Progress in Retinal and Eye Research 31 (2012) 43e64
Contents lists available at SciVerse ScienceDirect
Progress in Retinal and Eye Research
journal homepage: www.elsevier.com/locate/prer
Gene therapy in the Cornea: 2005epresent
Rajiv R. Mohan a, b, c, *, Jonathan C.K. Tovey a, b, Ajay Sharma a, b, Ashish Tandon a, b
a
Harry S. Truman Memorial Veterans’ Hospital, 800 Hospital Drive, Columbia, MO 65201, USA
Mason Eye Institute, University of Missouri, 1 Hospital Drive, Columbia, MO 65212, USA
c
College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65212, USA
b
a r t i c l e i n f o
a b s t r a c t
Article history:
Available online 28 September 2011
Successful restoration of vision in human patients with gene therapy affirmed its promise to cure ocular
diseases and disorders. The efficacy of gene therapy is contingent upon vector and mode of therapeutic
DNA introduction into targeted cells/tissues. The cornea is an ideal tissue for gene therapy due to its ease
of access and relative immune-privilege. Considerable progress has been made in the field of corneal
gene therapy in last 5 years. Several new gene transfer vectors, techniques and approaches have evolved.
Although corneal gene therapy is still in its early stages of development, the potential of gene-based
interventions to treat corneal abnormalities has begun to surface. Identification of next generation
viral and nanoparticle vectors, characterization of delivered gene levels, localization, and duration in the
cornea, and significant success in controlling corneal disorders, particularly fibrosis and angiogenesis, in
experimental animal disease models, with no major side effects have propelled gene therapy a step
closer toward establishing gene-based therapies for corneal blindness. Recently, researchers have
assessed the delivery of therapeutic genes for corneal diseases and disorders due to trauma, infections,
chemical, mechanical, and surgical injury, and/or abnormal wound healing. This review provides an
update on the developments in gene therapy for corneal diseases and discusses the barriers that hinder
its utilization for delivering genes in the cornea.
Published by Elsevier Ltd.
Keywords:
Cornea
Gene therapy
AAV
Nanoparticles
Decorin
Corneal scarring
Corneal neovascularization
Contents
1.
2.
3.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
Gene therapy vehicles for the cornea . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
2.1.
Viral vectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
2.1.1.
Adenovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
2.1.2.
Adeno-associated virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
2.1.3.
Retrovirus and lentivirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
2.2.
Nonviral vectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
2.2.1.
Microinjection technique . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
2.2.2.
Electroporation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
2.2.3.
Sonoporation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
2.2.4.
Gene gun . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
2.2.5.
Controlled corneal dehydration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
2.2.6.
Laser . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
2.2.7.
Chemicals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
2.3.
Next generation AAV and nanoparticle vectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
2.3.1.
Tyrosine-mutant AAV vectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49
2.3.2.
Nanoparticle vectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
2.3.3.
Dendrimer vectors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
Tissue-selective gene therapy tools . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
* Corresponding author. Mason Eye Institute, School of Medicine, University of Missouri-Columbia, 1 Hospital Dr., Columbia, MO 65212, USA. Tel.: þ1 573 884 1449;
fax: þ1 573 814 6551.
E-mail address:
[email protected] (R.R. Mohan).
1350-9462/$ e see front matter Published by Elsevier Ltd.
doi:10.1016/j.preteyeres.2011.09.001
44
4.
5.
6.
7.
8.
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
3.1.
Vector engineering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
3.2.
Custom vector-delivery techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
Gene therapy updates in various corneal disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
4.1.
Corneal graft rejection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
4.2.
Corneal scarring and wound healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
4.3.
Corneal neovascularization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
4.4.
Corneal alkali burn . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
4.5.
Other corneal disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
Lessons learned from corneal genetic studies: humans and animals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
Ocular gene therapy clinical trials: current status . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
Future directions: regenerative medicine, nanomedicine and other approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
1. Introduction
Gene therapy has advanced in leaps and bounds since its
introduction to medicine almost 20 years ago as a query on gene
therapy in the United States National Library of Medicine database
PubMed yields over 128,000 articles from almost all medical
disciplines. In the field of ophthalmology, gene therapy has shown
astounding success. Recently, a breakthrough was attained in the
restoration of vision in human patients suffering from Leber’s
congenital amaurosis using gene replacement therapy in the retinal
pigment epithelium (Bainbridge et al., 2008; Hauswirth et al., 2008;
Maguire et al., 2008, 2009; Simonelli et al., 2010). This plainly gives
credence to the potential of gene therapy to cure diseases of the
ocular system and prevent blindness.
Current treatments for corneal disorders include various pharmacological agents and surgical approaches depending on the cause,
extent, and type of corneal damage. Conventional drug therapies to
treat corneal disorders provide only short-term relief, require
repeated application, cause side effects, and are often ineffective.
More importantly they do not correct the cause of the problem but
merely suppress symptoms. Conversely, gene therapy fixes the root of
the problem, provides long-term cure, and does not require repeated
applications or clinic visits. However, at present no gene therapy
modalities are available for corneal diseases. The cornea is an ideal
tissue for gene therapy due to its ease of access and relative immuneprivilege. As it is a transparent tissue and devoid of blood vessels, it
can be readily monitored visually. These properties also allow for the
administration of therapeutic genes into corneal cells with relative
ease. The cornea is well suited for conducting ex vivo gene therapy
strategies because it can be maintained in culture for a long time. The
arduous task of developing novel gene-based modalities for the
cornea has greatly improved due to increased comprehension of
acquired and inherited corneal diseases in terms of molecular
mechanisms and pathogenesis. Numerous approaches employing
different viral and nonviral vectors and techniques to introduce genes
into the cornea in vitro, ex vivo, and in vivo have been tested. Among
viral vectors, adenovirus, adeno-associated virus (AAV), retrovirus,
and lentivirus vectors have been found to efficiently transport genes
into corneal tissue. However, concerns over safety and immunogenicity have limited their use. Nonviral vectors including plasmid DNA,
lipids, polymers, and nanoparticles are generally safe but often found
less efficient than their viral counterparts. Various physical techniques such as topical administration, gene gun, electroporation,
intrastromal injection, and iontophoresis have been used to augment
delivery of both viral and nonviral vectors. However, none of these
vectors or techniques is ideal, and each has its benefits and shortcomings. Herein we provide a comprehensive review of corneal gene
therapy approaches tested in the past six years and a brief general
overview of vectors. A detailed overview of gene therapy vectors and
their mode of action can be found in our previous corneal gene
therapy review (Mohan et al., 2005).
2. Gene therapy vehicles for the cornea
2.1. Viral vectors
Viruses have been used since the dawn of gene transfer technology to deliver genes into different cells and tissues. Viruses were
used as a vector in about 70% of gene therapy clinical trials (Young
et al., 2006). Adenovirus (AV), adeno-associated virus (AAV),
retrovirus, and lentivirus have been found to efficiently transport
genes into the cornea. Nevertheless, each of these vectors has its
own limitations. Adenovirus and retrovirus can successfully deliver
genes into the cornea for short periods of time with mild-to-severe
inflammatory responses. However, both of these vectors are of
limited use for corneal gene therapy because of their inability to
transduce low/non-dividing cells such as corneal endothelium and
keratocytes, and induction of immune reactions. AAV and disabled
lentivirus vectors offer better alternatives for delivering genes into
corneal keratocytes and endothelium because of their ability to
transduce slow/non-dividing cells and ability to provide long-term
transgene expression. The origin of lentivirus vectors (equine
infectious anemia virus and HIV) remains a major concern and
significantly dampens enthusiasm for its use in human patients.
