum bile acid test. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more of the patient’s bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) levels, as measured with a serum bile acid test, is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s cholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL). In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s cholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s chenodeoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL). In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s chenodeoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL). In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s deoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s deoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 PATENT ATTORNEY DOCKET NO.51037-075WO2 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s ursodeoxycholic acid level, as measured with a serum bile acid test, is greater than 2 nmol/mL (e.g., 2 nmol/mL, 3 nmol/mL, 4 nmol/mL, 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ursodeoxycholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. II. Liver Function Test In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia with an LFT. In some embodiments, a patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia with an LFT, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when a parameter (e.g., ASP level or AST level) of the patient’s LFT is greater than the age-adjusted norm, as described herein. IIa. Aspartate Aminotransferase In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s AST level in an LFT. In some embodiments, a patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s AST level in an LFT, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s AST level in an LFT and it is determined that a patient PATENT ATTORNEY DOCKET NO.51037-075WO2 exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). IIb. Alanine Aminotransferase In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s ALT level in an LFT. In some embodiments, a patient is monitored for the development of cholestasis, hyperbilirubinemia, or one or more symptoms thereof, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s ALT level in an LFT, and if the patient exhibits cholestasis or hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s ALT level in an LFT and it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). PATENT ATTORNEY DOCKET NO.51037-075WO2 Recommended Clinical Parameters for Monitoring a Patient for Development of Cholestasis I. Serum Bile Acid Test In some embodiments, a patient is monitored for the development of cholestasis. In some embodiments, a patient is monitored for the development of cholestasis with a serum bile acid test. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis with a serum bile acid test, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bile acids level, as measured with a serum bile acid test, is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bile acids level, as measured with a serum bile acid test, is greater than 14 µmol/L (e.g., 15 µmol/L, 16 µmol/L, 17 µmol/L, 18 µmol/L, 19 µmol/L, 20 µmol/L, 21 µmol/L, 22 µmol/L, 23 µmol/L, 24 µmol/L, 25 µmol/L, 26 µmol/L, 27 µmol/L, 28 µmol/L, 29 µmol/L, 30 µmol/L, 31 µmol/L, 32 µmol/L, 33 µmol/L, 34 µmol/L, 35 µmol/L, 36 µmol/L, 37 µmol/L, 38 µmol/L, 39 µmol/L, 40 µmol/L, 41 µmol/L, 42 µmol/L, 43 µmol/L, 44 µmol/L, 45 µmol/L, 46 µmol/L, 47 µmol/L, 48 µmol/L, 49 µmol/L, 50 µmol/L, 51 µmol/L, 52 µmol/L, 53 µmol/L, 54 µmol/L, 55 µmol/L, 56 µmol/L, 57 µmol/L, 58 µmol/L, 59 µmol/L, 60 µmol/L, 61 µmol/L, 62 µmol/L, 63 µmol/L, 64 µmol/L, 65 µmol/L, 66 µmol/L, 67 µmol/L, 68 µmol/L, 69 µmol/L, 70 µmol/L, 71 µmol/L, 72 µmol/L, 73 µmol/L, 74 µmol/L, 75 µmol/L, 76 µmol/L, 77 µmol/L, 78 µmol/L, 79 µmol/L, 80 µmol/L, 81 µmol/L, 82 µmol/L, 83 µmol/L, 84 µmol/L, 85 µmol/L, 86 µmol/L, 87 µmol/L, 88 µmol/L, 89 µmol/L, 90 µmol/L, 91 µmol/L, 92 µmol/L, 93 µmol/L, 94 µmol/L, 95 µmol/L, 96 µmol/L, 97 µmol/L, 98 µmol/L, 99 µmol/L, and 100 µmol/L). II. Blood Test In some embodiments, a patient is monitored for the development of cholestasis with a blood test (e.g., LFT or a bilirubin test). In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis with an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more parameters (e.g., GGT level, ASP level, AST level, ALT level, and bilirubin level) of the patient’s blood test (e.g., a LFT or a bilirubin test) is greater than the age-adjusted norm, as described herein. PATENT ATTORNEY DOCKET NO.51037-075WO2 IIa. Liver Function Test In some embodiments, a patient is monitored for the development of cholestasis with an LFT. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis with an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more parameters (e.g., GGT level, ASP level, AST level, and ALT level) of the patient’s LFT is greater than the age-adjusted norm, as described herein. IIai. Gamma-Glutamyl Transferase In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s GGT level in an LFT. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s GGT level in an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a GGT level that is greater than the age-adjusted norm. In some embodiments, the patient is a newborn (e.g., 0-6 months old), a toddler (e.g., 6-12 months old), or a child aged 1- ^5 years old. In some embodiments, the patient is a newborn of the age from 0-6 months old. In some embodiments, the patient is a toddler of the age from 6-12 months old. In some embodiments, the patient is a child of the age from 1-5 years old. In some embodiments, a patient is a newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 12-122 U/L (e.g., 12-122 U/L, 13-122 U/L, 14-122 U/L,15-122 U/L, 16-122 U/L, 17-122 U/L, 18-122 U/L, 19-122 U/L, 20-122 U/L, 25-122 U/L, 30-122 U/L, 40-122 U/L, 50-122 U/L, 60-122 U/L, 70-122 U/L, 80-122 U/L, 90-122 U/L, 100-122 U/L, 110-122 U/L, 120-122 U/L, or 121-122 U/L). In some embodiments, the patient is a male newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than 12 U/L (e.g., 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L). In some embodiments, the patient is a male newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 122 U/L (e.g., 123 U/L, 124 U/L, PATENT ATTORNEY DOCKET NO.51037-075WO2 125 U/L, 126 U/L, 127 U/L, 128 U/L, 129 U/L, 130 U/L, 135 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a male toddler (e.g., 6-12 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 1-39 U/L (e.g., 2-39 U/L, 3-39 U/L, 4-39 U/L, 5-39 U/L, 6-39 U/L, 7-39 U/L, 8-39 U/L, 9-39 U/L, 10-39 U/L, 11-39 U/L, 12-39 U/L, 13-39 U/L, 14-39 U/L, 15-39 U/L, 16-39 U/L, 17-39 U/L, 18-39 U/L, 19-39 U/L, 20-39 U/L, 21-39 U/L, 22-39 U/L, 23-39 U/L, 24-39 U/L, 25-39 U/L, 26-39 U/L, 27-39 U/L, 28-39 U/L, 29-39 U/L, 30-39 U/L, 31-39 U/L, 32-39 U/L, 33-39 U/L, 34-39 U/L, 35-39 U/L, 36-39 U/L, 37-39 U/L, or 38-39 U/L). In some embodiments, the patient is a male toddler (e.g., 6-12 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 39 U/L (e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/L, 48 U/L, 49 U/L, 50 U/L, 55 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a male child aged 1- ^5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 3-22 U/L (e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22 U/L, 13- 22 U/L, 14-22 U/L, 15-22 U/L, 16-22 U/L, 17-22 U/L, 18-22 U/L, 19-22 U/L, 20-22 U/L, and 21-22 U/L). In some embodiments, the patient is a male child aged 1- ^5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient is GGT level is less than 3 U/L (e.g., 2 U/L and 1 U/L). In some embodiments, the patient is a male child aged 1- ^5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 22 U/L (e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40 U/L, 50 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a female newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 15-132 U/L (e.g., 15-132 U/L, 16-132 U/L, 17-132 U/L, 18-132 U/L, 19-132 U/L, 20-132 U/L, 25-132 U/L, 30- 132 U/L, 40-132 U/L, 50-132 U/L, 60-132 U/L, 70-132 U/L, 80-132 U/L, 90-132 U/L, 100-132 U/L, 110-132 U/L, 120-132 U/L, 130-132 U/L, and 131-132 U/L). In some embodiments, the patient is a female newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than 15 U/L (e.g., 14 U/L, 13 U/L, 12 U/L, 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L). PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, the patient is a female newborn (e.g., 0-6 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 132 U/L (e.g., 133 U/L, 134 U/L, 135 U/L, 136 U/L, 137 U/L, 138 U/L, 139 U/L, 140 U/L, 145 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a female toddler (e.g., 6-12 months old) it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is outside of the normal range of about 1-39 U/L (e.g., 2-39 U/L, 3-39 U/L, 4-39 U/L, 5-39 U/L, 6-39 U/L, 7-39 U/L, 8-39 U/L, 9-39 U/L, 10-39 U/L, 11-39 U/L, 12-39 U/L, 13-39 U/L, 14-39 U/L, 15-39 U/L, 16-39 U/L, 17-39 U/L, 18-39 U/L, 19-39 U/L, 20-39 U/L, 21-39 U/L, 22-39 U/L, 23-39 U/L, 24-39 U/L, 25-39 U/L, 26-39 U/L, 27-39 U/L, 28-39 U/L, 29-39 U/L, 30-39 U/L, 31-39 U/L, 32-39 U/L, 33-39 U/L, 34-39 U/L, 35-39 U/L, 36-39 U/L, 37-39 U/L, or 38-39 U/L). In some embodiments, the patient is a female toddler (e.g., 6-12 months old) and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 39 U/L (e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/L, 48 U/L, 49 U/L, 50 U/L, 55 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a female child aged 1- ^5 years old it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 3-22 U/L (e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22 U/L, 13- 22 U/L, 14-22 U/L, 15-22 U/L, 16-22 U/L, 17-22 U/L, 18-22 U/L, 19-22 U/L, 20-22 U/L, and 21-22 U/L). In some embodiments, the patient is a female child aged 1- ^5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is less than 3 U/L (e.g., 2 U/L and 1 U/L). In some embodiments, the patient is a female child aged 1- ^5 years old and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is greater than 22 U/L (e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40 U/L, 50 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). IIaii. Alkaline Phosphatase In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s ASP level in an LFT. