[Skip to Navigation]
Sign In
Figure 1. 
Structure of dystrophin protein. The N-terminal contains the actin-binding domain. H indicates hinge; CR, cysteine-rich region; CT, C-terminal.

Structure of dystrophin protein. The N-terminal contains the actin-binding domain. H indicates hinge; CR, cysteine-rich region; CT, C-terminal.

Figure 2. 
Illustration shows the major components of the dystrophin glycoprotein complex. Within the cytoplasm of the muscle fiber, the N-terminal of dystrophin links to the actin cytoskeleton. The cysteine-rich C-terminal domain of dystrophin links to the membrane via the dystroglycan complex. Dystroglycan consists of β-dystroglycan, a transmembrane protein, and α-dystroglycan, a highly glycosylated extracellular membrane protein. Several G domains of laminin-2 bind α-dystroglycan to link the complex from the extracellular matrix through the membrane to the actin cytoskeleton.

Illustration shows the major components of the dystrophin glycoprotein complex. Within the cytoplasm of the muscle fiber, the N-terminal of dystrophin links to the actin cytoskeleton. The cysteine-rich C-terminal domain of dystrophin links to the membrane via the dystroglycan complex. Dystroglycan consists of β-dystroglycan, a transmembrane protein, and α-dystroglycan, a highly glycosylated extracellular membrane protein. Several G domains of laminin-2 bind α-dystroglycan to link the complex from the extracellular matrix through the membrane to the actin cytoskeleton.

Figure 3. 
Focal vascular delivery of recombinant adeno-associated virus serotype 8–green fluorescent protein (r-AAV8-GFP). Both heads of the gastrocnemius muscle in 6-week-old beagles were treated by focal vascular delivery of rAAV8-GFP through the femoral artery using a fluoroscopy guided catheter. A, At 6 weeks after transfer, more than 80% of the muscle fibers of the gastrocnemius muscle exhibited GFP expression. This section is from the central region of the medial head of the gastrocnemius muscle. B, The hamstring muscle from the same limb that was not targeted by focal vascular delivery shows no GFP expression.

Focal vascular delivery of recombinant adeno-associated virus serotype 8–green fluorescent protein (r-AAV8-GFP). Both heads of the gastrocnemius muscle in 6-week-old beagles were treated by focal vascular delivery of rAAV8-GFP through the femoral artery using a fluoroscopy guided catheter. A, At 6 weeks after transfer, more than 80% of the muscle fibers of the gastrocnemius muscle exhibited GFP expression. This section is from the central region of the medial head of the gastrocnemius muscle. B, The hamstring muscle from the same limb that was not targeted by focal vascular delivery shows no GFP expression.

Figure 4. 
Muscle mass increases after recombinant adeno-associated virus serotype 1 (rAAV1)–follistatin delivery. α-Sarcoglycan knockout animals were injected with 1 × 1011 vector genomes of rAAV1-follistatin bilaterally into the hamstring and gastrocnemius muscles at 3 weeks of age and analyzed 13 weeks later. For control, AAV1-GFP (green fluorescent protein) was used. Scale bars represent 10 cm.

Muscle mass increases after recombinant adeno-associated virus serotype 1 (rAAV1)–follistatin delivery. α-Sarcoglycan knockout animals were injected with 1 × 1011 vector genomes of rAAV1-follistatin bilaterally into the hamstring and gastrocnemius muscles at 3 weeks of age and analyzed 13 weeks later. For control, AAV1-GFP (green fluorescent protein) was used. Scale bars represent 10 cm.