Among viral vectors, AAV appears to be a good choice for corneal gene therapy because of its potency and safety profile.
Recombinant AAV vectors have shown great promise for ocular
gene therapy and restoring vision in patients with no major side
effects.
2.1.1. Adenovirus
Recombinant forms of AV have been engineered and utilized in
gene transfer studies in the past (Mohan et al., 2005). In sum, firstgeneration AV vectors lack the E1 gene region rendering them
unable to replicate although they can proliferate in cell lines that
provide E1 gene product. Second-generation AV vectors lack E1, E2,
and E4 viral genes leading to less immunogenicity than firstgeneration vectors. In third-generation AV vectors, the AV viral
genome is absent and only ITR sequences and packaging genes are
present thus giving the name gutless or high-capacity vectors.
Third-generation AV vectors are able to carry larger gene inserts
and are less immunogenic but require a helper virus. In order to
reduce helper virus contamination, Cre/lox-P system and 293 cells
stably transfected with Cre recombinase are utilized and have been
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
shown to decrease helper virus contamination to <0.01% (Palmer
and Ng, 2005). However, even this minimal level of contamination may be detrimental in clinical scenarios where large doses are
required.
Multiple studies have examined the efficiency of AV vectors for
corneal gene therapy (Mohan et al., 2005). AV vector was found
most efficient in transducing murine cornea compared to equine
immunodeficiency, lenti and bacculovirus vectors (Beutelspacher
et al., 2005). Recently, AV vectors have been shown to deliver
genes successfully into mouse corneal stroma and endothelium
under control of cytomegalovirus immediate-early promoter
(CMV) (Yu et al., 2007). The Ritter group has also reported cytokine
or growth factor gene transfer with AV vector in rodent corneas
(Ritter et al., 2007; Gong et al., 2007a,b). AV vectors provide shortlived transgene expression and also demonstrate moderate to
severe immune reaction apart from other side effects (Yu et al.,
2007; Ritter et al., 2007; Gong et al., 2007a,b; Sharma et al.,
2010a). Recombinant AV vectors have potential use in corneal
gene therapy to over-express proteins in the corneal epithelium
particularly in scenarios such as diabetes mellitus where transient
gene expression is desirable. However, the ephemeral nature of AV
vectors discourages its application in gene-based treatment for
inherited gene defects (Saghizadeh et al., 2010).
2.1.2. Adeno-associated virus
Among 110 identified AAV serotypes, to date only serotypes
1e10 have been used for gene therapy. AAV vectors have demonstrated high transduction efficiency and long-term transgene
expression in the retina, cornea, and many non-ocular tissues
in vivo (Alexander and Hauswirth, 2008; Gao et al., 2005; Surace
and Auricchio, 2008 and references therein). AAV2 has been
tested more in depth compared to other AAV serotypes for gene
therapy although each serotype has shown unique transduction
patterns for various cells/tissues. AAV6, -8 and -9, especially, have
the ability to mediate whole-body gene transfer efficiently (Ghosh
et al., 2007; Pacak et al., 2006). The variation in gene transfer by
different AAV serotypes is likely due to the interactions between
host-cell receptor and viral capsid (Van Vliet et al., 2008). Structural
variations in the capsid region of different AAV serotypes enable
each serotype to bind different cell surface receptors. For instance,
AAV serotypes 4, 5, and 6 use sialic acid (Wu et al., 2006) whereas
AAV 8 and 9 use laminin receptors to enter cells (Akache et al.,
2006). This led to the development of hybrid AAV vectors as
a variety of pseudopackaged vectors were produced using transencapsidation of the AAV2 genome into the capsid of AAV1e9.
These next generation hybrid vectors AAV2/1e9, displayed much
greater transduction efficiency compared to AAV2/2 in ocular
tissues of various animal models (Surace and Auricchio, 2008). It is
important to note that a great deal of variation in transduction
efficiency, tissue preference, first transgene appearance timing, and
duration was observed in studies performed with various hybrid
recombinant AAV vectors (Surace and Auricchio, 2008). Lebherz
et al. (2008) also evaluated the gene delivery efficiency and
expression stability of AAV2/7, 2/8, 2/9, with AAV2/1, 2/2, 2/5 using
enhanced green fluorescent protein (EGFP) and rhesus erythropoietin as transgenes. After vector introduction into the eye via
intravitreal and subretinal injection, the authors demonstrated that
AAV2/7 and 2/8 possess superior long-term transduction ability (up
to 6 months) in both retinal and anterior chamber tissues including
the iris, trabecular meshwork, and cornea, (Lebherz et al., 2008).
Subsequent studies with AAV2/6, AAV2/7, AAV2/8, and AAV2/9
vectors further reinforced these findings for retinal tissue (Surace
and Auricchio, 2008). In a similar fashion, AAV2/8 and 2/9
demonstrated 5e100 fold superior transduction efficiency in nonocular tissues such as heart (Wang et al., 2005), brain (Broekman
45
et al., 2006), skeletal muscle (Wang et al., 2005), lung (Limberis
and Wilson, 2006), and liver (Gao et al., 2006). We compared the
efficiency of AAV2/6, AAV2/8, and AAV2/9 serotypes for corneal
gene delivery using in vitro, in vivo, and ex vivo models to test
a hypothesis that the relative efficiency of AAV-mediated gene
transfer in the cornea depends on serotype. In our first study, the
cellular tropism and transduction efficiency of AAV2/6, AAV2/8, and
AAV2/9 vectors encoding alkaline phosphatase (AP) were tested
using primary human corneal fibroblasts cultures. We found the
tested serotypes efficiently transduced human corneal fibroblasts
but they differed in transduction efficiency. AAV2/6 displayed
30e50 fold higher transduction efficiency compared to AAV2/8 or
AAV2/9 (Fig. 1). A remarkably positive and important finding was
that none of the tested AAV serotypes induced significant cell death
or loss of cellular viability reaffirming that AAV vectors are safe for
the cornea (Sharma et al., 2010b). Interestingly, our subsequent
studies with these vectors using mouse cornea in vivo and human
cornea ex vivo showed very different transduction profiles
compared to our in vitro investigations (Sharma et al., 2010c).
Contrary to in vitro findings, the order of transduction efficiency for
the three tested vectors was found to be AAV2/9 AAV2/8 > AAV2/
6 (Fig. 2). These findings are not completely surprising as past
literature reports imply that in vitro gene delivery may differ from
in vivo transduction (Lipkowitz et al., 1999; Richter et al., 2000).
However, the important take home message from these studies is
that in vivo testing of gene therapy vectors for the cornea is
important before making any conclusive inferences about vector
efficacy. The one thing that remained unchanged during in vitro,
ex vivo and in vivo testing was the safety profile of tested AAV
serotypes. None of the three tested serotypes caused any significant
side effects such as cell death, loss of cellular viability, inflammation
and/or noticeable immune reaction in the cornea as measured with
TUNEL and CD11b or F4/80 immunostaining (Fig. 3). Recently,
transgene expression in the corneal epithelium was detected up to
8 months with AAV2 in the mouse eye suggesting that rAAVdelivered transgenes can persist for several months, and possibly
years, in vivo (Alexander and Hauswirth, 2008; Lai et al., 2007).