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s ASP level in an LFT, and if the patient PATENT ATTORNEY DOCKET NO.51037-075WO2 exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s ASP level in an LFT and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level that is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is outside of the normal range of about 50 to 300 U/L e.g., about 51 to 300 U/L, about 52 to U/L, about 53 to 300 U/L, about 54 to 300 U/L, about 55 to 300 U/L, about 56 to 300 U/L, about 57 to 300 U/L, about 58 to 300 U/L, about 59 to 300 U/L, about 60 to 300 U/L, about 65 to 300 U/L, about 70 to 300 U/L, about 80 to 300 U/L, about 90 to 300 U/L, about 100 to 300 U/L, about 125 to 300 U/L, about 150 to 300 U/L, about 175 to 300 U/L, about 200 to 300 U/L, about 225 to 300 U/L, about 250 to 300 U/L, or about 275 to 300 U/L). In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is less than 50 U/L (e.g., 50 U/L, 49 U/L, 48 U/L, 47 U/L, 46 U/L, 45 U/L, 44 U/L, 43 U/L, 42 U/L, 41 U/L, 40 U/L, 39 U/L, 38 U/L, 37 U/L, 36 U/L, 35 U/L, 34 U/L, 33 U/L, 32 U/L, 31 U/L, 30 U/L, 29 U/L, 28 U/L, 27 U/L, 26 U/L, 25 U/L, 24 U/L, 23 U/L, 22 U/L, 21 U/L, 20 U/L, 19 U/L, 18 U/L, 17 U/L, 16 U/L, 15 U/L, 14 U/L, 13 U/L, 12 U/L, 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, and 1 U/L). In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is greater than 300 U/L (e.g., 300 U/L, 301 U/L, 302 U/L, 303 U/L, 304 U/L, 305 U/L, 306 U/L, 307 U/L, 308 U/L, 309 U/L, 310 U/L, 311 U/L, 312 U/L, 313 U/L, 314 U/L, 315 U/L, 316 U/L, 317 U/L, 318 U/L, 319 U/L, 320 U/L, 321 U/L, 322 U/L, 323 U/L, 324 U/L, 325 U/L, 330 U/L, 340 U/L, 350 U/L, 400 U/L, and 500 U/L). IIaiii. Aspartate Aminotransferase In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s AST level in an LFT. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s AST level in an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s AST level in an LFT and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than the norm. PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). IIaiv. Alanine Aminotransferase In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s ALT level in an LFT. In some embodiments, a patient is monitored for the development of cholestasis, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s ALT level in an LFT, and if the patient exhibits cholestasis or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of cholestasis by measuring the patient’s ALT level in an LFT and it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). Recommended Clinical Parameters for Monitoring a Patient for Development of Hyperbilirubinemia Bilirubin Test In some embodiments, a patient is monitored for the development of hyperbilirubinemia. In some embodiments, a patient is monitored for the development of hyperbilirubinemia with a bilirubin test. In some embodiments, a patient is monitored for the development of hyperbilirubinemia, and if the patient exhibits hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, a patient is monitored for the development of hyperbilirubinemia with a bilirubin test, and if the patient exhibits hyperbilirubinemia or one or more symptoms thereof, the patient is administered an anti-cholestatic agent. In some embodiments, it is determined that a patient exhibits hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a bilirubin level that is greater than the norm. PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, it is determined that a patient exhibits hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bilirubin level is greater than 1.2 mg/dL (e.g., 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3. mg/dL, 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, and 100 mg/dL). In some embodiments, it is determined that a patient exhibits hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s direct bilirubin level is greater than 0.2 mg/dL (e.g., 0.2 mg/dL, 0.3 mg/dL, 0.4 mg/dL, 0.5 mg/dL, 0.6 mg/dL, 0.7 mg/dL, 0.8 mg/dL, 0.9 mg/dL, 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3. mg/dL, 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, and 100 mg/dL). In some embodiments, the patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a bilirubin level that is greater than 1 mg/dL (e.g., greater than 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3. mg/dL, 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, or 100 mg/dL) in a bilirubin test. Recommended Clinical Parameters for Determining that a Patient Exhibits Cholestasis or Hyperbilirubinemia or a Symptom Thereof In some embodiments, it is determined that the patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof by determining that one or more parameters (e.g., total bile acids level, GGT level, ASP level, AST level, and ALT level) of the patient’s serum bile acid test and/or blood test (e.g., an LFT) is greater than or less than the age-adjusted norm, as described herein, and the patient is administered an anti-cholestatic agent. In some embodiments, it is determined that the patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof by determining that one or more parameters (e.g., bilirubin level) of the patient’s blood test (e.g., bilirubin test) is greater than the norm, as described herein, and the patient is administered an anti-cholestatic agent. PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more of the patient’s bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) levels, as measured with a serum bile acid test, is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when a parameter (e.g., ASP level or AST level) of the patient’s LFT is greater than the age-adjusted norm, as described herein. I. Serum Bile Acid Test In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more of the patient’s bile acid (e.g., cholic acid, chenodeoxycholic acid, deoxycholic acid, or ursodeoxycholic acid) levels, as measured with a serum bile acid test, is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s cholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL). In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s cholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s chenodeoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL). In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s chenodeoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL). In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s deoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s deoxycholic acid level, as measured with a serum bile acid test, is greater than 6 nmol/mL (e.g., 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s ursodeoxycholic acid level, as measured with a serum bile acid test, is greater than 2 nmol/mL (e.g., 2 nmol/mL, 3 nmol/mL, 4 nmol/mL, 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ursodeoxycholic acid level, as measured with a serum bile acid test, is greater than 5 nmol/mL (e.g., 2 nmol/mL, 3 nmol/mL, 4 nmol/mL, 5 nmol/mL, 6 nmol/mL, 7 nmol/mL, 8 mol/mL, 9 nmol/mL, 10 nmol/mL, 15 nmol/mL, 20 nmol/mL, 30 nmol/mL, 40 nmol/mL, 50 nmol/mL, 60 nmol/mL, 70 nmol/mL, 80 nmol/mL, 90 nmol/mL, and 100 nmol/mL. II. Liver Function Test In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when a parameter (e.g., ASP level or AST level) of the patient’s LFT is greater than the age-adjusted norm, as described herein. IIa. Aspartate Aminotransferase In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s AST level in an LFT and it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). In some embodiments, it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). PATENT ATTORNEY DOCKET NO.51037-075WO2 IIb. Alanine Aminotransferase In some embodiments, a patient is monitored for the development of cholestasis or hyperbilirubinemia by measuring the patient’s ALT level in an LFT and it is determined that a patient exhibits cholestasis, hyperbilirubinemia, or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than the norm. In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). In some embodiments, it is determined that a patient exhibits cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). Recommended Clinical Parameters for Determining that a Patient Exhibits Cholestasis or a Symptom Thereof I. Serum Bile Acid Test In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits an acid bile level, as measured with a serum bile acid test, that is greater than the norm. In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bile acids level, as measured determined with a serum bile acid test, is greater than 14 µmol/L (e.g., 15 µmol/L, 16 µmol/L, 17 µmol/L, 18 µmol/L, 19 µmol/L, 20 µmol/L, 21 µmol/L, 22 µmol/L, 23 µmol/L, 24 µmol/L, 25 µmol/L, 26 µmol/L, 27 µmol/L, 28 µmol/L, 29 µmol/L, 30 µmol/L, 31 µmol/L, 32 µmol/L, 33 µmol/L, 34 µmol/L, 35 µmol/L, 36 µmol/L, 37 µmol/L, 38 µmol/L, 39 µmol/L, 40 µmol/L, 41 µmol/L, 42 µmol/L, 43 µmol/L, 44 µmol/L, 45 µmol/L, 46 µmol/L, 47 µmol/L, 48 µmol/L, 49 µmol/L, 50 µmol/L, 51 µmol/L, 52 µmol/L, 53 µmol/L, 54 µmol/L, 55 µmol/L, 56 µmol/L, 57 µmol/L, 58 µmol/L, 59 µmol/L, 60 µmol/L, 61 µmol/L, 62 µmol/L, 63 µmol/L, 64 µmol/L, 65 µmol/L, 66 µmol/L, 67 µmol/L, 68 µmol/L, 69 µmol/L, 70 µmol/L, 71 µmol/L, 72 µmol/L, 73 µmol/L, 74 µmol/L, 75 µmol/L, 76 µmol/L, 77 µmol/L, 78 µmol/L, 79 µmol/L, 80 µmol/L, 81 µmol/L, 82 µmol/L, 83 µmol/L, 84 µmol/L, 85 µmol/L, 86 µmol/L, 87 µmol/L, 88 µmol/L, 89 µmol/L, 90 µmol/L, 91 µmol/L, 92 µmol/L, 93 µmol/L, 94 µmol/L, 95 µmol/L, 96 µmol/L, 97 µmol/L, 98 µmol/L, 99 µmol/L, and 100 µmol/L). II. Blood Test In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more parameters (e.g., PATENT ATTORNEY DOCKET NO.51037-075WO2 GGT level, ASP level, AST level, ALT level, and bilirubin level) of the patient’s blood test (e.g., a LFT or a bilirubin test) is greater than the age-adjusted norm, as described herein. IIa. Liver Function Test In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when one or more parameters (e.g., GGT level, ASP level, AST level, and ALT level) of the patient’s LFT is greater than the age-adjusted norm, as described herein. IIai. Gamma-Glutamyl Transferase In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a GGT level, as measured in a LFT, that is greater than the age-adjusted norm. In some embodiments, the patient is a newborn (e.g., 0-6 months old), a toddler (e.g., 6-12 months old), or a child aged 1- ^5 years old. In some embodiments, the patient is a newborn of the age from 0-6 months old. In some embodiments, the patient is a toddler of the age from 6-12 months old. In some embodiments, the patient is a child of the age from 1-5 years old. In some embodiments, a patient is a newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 12-122 U/L (e.g., 12-122 U/L, 13-122 U/L, 14-122 U/L,15-122 U/L, 16-122 U/L, 17-122 U/L, 18-122 U/L, 19-122 U/L, 20-122 U/L, 25-122 U/L, 30-122 U/L, 40-122 U/L, 50-122 U/L, 60-122 U/L, 70-122 U/L, 80-122 U/L, 90-122 U/L, 100-122 U/L, 110-122 U/L, 120-122 U/L, or 121-122 U/L). In some embodiments, the patient is a male newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than 12 U/L (e.g., 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L). In some embodiments, the patient is a male newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 122 U/L (e.g., 123 U/L, 124 U/L, 125 U/L, 126 U/L, 127 U/L, 128 U/L, 129 U/L, 130 U/L, 135 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a male toddler (e.g., 6-12 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 1-39 U/L (e.g., 2-39 U/L, 3-39 U/L, 4-39 U/L, 5-39 U/L, 6-39 U/L, 7-39 U/L, 8-39 U/L, 9-39 U/L, 10-39 U/L, 11-39 U/L, 12-39 U/L, 13- 39 U/L, 14-39 U/L, 15-39 U/L, 16-39 U/L, 17-39 U/L, 18-39 U/L, 19-39 U/L, 20-39 U/L, 21-39 U/L, 22- 39 U/L, 23-39 U/L, 24-39 U/L, 25-39 U/L, 26-39 U/L, 27-39 U/L, 28-39 U/L, 29-39 U/L, 30-39 U/L, 31- 39 U/L, 32-39 U/L, 33-39 U/L, 34-39 U/L, 35-39 U/L, 36-39 U/L, 37-39 U/L, or 38-39 U/L). PATENT ATTORNEY DOCKET NO.51037-075WO2 In some embodiments, the patient is a male toddler (e.g., 6-12 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 39 U/L (e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/L, 48 U/L, 49 U/L, 50 U/L, 55 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a male child aged 1- ^5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 3-22 U/L (e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22 U/L, 13-22 U/L, 14-22 U/L, 15-22 U/L, 16-22 U/L, 17-22 U/L, 18-22 U/L, 19-22 U/L, 20-22 U/L, and 21-22 U/L). In some embodiments, the patient is a male child aged 1- ^5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than about 3 U/L (e.g., 2 U/L and 1 U/L). In some embodiments, the patient is a male child aged 1- ^5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 22 U/L (e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40 U/L, 50 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a female newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is outside of the normal range of about 15-132 U/L (e.g., 15-132 U/L, 16-132 U/L, 17-132 U/L, 18-132 U/L, 19-132 U/L, 20-132 U/L, 25-132 U/L, 30-132 U/L, 40-132 U/L, 50-132 U/L, 60-132 U/L, 70-132 U/L, 80-132 U/L, 90-132 U/L, 100-132 U/L, 110-132 U/L, 120-132 U/L, 130-132 U/L, and 131-132 U/L). In some embodiments, the patient is a female newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is less than about 15 U/L (e.g., 14 U/L, 13 U/L, 12 U/L, 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, or 1 U/L). In some embodiments, the patient is a female newborn (e.g., 0-6 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is greater than 132 U/L (e.g., 133 U/L, 134 U/L, 135 U/L, 136 U/L, 137 U/L, 138 U/L, 139 U/L, 140 U/L, 145 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a female toddler (e.g., 6-12 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is outside of the normal range of about 1-39 U/L (e.g., 2-39 U/L, 3-39 U/L, 4-39 U/L, 5-39 U/L, 6-39 U/L, 7-39 U/L, 8-39 U/L, 9-39 U/L, 10-39 U/L, 11-39 U/L, 12-39 U/L, 13-39 U/L, 14-39 U/L, 15-39 U/L, 16-39 U/L, 17-39 U/L, 18-39 U/L, 19-39 U/L, 20-39 U/L, 21-39 U/L, 22-39 U/L, 23-39 U/L, 24-39 U/L, 25-39 U/L, 26-39 U/L, 27-39 U/L, 28-39 U/L, 29-39 U/L, PATENT ATTORNEY DOCKET NO.51037-075WO2 30-39 U/L, 31-39 U/L, 32-39 U/L, 33-39 U/L, 34-39 U/L, 35-39 U/L, 36-39 U/L, 37-39 U/L, or 38-39 U/L). In some embodiments, the patient is a female toddler (e.g., 6-12 months old) and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti- cholestatic agent when the patient’s GGT level is greater than 39 U/L (e.g., 40 U/L, 41 U/L, 42 U/L, 43 U/L, 44 U/L, 45 U/L, 46 U/L, 47 U/L, 48 U/L, 49 U/L, 50 U/L, 55 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). In some embodiments, the patient is a female child aged 1- ^5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is outside of the normal range of about 3-22 U/L (e.g., about 3-22 U/L, 4-22 U/L, 5-22 U/L, 6-22 U/L, 7-22 U/L, 8-22 U/L, 9-22 U/L, 10-22 U/L, 11-22 U/L, 12-22 U/L, 13-22 U/L, 14-22 U/L, 15-22 U/L, 16-22 U/L, 17-22 U/L, 18-22 U/L, 19-22 U/L, 20-22 U/L, and 21-22 U/L). In some embodiments, the patient is a female child aged 1- ^5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is less than about 3 U/L (e.g., 2 U/L and 1 U/L). In some embodiments, the patient is a female child aged 1- ^5 years old and is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s GGT level is greater than 22 U/L (e.g., 23 U/L, 24 U/L, 25 U/L, 26 U/L, 27 U/L, 28 U/L, 29 U/L, 30 U/L, 35 U/L, 40 U/L, 50 U/L, 60 U/L, 70 U/L, 80 U/L, 90 U/L, 100 U/L, 110 U/L, 1120 U/L, 130 U/L, 140 U/L, 150 U/L, 160 U/L, 170 U/L, 180 U/L, 190 U/L, and 200 U/L). IIaii. Alkaline Phosphatase In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits an ASP level, as measured in a LFT, that is greater than the norm. In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is outside of the normal range of about 50 to 300 U/L (e.g., about 51 to 300 U/L, about 52 to U/L, about 53 to 300 U/L, about 54 to 300 U/L, about 55 to 300 U/L, about 56 to 300 U/L, about 57 to 300 U/L, about 58 to 300 U/L, about 59 to 300 U/L, about 60 to 300 U/L, about 65 to 300 U/L, about 70 to 300 U/L, about 80 to 300 U/L, about 90 to 300 U/L, about 100 to 300 U/L, about 125 to 300 U/L, about 150 to 300 U/L, about 175 to 300 U/L, about 200 to 300 U/L, about 225 to 300 U/L, about 250 to 300 U/L, or about 275 to 300 U/L). In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is less than about 50 U/L (e.g., 50 U/L, 49 U/L, 48 U/L, 47 U/L, 46 U/L, 45 U/L, 44 U/L, 43 U/L, 42 U/L, 41 U/L, 40 U/L, 39 U/L, 38 U/L, 37 U/L, 36 U/L, 35 U/L, 34 U/L, 33 U/L, 32 U/L, 31 U/L, 30 U/L, 29 U/L, 28 U/L, 27 U/L, 26 U/L, 25 U/L, 24 U/L, 23 U/L, 22 U/L, 21 U/L, 20 U/L, 19 U/L, 18 U/L, 17 U/L, 16 PATENT ATTORNEY DOCKET NO.51037-075WO2 U/L, 15 U/L, 14 U/L, 13 U/L, 12 U/L, 11 U/L, 10 U/L, 9 U/L, 8 U/L, 7 U/L, 6 U/L, 5 U/L, 4 U/L, 3 U/L, 2 U/L, and 1 U/L). In some embodiments, a is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ASP level is greater than 300 U/L (e.g., 300 U/L, 301 U/L, 302 U/L, 303 U/L, 304 U/L, 305 U/L, 306 U/L, 307 U/L, 308 U/L, 309 U/L, 310 U/L, 311 U/L, 312 U/L, 313 U/L, 314 U/L, 315 U/L, 316 U/L, 317 U/L, 318 U/L, 319 U/L, 320 U/L, 321 U/L, 322 U/L, 323 U/L, 324 U/L, 325 U/L, 330 U/L, 340 U/L, 350 U/L, 400 U/L, and 500 U/L). IIaiii. Aspartate Aminotransferase In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits an AST level, as measured in a LFT, that is greater than the norm. In some embodiments, a is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s AST level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). IIaiv. Alanine Aminotransferase In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits an ALT level, as measured in a LFT, that is greater than the norm. In some embodiments, a patient is determined to exhibit cholestasis or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s ALT level is greater than 50 U/L (e.g., 51 U/L, 52 U/L, 53 U/L, 54 U/L, 55 U/L, 56 U/L, 57 U/L, 58 U/L, 59 U/L, 60 U/L, 61 U/L, 62 U/L, 63 U/L, 64 U/L, 65 U/L, 66 U/L, 67 U/L, 68 U/L, 69 U/L, 70 U/L, 75 U/L, 80 U/L, 85 U/L, 90 U/L, 100 U/L, 110 U/L, 120 U/L, 130 U/L, 140 U/L, 150 U/L, 200 U/L, 300 U/L, 400 U/L, and 500 U/L). Recommended Clinical Parameters for Determining that a Patient Exhibits Hyperbilirubinemia or a Symptom Thereof Bilirubin Test In some embodiments, a patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a bilirubin level, as measured in a blood test (e.g., a bilirubin test), that is greater than the norm. In some embodiments, a patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s total bilirubin level is greater than 1.2 mg/dL (e.g., 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3. mg/dL, 3.4 mg/dL, 3.5 mg/dL, PATENT ATTORNEY DOCKET NO.51037-075WO2 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, and 100 mg/dL). In some embodiments, a patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient’s direct bilirubin level is greater than 0.2 mg/dL (e.g., 0.2 mg/dL, 0.3 mg/dL, 0.4 mg/dL, 0.5 mg/dL, 0.6 mg/dL, 0.7 mg/dL, 0.8 mg/dL, 0.9 mg/dL, 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3. mg/dL, 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, and 100 mg/dL). In some embodiments, the patient is determined to exhibit hyperbilirubinemia or one or more symptoms thereof and is administered an anti-cholestatic agent when the patient exhibits a bilirubin level that is greater than 1 mg/dL (e.g., greater than 1 mg/dL, 1.1 mg/dL, 1.2 mg/dL, 1.3 mg/dL, 1.4 mg/dL, 1.5 mg/dL, 1.6 mg/dL, 1.7 mg/dL, 1.8 mg/dL, 1.9 mg/dL, 2 mg/dL, 2.1 mg/dL, 2.2 mg/dL, 2.3 mg/dL, 2.4 ]mg/dL, 2.5 mg/dL, 2.6 mg/dL, 2.7 mg/dL, 2.8 mg/dL, 2.9 mg/dL, 3 mg/dL, 3.1 mg/dL, 3.2 mg/dL, 3.3. mg/dL, 3.4 mg/dL, 3.5 mg/dL, 3.6 mg/dL, 3.7 mg/dL, 3.8 mg/dL, 3.9 mg/dL, 4 mg/dL, 4.1 mg/dL, 4.2 mg/dL, 4.3 mg/dL, 4.4 mg/dL, 4.5 mg/dL, 4.6 mg/dL, 4.7 mg/dL, 4.8 mg/dL, 4.9 mg/dL, 5 mg/dL, 10 mg/dL, 15 mg/dL, 20 mg/dL, 30 mg/dL, 40 mg/dL, 50 mg/dL, 60 mg/dL, 70 mg/dL, 80 mg/dL, 90 mg/dL, or 100 mg/dL) in a bilirubin test. Examples The following examples are put forth so as to provide those of ordinary skill in the art with a description of how the compositions and methods described herein may be used and evaluated and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. Example 1. Evaluating safety and efficacy of a pseudotyped AAV8 vector including a nucleic acid sequence encoding a myotubularin 1 gene in a murine model of X-Linked myotubular myopathy Objective and Study Methodology The objective of this study was to perform an efficacy and sampling study using myotubularin 1 (MTM1) hemizygous (“HEMI”) knock out (KO) mice and determine the possible effects of liver- specific MTM1 expression in mice. The mice were dosed with different test articles intravenously at ~4 weeks of age. Mice were sampled at ~16 weeks of age (~12 weeks post-dosing). Tissue samples were used for myotubularin and bile salt transporter expression, bile acid level measurements, and histopathology. Whole blood samples were used for hematology analysis and serum samples were PATENT ATTORNEY DOCKET NO.51037-075WO2 used for clinical chemistry analysis. In addition, a group of naïve MTM1 KO mice were sampled at ~5 weeks of age, and tissues were used for histopathology. This study was performed according to appropriate methodologies and standard operating procedures (SOPs) at the testing facility or test sites. This study was compliant with standard ethics of animal welfare. Materials and Methods This study used MTM1 KO mice, with 28 male wild type (WT) mice and 70 male KO mice. Animals were sampled at ~5 and ~16 weeks. Each animal was permanently identified according to SOPs of the Testing Facility with unique permanent identification numbers. Animals were identified using toe and tail numbering and ear marks. There was a ~3-4-day