1.
Emery  AE Population frequency of inherited neuromuscular diseases: a world survey.  Neuromuscul Disord 1991;1 (1) 19- 29PubMedGoogle Scholar
2.
Hoffman  EPBrown  RH  JrKunkel  LM Dystrophin: the protein product of the Duchenne muscular dystrophy locus.  Cell 1987;51 (6) 919- 928PubMedGoogle Scholar
3.
Koenig  MHoffman  EPBertelson  CJMonaco  APFeener  CKunkel  LM Complete cloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals.  Cell 1987;50 (3) 509- 517PubMedGoogle Scholar
4.
Sinha  SMishra  SSingh  VMittal  RDMittal  B High frequency of new mutations in North Indian Duchenne/Becker muscular dystrophy patients.  Clin Genet 1996;50 (5) 327- 331PubMedGoogle Scholar
5.
Mendell  JRMoxley  RTGriggs  RC  et al.  Randomized, double-blind six-month trial of prednisone in Duchenne's muscular dystrophy.  N Engl J Med 1989;320 (24) 1592- 1597PubMedGoogle Scholar
6.
Tinsley  JDeconinck  NFisher  R  et al.  Expression of full-length utrophin prevents muscular dystrophy in mdx mice.  Nat Med 1998;4 (12) 1441- 1444PubMedGoogle Scholar
7.
Bogdanovich  SKrag  TOBarton  ER  et al.  Functional improvements of dystrophic muscle by myostatin blockade.  Nature 2002;420 (6914) 418- 421PubMedGoogle Scholar
8.
Biggar  WDGingras  MFehlings  DLHarris  VASteele  CA Deflazacort treatment of Duchenne muscular dystrophy.  J Pediatr 2001;138 (1) 45- 50PubMedGoogle Scholar
9.
Biggar  WDHarris  VAEliasoph  LAlman  B Long-term benefits of deflazacort treatment for boys with Duchenne muscular dystrophy in their second decade [published online ahead of print March 20, 2006].  Neuromuscul Disord2006164249255 doi:10.1016/j.nmd.2006.01.010PubMedGoogle Scholar
10.
Balaban  BMatthews  DJClayton  GHCarry  T Corticosteroid treatment and functional improvement in Duchenne muscular dystrophy: long-term effect.  Am J Phys Med Rehabil 2005;84 (11) 843- 850PubMedGoogle Scholar
11.
Aartsma-Rus  AKaman  WEWeij  Rden Dunnen  JTvan Ommen  GTvan Deutekom  JC Exploring the frontiers of therapeutic exon skipping for Duchenne muscular dystrophy by double targeting within one or multiple exons.  Mol Ther 2006;14 (3) 401- 407PubMedGoogle Scholar
12.
McClorey  GMoulton  HMIversen  PLFletcher  SWilton  SD Antisense oligonucleotide-induced exon skipping restores dystrophin expression in vitro in a canine model of DMD [published online ahead of print May 25, 2006].  Gene Ther 2006;13 (19) 1373- 1381PubMedGoogle Scholar
13.
Barton-Davis  ERCordier  LShoturma  DILeland  SESweeney  HL Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice.  J Clin Invest 1999;104 (4) 375- 381PubMedGoogle Scholar
14.
Hamed  SA Drug evaluation: PTC-124—a potential treatment of cystic fibrosis and Duchenne muscular dystrophy.  IDrugs 2006;9 (11) 783- 789PubMedGoogle Scholar
15.
Liang  KWNishikawa  MLiu  FSun  BYe  OHuang  L Restoration of dystrophin expression in mdx mice by intravascular injection of naked DNA containing full-length dystrophin cDNA.  Gene Ther 2004;11 (11) 901- 908PubMedGoogle Scholar
16.
Scott  JMLi  SHarper  SQ  et al.  Viral vectors for gene transfer of micro-, mini-, or full-length dystrophin.  Neuromuscul Disord 2002;12(suppl 1)S23- S29PubMedGoogle Scholar
17.
Tripathy  SKBlack  HBGoldwasser  ELeiden  JM Immune responses to transgene-encoded proteins limit the stability of gene expression after injection of replication-defective adenovirus vectors.  Nat Med 1996;2 (5) 545- 550PubMedGoogle Scholar
18.
Hartigan-O’Connor  DBarjot  CCrawford  RChamberlain  JS Efficient rescue of gutted adenovirus genomes allows rapid production of concentrated stocks without negative selection.  Hum Gene Ther 2002;13 (4) 519- 531PubMedGoogle Scholar
19.
Verma  IM A tumultuous year for gene therapy [editorial].  Mol Ther 2000;2 (5) 415- 416PubMedGoogle Scholar
20.
Gao  GVandenberghe  LHWilson  JM New recombinant serotypes of AAV vectors.  Curr Gene Ther 2005;5 (3) 285- 297PubMedGoogle Scholar
21.
Schnepp  BCJensen  RLChen  CLJohnson  PRClark  KR Characterization of adeno-associated virus genomes isolated from human tissues.  J Virol 2005;79 (23) 14793- 14803PubMedGoogle Scholar
22.
Gregorevic  PBlankinship  MJAllen  JM  et al Systemic delivery of genes to striated muscles using adeno-associated viral vectors [published online ahead of print July 25, 2004].  Nat Med2004108828834 doi:10.1038/nm1085PubMedGoogle Scholar
23.
Xiao  WChirmule  NBerta  SCMcCullough  BGao  GWilson  JM Gene therapy vectors based on adeno-associated virus type 1.  J Virol 1999;73 (5) 3994- 4003PubMedGoogle Scholar
24.
Wang  ZZhu  TQiao  C  et al Adeno-associated serotype 8 efficiently delivers genes to muscle and heart [published online ahead of print February 27, 2005].  Nat Biotechnol2005233321328 doi:10.1038/nbt1073PubMedGoogle Scholar
25.
Schnepp  BCClark  KRKlemanski  DLPacak  CAJohnson  PR Genetic fate of recombinant adeno-associated virus vector genomes in muscle.  J Virol 2003;77 (6) 3495- 3504PubMedGoogle Scholar