Thus, using AAV2/5 vector and a mouse model we performed timedependent long-term animal experiments to characterize delivered
transgene expression and duration for the cornea in vivo. Our
ongoing studies showed different levels of transgene expression in
the cornea in vivo up to 10 months following topical vector application (Mohan et al., unpublished data).
The alterations in function, host range, and tissue tropism
shown by different AAV serotypes may be due to a degree of
sequence variation within the capsid region (Van Vliet et al., 2008).
Furthermore, the differences in transduction profile are likely
related to distinct uptake and intracellular trafficking mechanisms
of the various serotypes. In addition, different serotype-specific ITR
elements may also influence transgene expression. Various studies
report that the use of different serotypes or cross-packaging
expands the tissue tropism of rAAV vectors. Moreover, selective
use of serotypes might allow for targeting specific tissues. For
example, an rAAV2 genome packaged in an AAV5 capsid (AAV2/5)
transduced photoreceptors and retinal pigment epithelial cells in
the eye (Gao et al., 2005; Surace and Auricchio, 2008) whereas
AAV2-encapsidated vector transduced primarily photoreceptors,
and an AAV1-encapsidated vector transduced mainly retinal
pigment epithelial cells. Additionally, the utilization of different
AAV serotypes may overcome the problems associated with vector
re-administration and preexisting immunity to rAAV2 in humans
(Adriaansen et al., 2006).
AAV vectors have emerged to the forefront in ocular gene
therapy as they have been used in a ground-breaking clinical trial to
treat retinal diseases. Despite the proven potency and safety of AAV
46
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
Fig. 1. Representative cytochemical staining images showing transduction efficiency of AAV2/6, AAV2/8 and AAV2/9 vectors for human corneal fibroblasts. The alkaline phosphatase
marker gene (stained purple) and 30 h time point were used for comparison.
vectors, relatively few studies have been performed to evaluate the
potential of AAV vectors for ocular surface gene delivery including
the cornea. A recent study compared rAV and rAAV tropism to
human corneal cells in vitro and rabbit corneas in vivo. It was noted
that rAAV transduced more keratocytes while rAV transduction was
detected mostly in the epithelium. Interestingly, rAAV-mediated
gene delivery led to lower GFP expression compared to rAV. This
may have specific clinical ramifications as choice of vector may be
dependent on transgene expression level or number of corneal cells
requiring gene transfer. For example, in corneal disorders of the
Fig. 2. Quantification of AAV2/6, AAV2/8 or AAV2/9 mediated transgene delivery in
mouse stroma in vivo. The delivered alkaline phosphatase gene expression in the
corneal tissue sections was quantified digitally by measuring pixels of purple stained
tissue in 4 104 mm2 tissue area. *p < 0.05 AAV6/AAV8/AAV9 compared to control,
4 ¼ p < 0.05 AAV8 to compared to AAV6, U ¼ p < 0.01 AAV9 compared to AAV6.
epithelium like diabetes mellitus, rAV may be more useful whereas
rAAV may be more beneficial in stromal keratopathies where keratocytes are targeted (Liu et al., 2008a). Using AAV2, RPE65 gene
was delivered to the retina of patients with Leber’s congenital
amaurosis and successfully restored vision (Bainbridge et al., 2008;
Hauswirth et al., 2008; Maguire et al., 2008, 2009; Simonelli et al.,
2010). These studies clearly outline the safety and efficiency of AAVbased gene therapy for the retina leading the way for gene therapy
for other ocular tissues including the cornea.
2.1.3. Retrovirus and lentivirus
Limited progress has occurred in the development and/or
testing of retro- and lentivirus vectors. The “gutless” retroviral
vectors were developed by deleting the majority of the viral
genome not required for infection. Further, the risk of replication
was curtailed by using a packaging cell line that employs gag/pol
and env from separate constructs, and by replacing the 50 long
terminal repeat (LTR) U3 region in the viral genome with the CMV
promoter (Lech and Somaia, 2008). LTRs, the control center for gene
expression, are sequences of DNA that flank functional genes found
in retroviral DNA and retrotransposons. Retroviruses such as HIV
use LTRs to insert their genetic sequences into the host genome.
Even though modifications in these sequences have been tested,
the risk of insertional oncogenesis due to their random integration
remains. During a clinical trial for X-linked severe combined
immunodeficiency treatment, retroviral vector integration was
noted near proto-oncogenes, and led to the development of T-cell
leukemia in 4 patients (Hacein-Bey-Abina et al., 2008), and the
death of one of those individuals (Herzog, 2010). This clearly
reduces enthusiasm for adopting retrovirus for gene therapy.
The human immunodeficiency virus (HIV)-based lentiviral
vectors have also been investigated for corneal gene transfer.
Lentivirus appears to be less toxic to the cell’s genome than
retrovirus (Montini et al., 2006). Current lentiviral vectors for gene
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
therapy lack the entire viral genome, and contain only the packaging signal and the cis-acting elements, including the LTR. In an
attempt to generate safer lentiviral vectors, the U3 region of the 30
LTR has been deleted. In recently generated lentiviral vectors, the
endogenous promoter has been replaced with a CMV/LTR hybrid
promoter in an effort to improve safety. Currently, glycoprotein of
vesicular stomatitis virus is the most commonly used env protein
for lentiviral pseudotyping (Lech and Somaia, 2008). Not long ago it
was reported that transcriptionally active LTRs are chief contributors to genotoxicity and that self-inactivating LTRs decrease the
oncogenic potential thus improving the safety of lentiviral vectors
(Montini et al., 2009). Clinically, lentiviral vectors have yielded no
reports of serious adverse events (Williams and Coster, 2010).
Selective studies have been performed to evaluate the efficacy of
lentiviral vectors for corneal gene therapy during the past 6 years.
Beutelspacher et al. (2005) compared self-inactivating HIV-1 and
Equine Infectious Anemia Virus (EIAV) transduction efficiency in
murine, rabbit, and human corneal endothelial cells and found EIAV
to be more effective than HIV-1 for corneal endothelial gene
therapy. Another study reported that a single injection of HIVbased lentivirus into the anterior chamber of the rodent eye efficiently delivered transgene into the corneal endothelium (Challa
et al., 2005). Parker et al. (2007) demonstrated 80e90% transduction of rat, bovine, and human corneal endothelial cells with
lentiviral vector expressing enhanced yellow fluorescent protein
under control of the Simian virus type 40 early promoter.
Bemelmans et al. (2009) demonstrated efficient transduction of
keratocytes up to 3 weeks in a pig model by injecting HIV-1
expressing GFP into a stromal pocket created with a femtosecond
laser.
