Gene Therapy for Human Disease: Clinical Advances and Challenges
Review Article
Gene therapy for retinal disease

https://doi.org/10.1016/j.trsl.2012.12.007Get rights and content

Gene therapy strategies for the treatment of inherited retinal diseases have made major advances in recent years. This review focuses on adeno-associated viral (AAV) vector approaches to treat retinal degeneration and, thus, prevent or delay the onset of blindness. Data from human clinical trials of gene therapy for retinal disease show encouraging signs of safety and efficacy from AAV vectors. Recent progress in enhancing cell-specific targeting and transduction efficiency of the various retinal layers plus the use of AAV-delivered growth factors to augment the therapeutic effect and limit cell death suggest even greater success in future human trials is possible.

Section snippets

Cell-Specific Targeting Within the Retina: Serotype Tropism

The majority of IRDs are caused by mutations in genes specifically expressed in the photoreceptor of RPE cells, therefore, the ideal gene therapy would provide efficient transduction with restricted transgene expression to these cell types. Adeno-associated virus (AAV) is currently the favored vector for retinal gene therapy but lentiviral (LV) vectors are also being pursued. Whilst LV vectors have been shown to be efficient in targeting murine RPE cells,16 transduction of photoreceptors has

Promoter Choice in Retinal Gene Therapy

The use of ubiquitous promoters has helped reveal transduction patterns of AAV vectors (as described above), but as human gene therapy trials are becoming more frequent, the development of vectors that specifically target the cell of interest will increase safety and efficacy of the treatment. In retinal gene therapy animal studies, the commonly used ubiquitous promoters have been immediate-early cytomegalovirus (CMV) enhancer-promoter37 and the CAG promoter, which combines the CMV enhancer

Self-Complementary AAV Vector use in Retinal Gene Therapy Studies

The onset of expression following AAV infection is delayed by numerous factors but the rate-limiting step is the synthesis of the complementary strand to the single stranded DNA AAV genome.59, 60 Self-complementary AAV (scAAV) vectors have been developed that can bypass this critical step. These vectors package a single-stranded transgene that contains both a forward and reverse copy of the therapeutic gene separated by an altered inverted terminal repeat. Following entry into the target cell

AAV Treatment of Autosomal Recessive Models of Retinal Disease

The majority of animal models of IRD exhibit null mutations, relevant to the study of autosomal recessive IRDs. Many studies have shown the feasibility of gene replacement therapy in such animal models. Peripherin2 is a rod photoreceptor-specific structural protein, mutations in which lead to autosomal recessive RP or macular dystrophy.64 The retinal degeneration slow (rds) mouse is homozygous for a null mutation in the rds/peripherin-2 gene, completely lacking functional peripherin2.65 Gene

AAV Treatment of Autosomal Dominant Models of Retinal Disease

The majority of animal models available for study of IRD carry null mutations, yet, there are numerous IRDs that do not show a complete lack of protein. Dominant mutations often yield mutated versions of a protein; if these impart a dominant-negative effect then provision of the wild-type gene and subsequent protein should resolve the phenotype as in the gene replacement strategies above. Dominant-negative mutations tend to eliminate 1 or more functions of the encoded protein, for example, the

Human AAV Clinical Trials for the Treatment of Retinal Disease

In 2008, data from 3 independent trials revealed evidence of improved visual function following rAAV2/2 subretinal delivery of hRPE65 to LCA patients (reviewed in reference147). RPE65 (retinal pigment epithelium-specific protein 65kDa) is expressed in the RPE and is essential in the visual pathway for its role in regenerating the active chromophore required for light absorption, loss of function of RPE65, therefore, leads to loss of vision.148 Whilst all 3 clinical trials were consistent in

Neuroprotection of the Retina

AAV delivery of growth and anti-apoptotic factors can be used to prevent cell loss when retinal degeneration is advanced and the causative mutation unknown and also to enhance the effects of a co-delivered therapeutic gene. Apoptosis of photoreceptor cells is common to most forms of IRD, therefore, preventing cell death could significantly slow degeneration and allow a greater window for gene therapy intervention. Minimizing rod photoreceptor death would also prolong cone survival and AAV

Summary

Gene therapy of retinal diseases is an advancing field. Identification of serotype tropism and enhancement of transduction efficiencies of the various retinal cell layers is allowing for targeted AAV treatment in various forms: growth factors and anti-apoptotic factors to prevent cell loss and also to enhance gene therapy in addition to gene-replacement and suppression-replacement therapies. All are showing success in animal models of retinal disease. Clinical trials of AAV-based gene therapy

References (164)

  • J.A. Sawicki et al.