26.
Kotin  RMSiniscalco  MSamulski  RJ  et al.  Site-specific integration by adeno-associated virus.  Proc Natl Acad Sci U S A 1990;87 (6) 2211- 2215PubMedGoogle Scholar
27.
Davé  UPJenkins  NACopeland  NG Gene therapy insertional mutagenesis insights.  Science 2004;303 (5656) 333PubMedGoogle Scholar
28.
Jiang  HCouto  LBPatarroyo-White  S  et al.  Effects of transient immunosuppression on adeno associated virus-mediated, liver-directed gene transfer in rhesus macaques and implications for human gene therapy.  Blood 2006;108 (10) 3321- 3328PubMedGoogle Scholar
29.
Scallan  CDJiang  HLiu  T  et al Human immunoglobulin inhibits liver transduction by AAV vectors at low AAV2 neutralizing titers in SCID mice [published online ahead of print October 25, 2005].  Blood2006107518101817 doi:10.1182/blood-2005-08-3229PubMedGoogle Scholar
30.
Hodges  BLTaylor  KMChu  Q  et al Local delivery of a viral vector mitigates neutralization by antiviral antibodies and results in efficient transduction of rabbit liver [published online ahead of print August 31, 2005]  Mol Ther200512610431051 doi:10.1016/j.ymthe.2005.06.475PubMedGoogle Scholar
31.
Manno  CSPierce  GFArruda  VR  et al Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response [published correction appears in Nat Med. 2006;12(5):592] [published online ahead of print February 12, 2006].  Nat Med2006123342347 doi:10.1038/nm1358PubMedGoogle Scholar
32.
Rock  KL Exiting the outside world for cross-presentation.  Immunity 2006;25 (4) 523- 525PubMedGoogle Scholar
33.
Straub  VCampbell  KP Muscular dystrophies and the dystrophin-glycoprotein complex.  Curr Opin Neurol 1997;10 (2) 168- 175PubMedGoogle Scholar
34.
Blake  DJTinsley  JMDavies  KE Utrophin: a structural and functional comparison to dystrophin.  Brain Pathol 1996;6 (1) 37- 47PubMedGoogle Scholar
35.
England  SBNicholoson  LVJohnson  MA  et al.  Very mild muscular dystrophy associated with the deletion of 46% of dystrophin.  Nature 1990;343 (6254) 180- 182PubMedGoogle Scholar
36.
Harper  SQHauser  MADelloRusso  C  et al.  Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy.  Nat Med 2002;8 (3) 253- 261PubMedGoogle Scholar
37.
Crawford  GEFaulkner  JACrosbie  RHCampbell  KPFroehnerf  SCChamberlain  JS Assembly of the dystrophin-associated protein complex does not require the dystrophin COOH-terminal domain.  J Cell Biol2000150613991410 doi:10.1083/jcb.150.6.1399PubMedGoogle Scholar
38.
Wang  BLi  JXiao  X Adeno-associated virus vector carrying human mini-dystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model.  Proc Natl Acad Sci U S A 2000;97 (25) 13714- 13719PubMedGoogle Scholar
39.
Watchko  JO’Day  TWang  B  et al Adeno-associated virus vector-mediated minidystrophin gene therapy improves dystrophic muscle contractile function in mdx mice.  Hum Gene Ther2002131214511460 doi:10.1089/10430340260185085PubMedGoogle Scholar
40.
Liu  MHarper  SQGrange  RWChamberlin  JSDuan  D Adeno-associated virus-mediated microdystrophin expression protects young mdx muscle from contraction-induced injury.  Mol Ther 2005;11 (2) 245- 256PubMedGoogle Scholar
41.
Abmayr  SGregorevic  PAllen  JMChamerlain  JS Phenotypic improvement of dystrophic muscles by rAAV/microdystrophin vectors is augmented by Igf1 codelivery.  Mol Ther 2005;12 (3) 441- 450PubMedGoogle Scholar
42.
Nguyen  HHJayasinha  VXia  BHoyte  KMartin  PT Overexpression of the cytotoxic T cell GalNAc transferase in skeletal muscle inhibits muscular dystrophy in mdx mice.  Proc Natl Acad Sci U S A200299856165621 doi:10.1073/pnas.082613599PubMedGoogle Scholar
43.
Grimm  DKern  ARittner  KKleinschmidt  JA Novel tools for production and purification of recombinant adeno-associated virus vectors.  Hum Gene Ther 1998;9 (18) 2745- 2760PubMedGoogle Scholar
44.
Clark  KRVoulgaropoulou  FFraley  DMJohnson  PR Cell lines for the production of recombinant adeno-associated virus.  Hum Gene Ther 1995;6 (10) 1329- 1341PubMedGoogle Scholar
45.
Inoue  NRussell  DW Packaging cells based on inducible gene amplification for the production of adeno-associated virus vectors.  J Virol 1998;72 (9) 7024- 7031PubMedGoogle Scholar
46.
Tamayose  KHirai  YShimada  T A new strategy for large-scale preparation of high-titer recombinant adeno-associated virus vectors by using packaging cell lines and sulfonated cellulose column chromatography.  Hum Gene Ther 1996;7 (4) 507- 513PubMedGoogle Scholar
47.
Wagner  JAMessner  AHMoran  ML  et al.  Safety and biological efficacy of an adeno-associated virus vector-cystic fibrosis transmembrane regulator (AAV-CFTR) in the cystic fibrosis maxillary sinus.  Laryngoscope 1999;109 (2, pt 1) 266- 274PubMedGoogle Scholar
48.
Wagner  JAReynolds  TMoran  ML  et al.  Efficient and persistent gene transfer of AAV-CFTR in maxillary sinus.  Lancet 1998;351 (9117) 1702- 1703PubMedGoogle Scholar
Neurological Review
September 2007