47
2.2.1. Microinjection technique
Introduction of plasmid via microinjection has led to successful
delivery of genes including GFP, interleukin (IL)18, Flt23k, endostatin, MMP14, and vasohibin into various cells of the cornea (Kim
et al., 2005; Jani et al., 2007; Lai et al., 2007; Galiacy et al., 2011;
Zhou et al., 2010; Sharma A, et al. IOVS 2010;51:ARVO E-Abstract
2839). Microinjections targeting different layers of the cornea have
been performed at various anatomic locations and include intrastromal, subconjunctival, and directly into the anterior chamber
(Singh et al., 2005; Kim et al., 2005; Lai et al., 2007; Zhou et al.,
2010; Yu et al., 2007; Sharma A, et al. IOVS 2010;51:ARVO EAbstract 2839). A recent study performed in mice demonstrated
that bevacizumab treatment given via intraocular injection was
more efficacious than the subconjunctival (periocular) route
(Dratviman-Storobinsky et al., 2009). Intrastromal injection may be
appropriate in the treatment of acute corneal diseases as shortlived gene expression was detected in the cornea following intrastromal injection (Hao et al., 2010). Conversely, subconjunctival
injection showed long-term stable transgene expression and may
help to avoid the endophthalmitis and cataract formation associated with intracameral injection (Kuo et al., 2009). Microsurgical
techniques also offer a feasible method of exploring gene function
in corneal disorders. For example, Kuo et al. (2008) showed
successful transgene delivery in the cornea in vivo using intrastromal lamellar implantation of a partially dried p-bFGFeSAINT-18
complex composed of SAINT-18 and plasmid vector encoding
reporter or FGF2 gene. This corneal gene transfer method permitted
localized transgene delivery in the cornea. It is thought that except
for topical and subconjunctival administration, all other gene
delivery strategies to the cornea are invasive and compromise
corneal integrity (Cheng et al., 2007).
2.2. Nonviral vectors
The introduction of plasmid DNA expressing therapeutic genes
into target cells without the use of viruses falls under the broad
category of nonviral gene transfer methods. Nonviral gene therapy
is considered safer than viral gene therapy due to low toxicity,
immunogenicity, and pathogenicity. Additionally, plasmid vector
production is straightforward and cost-effective. Nevertheless, low
transfection efficiency is a major challenge. Many strategies have
been developed and are in the preclinical pipeline to improve
plasmid delivery in cells. A brief description of popular techniques
is given below.
2.2.2. Electroporation
Electroporation, also known as electrogenetherapy or electropermeabilization, makes use of high-intensity electrical pulses to
form transient pores in the cell membrane and is useful for gene
delivery in both cultured eye cells and ocular surface tissues in vivo.
An advantage is large DNA constructs can be transported into cells
although specialized equipment is necessary (Williams and Coster,
2010). However, very few studies have been reported in the cornea
since 2005. Electroporation has the ability to deliver foreign genes
into the corneal epithelium as well as keratocytes (Zhou and Dean,
2007). Electrical current of 200 V/cm did not cause trauma, corneal
Fig. 3. Representative images showing AAV9 effects on immune reaction (measured by Cd11b) and cell death (measured by TUNEL) in mouse corneas. Like control corneas (no AAV
treatment), few Cd11bþ and TUNELþ cells were observed in AAV-treated mouse stroma. The TUNEL þ cells detected in the corneal epithelium were because of its replenishment via
apoptosis. AAV6 and AAV8 showed similar results (data not shown). Nuclei are stained blue with DAPI.
48
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
edema, or inflammation but introduced transgene at low levels.
Higher electrical current resulted in enhanced gene transfer but
also led to considerable corneal damage. Electrical current can
cause irreversible tissue damage as a result of thermal heating or
Ca2þ influx due to disruption of cell membranes. Electrically
assisted gene delivery to the endothelium of ex vivo human corneas
was recently described (He et al., 2010). Using custom-designed
electrodes, two reporter genes, EGFP and beta-galactosidase
(bgal), were successfully transported into human corneas in
organ culture using eight 1-Hz 100-ms pulses of 125 mA square
current. Although efficiency was much lower than viral vector, low
cell death and no remarkable change in tight junction integrity of
endothelial cells show its potential clinical application (He et al.,
2010). The electrotransfer of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) small interfering RNA (siRNA) and dextran
macromolecules into mice corneal epithelium in vivo using iontophoresis and electroporation individually and in combination has
been reported (Hao et al., 2009). Although both iontophoresis and
electroporation independently delivered macromolecules into the
cornea, iontophoresis was found to be more efficient and the
combination of iontophoresis followed by electroporation was
more effective than both methods alone (Hao et al., 2009).
2.2.3. Sonoporation
Sonoporation employs ultrasound waves to create pores in the
plasma membrane in order to deliver DNA to the nucleus. Ultrasound is effective for cell transfection in vitro and in vivo (Liu et al.,
2006). Transfection efficiency of this approach is dependent on the
transducer frequency, acoustic pressure, output strength, and pulse
duration of ultrasound treatment in addition to the use of contrast
agents such as microbubbles. Microbubbles were generated as
contrast agents to not only enhance imaging but also improve gene
delivery efficiency by boosting cell permeability (Mitragotri, 2005).
Ultrasound-targeted microbubble destruction may hold great
potential as a site-specific gene transfer approach and has been used
successfully in AAV-mediated gene transfection of human RPE cells
in vitro and rat retina in vivo (Li et al., 2009a,b). Microbubbles
generally measure approximately 3 mm in diameter and are
composed of a shell which houses a gas core. Available commercially, microbubbles differ in shell composition (i.e. albumin,
galactose, lipid, polymers) and gas core (i.e. air, perfluorocarbon,
nitrogen) (Lindner, 2004). A notable advantage of microbubbles is
that they may be targeted to specific areas of interest. One way to
accomplish this is the addition of a tiered polymer coat to the shell
and subsequent covalent (i.e. carbodiimide-mediated amide) or
noncovalent (i.e. biotineavidin) coupling of targeting ligands to the
polymer coat (Klibanov, 2006). The precise mechanism by which
gene transfection is enhanced by ultrasound-mediated destruction
of carrier microbubbles is not clear but may include shell
fragmentation-induced cellular microporation and microenvironment changes brought about by high-velocity pressure jets
or heat and free radical production (Lindner, 2004). Ultrasound
facilitates gene transfer by way of passive diffusion (Gao et al., 2007).
The Sakamoto group demonstrated successful gene delivery into
rabbit cornea in vitro and in vivo by combining ultrasound and
microbubbles (Sonoda et al., 2006). The central cornea was injected
with plasmid mixed with perflutren protein and a probe was placed
on the ocular surface for ultrasound (1 MHz, 120 s, 50% duty cycle,
1e2 W/cm2) exposure. The ultrasound and microbubbles greatly
increased transduction efficiency into keratocytes without tissue
damage. GFP was expressed in the stroma for up to 30 days and was
limited to the area exposed to ultrasound (Sonoda et al., 2006).
However, multiple safety concerns of microbubble use including
instability in serum and infection risk of components such as human
albumin limit its use. Subsequent studies entailed the utilization of
ultrasound with a newly developed contrast agent, a bubble liposome made of polyethylenglycol modified liposome containing
perfluoropropane gas. Sonoporation coupled with the bubble liposome showed enhanced transgene delivery into rabbit corneal
epithelial cells and rat subconjunctiva (Yamashita et al., 2007).
Although these studies outline the potential of sonoporation for
corneal gene therapy, many parameters remain to be investigated.