    A composite CMV-IE enhancer/beta-actin promoter is ubiquitously expressed in mouse cutaneous epithelium

    Experimental Cell Res

    (1998)
  • N. Esumi et al.

    Analysis of the VMD2 promoter and implication of E-box binding factors in its regulation

    J Biol Chem

    (2004)
  • L. Petit et al.

    Restoration of Vision in the pde6β-deficient Dog, a Large Animal Model of Rod-cone Dystrophy

    Mol Ther

    (2012)
  • J.-J. Pang et al.

    Long-term retinal function and structure rescue using capsid mutant AAV8 vector in the rd10 mouse, a model of recessive retinitis pigmentosa

    Mol Ther

    (2011)
  • A.J. Smith et al.

    AAV-Mediated gene transfer slows photoreceptor loss in the RCS rat model of retinitis pigmentosa

    Mol Ther

    (2003)
  • M.M. Sohocki et al.

    Prevalence of AIPL1 mutations in inherited retinal degenerative disease

    Mol Genetics Metab

    (2000)
  • T.P. Dryja et al.

    Null RPGRIP1 alleles in patients with Leber congenital amaurosis

    Am J Hum Genetics

    (2001)
  • S. Kohl et al.

    Mutations in the cone photoreceptor G-protein alpha-subunit gene GNAT2 in patients with achromatopsia

    Am J Hum Genetics

    (2002)
  • S. Michalakis et al.

    Restoration of cone vision in the CNGA3−/− mouse model of congenital complete lack of cone photoreceptor function

    Molecular therapy : the journal of the American Society of Gene Therapy

    (2010)
  • D. Hicks et al.

    The implications of rod-dependent cone survival for basic and clinical research

    Invest Ophthalmol Vis Sci

    (1999)
  • T. Léveillard et al.

    Identification and characterization of rod-derived cone viability factor

    Nat Genetics

    (2004)
  • A. Rattner et al.

    Molecular genetics of human retinal disease

    Annu Rev Genetics

    (1999)
  • M.M. Hims et al.

    Retinitis pigmentosa: genes, proteins and prospects

    Dev Ophthalmol

    (2003)
  • I. Voo et al.

    Update on the genetics of macular dystrophies

    Retina (Philadelphia, Pa)

    (2004)
  • M.A. Maw et al.

    Mutation of the gene encoding cellular retinaldehyde-binding protein in autosomal recessive retinitis pigmentosa

    Nat Genetics

    (1997)
  • X. Sun et al.

    Gene therapy with a promoter targeting both rods and cones rescues retinal degeneration caused by AIPL1 mutations

    Gene Ther

    (2010)
  • E. Ivanova et al.

    Evaluation of the adeno-associated virus mediated long-term expression of channelrhodopsin-2 in the mouse retina

    Mol Vis

    (2009)
  • K.D. Kolstad et al.

    Changes in adeno-associated virus-mediated gene delivery in retinal degeneration

    Hum Gene Ther

    (2010)
  • C. Kostic et al.

    Gene therapy regenerates protein expression in cone photoreceptors in Rpe65(R91W/R91W) mice

    PLOS One

    (2011)
  • K.S. Balaggan et al.

    Stable and efficient intraocular gene transfer using pseudotyped EIAV lentiviral vectors

    J Gene Med

    (2006)
  • A. Auricchio et al.

    Exchange of surface proteins impacts on viral vector cellular specificity and transduction characteristics: the retina as a model

    Hum Mol Genetics

    (2001)
  • O. Grüter et al.

    Lentiviral vector-mediated gene transfer in adult mouse photoreceptors is impaired by the presence of a physical barrier

    Gene Ther

    (2005)
  • M. Calame et al.

    Retinal degeneration progression changes lentiviral vector cell targeting in the retina

    PLOS One

    (2011)
  • R.R. Ali et al.

    Gene transfer into the mouse retina mediated by an adeno-associated viral vector

    Hum Mol Genetics

    (1996)
  • C. Lebherz et al.

    Novel AAV serotypes for improved ocular gene transfer

    J Gene Med

    (2008)
  • G.S. Yang et al.

    Virus-mediated transduction of murine retina with adeno-associated virus: effects of viral capsid and genome size

    J Virol

    (2002)
  • M. Allocca et al.