Gene Therapy for Duchenne Muscular Dystrophy: Expectations and Challenges

Author Affiliations

Author Affiliations: Center for Gene Therapy, Columbus Children's Research Institute and Columbus Children's Hospital, and the Department of Pediatrics, Ohio State University, Columbus.

 

DAVID EPLEASUREMD

Arch Neurol. 2007;64(9):1236-1241. doi:10.1001/archneur.64.9.1236
Abstract

Duchenne muscular dystrophy is a debilitating X-linked disease with limited treatment options. We examined the possibility of moving forward with gene therapy, an approach that demonstrates promise for treating Duchenne muscular dystrophy. Gene therapy is not limited to replacement of defective genes but also includes strategies using surrogate genes with alternative but effective means of improving cellular function or repairing gene mutations. The first viral-mediated gene transfer for any muscle disease was carried out at Columbus Children's Research Institute and Ohio State University for limb girdle muscular dystrophy type 2D, and the first viral-mediated trial of gene transfer for Duchenne muscular dystrophy is under way at the same institutions. These studies, consisting of intramuscular injection of virus into a single muscle, are limited in scope and represent phase 1 clinical trials with safety as the primary end point. These initial clinical studies lay the foundation for future studies, providing important information about dosing, immunogenicity, and viral serotype in humans. This article highlights the challenges and potential pitfalls as the field advances this treatment modality to clinical reality.