2.2.4. Gene gun
Gene gun is a ballistic (also called bioballistic) gene transfer
method. It utilizes micron-sized biologically inert heavy metal (gold,
silver or tungsten) particles and mechanical or macroprojectile
(centripetal, magnetic or electrostatic) force. The bombarding of
DNA-coated particles on cells/tissues with high velocity results in
gene transfer. Gene delivery with gene gun depends on many factors
such as amount of DNA-coated on particles, temperature, amount of
cells, amount of force, number of DNA-coated particles, etc. Shallow
penetration of particles, substantial cell damage, uncontrolled gene
transfer, high cost, access to internal organs, etc. are few among
many limitations of this method. Successful gene delivery in the
cornea with gene gun has been reported (Hao et al., 2010). Genes
such as IL4 and IL10 plasmid DNA and opioid growth factor receptor
(OGFr) have been introduced into the corneal epithelium with this
method (Bauer et al., 2006; Zagon et al., 2006). Zagon and cohorts
showed delayed corneal abrasion healing after gene gun-mediated
delivery of sense OGFr cDNA in rat eyes. Conversely, they found
that antisense OGFr cDNA over-expression by the same delivery
method led to accelerated corneal wound healing (Zagon et al.,
2006). This study established the autocrine behavior of the OGFOGFr axis, its regulatory role in ocular surface wound healing, and
its potential in gene therapy approaches for corneal diseases where
wound healing is impaired such as diabetic keratopathy. In addition,
it highlighted the gene gun technique as a valuable tool in examining the role of genes in epithelial abnormalities (Zagon et al.,
2006). Another research group targeted plasmids expressing IL10
and IL4 to the mouse cornea in order to determine immune
modulation of HSV-1 infection. The expression of delivered genes
was limited to corneal epithelial cells and attenuated the clinical
course of infection although small numbers of F4/80þ and CD11bþ
cells were noted with treatment (Bauer et al., 2006). Thus the gene
gun method may be of great benefit in the focal treatment of corneal
epithelial disorders as minimal transfection occurs in neighboring
tissues. However, the technique, in its present form, is in need of
additional optimization before being adopted for use in the clinic
(Bauer et al., 2006). Furthermore, due to high corneal epithelial cell
turnover, transgene expression is transient.
2.2.5. Controlled corneal dehydration
Recently we reported that controlled corneal dehydration
increases vector absorption in mouse and rabbit corneas in vivo and
human cornea ex vivo using a hair dryer following epithelial
removal. Vectors expressing marker gene were used to study the
effect of corneal drying on gene transfer. As evident from the data
presented in Fig. 4, increased corneal drying with a hair dryer
showed increased transgene delivery in the mouse cornea in vivo.
However, excessive dehydration may have a negative impact on
corneal integrity. Detection of significantly higher transgene
delivery in vivo after 50 s of corneal drying with moderate changes
in corneal morphology affirmed the promise of this simple technique for corneal gene therapy. High transgene delivery was also
observed after 30 s of drying without jeopardizing corneal
morphology and moderate gene delivery was noted after 10 or 20 s
of drying with no altered corneal morphology. This study revealed
that controlled corneal drying modulates gene delivery in the
cornea presumably due to a change in corneal hydration. Thus, we
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
49
Fig. 4. Representative H&E staining images showing histology of mouse corneas subjected to air-drying and collected 14 days after AAV8 application. A: 0 s air-drying. B: 50 s airdrying. Mouse corneas subjected to 50 s drying showed moderate or higher levels of morphological changes in anterior stroma. C: shows digital quantification of delivered marker
gene expression detected at day 14 in mouse corneas in vivo. These corneas received 2 ml of AAV8 vector immediately after 0, 20, 30, or 50 s of drying. *p < 0.05 as compared to 20 s;
4 p < 0.05 as compared to 30 s.
postulated that administration of efficient vector employing simple
minimally invasive techniques is a novel approach for delivering
therapeutic levels of genes in the cornea in vivo (Mohan et al.,
2010b).
2.2.6. Laser
Recently, a stromal pocket technique involving the use of the
femtosecond laser to introduce genes into the pig cornea ex vivo
was reported (Bemelmans et al., 2009). A stromal pocket 110 mm in
depth was produced with a femtosecond laser and lentiviral vector
expressing GFP was injected. Histology of corneal tissue performed
5 days after vector application showed wound closure and marker
gene expression in the cells (most likely keratocytes) around the
corneal pocket. Interestingly, the levels of transgene expression
noted at day-5 remained up to 3 weeks (Bemelmans et al., 2009).
This method facilitates gene delivery into cells in a specific, targeted corneal region. However, femtosecond laser is known to
induce intense wound healing and infiltration of inflammatory cells
in the rabbit cornea (Netto et al., 2007). Thus, the next step would
entail the translation of this procedure from an ex vivo model to an
in vivo model and the establishment of a safety profile.
2.2.7. Chemicals
Scores of natural and synthetic chemicals have been tested to
introduce genes into corneal cells. In this review we provide a short
description of more widely tested lipids and polymers for delivering
genes in the cornea. Among many lipids dioleoyltrimethylammonium
chloride (DOTMA), dioleoylphosphatidyl-ethanolamine (DOPE),
1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), dimethyl
dioctadecyl ammonium bromide (DDAB), 3b-[N-(N0 ,N0 -dimethylaminoethane) carbamoyl]cholesterol (DC-cholesterol), and N-methyl4(dioleyl) methylpyridinium chloride (SAINT-2) showed promise for
corneal gene therapy. A transfection solution prepared from five
different lipids showed 7e17% transgene delivery in corneal endothelial cells in vitro (Dannowski et al., 2005). Our laboratory formulated transfection solutions manipulating ratios of DOPE and DDAB,
and observed 12e15% gene delivery in human corneal fibroblasts
in vitro, and rodent and rabbit stroma in vivo after corneal
administration via a defined delivery technique (Mohan et al.,
unpublished data). Gene delivery in cultured human epithelial cells
and in rabbit corneal epithelium in vivo has also been reported with
DOTAP/DOPE transfection mixture (Toropainen et al., 2007). A
formulation of liposomes and transferrin was shown to modulate
chemokine expression with viral macrophage inflammatory protein II
in a murine model of corneal allograft rejection (Pillai et al., 2008).
Transferrin was used to enhance transfection efficiency by promoting
endocytosis. Among cationic lipids/polymers, polyethyleneimine
(PEI), poly-lactide-co-glycolide (PLGA), polylactic acid (PLA), and chitosan have been evaluated for corneal gene transfer. PEI showed
significant transgene delivery in cultured human corneal epithelial
cells and rabbit cornea in vivo (Hornof et al., 2008; Yuan et al., 2006).
Chitosan, a naturally occurring aminopolysaccharide resulting from
alkaline deacetylation of chitin, has been examined widely for gene
transfer because of its nontoxic nature at high concentrations, high
availability, low cost, and biodegradable character. Several studies
have reported successful delivery of genes in ocular tissues, including
the cornea, using chitosan (de la Fuente et al., 2008a,b). Nonetheless,
many challenges such as poor transfection, significant immune reaction, cell-targeting, etc. remain to be resolved for its wider application
as a useful delivery system (Xu et al., 2010).
2.3. Next generation AAV and nanoparticle vectors
To overcome the obstacles presented by conventional viral and
nonviral vehicles many new vectors considered next generation
were developed. Discussing all of these is beyond the scope of this
review. Herein we focus our attention on tyrosine-mutant AAV,
nanoparticles, and dendrimers which have shown promise for
corneal gene therapy in preclinical animal studies.