    Novel adeno-associated virus serotypes efficiently transduce murine photoreceptors

    J Virol

    (2007)
  • F. Rolling

    Recombinant AAV-mediated gene transfer to the retina: gene therapy perspectives

    Gene Ther

    (2004)
  • J. Bennett et al.

    Stable transgene expression in rod photoreceptors after recombinant adeno-associated virus-mediated gene transfer to monkey retina

    Proc Natl Acad Sci U S A

    (1999)
  • A.J. Lotery et al.

    Adeno-associated virus type 5: transduction efficiency and cell-type specificity in the primate retina

    Hum gene Ther

    (2003)
  • K. Mancuso et al.

    Recombinant adeno-associated virus targets passenger gene expression to cones in primate retina

    J Opt Soc Am A Opt Image Sci Vis

    (2007)
  • M. Agbandje-McKenna et al.

    AAV capsid structure and cell interactions

    Methods Mol Biol (Clifton, NJ)

    (2011)
  • C.E. Thomas et al.

    Rapid uncoating of vector genomes is the key to efficient liver transduction with pseudotyped adeno-associated virus vectors

    J Virol

    (2004)
  • Z. Yan et al.

    Ubiquitination of both adeno-associated virus type 2 and 5 capsid proteins affects the transduction efficiency of recombinant vectors

    J Virol

    (2002)
  • L. Zhong et al.

    Next generation of adeno-associated virus 2 vectors: point mutations in tyrosines lead to high-efficiency transduction at lower doses

    Proc Natl Acad Sci U S A

    (2008)
  • C.A. Grant et al.

    Evaluation of recombinant adeno-associated virus as a gene transfer vector for the retina

    Curr Eye Res

    (1997)
  • G. Le Meur et al.

    Restoration of vision in RPE65-deficient Briard dogs using an AAV serotype 4 vector that specifically targets the retinal pigmented epithelium

    Gene Ther

    (2007)
  • J.W.B. Bainbridge et al.

    Effect of gene therapy on visual function in Leber's congenital amaurosis

    N Engl J Med

    (2008)
  • H. Sun et al.

    The vitelliform macular dystrophy protein defines a new family of chloride channels

    Proc Natl Acad Sci U SA

    (2002)
  • R.C. Ryals et al.

    Quantifying transduction efficiencies of unmodified and tyrosine capsid mutant AAV vectors in vitro using two ocular cell lines

    Mol Vis

    (2011)
  • Cited by (59)

    • RPE based gene and cell therapy for inherited retinal diseases: A review

      2022, Experimental Eye Research
      Citation Excerpt :

      The health of the inner cell layer of the retina plays a vital role in the proper functioning of vision. Mutations in certain genes of these cells can lead to IRD, but in most cases, it is caused by mutations in genes expressed in photoreceptors or RPE cells (McClements and MacLaren, 2013). Since the function of RPE plays an important role in the survival of rod and cone photoreceptors, gene mutations in RPE cells can lead to photoreceptor death and retinal degradation (Veleri et al., 2015).

    • The Location of Exon 4 Mutations in RP1 Raises Challenges for Genetic Counseling and Gene Therapy

      2019, American Journal of Ophthalmology
      Citation Excerpt :

      With the advent of gene therapy, disease inheritance patterns must be assessed to configure the best therapeutic approach. For disease caused by a dominant negative mechanism, a gene therapy strategy would require silencing of the expression of the mutated allele.21 As it is apparent in RP1 disease that a single allele is adequate for normal functionality, further gene supplementation would not be necessary.

    • Current strides in AAV-derived vectors and SIN channels further relieves the limitations of gene therapy

      2018, Egyptian Journal of Medical Human Genetics
      Citation Excerpt :

      For instance, Leber’s congenital amaurosis (LCA) (an early onset retinal dystrophy), characterized by a malfunction and deterioration of photoreceptors is caused by mutations in retinal pigment epithelium-specific protein 65 kDa (RPE65) gene [29–31]. This gene codes for a retinoid isomerase; a key factor in retinol function in the visual cycle [28,31,32], by using specific hRPE65 (resulting in a lower expression) or ubiquitously promoters (higher expression), after the injection of AAV2 vectors to the subretina, the over-expression of a functional copy of the gene RPE65 could improve vision [30]. As shown from the low immunogenicity, safety, clinical benefit and good tolerability, in three previous clinical trials, the immune system is an important factor that must be considered in all kinds of experimental and clinical research practices [28].

    View all citing articles on Scopus

    Conflict of interests: All authors have read the journal’s policy on conflicts of interest and have none to declare.

    View full text