Duchenne muscular dystrophy (DMD) is the most common life-threatening childhood form of muscular dystrophy.1 It is an X-linked recessive disorder with an incidence estimated to be 1 in 3500 live male newborns. Mutations in the dystrophin gene,2 encoding a large (427-kDa) cytoskeletal protein found in skeletal and cardiac muscle, as well as smooth muscle, brain, and retina, cause DMD. The dystrophin gene is the largest gene identified to date,3 and because of its size, it is susceptible to a high sporadic mutation rate, ensuring that the disease can never be eliminated. It is estimated that 1 in 10 000 germ cells show de novo mutations, with some estimates as high as twice this number in certain populations.4 The consequences of the disease are debilitating, usually resulting in death in the early 20s in affected individuals; thus, medicine is challenged to find a treatment. Only corticosteroids have a salutary effect on DMD. Unequivocal proof was established in 1989 in a double-blind, randomized, controlled trial.5 The wide range of therapeutic prospects includes growth-modulating agents that increase muscle regeneration and delay muscle fibrosis,6,7 anti-inflammatory or second-messenger signal-modulating agents that affect immune responses,8-10 powerful antisense oligonucletotides (2′-O-methyl phosphorothioate backbone or morpholinos) with exon skipping capacity,11,12 and agents designed to suppress stop codon mutations (aminoglycosides and other agents such as PTC-124 [PTC Therapeutics Inc, South Plainfield, New Jersey]).13,14 In this brief review, we examine the possibility of moving forward with gene therapy for muscular dystrophy, addressing topics that will be relevant to musculoskeletal disorders and gene therapy in general.

Choice of virus for gene therapy

The delivery of “naked” DNA, a rudimentary form of gene therapy, proved to be inefficient and incapable of sustained transgene translation in DMD.15 In contrast, viruses are ideal vehicles for therapeutic gene transfer, having evolved to infect specific cell populations with high efficiency. The choice of virus is guided by factors including the target cell, immunogenicity, and required duration of transgene expression. Adenovirus was the early favorite for gene transfer to muscle and other tissues including lung (cystic fibrosis) and liver (infectious, neoplastic, and metabolic disorders). Disabling regions for replication (E1 and E2) and regulatory functions (E3 and E4) resulted in recombinant adenoviruses with presumed reduced ability to stimulate immune responses while achieving sustained transgene expression. An extreme modification of adenovirus was the deletion of all viral genes (“gutted”), enabling insertion of as much as 35 kilobases (kb) of DNA and, thus, more than enough for the entire dystrophin complementary DNA. This approach gained momentum16 despite several drawbacks including challenges in vector production and concerns about the emergence of replication-competent virus.17,18 However, substantial uneasiness about the use of this vector was the outgrowth of the unexpected death of Jesse Gelsinger in 1999 after transfer of the ornithine carbamylase gene by adenovirus.19 Within hours of intrahepatic administration, Gelsinger experienced severe complications and died 2 days later. The exact cause of his death has never been fully delineated, but the clinical findings overlapped with features of toxic shock syndrome.19

After the death of Gelsinger, a member of the parvovirus family, adeno-associated virus (AAV), a small single-stranded DNA virus not associated with human disease, took center stage for viral gene delivery. Adeno-associated virus, a member of the Parvoviridae family, is a Dependovirus, requiring helper functions from other sources, usually viruses, to complete its life cycle. The AAV genome is composed of approximately 4.7 kb containing the replication and encapsidation genes. This viral genome is flanked by two 145–base pair inverted terminal repeats. Most often, adenoviral genes provide the helper functions to activate and express the replication and encapsidation genes required for rescue, replication, and packaging. Herpesvirus can have a similar role in activating the helper genes. A further attraction of the parvovirus family is the recognition of the broad diversity of AAV serotypes and genomic variants. Well over 120 AAV variants have been isolated from various species including nonhuman primates.20,21 As of 2006, 11 AAV serotypes (AAV1-11) had been described as candidates for gene therapy. Advantages of each are under intense investigation to establish specific tropism relevant to the target tissue. The most promising serotypes for transducing skeletal muscle are AAV1, AAV2, AAV6, and AAV8.22-25

Adeno-associated virus vectors have a relatively spotless safety profile. An early concern was site-specific integration on chromosome 19. While this attribute is characteristic of wild-type AAV, it is a rare event with the recombinant form of the virus (rAAV).26 This was thought to be a potential disadvantage, forcing transgene expression from an episomal location. However, since the observed complications of insertional mutagenesis in patients with severe combined immunodeficiency syndrome, genome integration with the potential to disrupt nearby genes is considered less desirable.27 Loss of this attribute has not diminished the potential for long duration of transgene expression best explained by the stability of the rAAV vector genome to form stable episomal concatemers.