acclimation period before dosing. Animals were housed in groups of up to 3-4 per cage, and at least one WT mouse was placed in each cage for socializing purposes. Individually ventilated caging systems (IVC) and polycarbonate Type II Long cages were used to house the mice. Veterinary care was available throughout the course of the study and animals were examined by the responsible trained personnel and supervised by veterinarian as warranted by clinical signs or other changes. Before the initiation of the in-life phase, any animals considered unsuitable for use in the study were replaced by alternate animals obtained from the same shipment and maintained under the same environmental conditions. Animals in poor health or at extremes of body weight range were not assigned to groups. In setting up groups for this study, KO mice were randomized into groups so that whole litters of mice did not end up in a single testing group to avoid a “litter effect”. Mice were housed in groups of up to 4-5. Each cage included 3-4 KO mice, and 1-2 WT mice for socializing purposes. None of the KO mice were housed individually. Four articles were tested: 1) an AAV8 vector containing a mouse MTM1 (mMTM1) gene under the control of a desmin promoter (AAV8-Des-mMTM1), 2) an AAV8 vector containing mMTM1 under the control of an APoE-A1AT promoter (AAV8-APoE-A1AT-mMTM1), 3) an AAV8 vector containing a human MTM1 (hMTM1) gene with a stop codon under the control of a desmin promoter (AAV8-Des-hMTM1-STOP), and 4) an AAV8 empty capsid. A vehicle control was used containing a placebo of Ringer’s Lactate solution with 0.01% pluronic. Dose levels were selected based on previous studies with mice. The high dose was a multiple of the previous high dose in mice, as the previous high dose showed significant pharmacological effect without toxicity. Animals were dosed by slow intravenous (IV) bolus injection into a tail vein. Dose volumes (total 16.33 mL/kg) were administered as a split IV bolus dose with 2 to 3 hours between administrations. The dose site, end time, and dose volume were documented. Animals were anesthetized with isoflurane, if necessary. This administration route is consistent with the proposed route of administration in humans and is expected to provide appropriate systemic exposure for investigation and associated pharmacological activity. The administration frequency was one dose on day 1. Dose levels were anticipated to identify achievable associated pharmacological activity. Necropsy occurred ~12 weeks post dosing. PATENT ATTORNEY DOCKET NO.51037-075WO2 Treatment/Study groups are summarized in Table 4, below. Additionally, a group of 10 naïve MTM1 KO mice were sampled at ~5 (5.3) weeks of age. Table 4. Mouse study groups
Figure imgf000080_0001
PATENT ATTORNEY DOCKET NO.51037-075WO2 Test article volumes used prior to dosing are summarized in Table 5, below. Table 5. Test article volumes prior to dosing
Figure imgf000081_0001
PATENT ATTORNEY DOCKET NO.51037-075WO2 The following humane endpoint criteria are applicable for the study. If the mice met the predefined humane endpoint criteria, they underwent a planned euthanasia for welfare reasons. If any of the mice needed to be euthanized for welfare reasons, they were opened, and macroscopic observations were recorded. In case the general health status of an animal deteriorated significantly, a shortened tissue isolation protocol was applied, if possible. The shortened tissue isolation protocol consisted of collecting a skeletal muscle (quadriceps) and 1 liver lobe in liquid nitrogen and 1 liver lobe and heart (dorsal portion) tissue in formalin. The skeletal muscle and liver lobe were stored at - 80ºC while the liver lobe and heart (dorsal portion) in formalin were stored at room temperature. Due to a limited staff capacity during evenings and weekends to perform sampling on acute cases needing immediate action, mice were euthanized by an overdose of CO2 and decapitated. In this case, no sampling was performed. Body weight was measured three times a week and mortality were recorded. If surviving to the end of the study, at endpoint, at ~16-17 weeks of age (~12-13 weeks after dosing), the mice were euthanized by deep anesthetization with sodium pentobarbital (180 mg/kg). As much whole blood was collected as possible via cardiac puncture. Blood was first collected for hematology (100 μL of EDTA whole blood), and then for clinical chemistry (200 μL of whole blood to separate minimum 80 μL of serum). Any remaining serum was collected. Then, the mice were transcardially perfused with heparinized saline to remove blood from the tissues. The group of 10 naïve MTM1 KO mice were sampled at ~5 wks of age. Sampling included blood sampling (whole blood and serum), perfusion, and dissection of samples for histology and histopathology. No samples were taken for other analyses. Tissue samples were used for myotubularin and bile salt transporter expression, bile acid level measurements, and histopathology. In-life and endpoint whole blood samples were used for hematology analysis and serum samples for clinical chemistry analysis. The same portion of the sample from each mouse was used for each analysis. The heart was collected with the left and right atria, then halved in the frontal plane. Tissue collection and analysis is summarized in Table 6, below. Table 6. Mouse tissue collection and analysis
Figure imgf000082_0001
PATENT ATTORNEY DOCKET NO.51037-075WO2
Figure imgf000083_0001
Histology and Histopathology The following tissue samples were dissected, and placed in 10% neutral buffered formalin: • Quadriceps (L. muscle, ½) • Diaphragm (1 sample) • Liver (left lateral lobe and a small part of the right medial lobe (with gall bladder)) • Heart (1/2, dorsal) • Lungs (1 sample) • Kidneys (L + R, 2 samples) • Spleen (1 sample) • Brain (1 sample) Samples were embedded in paraffin, sectioned to slide, and stained with hematoxylin and eosin (H&E) for microscopic evaluation. Sample Collection for: Myotubularin (ELISA), Vector Copy Number (VCN), and/or Bile Acid Analysis (ELISA); Myotubularin and Bile Acid Salt Transporter Analysis by Immunohistochemistry (IHC); and Reserve Tissues Sterile Collection for bioanalytical analysis (Myotubularin (ELISA), Vector Copy Number (VCN) and/or Bile Acid Analysis (ELISA); Myotubularin and Bile Acid Salt Transporter Analysis by IHC; and Reserve Tissues): Each tissue was collected in RNAse-free screw top polypropylene tubes, flash frozen in liquid nitrogen, under sterile conditions, and stored at -80°C according to Testing Facility SOPs. Samples were stored on dry ice prior to storage. Scalpel blades, weigh boats (small dish), and dissecting instruments were replaced/cleaned prior to handling a new mouse. All instruments were wiped with RNAse Zap (or similar) between collection of each tissue. Precautionary measures were in place to prevent cross-contamination during tissue collection. Myotubularin (ELISA), Vector Copy Number (VCN) and/or Bile Acid Analysis (ELISA) The following tissue samples were dissected and frozen in liquid nitrogen: • Quadriceps (R. muscle) • Diaphragm (1 sample) • Liver (right lobe, right lateral lobe, caudate lobe) • Heart (1/2, ventral) (split in two samples) PATENT ATTORNEY DOCKET NO.51037-075WO2 Myotubularin and Bile Acid Salt Transporter Analysis by IHC The following tissue samples were dissected and frozen in liquid nitrogen: • Quadriceps (L. muscle, ½) • Diaphragm (1 sample) • Liver (left medial lobe and remaining right medial lobe without gall bladder) Reserve Tissues The following tissue samples were dissected and frozen in liquid nitrogen: • Triceps (R. muscle) • Gastrocnemius (R. muscle) • Tibialis anterior (R. muscle) Clinical Chemistry Panel At in-life sampling and terminal necropsy, a clinical biochemistry panel of markers was analyzed in unhemolysed serum for the following parameters: alanine aminotransferase (ALAT), aspartate aminotransferase (ASAT), alkaline phosphatase (AFOS), gamma-glutamyl transferase (GGT), lactate dehydrogenase (LDH), creatine kinase (CK), albumin, total bilirubin, total protein, creatinine, bile acids, calculated globulin, calculated albumin/globulin ratio, and urea nitrogen. If the blood sample was limited, analysis priority was given in the following order: 1. Alanine aminotransferase (ALAT) 2. Total/Direct bilirubin 3. Bile acids 4. Aspartate aminotransferase (ASAT) 5. Alkaline phosphatase (AFOS) 6. Gamma-glutamyl transferase (GGT) 7. Lactate dehydrogenase (LDH) 8. Creatine kinase (CK) 9. Albumin 10. Total protein 11. Creatinine 12. Urea Nitrogen Only ALAT was analyzed from in-life samples due to limited serum sample. Hematology Panel At in-life sampling and terminal necropsy, a hematology panel of markers was analyzed in EDTA whole blood for the following parameters: hemoglobin, red blood cell counts, white blood cell counts including relative and absolute differential counts, absolute reticulocyte counts, reticulocyte%, and thrombocyte counts. Additional parameters, if assay quantity of whole blood allowed, included: hematocrit, mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular PATENT ATTORNEY DOCKET NO.51037-075WO2 hemoglobin concentration. Genotyping Samples A tail sample was taken at the endpoint sampling for possible regenotyping with regular PCR. Statistical Analysis and Data Graphical Presentation Prior to further statistical analysis, data quality checks and validations were performed. During that process, potential outliers were identified and assessed. No outliers were removed from the data without a clear justification for removal (e.g., identified measurement error in the laboratory notes). The planned comparisons in this study were: • Group 1 vs. Group 2 and Group 7 • Group 2 vs. Groups 3-6 The assumption of normality of each data set was primarily based on experience (e.g., data within a population is known to be approximately Gaussian) and observations during the validation phase. Some biological variables are known to follow lognormal distributions – in these cases, the data is first transformed to logarithms, and then a parametric statistical test can be used. The same normality assumption was used for a series of experiments or a group of similar readouts from an assay. In addition, the D’Agostino-Pearson omnibus normality test was used for clinical chemistry and hematology data to support the decision whether parametric or nonparametric tests were to be used. The summary of readouts for statistical analysis and visualization is summarized in Table 7, below. Table 7. Summary of readouts for statistical analysis and visualization
Figure imgf000085_0001