2.3.1. Tyrosine-mutant AAV vectors
The AAV capsid surface is a fundamental element involved in
host receptor binding, cellular uptake, and intracellular trafficking
(Van Vliet et al., 2008) thus affecting transduction efficiency. Trafficking within the target cell renders the AAV vector susceptible to
natural cellular degradation mechanisms such as the
50
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
ubiquitineproteasome pathway (Douar et al., 2001). Phosphorylation of AAV surface exposed tyrosine residues marks the vector for
ubiquitination and subsequent proteasome-mediated degradation
prior to entering the nucleus (Petrs-Silva et al., 2009). A recent
study presented evidence that phosphorylation of surface exposed
tyrosine residues by epidermal growth factor receptor protein
tyrosine kinase (EGFR-PTK) significantly reduces transduction
efficiency of both single-stranded and self-complementary AAV2
vectors by w68% and w74%, respectively. In addition, intracellular
trafficking is retarded due to AAV ubiquitination followed by
degradation mediated by proteasomes (Zhong et al., 2008). Sitedirected mutagenesis of each of the seven AAV2 capsid tyrosine
residues (Y252, Y272, Y444, Y500, Y700, Y704, and Y730) by
phenylalanine residue substitution leads to increased vector
transduction and transgene expression by circumventing EGFR-PTK
phosphorylation and the ubiquitineproteasome pathway in human
cells in vitro and murine hepatocytes in vivo (Zhong et al., 2008).
These so-called next generation tyrosine-mutant AAV2, AAV8, and
AAV9 vectors have several-fold higher transgene delivery in retinal
cells after subretinal or intravitreal injection compared to their
wild-type counterparts (Petrs-Silva et al., 2009). These results are
also clinically applicable in the context of vector dose. AAV2 vectors
have been used in many Phase I/II clinical trials (Zhong et al., 2008).
However, relatively large vector doses are required to attain therapeutic levels and may trigger an immune reaction. Neutralizing
antibody levels are thought to be proportional to virus dose and
preexisting antibodies may reduce transduction efficiency upon
vector re-administration (Petry et al., 2008). Tyrosine-mutant AAV
vectors were shown to decrease vector dosage needed for transduction. A potent tyrosine-mutant AAV2 Y444F vector showed
superior transduction even at 10,000-fold lower titer compared to
wild-type AAV2. The lower AAV dose also leads to a decreased
immune response (Petrs-Silva et al., 2009). Double, triple, and
quadruple tyrosine-mutants have been generated to evaluate
potential augmentation of AAV transduction efficiency. In bone
marrow-derived primary murine cells and human mesenchymal
stem cells, a triple AAV2 mutant consisting of Y444F, Y500F, and
Y730F mutations increased transduction efficiency by almost 130fold and significantly improved viral intracellular trafficking (Li
et al., 2010). Our laboratory examined the efficacy of tyrosinemutant AAV2, AAV8, and AAV9 vectors in delivering genes to the
stroma and tyrosine-mutant AAV8 to the endothelium of the mouse
cornea in vivo. In our studies, we utilized hair dryer-based topical
application technique for achieving targeted delivery into the
mouse stroma (Mohan et al., unpublished data) and specialized
microinjection techniques to introduce the AAV8 vector encoding
GFP into the anterior chamber of mice eyes (Sharma A, et al. IOVS
2010;51:ARVO E-Abstract 2839). All the three tested tyrosinemutant AAV serotypes showed significantly higher transgene
delivery in the mouse stroma in vivo compared to their corresponding wild type in preliminary studies (Mohan et al., unpublished data). The tyrosine-mutant AAV8 vector demonstrated
substantially greater marker gene delivery than the wild-type AAV8
vector (Fig. 5) into the corneal endothelium of mouse eye in vivo
with no apparent adverse effects. Stereomicroscopy, immunocytochemistry and slit-lamp biomicroscopy were used to analyze the
efficacy and safety of screened vectors. Our recent studies showing
substantial targeted gene delivery into corneal endothelium are
particularly exciting as it has potential as an effective therapy for
endothelial dystrophies in the future.
2.3.2. Nanoparticle vectors
The National Institutes of Health coined the term nanoparticles
(NPs) for materials 1e100 nm in size used in medicine for disease
diagnosis and therapy. The advances in nanotechnology have
supplied a large reservoir of NPs that may well revolutionize the
future of medicine. Smaller than 100 nm in size, NPs are minuscule
and thus able to access anywhere within the in vivo realm, as living
cells are 10,000e20,000 nm in diameter (Sharma et al., 2010a).
Additionally, the ability of NPs to multiplex and incorporate
a myriad of ligands such as DNA, antibodies, peptides, and probes
can give rise to an array of therapeutic modalities. Internalization of
NPs in mammalian cells is carried out by phagocytosis, macropinocytosis, clathrin- or caveolae-mediated endocytosis, and other
clathrin- and caveolae-independent endocytic pathways (Ragusa
et al., 2007; Hillaireau and Couvreur, 2009).
Many studies reported gene transfer in non-ocular cells with
metallic (gold), polymeric (PEI, poly-L-lysine, PLGA), or hybrid
(metallic-polymeric) NPs (Ragusa et al., 2007; Ghosh et al., 2008).
Limited studies investigated the prospective of NPs for delivering
genes in the eye, including the cornea (Cai et al., 2008; de la Fuente
et al., 2008a,b; Sharma et al., 2011). An in vivo study showed marker
gene delivery in the retina, lens, trabecular meshwork, and cornea
with PEG-substituted-30mer lysine NPs (Cai et al., 2008). These NPs
provided short-term transgene expression, and may necessitate
repeated applications for sustained effect. NPs made from PLGA
have been shown to deliver significant transgene in cultured rabbit
conjunctival epithelial cells (Qaddoumi et al., 2004). Chitosan NPs
have been shown to transfect up to 15% of human corneal epithelial
and normal human conjunctival cells in vitro without affecting
Fig. 5. Representative stereomicroscopy (A) and immunocytochemistry in tissue sections (B) showing tyrosine-mutant AAV8-mediated transgene delivery in corneal endothelium
of mouse eye in vivo. Dispensing of vector via custom delivery-technique fetched selective gene delivery into corneal endothelium. Cell nuclei were stained blue with DAPI while
cells expressing GFP gene stained green.
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
cellular viability (de la Fuente et al., 2008a,b). Recently, a nanodendritic compound was used as an in situ polymerizable adhesive
in repairing a variety of corneal wounds in porcine and human eyes
ex vivo without major side effects (Grinstaff MW. IOVS
2008;49:ARVO E-Abstract 4801). A follow up in vivo study using
chicken eyes reaffirmed biodendrimer efficacy and suggested that
it may be superior to suturing (Berdahl et al., 2009). Our ongoing
experiments with DOTAP NPs showed significant delivery of GFP
marker gene in cultured human corneal fibroblasts (up to 20%) and
human corneal endothelial cells (up to 14%), without compromising
cellular viability and proliferation (Fig. 6; Mohan RR, et al. IOVS
2007;48:ARVO E-Abstract 2733). One in vitro study investigated
a nanocarrier, comprised of biocompatible and bioadhesive
compounds hyaluronic acid (HA) and chitosan, as a vehicle for gene
delivery to human corneal epithelial and conjunctival cells (de la
Fuente et al., 2008b). The NPs loaded with plasmid encoding
reporter gene, were internalized by ocular cells via hyaluronan
receptor CD44-mediated endocytosis as visualized by confocal
microscopy and showed high levels of EGFP expression (up to 15%)
with undetectable negative impacts on cellular viability. Reporter
gene expression was directly proportional to HA content possibly
due to improved internalization, trafficking, and transcription
brought about by the polysaccharide. Interestingly, NPs composed
of oligomeric HA yielded lower transfection than polymeric HA
NPs, although the opposite effect was noted for chitosan. The HA
quantity in NPs also affected cytotoxicity as increased HA levels
were associated with a reduction in cellular toxicity. Thus NPs made
of high HA content combined with chitosan oligomers may greatly
improve gene transfer in ocular surface tissues. Later this group
demonstrated transgene expression in the cornea and conjunctiva
in vivo for 7 days with HA-chitosan NPs using a rabbit model (de la
Fuente et al., 2008b). NPs were found to transfect more peripheral
corneal cells possibly as a result of NP accumulation in those
regions. The results of this study may have clinical application in
eye drops for ocular surface abnormalities in the near future.