Immunogenicity of aav and potential impact on gene transfer

A key issue evolving with the transfer of any gene delivery vehicle is the potential for an immune response. While low immunogenicity was considered a major strength supporting the use of rAAV in clinical trials, a number of observations have recently provided a more realistic view. An obvious barrier to AAV transduction is the presence of circulating neutralizing antibodies that preclude binding of the virion to its cellular receptor.28,29 This potential threat can be reduced by prescreening patients for AAV serotype-specific neutralizing antibodies or through the application of therapeutic maneuvers such as plasmapheresis before gene transfer. Alternatively, delivering the virus through an intravascular balloon catheter imparts a protected environment enhanced by the removal of blood from the gene-targeted area, providing a hospitable environment for transduction.30 Another challenge recently uncovered is the development of a cell-mediated cytotoxic T-cell (CT) response to AAV capsid peptides, limiting transgene expression. In the human factor IX gene therapy trial in which rAAV was delivered to liver, only short-term transgene expression was achieved and levels of therapeutic protein declined to baseline levels 10 weeks after vector infusion.31 This was accompanied by signs of hepatocellular damage (elevation of serum transaminase enzyme values) and a CT response as measured by release of interferon-γ directed toward specific AAV capsid peptides.31 In this setting, AAV capsid sequences were displayed on the surface of transduced liver cells by major histocompatibility complex class I molecules, leading to invasion by activated CD8-positive CTs. To overcome this response, transient immunosuppression may be required; at least until AAV capsids are completely cleared.

Although data from the hemophilia B trial raise concerns that cannot be ignored, additional findings suggest that T-cell activation requires capsid binding to the heparan sulfate proteoglycan (HSPG) receptor, permitting virion shuttling into a dendritic cell pathway (referred to as cross-presentation).32 This is typical of the AAV2 (used in the clinical factor IX gene therapy trial) but not for AAV serotypes independent of HSPG binding. Further studies are under way to confirm the privileged immune status of virions deficient in HSPG binding. This is not a trivial issue because gene transfer through a vascular approach with HSPG-deficient AAV serotypes, such as AAV8 (see “Gene Therapy by Vascular Delivery” section), seems promising. If AAV8 falls below the window of detection of the immune system, its potential as a safe and effective gene transfer vehicle is substantially enhanced.

Modifying dystrophin dna for gene transfer

The dystrophin protein has 4 major domains (Figure 1),2,33,34 as follow: the N-terminal, which binds to cytoskeletal F-actin; a central rod domain composed of 24 spectrinlike repeats; the cysteine-rich domain, which binds to the transmembrane protein α-dystroglycan, linking noncovalently to β-dystroglycan, which together represent a vital component of the dystrophin glycoprotein complex linking the extracellular matrix to the actin cytoskeleton (Figure 2); and the C-terminal, which is not a prerequisite for maintaining stability of the muscle membrane. The full-length dystrophin complementary DNA is 14 kb, posing a substantial hurdle to the small packaging capacity of AAV. The concept of small dystrophins that easily fit into rAAV vectors is potentially useful and is based on clinical observations in patients with large gene deletions of the rod domain accompanied by a mild phenotype. The seminal example involves a 61-year-old ambulatory patient with Becker dystrophy with a rod domain deletion (exons 17-48), equivalent to almost half (48%) of the coding region of dystrophin.35 Such findings encouraged a systematic study in dystrophin-deficient (mdx) mice to find the optimal complementary DNA that would both stabilize the sarcolemmal membrane and fit the restrictive capacity of AAV.36 A consensus construct has emerged that permits deletion of the C-terminal, and a rod domain consisting of either 4 (microdystrophin) or 5 or more (minidystrophin) spectrin repeats, combined with removal of the 5′ and 3′ untranslated regions.35-37 An rAAV encoding minidystrophin with these features38 is undergoing clinical testing in a phase 1 gene therapy study by intramuscular injection in the biceps muscle of boys with DMD at Columbus Children's Hospital and Research Institute, Columbus, Ohio.

Gene therapy by vascular delivery

One challenge with gene therapy to the muscle is achieving widespread delivery to multiple muscle groups. We and others have been working on vascular delivery methods to transfer therapeutic genes with the intent of attaining clinically meaningful results. Several AAV serotypes are more suitable for vascular delivery, particularly AAV6 and AAV8. These serotypes demonstrate a preferential capability for crossing the endothelial vascular barrier and transducing muscle with a high degree of efficiency, obviating the use of permeability-enhancing agents or methods (Figure 3). There are several important implications from such findings. Using AAV serotypes with selective capacity to traverse the endothelium for a clinical study avoids drugs (eg, vascular endothelial growth factor, histamine, or papaverine), high infusion pressures, or excessive viral dosing, any of which adds potential for an adverse event. A further implication of selective serotype transit across the vascular barrier is that AAV8 does not rely on HSPG receptor binding, potentially mitigating the need for immunosuppression. This point is currently under additional study and has important inferences for the planning of clinical trials.

Use of surrogate genes to modify the dystrophic phenotype

Evidence from experimental gene replacement to the mdx mouse shows that despite substantial correction of the membrane defect by morphologic criteria (eg, protection against Evans blue dye permeability) and reversal of the phenotype (reduced central nucleation) in animals treated at an early age, there remains a gap in full restoration of contractile properties (protection against eccentric contraction) using minidystrophins and microdystrophins.39,40 This raises concerns that small dystrophin delivery may not restore the phenotype to normal in clinical studies. The extent of correction remains to be established in clinical trials. Other approaches under development may augment the therapeutic effect of the small dystrophins.