Descriptive statistics, including group size, mean, SD, and SEM were provided for each parameter. Statistical Analysis of Single Time Point Data Simple comparisons between two groups were performed using unpaired Welch’s t-test, or, when the assumption of normality or lognormality was not met, by the Mann-Whitney U-test. Repeated Observations of the Same Subjects (Longitudinal Data) Comparisons between the two groups were performed using a two-way Mixed-effects model (Mixed Anova) with Geisser-Greenhouse correction (not assuming sphericity), followed by Fisher’s LSD test between the two groups performed for each time point. Time, group, and the interaction of Time x Group were the fixed effects, and the individual subject was the random effect. PATENT ATTORNEY DOCKET NO.51037-075WO2 In the case of longitudinal comparison of three or more groups, the two-way Mixed-effects model with Geisser-Greenhouse correction, followed by Dunnett’s multiple comparisons test was performed. One family per time point was set. Time, group, and the interaction of Time x Group were the fixed effects, and the individual subject was the random effect. The following sections provide detailed results for certain of the parameters mentioned in Table 6 above. Results Results of this study provided an indication of safety and efficacy of the compositions and methods described above. Body Weight Body weight was significantly lower in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice (Two-Way-ANOVA: Group: p < 0.0001) at all time points (Fisher’s LSD multiple comparison: p < 0.001, for all) (FIG.5). Body weight was significantly less in WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP mice compared to WT Vehicle mice (Two-Way-ANOVA: Group: p < 0.05) on weeks 7-16 (Fisher’s LSD multiple comparison: p < 0.05, for all) (FIG.5). Body weight was significantly altered among HEMI mice (Two-Way-ANOVA: Group effect: NS, Group x Time interaction effect: p < 0.0001). Post-hoc comparisons revealed that body weight of HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice were significantly decreased on weeks 4 and 5 (Dunnett’s multiple comparison: p < 0.05, for both) and body weight of HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP mice were significantly decreased on weeks 5-6 and 8-9 compared to HEMI AAV8-Des-mMTM1 mice (Dunnett’s multiple comparison: p < 0.05, for all). Mortality No data from animals that died or were euthanized prior to dosing is shown. No mortality was observed in groups 1-3, 5, and 7. Two mice in each of groups 4, 6, and naive were found dead prior to dosing due to factors unrelated to the test articles. Clinical Chemistry All the clinical chemistry assessments were performed only from terminal samples (week 16 or week 5 for naïve mice) for all groups, with the exception of ALAT that was analyzed from both in- life and terminal samples (weeks 4-10). Data from clinical chemistry analysis is shown in FIGS.6A- 19B. Alkaline Phosphatase (AFOS) There were no statistically significant differences between the groups in AFOS levels at 16 weeks of age (FIG.6A). PATENT ATTORNEY DOCKET NO.51037-075WO2 AFOS levels from naïve mice at 5 weeks of age are shown in FIG.6B. Alanine Aminotransferase (ALAT) ALAT levels were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 6- and 16-week time points (Two-Way-ANOVA: Group effect: p = 0.0005) (Fisher’s LSD multiple comparison: p < 0.05, for both) (FIG.7A). ALAT levels were significantly increased at 10 weeks of age in HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 mice compared to HEMI AAV8-Des-mMTM1 mice (One-Way ANOVA: Group effect: p = 0.0026, Dunnett’s post-hoc comparison: p < 0.05) (FIG.7A). ALAT levels from naïve mice at 5 weeks of age are shown in FIG.7B. Aspartate Aminotransferase (ASAT) ASAT levels were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test: p < 0.01) (FIG.8A). ASAT levels from naïve mice at 5 weeks of age are shown in FIG.8B. Albumin There were no statistically significant differences between the groups in albumin levels at 16 weeks of age (FIG.9A). Albumin levels from naïve mice at 5 weeks of age are shown in FIG.9B. Gamma-Glutamyl Transferase (GGT) GGT levels were significantly increased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice and HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP mice compared to HEMI AAV8-Des-mMTM1 mice at 16 weeks of age (One-Way ANOVA: Group effect: p = 0.0013, Dunnett’s post-hoc comparison: p < 0.05, for both) (FIG.10A). GGT levels from naïve mice at 5 weeks of age are shown in FIG.10B. Total Protein There were no statistically significant differences between the groups in total protein levels at 16 weeks of age (FIG.11A). Total protein levels from naïve mice at 5 weeks of age are shown in FIG.11B. Urea There were no statistically significant differences between the groups in urea levels at 16 weeks of age (FIG.12A). Urea levels from naïve mice at 5 weeks of age are shown in FIG.12B. Lactate Dehydrogenase (LDH) There were no statistically significant differences between the groups in LDH levels at 16 PATENT ATTORNEY DOCKET NO.51037-075WO2 weeks of age (FIG.13A). LDH levels from naïve mice at 5 weeks of age are shown in FIG.13B. Bile Acids Bile acids levels were significantly increased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice compared to HEMI AAV8-Des-mMTM1 mice at 16 weeks of age (One-Way ANOVA: Group effect: p = 0.0258, Dunnett’s post-hoc comparison: p < 0.05) (FIG.14A). However, it should be noted that most of the bile acids values were under the detection level in all treatment groups. Bile acid levels from naïve mice at 5 weeks of age are shown in FIG.14B. Total Bilirubin There were no statistically significant differences between the groups in total bilirubin levels at 16 weeks of age (FIG.15). No total bilirubin data was available from naïve mice at 5 weeks of age (all values were below the detection level). Calculated Globulin Calculated globulin levels were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test: p < 0.05) (FIG.16A). Calculated globulin levels from naïve mice at 5 weeks of age are shown in FIG.16B. Calculated Albumin/Globulin Ratio There were no statistically significant differences between the groups in calculated albumin/globulin ratio levels at 16 weeks of age (FIG.17A). Albumin/Globulin ratio levels from naïve mice at 5 weeks of age are shown in FIG.17B. Creatine Kinase CK levels were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test: p < 0.05) (FIG.18A). CK levels from naïve mice at 5 weeks of age are shown in FIG.18B. Creatinine There were no statistically significant differences between the groups in creatinine levels at 16 weeks of age (FIG.19A). Creatinine levels from naïve mice at 5 weeks of age are shown in FIG.19B. Hematology Due to different blood sampling techniques between in-life sampling (saphenous vein, no anesthesia) and terminal sampling (cardiac puncture, pentobarbital anesthesia), statistical analysis was performed separately for in-life samples (weeks 4-10) and terminal samples (week 16). It has PATENT ATTORNEY DOCKET NO.51037-075WO2 been reported that significant differences in blood parameters, for example, the number of total white blood cells, result from different blood sampling techniques (Hoggatt et al. Exp Hematol.44(2):132– 137.e1. (2016)). Data from hematology analysis is shown in FIGS.20A-41B. White Blood Cell Count (WBC) White blood cell counts were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test, p < 0.05) (FIG.20A). White blood cell counts were significantly increased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice at 7 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: NS, Group x Time interaction effect: p < 0.05, Dunnett’s post-hoc comparison: p < 0.05) (FIG.20A). White blood cell counts from naïve mice at 4 and 5 weeks of age are shown in FIG.20B. Red Blood Cell Count (RBC) There were no statistically significant differences between the groups in red blood cell counts (FIG.21A). Red blood cell counts from naïve mice at 4 and 5 weeks of age are shown in FIG.21B. Hemoglobin (HGB) There were no statistically significant differences between the groups in hemoglobin levels (FIG.22A). Hemoglobin levels from naïve mice at 4 and 5 weeks of age are shown in FIG.22B. Hematocrit (HCT) Hematocrit levels were significantly increased in HEMI AAV8-Des-mMTM1 + AAV8-APoE- A1AT-mMTM1 mice compared to HEMI AAV8-Des-mMTM1 mice at 6 weeks of age (Two-Way ANOVA: Group effect: p < 0.05, Dunnett’s post-hoc comparison: p < 0.05) (FIG.23A). Hematocrit levels from naïve mice at 4 and 5 weeks of age are shown in FIG.23B. Mean Corpuscular Volume (MCV) MCV levels were significantly decreased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 4 and 5 weeks of age (t-test, p < 0.01, for all) (FIG.24A). MCV levels from naïve mice at 4 and 5 weeks of age are shown in FIG.24B. Mean Corpuscular Hemoglobin (MCH) There were no statistically significant differences between the groups in MCH levels (FIG. 25A). MCH levels from naïve mice at 4 and 5 weeks of age are shown in FIG.25B. PATENT ATTORNEY DOCKET NO.51037-075WO2 Mean Corpuscular Hemoglobin Concentration (MCHC) MCHC levels were significantly decreased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice at 5 and 16 weeks of age and in HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 mice at 6 weeks of age, and increased in HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8- Des-hMTM1-STOP mice at 7 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: NS, Group x Time interaction effect: p < 0.01, Dunnett’s post-hoc comparison: p < 0.05, for all) (FIG.26A). MCHC levels from naïve mice at 4 and 5 weeks of age are shown in FIG.26B. Thrombocyte Count (PLT) Thrombocyte or platelet counts were significantly decreased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 6 weeks of age (Two-Way ANOVA: Group effect: p < 0.05, Dunnett’s post-hoc comparison: p < 0.01) (FIG.27A). Thrombocyte or platelet counts from naïve mice at 4 and 5 weeks of age are shown in FIG. 27B. Neutrophil Count (Relative) (Neut(%)) There were no statistically significant differences between the groups in relative neutrophil counts (FIG.28A). Relative neutrophil counts from naïve at 4 and 5 weeks of age mice are shown in FIG.28B. Neutrophil Count (Absolute) (Neut) There were no statistically significant differences between the groups in absolute neutrophil counts (FIG.29A). Absolute neutrophil counts from naïve mice at 4 and 5 weeks of age are shown in FIG.29B. Lymphocyte Count (Relative) (Lymph(%)) Relative lymphocyte counts were significantly decreased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice at 5 and 6 