Recently marketed ultrapure chitosan oligomers (NOVAFECT) were
used by Klausner et al. (2010) as a blueprint in designing novel
chitosan-based NPs with different polycation nitrogen to DNA
phosphate group (N/P) ratios as gene carriers for the cornea. The
chitosan-based NPs were intrastromally injected into rat corneas
and successfully transfected keratocytes in vivo (Klausner et al.,
2010). The transfection efficiency of chitosan and its derivatives is
highly dependent on degree of deacetylation, molecular weight of
chitosan, pH, serum, cell type, and N/P ratio (Kim et al., 2007). The
utility of PEI nanometallic-conjugates for gene delivery has also
been reported (Thomas and Klibanov, 2003). Gold nanoparticles
51
(GNPs) are highly attractive as biocarriers because they are
nontoxic, inert, easy to synthesize, and can condense DNA efficiently (Pissuwan et al., 2011). As gene transporters, GNPs have
displayed significant marker gene delivery and expression into
many mammalian cells (Ghosh et al., 2008; Li et al., 2009a,b; Zhou
et al., 2008; Thomas and Klibanov, 2003). PEI-gold nanoconjugate
transfection efficiency and toxicity largely depends on PEI molecular weight. These authors used 2 kDa PEI (PEI2) conjugated to
GNPs and found that low molecular weight PEI increased nanoconstruct transfection efficiency of COS-7 cells six times more than
PEI25 (Thomas et al., 2005). In addition, the synergistic action of
PEI2-GNPs with N-dodecyl-PEI2 was also demonstrated and
showed increased gene delivery by 11 fold compared to PEI25. An
observation of paramount importance was the transfection of up to
50% cells with ternary complex, whereas unmodified PEI2 and
PEI25 showed 4% and 8% transfection, respectively. The use of PEI2GNPs also resulted in higher cytotoxicity and was linked to the
ability of PEI to enter the nucleus. Subsequently these authors
suggested that optimal N/P ratio is required for high transfection
and is dependant on several factors such as polycation molecular
weight, hydrophobicity, and degradability. The lower N/P ratio
values are indicative of a polycation’s superior ability to condense
DNA and better in vivo gene delivery (Thomas et al., 2005). PEI has
been found to be toxic in vivo (Tiyaboonchai et al., 2003) and
research is underway to minimize its toxicity by chemical modifications (Lungwitz et al., 2005).
Recently we assessed the efficiency of PEI2-GNPs for delivering
genes in the human cornea using an in vitro model. This was the
first time hybrid GNPs were investigated for delivering therapeutics
in the cornea. In these studies PEI2-GNPs with N/P ratios of 60, 90,
120, and 180 showed significant gene transport in human corneal
fibroblasts with no negative effects on cellular viability or phenotype (Mohan et al., unpublished data). Sensing the potential of this
nanoconstruct for corneal nanomedicine, we performed an elaborate study to evaluate PEI2-GNPs toxicity and safety for the cornea
in vivo using a rabbit model. PEI2-GNPs were topically applied onto
the cornea using a cloning cylinder with and without removing
corneal epithelium once for a brief period of time ranging from 2 to
5 min (Sharma et al., 2011). Cloning cylinders are available
commercially and are made in various sizes from a variety of
materials such as glass, porcelain, stainless steel, etc (see Fig. 8A for
appearance). The cloning cylinder was used based on our previous
experience as it enhances vector delivery to corneal cells in a targeted region limiting its contact with neighboring ocular tissues,
and thus preventing unwanted NP entry into adjacent ocular cells.
The rabbit corneal tissues collected at 12 h, 72 h, or 7 d post-PEI2-
Fig. 6. Representative images showing GFP gene delivery by DOTAP nanoparticles in cultured human corneal fibroblasts (A) and HPV16-E6/E7 transformed human corneal
endothelial cells (B). Cell nuclei were stained blue with DAPI while cells expressing GFP gene stained green.
52
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
GNP application displayed substantial gold uptake in the rabbit
cornea with a slow clearance of GNP over time (Sharma et al., 2011).
Transmission electron microscopy detected GNP in the extracellular matrix (ECM) and keratocytes of rabbit corneas. Slit-lamp
biomicroscopy performed 7 days after topical PEI2-GNP application
revealed no corneal inflammation, redness, or edema in rabbit eyes
in vivo although some degree of discoloration in the eye was noted
due to the presence of GNPs (Fig. 7A). The effect of this discoloration on optical properties of the eye is still unknown and requires
further investigation. The rabbit corneas collected 12 h after PEI2GNP application without removing corneal epithelium showed
few TUNELþ cells in the stroma (Fig. 7B) whereas corneas that
received GNPs after removing corneal epithelium showed significant TUNELþ cells (Fig. 7C). These results suggest that PEI2-GNPs
are safe for the cornea and have potential for delivering gene
therapy to the cornea but certainly demand more research in the
area of GNPs clearance from the cornea and its effects on the optical
properties of the eye (Sharma et al., 2011). Past reports point to
renal excretion as the chief route of GNP clearance from the body
(Hainfeld et al., 2006; De Jong et al., 2008). Since the cornea lacks
a vascular supply, this route of clearance is not available. We
postulate that GNPs are cleared from the cornea via slow diffusion
into the tear fluid or aqueous humor (Sharma et al., 2011).
NPs composed of human serum albumin (HSA), the most
abundant plasma protein, have been studied for gene delivery in
the cornea (Jani et al., 2007). HSA complexed with plasmid
encoding a soluble trap for vascular endothelial growth factor
(VEGF) was injected into the mouse stroma three weeks prior to
alkali injury. HSA NPs were detected in keratocytes, showed
transgene expression up to 5 weeks post-injection, and reduced
corneal neovessel formation. Furthermore, no corneal inflammation, edema, opacity, or other toxicity in corneas injected with HSA
NPs was noticed during a three week observatory period (Jani et al.,
2007). It was reported that HSA NPs enter cells via caveolae- or
clathrin-mediated endocytosis, safeguard plasmid from DNase Iand vitreous humor-mediated biodegradation, permit plasmid
escape from lysosome entrapment, and lead to sustained release of
plasmid (>6 d in vitro) resulting in increased gene expression and
activity (Mo et al., 2007).
2.3.3. Dendrimer vectors
Dendrimers, as the name suggests, are highly branched molecules with the ability to envelope or complex other molecules such
as DNA (Mintzer and Simanek, 2009). The potential of dendrimers
as drug delivery systems, antibacterial agents, and bioadhesives in
wound repair for the cornea have been investigated (Durairaj et al.,
2010; Degoricija et al., 2007; Berdahl et al., 2009; Calabretta et al.,
2007). Activated polyamidoamine dendrimers complexed with
plasmid DNA have been shown to transfect 6e10% of corneal
endothelial cells after direct application to the cornea (Hudde et al.,
1999). Marano and colleagues reported inhibition of laser-induced
new blood vessel formation in rat eyes with no adverse effects by
dendrimer-mediated delivery of anti-VEGF oligonucleotide
(Marano et al., 2005). However, the potential of dendrimers for
corneal gene transfer has not been investigated extensively.