The field is beginning to test “booster” genes as a therapeutic strategy. Such approaches include combinations of small dystrophins with overexpression of insulin growth factor-1, in particular, the muscle isoform (mIgf-1),41 or inhibition of the negative muscle growth regulatory factor myostatin. This approach has recently been illustrated in the mdx mouse, in which coinjection of the rAAV-microdystrophin and rAAV–mIgf-1 vectors resulted in a degree of protection against contraction-induced injury not possible without dual gene therapy.41 Similarly, the inhibition of myostatin results in larger and stronger muscles with conservation of benefits, from mice to humans. In addition to inhibiting myostatin with specific antibodies (as applied in a current clinical trial for muscular dystrophies), follistatin is a potent myostatin antagonist. Adeno-associated virus can deliver the gene for follistatin and, because it is a secretory peptide, very high serum levels can be reached, resulting in muscle hypertrophy at regional and remote sites. When virus is injected into the gastrocnemius and hamstring muscles in animals, substantial increases in muscle mass are noted at the site of injection and at remote sites (Figure 4). We are exploring AAV-mediated gene therapy as a form of combinational treatment of muscular dystrophy that would also be applicable to other forms of muscle disease.

Another novel surrogate gene strategy for muscular dystrophy uses the enzyme CT GalNAc transferase, preferably called Galgt2, normally restricted to the neuromuscular junction. At this site, Galgt2 is responsible for glycosylating (adding a sugar moiety, the CT carbohydrate) α-dystroglycan. The CT carbohydrate (a name derived from a common epitope shared with activated CTs) bolsters α-dystroglycan in its linkage to the laminins of the extracellular matrix, further ensuring membrane stability. Overexpression of Galgt2 in transgenic mice42 or by gene transfer using AAV results in the ectopic expression of glycosylated proteins (eg, CT carbohydrate–α-dystroglycan), laminins usually confined to the neuromuscular junction (laminin α4 and laminin α5) and utrophin along the entire myofiber. The net result is membrane stability by the substitution of utrophin for dystrophin, creating a utrophin-glycoprotein complex. Studies by colleagues in our Gene Therapy Center42 are laying the groundwork for a clinical trial using the GALGT2 gene. Another advantage accruing to this approach is that overexpression of this normal gene averts the potential for transgene immunogenicity.

Vector production methods

Extending gene therapy studies beyond small phase 1 safety trials is an important challenge for vector production. To illustrate this point, if a single human dose is determined to be 1013 particles for a given application and 1000 patients with DMD are candidates for treatment, 1016 particles would be required. Based on current vector production standards, each cell produces approximately 104 AAV particles, necessitating 1012 cells to meet the needs for a clinical trial. In practical terms, this would translate to approximately 100 000 flasks or 1000 L of cells in suspension, emphasizing both the limitations and importance of scalable production methods.

Genetically engineered cell lines can be adapted to overcome this potential barrier to clinical applications.43-48 Three essential components are required for rAAV production: the rAAV vector genome, the AAV trans-helper genes (replication and encapsidation), and adenovirus or herpesvirus helper genes delivered in plasmid DNA form or via virus infection. Based on our experience, HeLa cells yield the highest titers of rAAV per cell that we have observed to date.

Conclusions

Multiple strategies are being developed for gene therapy for muscular dystrophy. Clinically meaningful results are anticipated through optimization of a vascular delivery route using replacement, surrogate, or booster genes. Additional studies are required to further define the immunogenicity of AAV serotypes and transgenes and the potential need for immunosuppression. Successful gene therapy as a treatment for DMD is clearly on the horizon, with the potential to improve the lives of patients with this devastating disease.

Back to top
Article Information

Correspondence: Jerry R. Mendell, MD, Center for Gene Therapy, Columbus Children's Research Institute, 700 Children's Dr, Columbus, OH 43205 (mendellj@ccri.net).

Accepted for Publication: February 26, 2007.

Author Contributions:Study concept and design: Rodino-Klapac, Chicoine, Kaspar, and Mendell. Acquisition of data: Rodino-Klapac, Chicoine, Kaspar, and Mendell. Analysis and interpretation of data: Rodino-Klapac, Chicoine, Kaspar, and Mendell. Drafting of the manuscript: Rodino-Klapac, Chicoine, Kaspar, and Mendell. Critical revision of the manuscript for important intellectual content: Rodino-Klapac, Chicoine, Kaspar, and Mendell. Obtained funding: Kaspar and Mendell. Administrative, technical, and material support: Rodino-Klapac, Chicoine, Kaspar, and Mendell. Study supervision: Chicoine, Kaspar, and Mendell.

Financial Disclosure: None reported.

Funding/Support: This study was supported by grant 5 U54 AR050733 from the Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Centers, National Institutes of Health.