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: NS, Group x Time interaction effect: p < 0.05, Dunnett’s post-hoc comparison: p < 0.05, for all) (FIG.30A). Relative lymphocyte counts from naïve mice are shown in FIG.30B. Lymphocyte Count (Absolute) (Lymph) Absolute lymphocyte counts were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test, p < 0.01) (FIG.31A). Absolute lymphocyte counts from naïve mice at 4 and 5 weeks of age are shown in FIG.31B. Monocyte Count (Relative) (Mono(%)) Relative monocyte counts were significantly increased in WT AAV8-Des-mMTM1 + AAV8- PATENT ATTORNEY DOCKET NO.51037-075WO2 Des-hMTM1-STOP mice compared to WT Vehicle mice at 16 weeks of age (t-test, p < 0.05) (FIG. 32A). Relative monocyte counts from naïve mice at 4 and 5 weeks of age are shown in FIG.32B. Monocyte Count (Absolute) (Mono) Absolute monocyte counts were significantly increased in HEMI AAV8-Des-mMTM1 mice compared to WT Vehicle mice at 16 weeks of age (t-test, p < 0.01) (FIG.33A). Absolute monocyte counts from naïve mice at 4 and 5 weeks of age are shown in FIG.33B. Eosinophil Count (Relative) (Eos(%)) Eosinophil counts were significantly increased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice at 4 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: p < 0.05, Dunnett’s post-hoc comparison: p < 0.01) (FIG.34A). Eosinophil counts from naïve mice at 4 and 5 weeks of age are shown in FIG.34B. Eosinophil Count (Absolute) (Eos) Absolute eosinophil counts were significantly increased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice at 5 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: p = 0.0001, Dunnett’s post-hoc comparison: p < 0.05) (FIG.35A). Absolute eosinophil counts from naïve mice at 4 and 5 weeks of age are shown in FIG.35B. Basophil Count (Relative) (Baso(%)) Relative basophil counts significantly decreased in HEMI AAV8-Des-mMTM1 + AAV8 empty capsid mice and in HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1- STOP mice at 5 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: NS, Group x Time interaction effect: p < 0.01, Dunnett’s post-hoc comparison: p < 0.05) (FIG. 36A). Relative basophil counts from naïve mice at 4 and 5 weeks of age are shown in FIG.36B. Basophil Count (Absolute) (Baso) There were no statistically significant differences between the groups in absolute basophil counts (FIG.37A). Absolute basophil counts from naïve mice at 4 and 5 weeks of age are shown in FIG.37B. Large Unstained Cell Count (Relative) (LUC(%)) Relative count of large unstained cells was significantly increased in WT AAV8-Des-mMTM1 + AAV8-Des-hMTM1-STOP mice at 7 weeks of age compared to WT Vehicle mice (Two-Way ANOVA: Group effect: p < 0.05, Dunnett’s post-hoc comparison: p < 0.01) (FIG.38A). Relative count of large unstained cells from naïve mice at 4 and 5 weeks of age are shown in FIG.38B. PATENT ATTORNEY DOCKET NO.51037-075WO2 Large Unstained Cell Count (Absolute) (LUC) Absolute count of large unstained cells was significantly decreased in HEMI AAV8-Des- mMTM1 + AAV8-APoE-A1AT-mMTM1 + AAV8-Des-hMTM1-STOP mice at 6 weeks of age and in HEMI AAV8-Des-mMTM1 + AAV8-APoE-A1AT-mMTM1 mice at 10 weeks of age compared to HEMI AAV8-Des-mMTM1 mice (Two-Way ANOVA: Group effect: NS, Group x Time interaction effect: p < 0.05, Dunnett’s post-hoc comparison: p < 0.05) (FIG.39A). Absolute count of large unstained cells from naïve mice at 4 and 5 weeks of age are shown in FIG.39B. Reticulocyte Count (Relative) (Retic(%)) There were no statistically significant differences between the groups in relative reticulocyte counts (FIG.40A). Relative reticulocyte counts from naïve mice at 4 and 5 weeks of age are shown in FIG.40B. Reticulocyte Cell Count (Absolute) (Retic) There were no statistically significant differences between the groups in absolute reticulocyte cell counts (FIG.41A). Absolute reticulocyte cell counts from naïve mice at 4 and 5 weeks of age are shown in FIG. 41B. Conclusion The results of the experiments described above support the safety and efficacy of AAV vectors encoding an MTM1 transgene under the control of a liver-directed regulatory element, optionally in combination with AAV vectors encoding the MTM1 transgene under the control of a muscle-directed regulatory element, for the treatment of XLMTM. Example 2. Treatment of X-Linked myotubular myopathy in human patients by concurrent administration of a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding a myotubularin 1 gene operably linked to a liver-specific promoter and a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding a myotubularin 1 gene operably linked to a desmin promoter Using the compositions and methods of the disclosure, a patient having XLMTM may be concurrently administered a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter (FIG.1 and FIG.2) and a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec), each in a dose of less than about 3 x 1014 vg/kg (e.g., in an amount of less than about 3 x 1014 vg/kg, 2.9 x 1014 vg/kg, 2.8 x 1014 vg/kg, 2.7 x 1014 vg/kg, 2.6 x 1014 vg/kg, 2.5 x 1014 vg/kg, 2.4 x 1014 vg/kg, 2.3 x 1014 vg/kg, 2.2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 PATENT ATTORNEY DOCKET NO.51037-075WO2 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). Upon concurrently administering the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter (FIG.1 and FIG.2) and the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient, the patient displays a change from baseline in maximal inspiratory pressure. For example, the patient displays the change from baseline in maximal inspiratory pressure by about 24 weeks (e.g., by about 20 weeks, 16 weeks, 12 weeks, 8 weeks, or 4 weeks) after concurrent administration of the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a liver- specific promoter (FIG.1 and FIG.2) and the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient. Example 3. Treatment of X-Linked myotubular myopathy in human patients by a pseudotyped AAV2/8 vector including both a nucleic acid sequence encoding a myotubularin 1 gene operably linked to a liver-specific promoter and a nucleic acid sequence encoding a myotubularin 1 gene operably linked to a desmin promoter Using the compositions and methods of the disclosure, a patient having XLMTM may be administered a pseudotyped AAV2/8 vector including both a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter and a nucleic acid sequence encoding an MTM1 gene operably linked to a muscle-specific promoter (FIG.3) in a dose of less than about 3 x 1014 vg/kg (e.g., in an amount of less than about 3 x 1014 vg/kg, 2.9 x 1014 vg/kg, 2.8 x 1014 vg/kg, 2.7 x 1014 vg/kg, 2.6 x 1014 vg/kg, 2.5 x 1014 vg/kg, 2.4 x 1014 vg/kg, 2.3 x 1014 vg/kg, 2.2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). Upon administering the AAV2/8 vector including both a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter and a nucleic acid sequence encoding an MTM1 gene operably linked to a muscle-specific promoter (FIG.3) to the patient, the patient exhibits a change from baseline in hours of mechanical ventilation support over time. For example, the patient exhibits the change from baseline in hours of mechanical ventilation support over time by about 24 weeks (e.g., by about 20 weeks, 16 weeks, 12 weeks, 8 weeks, or 4 weeks) after administration of the AAV2/8 vector including both a nucleic acid sequence encoding an MTM1 gene operably linked to a liver-specific promoter and a nucleic acid sequence encoding an MTM1 gene operably linked to a muscle-specific promoter (FIG.3) to the patient. PATENT ATTORNEY DOCKET NO.51037-075WO2 Example 4. Treatment of X-Linked myotubular myopathy in human patients by a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding a myotubularin 1 gene operably linked to a ubiquitous promoter Using the compositions and methods of the disclosure, a patient having XLMTM may be administered a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a ubiquitous promoter (FIG.4) in a dose of less than about 3 x 1014 vg/kg (e.g., in an amount of less than about 3 x 1014 vg/kg, 2.9 x 1014 vg/kg, 2.8 x 1014 vg/kg, 2.7 x 1014 vg/kg, 2.6 x 1014 vg/kg, 2.5 x 1014 vg/kg, 2.4 x 1014 vg/kg, 2.3 x 1014 vg/kg, 2.2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). Upon administering the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a ubiquitous promoter (FIG.4) to the patient, the patient achieves functionally independent sitting for at least 30 seconds. For example, the patient achieves the functionally independent sitting by about 24 weeks (e.g., by about 20 weeks, 16 weeks, 12 weeks, 8 weeks, or 4 weeks) after administration of the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a ubiquitous promoter (FIG.4) to the patient. Example 5. Treatment of X-Linked myotubular myopathy in human patients by a non-viral vector including a liver-expressing construct in combination with a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding a myotubularin 1 gene operably linked to a desmin promoter Using the compositions and methods of the disclosure, a patient having XLMTM may be administered a non-viral vector (e.g. a lipid nanoparticle) including a liver-expressing construct in combination with a pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec), with the AAV2/8 vector administered in a dose of less than about 3 x 1014 vg/kg (e.g., in an amount of less than about 3 x 1014 vg/kg, 2.9 x 1014 vg/kg, 2.8 x 1014 vg/kg, 2.7 x 1014 vg/kg, 2.6 x 1014 vg/kg, 2.5 x 1014 vg/kg, 2.4 x 1014 vg/kg, 2.3 x 1014 vg/kg, 2.2 x 1014 vg/kg, 2.1 x 1014 vg/kg, 2 x 1014 vg/kg, 1.9 x 1014 vg/kg, 1.8 x 1014 vg/kg, 1.7 x 1014 vg/kg, 1.6 x 1014 vg/kg, 1.5 x 1014 vg/kg, 1.4 x 1014 vg/kg, 1.3 x 1014 vg/kg, 1.2 x 1014 vg/kg, 1.1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1014 vg/kg, 1 x 1013 vg/kg, 1 x 1012 vg/kg, 1 x 1011 vg/kg, 1 x 1010 vg/kg, 1 x 109 vg/kg, 1 x 108 vg/kg, or less). Upon administering the non-viral vector (e.g. a lipid nanoparticle) including a liver-expressing construct in combination with the pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient, the patient achieves functionally independent sitting for at least 30 seconds. For example, the patient achieves functionally independent sitting for at least 30 seconds by about 24 weeks (e.g., by about 20 weeks, 16 weeks, 12 weeks, 8 weeks, or 4 weeks) after administration of the non-viral vector (e.g., a lipid nanoparticle) including a liver-expressing construct in combination with the PATENT ATTORNEY DOCKET NO.51037-075WO2 pseudotyped AAV2/8 vector including a nucleic acid sequence encoding an MTM1 gene operably linked to a desmin promoter (e.g., resamirigene bilparvovec) to the patient.