References
1.
Emery  AE Population frequency of inherited neuromuscular diseases: a world survey.  Neuromuscul Disord 1991;1 (1) 19- 29PubMedGoogle Scholar
2.
Hoffman  EPBrown  RH  JrKunkel  LM Dystrophin: the protein product of the Duchenne muscular dystrophy locus.  Cell 1987;51 (6) 919- 928PubMedGoogle Scholar
3.
Koenig  MHoffman  EPBertelson  CJMonaco  APFeener  CKunkel  LM Complete cloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals.  Cell 1987;50 (3) 509- 517PubMedGoogle Scholar
4.
Sinha  SMishra  SSingh  VMittal  RDMittal  B High frequency of new mutations in North Indian Duchenne/Becker muscular dystrophy patients.  Clin Genet 1996;50 (5) 327- 331PubMedGoogle Scholar
5.
Mendell  JRMoxley  RTGriggs  RC  et al.  Randomized, double-blind six-month trial of prednisone in Duchenne's muscular dystrophy.  N Engl J Med 1989;320 (24) 1592- 1597PubMedGoogle Scholar
6.
Tinsley  JDeconinck  NFisher  R  et al.  Expression of full-length utrophin prevents muscular dystrophy in mdx mice.  Nat Med 1998;4 (12) 1441- 1444PubMedGoogle Scholar
7.
Bogdanovich  SKrag  TOBarton  ER  et al.  Functional improvements of dystrophic muscle by myostatin blockade.  Nature 2002;420 (6914) 418- 421PubMedGoogle Scholar
8.
Biggar  WDGingras  MFehlings  DLHarris  VASteele  CA Deflazacort treatment of Duchenne muscular dystrophy.  J Pediatr 2001;138 (1) 45- 50PubMedGoogle Scholar
9.
Biggar  WDHarris  VAEliasoph  LAlman  B Long-term benefits of deflazacort treatment for boys with Duchenne muscular dystrophy in their second decade [published online ahead of print March 20, 2006].  Neuromuscul Disord2006164249255 doi:10.1016/j.nmd.2006.01.010PubMedGoogle Scholar
10.
Balaban  BMatthews  DJClayton  GHCarry  T Corticosteroid treatment and functional improvement in Duchenne muscular dystrophy: long-term effect.  Am J Phys Med Rehabil 2005;84 (11) 843- 850PubMedGoogle Scholar
11.
Aartsma-Rus  AKaman  WEWeij  Rden Dunnen  JTvan Ommen  GTvan Deutekom  JC Exploring the frontiers of therapeutic exon skipping for Duchenne muscular dystrophy by double targeting within one or multiple exons.  Mol Ther 2006;14 (3) 401- 407PubMedGoogle Scholar
12.
McClorey  GMoulton  HMIversen  PLFletcher  SWilton  SD Antisense oligonucleotide-induced exon skipping restores dystrophin expression in vitro in a canine model of DMD [published online ahead of print May 25, 2006].  Gene Ther 2006;13 (19) 1373- 1381PubMedGoogle Scholar
13.
Barton-Davis  ERCordier  LShoturma  DILeland  SESweeney  HL Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice.  J Clin Invest 1999;104 (4) 375- 381PubMedGoogle Scholar
14.
Hamed  SA Drug evaluation: PTC-124—a potential treatment of cystic fibrosis and Duchenne muscular dystrophy.  IDrugs 2006;9 (11) 783- 789PubMedGoogle Scholar
15.
Liang  KWNishikawa  MLiu  FSun  BYe  OHuang  L Restoration of dystrophin expression in mdx mice by intravascular injection of naked DNA containing full-length dystrophin cDNA.  Gene Ther 2004;11 (11) 901- 908PubMedGoogle Scholar
16.
Scott  JMLi  SHarper  SQ  et al.  Viral vectors for gene transfer of micro-, mini-, or full-length dystrophin.  Neuromuscul Disord 2002;12(suppl 1)S23- S29PubMedGoogle Scholar
17.
Tripathy  SKBlack  HBGoldwasser  ELeiden  JM Immune responses to transgene-encoded proteins limit the stability of gene expression after injection of replication-defective adenovirus vectors.  Nat Med 1996;2 (5) 545- 550PubMedGoogle Scholar
18.
Hartigan-O’Connor  DBarjot  CCrawford  RChamberlain  JS Efficient rescue of gutted adenovirus genomes allows rapid production of concentrated stocks without negative selection.  Hum Gene Ther 2002;13 (4) 519- 531PubMedGoogle Scholar
19.
Verma  IM A tumultuous year for gene therapy [editorial].  Mol Ther 2000;2 (5) 415- 416PubMedGoogle Scholar
20.
Gao  GVandenberghe  LHWilson  JM New recombinant serotypes of AAV vectors.  Curr Gene Ther 2005;5 (3) 285- 297PubMedGoogle Scholar
21.
Schnepp  BCJensen  RLChen  CLJohnson  PRClark  KR Characterization of adeno-associated virus genomes isolated from human tissues.  J Virol 2005;79 (23) 14793- 14803PubMedGoogle Scholar
22.
Gregorevic  PBlankinship  MJAllen  JM  et al Systemic delivery of genes to striated muscles using adeno-associated viral vectors [published online ahead of print July 25, 2004].  Nat Med2004108828834 doi:10.1038/nm1085PubMedGoogle Scholar
23.
Xiao  WChirmule  NBerta  SCMcCullough  BGao  GWilson  JM Gene therapy vectors based on adeno-associated virus type 1.  J Virol 1999;73 (5) 3994- 4003PubMedGoogle Scholar
24.
Wang  ZZhu  TQiao  C  et al Adeno-associated serotype 8 efficiently delivers genes to muscle and heart [published online ahead of print February 27, 2005].  Nat Biotechnol2005233321328 doi:10.1038/nbt1073PubMedGoogle Scholar
25.
Schnepp  BCClark  KRKlemanski  DLPacak  CAJohnson  PR Genetic fate of recombinant adeno-associated virus vector genomes in muscle.  J Virol 2003;77 (6) 3495- 3504PubMedGoogle Scholar
26.
Kotin  RMSiniscalco  MSamulski  RJ  et al.  Site-specific integration by adeno-associated virus.  Proc Natl Acad Sci U S A 1990;87 (6) 2211- 2215PubMedGoogle Scholar
27.
Davé  UPJenkins  NACopeland  NG Gene therapy insertional mutagenesis insights.  Science 2004;303 (5656) 333PubMedGoogle Scholar
28.
Jiang  HCouto  LBPatarroyo-White  S  et al.  Effects of transient immunosuppression on adeno associated virus-mediated, liver-directed gene transfer in rhesus macaques and implications for human gene therapy.  Blood 2006;108 (10) 3321- 3328PubMedGoogle Scholar
29.
Scallan  CDJiang  HLiu  T  et al Human immunoglobulin inhibits liver transduction by AAV vectors at low AAV2 neutralizing titers in SCID mice [published online ahead of print October 25, 2005].  Blood2006107518101817 doi:10.1182/blood-2005-08-3229PubMedGoogle Scholar
30.
Hodges  BLTaylor  KMChu  Q  et al Local delivery of a viral vector mitigates neutralization by antiviral antibodies and results in efficient transduction of rabbit liver [published online ahead of print August 31, 2005]  Mol Ther200512610431051 doi:10.1016/j.ymthe.2005.06.475PubMedGoogle Scholar
31.
Manno  CSPierce  GFArruda  VR  et al Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response [published correction appears in Nat Med. 2006;12(5):592] [published online ahead of print February 12, 2006].  Nat Med2006123342347 doi:10.1038/nm1358PubMedGoogle Scholar
32.
Rock  KL Exiting the outside world for cross-presentation.  Immunity 2006;25 (4) 523- 525PubMedGoogle Scholar
33.
Straub  VCampbell  KP Muscular dystrophies and the dystrophin-glycoprotein complex.  Curr Opin Neurol 1997;10 (2) 168- 175PubMedGoogle Scholar
34.
Blake  DJTinsley  JMDavies  KE Utrophin: a structural and functional comparison to dystrophin.  Brain Pathol 1996;6 (1) 37- 47PubMedGoogle Scholar
35.
England  SBNicholoson  LVJohnson  MA  et al.  Very mild muscular dystrophy associated with the deletion of 46% of dystrophin.  Nature 1990;343 (6254) 180- 182PubMedGoogle Scholar
36.
Harper  SQHauser  MADelloRusso  C  et al.  Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy.  Nat Med 2002;8 (3) 253- 261PubMedGoogle Scholar
37.
Crawford  GEFaulkner  JACrosbie  RHCampbell  KPFroehnerf  SCChamberlain  JS Assembly of the dystrophin-associated protein complex does not require the dystrophin COOH-terminal domain.  J Cell Biol2000150613991410 doi:10.1083/jcb.150.6.1399PubMedGoogle Scholar
38.
Wang  BLi  JXiao  X Adeno-associated virus vector carrying human mini-dystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model.  Proc Natl Acad Sci U S A 2000;97 (25) 13714- 13719PubMedGoogle Scholar
39.
Watchko  JO’Day  TWang  B  et al Adeno-associated virus vector-mediated minidystrophin gene therapy improves dystrophic muscle contractile function in mdx mice.  Hum Gene Ther2002131214511460 doi:10.1089/10430340260185085PubMedGoogle Scholar
40.
Liu  MHarper  SQGrange  RWChamberlin  JSDuan  D Adeno-associated virus-mediated microdystrophin expression protects young mdx muscle from contraction-induced injury.  Mol Ther 2005;11 (2) 245- 256PubMedGoogle Scholar
41.
Abmayr  SGregorevic  PAllen  JMChamerlain  JS Phenotypic improvement of dystrophic muscles by rAAV/microdystrophin vectors is augmented by Igf1 codelivery.  Mol Ther 2005;12 (3) 441- 450PubMedGoogle Scholar
42.
Nguyen  HHJayasinha  VXia  BHoyte  KMartin  PT Overexpression of the cytotoxic T cell GalNAc transferase in skeletal muscle inhibits muscular dystrophy in mdx mice.  Proc Natl Acad Sci U S A200299856165621 doi:10.1073/pnas.082613599PubMedGoogle Scholar
43.
Grimm  DKern  ARittner  KKleinschmidt  JA Novel tools for production and purification of recombinant adeno-associated virus vectors.  Hum Gene Ther 1998;9 (18) 2745- 2760PubMedGoogle Scholar
44.
Clark  KRVoulgaropoulou  FFraley  DMJohnson  PR Cell lines for the production of recombinant adeno-associated virus.  Hum Gene Ther 1995;6 (10) 1329- 1341PubMedGoogle Scholar
45.
Inoue  NRussell  DW Packaging cells based on inducible gene amplification for the production of adeno-associated virus vectors.  J Virol 1998;72 (9) 7024- 7031PubMedGoogle Scholar
46.
Tamayose  KHirai  YShimada  T A new strategy for large-scale preparation of high-titer recombinant adeno-associated virus vectors by using packaging cell lines and sulfonated cellulose column chromatography.  Hum Gene Ther 1996;7 (4) 507- 513PubMedGoogle Scholar
47.
Wagner  JAMessner  AHMoran  ML  et al.  Safety and biological efficacy of an adeno-associated virus vector-cystic fibrosis transmembrane regulator (AAV-CFTR) in the cystic fibrosis maxillary sinus.  Laryngoscope 1999;109 (2, pt 1) 266- 274PubMedGoogle Scholar
48.
Wagner  JAReynolds  TMoran  ML  et al.  Efficient and persistent gene transfer of AAV-CFTR in maxillary sinus.  Lancet 1998;351 (9117) 1702- 1703PubMedGoogle Scholar
×