Claims

PATENT ATTORNEY DOCKET NO.51037-075WO2 CLAIMS 1. A recombinant adeno-associated viral (AAV) vector comprising a transgene encoding myotubularin 1 (MTM1), wherein the transgene is operably linked to a promoter that is active in liver tissue. 2. The AAV vector of claim 1, wherein the promoter is selectively active in liver tissue. 3. The AAV vector of claim 2, wherein upon separately contacting the vector with one or more liver cells and one or more non-liver cells (e.g., in vitro or in vivo) under equivalent conditions, the vector results in a level of expression of the transgene in the one or more liver cells that is greater than the level of expression of the transgene in the one or more non-liver cells. 4. The AAV vector of claim 3, wherein upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is from 2-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells. 5. The AAV vector of claim 4, wherein upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is from 50-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells. 6. The AAV vector of claim 5, wherein upon separately contacting the vector with the one or more liver cells and the one or more non-liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is from 100-fold to 1,000-fold greater than the level of expression of the transgene in the one or more non-liver cells. 7. The AAV vector of claim 2, wherein upon contacting the vector with the one or more liver cells, the vector results in a level of expression of the transgene in the one or more liver cells that is at least 0.2-fold, at least 0.5-fold, at least 1-fold, at least 2-fold, at least 5-fold, or at least 10-fold the level of wild-type MTM1 expression in a healthy liver cell. 8. The AAV vector of any one of claims 3-6, wherein the non-liver cells are muscle cells or neural cells. 9. The AAV vector of any one of claims 3-6, wherein the non-liver cells are cardiac cells. 10. The AAV vector of any one of claims 1-9, wherein the promoter comprises an LP1 promoter. 11. The AAV vector of claim 10, wherein the LP1 promoter comprises a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 3. PATENT ATTORNEY DOCKET NO.51037-075WO2 12. The AAV vector of claim 11, wherein the LP1 promoter comprises a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 3, optionally wherein the LP1 promoter comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 3. 13. The AAV vector of claim 12, wherein the LP1 promoter comprises the nucleic acid sequence of SEQ ID NO: 3. 14. The AAV vector of any one of claims 1-13, wherein the promoter comprises an apolipoprotein E (ApoE) promoter. 15. The AAV vector of claim 14, wherein the ApoE promoter comprises a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 8. 16. The AAV vector of claim 15, wherein the ApoE promoter comprises a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 8, optionally wherein the ApoE promoter comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 8. 17. The AAV vector of claim 16, wherein the ApoE promoter comprises the nucleic acid sequence of SEQ ID NO: 8. 18. The AAV vector of any one of claims 1-17, wherein the promoter comprises an alpha-1- antitrysin (A1AT) promoter. 19. The AAV vector of claim 18, wherein the A1AT promoter comprises a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 9. 20. The AAV vector of claim 19, wherein the A1AT promoter comprises a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 9, optionally wherein the A1AT promoter comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 9. 21. The AAV vector of claim 20, wherein the A1AT promoter comprises the nucleic acid sequence of SEQ ID NO: 9. 22. The AAV vector of any one of claims 1-21, wherein the promoter is a chimeric promoter comprising an ApoE promoter and an A1AT promoter. 23. The AAV vector of claim 22, wherein the chimeric promoter comprises a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 2. PATENT ATTORNEY DOCKET NO.51037-075WO2 24. The AAV vector of claim 23, wherein the chimeric promoter comprises a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 2, optionally wherein the chimeric promoter comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 2. 25. The AAV vector of claim 24, wherein the chimeric promoter comprises the nucleic acid sequence of SEQ ID NO: 2. 26. The AAV vector of any one of claims 1-25, wherein the promoter comprises a constitutive promoter. 27. The AAV vector of claim 26, wherein the constitutive promoter is a phosphoglycerate kinase (PGK) promoter, an elongation factor-1 alpha (EF1alpha) promoter, a glyceraldehyde 3- phosphate dehydrogenase (GAPDH) promote, a cytomegalovirus (CMV) promoter, or a chicken-β- actin (CBA) promoter. 28. The AAV vector of any one of claims 1-27, wherein the vector further comprises a second transgene encoding MTM1. 29. The AAV vector of claim 28, wherein the second transgene encoding MTM1 is operably linked to a promoter that is active in muscle tissue. 30. The AAV vector of claim 29, wherein the promoter that is active in muscle tissue is selectively active in muscle tissue. 31. The AAV vector of claim 30, wherein the promoter that is selectively active in muscle tissue effectuates (e.g., in vitro or in vivo) a level of expression of the MTM1 transgene in muscle cells that is from 2-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. 32. The AAV vector of claim 31, wherein the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 10-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. 33. The AAV vector of claim 32, wherein the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 50-fold to 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. 34. The AAV vector of claim 33, wherein the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is from 100-fold to PATENT ATTORNEY DOCKET NO.51037-075WO2 1,000-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. 35. The AAV vector of claim 34, wherein the promoter that is selectively active in muscle tissue effectuates a level of expression of the MTM1 transgene in muscle cells that is at least 2-fold, at least 5-fold, at least 10-fold, at least 50-fold, or at least 100-fold greater than the level of expression of the MTM1 transgene effectuated by the same promoter in non-muscle cells. 36. The AAV vector of any one of claims 28-35, wherein the second transgene encoding MTM1 is operably linked to a muscle creatine kinase (MCK) promoter or a desmin (DES) promoter. 37. The AAV vector of any one of claims 1-36, wherein each transgene encoding MTM1, independently, comprises: a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 6; or a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 7. 38. The AAV vector of claim 37, wherein each transgene encoding MTM1, independently, comprises: a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1, optionally wherein each transgene encoding MTM1, independently, comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 6, optionally wherein each transgene encoding MTM1, independently, comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 6; or a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 7, optionally wherein each transgene encoding MTM1, independently, comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 7. 39. The AAV vector of claim 38, wherein each transgene encoding MTM1 comprises: the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or the nucleic acid sequence of SEQ ID NO: 6; or the nucleic acid sequence of SEQ ID NO: 7. 40. The AAV vector of any one of claims 1-39, wherein the AAV is an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAVrh10, or AAVrh74 serotype. PATENT ATTORNEY DOCKET NO.51037-075WO2 41. The AAV vector of any one of claims 1-40, wherein the AAV vector is a pseudotyped AAV. 42. The AAV vector of claim 41, wherein the pseudotyped AAV is AAV2/8 or AAV2/9, optionally wherein the pseudotyped AAV is AAV2/8. 43. A method of treating X-linked myotubular myopathy (XLMTM) in a human patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of the AAV vector of any one of claims 1-42. 44. The method of claim 43, wherein the patient is five years old or younger at the time of administration of the AAV vector. 45. The method of claim 44, wherein the patient is four years old or younger at the time of administration of the AAV vector, optionally wherein the patient is three years old or younger, two years old or younger, one year old or younger, or six months old or younger. 46. The method of any one of claims 43-45, wherein the AAV vector is administered to the patient in an amount of less than 3 x 1014 vg/kg. 47. The method of claim 46, wherein the AAV vector is administered to the patient in an amount of less than 2.5 x 1014 vg/kg, optionally wherein the AAV vector is administered to the patient in an amount of less than 2 x 1014 vg/kg, less than 1.5 x 1014 vg/kg, or less than 1.4 x 1014 vg/kg. 48. The method of any one of claims 43-45, wherein the AAV vector is administered to the patient in an amount of from 3 x 1013 vg/kg to 2.3 x 1014 vg/kg, optionally wherein the AAV vector is administered to the patient in an amount of from 8 x 1013 vg/kg to 1.8 x 1014 vg/kg, from 1 x 1014 vg/kg to 1.6 x 1014 vg/kg, from 1.1 x 1014 vg/kg to 1.5 x 1014 vg/kg, or from 1.2 x 1014 vg/kg to 1.4 x 1014 vg/kg. 49. The method of any one of claims 43-48, wherein the AAV vector is administered to the patient in an amount of about 1.3 x 1014 vg/kg. 50. The method of any one of claims 43-49, wherein the AAV vector is administered to the patient by way of intravenous, intramuscular, intrahepatic, intradermal, or subcutaneous administration. 51. The method of any one of claims 43-50, wherein the patient is further administered an anti-cholestatic agent. 52. The method of claim 51, wherein the anti-cholestatic agent is selected from the group consisting of a bile acid, a farnesoid X receptor (FXR) ligand, a fibroblast growth factor 19 (FGF-19) mimetic, a Takeda-G-protein-receptor-5 (TGR5) agonist, a peroxisome proliferator-activated receptor (PPAR) agonist, a PPAR-alpha agonist, a PPAR-delta agonist, a dual PPAR-alpha and PPAR-delta agonist, an apical sodium-dependent bile acid transporter (ASBT) inhibitor, an immunomodulatory PATENT ATTORNEY DOCKET NO.51037-075WO2 drug, an antifibrotic therapy, and a nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitor. 53. The method of claim 52, wherein: (i) the FXR ligand is obeticholic acid, cilofexor, tropifexor, tretinoin, or EDP-305; (ii) the FGF-19 mimetic is aldafermin; (iii) the TGR5 agonist is INT-777 or INT-767; (iv) the PPAR agonist is bezafibrate, seladelpar, or elafibrinor; (v) the PPAR-alpha agonist is fenofibrate; (vi) the PPAR-delta agonist is seladelpar; (vii) the dual PPAR-alpha and PPAR-delta agonist is elafibranor; (viii) the ASBT inhibitor is odevixibat, maralixibat, or linerixibat; (ix) the immunomodulatory drug is rituximab, abatacept, ustekinumab, infliximab, baricitinib, or FFP-104; (x) the antifibrotic therapy is a vitamin D receptor agonist or simtuzumab; and/or (xi) the NOX inhibitor is setanaxib. 54. The method of claim 52, wherein the bile acid is ursodeoxycholic acid, nor- ursodeoxycholic acid, or a pharmaceutically acceptable salt thereof. 55. The method of any one of claims 43-54, wherein the patient does not have a history of cholestasis or hyperbilirubinemia. 56. The method of any one of claims 43-55, wherein the patient does not have a history of any underlying liver disease. 57. The method of any one of claims 43-56, wherein the method comprises administering to the patient therapeutically effective amounts of: (i) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in liver tissue, and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue. 58. The method of claim 57, wherein the promoter that is active in liver tissue comprises an LP1 promoter. 59. The method of claim 58, wherein the LP1 promoter comprises a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 3. 60. The method of claim 59, wherein the LP1 promoter comprises a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 3, optionally wherein the LP1 promoter comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 3. PATENT ATTORNEY DOCKET NO.51037-075WO2 61. The method of claim 60, wherein the LP1 promoter comprises the nucleic acid sequence of SEQ ID NO: 3. 62. The method of any one of claims 57-61, wherein the promoter that is active in liver tissue comprises an ApoE promoter. 63. The method of claim 62, wherein the ApoE promoter comprises a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 8. 64. The method of claim 63, wherein the ApoE promoter comprises a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 8, optionally wherein the ApoE promoter comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 8. 65. The method of claim 64, wherein the ApoE promoter comprises the nucleic acid sequence of SEQ ID NO: 8. 66. The method of any one of claims 57-65, wherein the promoter that is active in liver tissue comprises an A1AT promoter. 67. The method of claim 66, wherein the A1AT promoter comprises a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 9. 68. The method of claim 67, wherein the A1AT promoter comprises a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 9, optionally wherein the A1AT promoter comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 9. 69. The method of claim 68, wherein the A1AT promoter comprises the nucleic acid sequence of SEQ ID NO: 9. 70. The method of any one of claims 57-69, wherein the promoter that is active in liver tissue is a chimeric promoter comprising an ApoE promoter and an A1AT promoter. 71. The method of claim 70, wherein the chimeric promoter comprises a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 2. 72. The method of claim 71, wherein the chimeric promoter comprises a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 2, optionally wherein the chimeric promoter comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 2. PATENT ATTORNEY DOCKET NO.51037-075WO2 73. The method of claim 72, wherein the chimeric promoter comprises the nucleic acid sequence of SEQ ID NO: 2. 74. The method of any one of claims 57-73, wherein the promoter that is active in muscle tissue comprises an MCK promoter or a DES promoter. 75. A method of treating XLMTM in a human patient in need thereof, the method comprising administering to the patient therapeutically effective amounts of: (i) a non-viral composition comprising a nucleic acid encoding MTM1 operably linked to a promoter that is active (e.g., selectively active) in liver tissue, and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue. 76. The method of claim 75, wherein the non-viral composition is a liposome, vesicle, synthetic vesicle, exosome, synthetic exosome, dendrimer, or nanoparticle. 77. The method of claim 76, wherein the nanoparticle is a lipid nanoparticle. 78. The method of any one of claims 75-77, wherein the promoter is a DES promoter. 79. The method of any one of claims 75-78, wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises: a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 6; or a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 7. 80. The method of claim 79, wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises: a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1, optionally wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 6, optionally wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 6; or a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 7, optionally wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, PATENT ATTORNEY DOCKET NO.51037-075WO2 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 7. 81. The method of claim 80, wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each comprises: the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or the nucleic acid sequence of SEQ ID NO: 6; or the nucleic acid sequence of SEQ ID NO: 7. 82. The method of any one of claims 75-81, wherein the AAV vector is resamirigene bilparvovec. 83. A kit comprising the AAV vector of any one of claims 1-42, wherein the kit further comprises a package insert instructing a user to administer the AAV vector to a patient diagnosed as having XLMTM. 84. A kit comprising: (i) a non-viral composition comprising a nucleic acid encoding MTM1, and (ii) an AAV vector comprising a transgene encoding MTM1 under the control of a promoter that is active (e.g., selectively active) in muscle tissue, wherein the kit further comprises a package insert instructing a user to administer the non-viral composition and the AAV vector to a patient diagnosed as having XLMTM. 85. The kit of claim 84, wherein the non-viral composition is a liposome, vesicle, synthetic vesicle, exosome, synthetic exosome, dendrimer, or nanoparticle. 86. The kit of claim 85, wherein the nanoparticle is a lipid nanoparticle. 87. The kit of any one of claims 84-86, wherein the promoter is a DES promoter. 88. The kit of any one of claims 84-87, wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises: a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 6; or a nucleic acid sequence that is at least 75% identical to the nucleic acid sequence of SEQ ID NO: 1. 89. The kit of claim 88, wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises: a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1, optionally wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises a nucleic acid sequence that is at least 81%, 82%, PATENT ATTORNEY DOCKET NO.51037-075WO2 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 6, optionally wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 6; or a nucleic acid sequence that is at least 80% identical to the nucleic acid sequence of SEQ ID NO: 7, optionally wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each, independently, comprises a nucleic acid sequence that is at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleic acid sequence of SEQ ID NO: 7. 90. The kit of claim 89, wherein the nucleic acid encoding MTM1 and the transgene encoding MTM1 each comprises: the nucleic acid sequence of residues 4927-6748 of SEQ ID NO: 1; or the nucleic acid sequence of SEQ ID NO: 6; or the nucleic acid sequence of SEQ ID NO: 7. 91. The kit of any one of claims 84-90, wherein the AAV vector is resamirigene bilparvovec.
PCT/US2023/074909 2022-09-23 2023-09-22 Compositions and methods for the treatment of neuromuscular disorders WO2024064895A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263409423P 2022-09-23 2022-09-23
US63/409,423 2022-09-23

Publications (2)

Publication Number Publication Date
WO2024064895A2 true WO2024064895A2 (en) 2024-03-28
WO2024064895A3 WO2024064895A3 (en) 2024-05-10

Family

ID=90455301

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/074909 WO2024064895A2 (en) 2022-09-23 2023-09-22 Compositions and methods for the treatment of neuromuscular disorders

Country Status (1)

Country Link
WO (1) WO2024064895A2 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11072801B2 (en) * 2014-01-21 2021-07-27 Vrije Universiteit Brussel Muscle-specific nucleic acid regulatory elements and methods and use thereof
SG11202000764RA (en) * 2017-08-09 2020-02-27 Bioverativ Therapeutics Inc Nucleic acid molecules and uses thereof
JP2022507402A (en) * 2018-11-19 2022-01-18 ユニキュアー アイピー ビー.ブイ. Liver-specific virus promoter and how to use it
CA3210955A1 (en) * 2021-02-09 2022-08-18 Affinia Therapeutics Inc. Recombinant aavs with improved tropism and specificity

Also Published As

Publication number Publication date
WO2024064895A3 (en) 2024-05-10

Similar Documents

Publication Publication Date Title
KR102526711B1 (en) Adeno-associated virus vector variants for high efficiency genome editing and methods thereof
CA2328447C (en) Adeno-associated virus vectors and uses thereof
US7241447B1 (en) Adeno-associated virus vectors and uses thereof
CA2386546A1 (en) Adeno-associated viruses and uses thereof
JP2018509164A (en) Recombinant Glut1 adeno-associated virus vector construct and method for restoring Glut1 expression
JP2021512649A (en) Transcriptional regulatory elements and their use
CA3005474A1 (en) Compositions and methods for correction of heritable ocular disease
WO2024094044A1 (en) Optimized pah gene and expression cassette and use thereof
EP1849872A1 (en) Adeno-associated virus vectors and uses thereof
EP4090382A2 (en) Adeno associated virus based gene therapy for phenylketonuria
JP2023505851A (en) Adeno-associated virus vectors for the treatment of Hunter&#39;s disease
WO2024064895A2 (en) Compositions and methods for the treatment of neuromuscular disorders
WO2023150051A1 (en) Compositions and methods of using two-promoter vector for treatment of lysosomal storage disorders
EP4374880A2 (en) Adeno-associated virus vector delivery of alpha-sarcoglycan and the treatment of muscular dystrophy
CN116887868A (en) Treatment of darinopathies
TW202116359A (en) Compositions and methods for treating glycogen storage disorders
WO2020210592A1 (en) Recombinant aav gene therapy for ngyl1 deficiency
US20240148906A1 (en) Compositions and methods for treating x-linked myotubular myopathy
AU2022283262A1 (en) Compositions and methods for improved treatment of x-linked myotubular myopathy
US20220403417A1 (en) Aav-based delivery of thymine kinase 2
WO2023196863A1 (en) Adeno-associated virus potency assay and uses thereof
AU2022327583A1 (en) Compositions and methods for improved treatment of disorders affecting the central nervous system
CN117947040A (en) Expression cassette for target gene and application thereof
JP2024515823A (en) Gene Therapy for BCAA Modification in Maple Syrup Urine Disease (MSUD)
AU8003200A (en) Adeno-associated viruses and uses thereof

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: 23869232

Country of ref document: EP

Kind code of ref document: A2