3. Tissue-selective gene therapy tools
3.1. Vector engineering
Different vectors have been engineered to attain tissue-selective
targeted gene delivery to corneal cells. A logical approach is to
develop corneal cell-specific vectors. However, such vector engineering is a challenging undertaking and demands active research.
Ideally, cornea cell-specific (epithelial, keratocyte, endothelial)
vectors drive therapeutic gene expression in a controlled and targeted fashion in the desired cells of the cornea making them safe
and effective for clinical applications of gene therapy. The use of
tissue-specific promoters cloned into viral and nonviral vectors can
accomplish tissue selectivity (Klausner et al., 2007). Although
epithelial-specific (Shiraishi et al., 1998) and keratocyte-specific
(Carlson et al., 2004) promoters for the cornea have been
described earlier in murine and rabbit models, promoters with
corneal endothelial cell specificity for gene therapy in vivo have yet
to be defined. The keratin 12 (epithelial-specific) and keratocan
(keratocyte-specific) promoters in conjunction with polymeric
micelles were tested to deliver b-galactosidase marker gene into
the corneas of mice and rabbits in a non-invasive manner using eye
drops (Tong et al., 2007). This approach of using cornea-specific
Fig. 7. Representative slit-lamp biomicroscopy (A) and TUNEL assay images (B and C) showing PEI2-GNP toxicity to rabbit cornea in vivo. No opacity, redness, or inflammation was
seen but PEI2-GNP-treated corneas exhibited mild purple coloration (pointed with arrowhead) due to GNP uptake. The rabbit corneas in which PEI2-GNP was applied after
epithelial removal (B) showed many TUNELþ cells compared to the corneas that received transfection solution without epithelial removal (C). Nuclei are stained blue with DAPI and
TUNELþ cells in red.
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
promoters in plasmids has significantly enhanced transgene
expression in targeted corneal cells (Tong et al., 2007). Another
approach for targeted gene therapy in the cornea includes usage of
an inducible strong promoter with a switch-on and -off mechanism
(Williams and Coster, 2010). These inducible promoters regulate
activation and duration of gene expression, which is triggered in
the presence of a supplementary factor that is either released
endogenously under particular physiological conditions (e.g.
ischemia) or administered exogenously (Bainbridge et al., 2006).
Bitransgenic mouse lines that over-express b-galactosidase in the
corneal epithelium upon induction with doxycycline have been
characterized in the past (Chikama et al., 2005). A gene-targeting
construct with an internal ribosomal entry site-reverse tetracycline transcription activator cassette was introduced into the
keratin 12 gene (Krt12) to generate a knock-in Krt12rtTA/þ mouse
line. These knock-in mice were then bred with tet-O-LacZ reporter
mice to produce Krt12rtTA/þ/tet-O-LacZ bitransgenic mice. Doxycycline ingestion by these bitransgenic mice increased corneal
reporter gene expression 15-fold. In addition, b-galactosidase
enzyme activity was detected 24 h post-antibiotic induction, leveled out at 2 d, and returned to basal levels 4 wks after doxycycline
was eliminated from the diet (Chikama et al., 2005). Not only are
studies of this nature extremely useful in expounding signaling
mechanisms of different growth factors and cytokines, but also in
clarifying the roles of various corneal genes under homeostatic and
pathologic conditions (Chikama et al., 2005). More recently, Parker
et al. (2009) studied a glucocorticosteroid-inducible promoter
(GRE5) in a lentiviral vector encoding IL10 in A549 cells, and ovine
and human corneas in vitro (Parker et al., 2009). A549 cells cultured
with dexamethasone displayed a 30e40-fold increase in IL10
compared to controls while ovine and human corneas demonstrated 9e10-fold increase as quantified with enzyme-linked
immunosorbent assay. This study outlines the efficacy of
a steroid-inducible promoter in corneal gene transfer and paves the
way for its application in future studies involving the modulation of
transgene expression in donor corneal allografts (Parker et al.,
2009). Even now, no ideal non-leaky cell-specific or inducible
vectors are available for corneal gene therapy although considerable progress has been made in this area.
3.2. Custom vector-delivery techniques
Clinicians routinely perform simple surgical procedures such as
epithelial scrape, microinjection, etc in the cornea in the general
eye clinic to treat corneal abnormalities. We postulated a few years
ago that an appropriate combination of vector and vector-delivery
techniques could be used for developing targeted gene therapies.
The ideal vector-delivery technique would involve minimal corneal
injury during and/or following vector application and have the
ability to introduce the vector to a precise location in a controlled
53
manner. Methods such as intracameral delivery, intrastromal
application, topical, and subconjunctival administration have all
shown promising outcomes. However, with the exception of topical
application and subconjunctival injection, all corneal gene transfer
approaches have been deemed invasive with damaging impacts on
the integrity of the ocular surface (Cheng et al., 2007). Even though
topical application is the most acceptable approach to deliver
therapeutics to the eye due to its non-invasive nature, preservation
of corneal integrity, and minimal systemic side effects, the use of
eye drops containing macromolecules including genes for corneal
delivery is ineffective without opening tight junctions to enhance
epithelial layer permeability (Cheng et al., 2007; Hao et al., 2010).
Our recent studies demonstrated that vector-delivery techniques
play a role in gene delivery in the cornea, and could be used as a tool
to reduce contact of vector to unwanted tissue and enhance targetspecificity (Mohan et al., 2010b). A few strategies we have used for
this purpose are shown in Fig. 8 and include cloning cylinder (A)
outlined previously, soaked circular sponge (B), and glass-capillary
(C). It is well known that corneal permeability and hydration affect
corneal transparency (Maurice, 1984). The dehydration of the
cornea with a hair dryer is a conventional method to treat Fuchs’
dystrophy in human patients (Bainbridge et al., 2008). We incorporated this technique currently in clinical use to dehydrate the
cornea in a controlled fashion to increase vector absorption (Mohan
et al., 2010b). After removing the epithelium via gentle scraping
with a beaver blade, a technique commonly used in refractive laser
surgery, a state of corneal dehydration was promoted by applying
a hair dryer once, twice, thrice, or five times to blow warm air on
the ocular surface for 10 s with 5 s intervals. Balanced salt solution
(BSS) or AAV8 vector encoding alkaline phosphatase gene was then
immediately applied topically for 2 min with a custom-designed
cloning cylinder to mouse and rabbit corneas in vivo and human
cornea ex vivo. We found a statistically significant BSS/vector
absorption ranging from 14 to 27% in corneas dried for 20, 30, and
50 s. AAV8-mediated transgene delivery was also increased with
longer exposure to the hair dryer. However, corneas that underwent prolonged drying for 50 s demonstrated amplified infiltration
of activated granulocytes as detected with CD11b immunostaining
whereas 30 s of hair dryer-supplied warm air did not trigger severe
immune reaction or jeopardize corneal morphology (Mohan et al.,
2010b).
It has been long known that corneal epithelial injury induces
apoptosis of keratocytes, inflammation, and wound healing in the
cornea (Mohan et al., 2003; Wilson et al., 2002; Jester et al., 1999).
Several researchers have demonstrated a cascade of cellular events
including cytokine and growth factor release following epithelial
injury leading to varied levels of corneal wound healing response
(Mohan et al., 2003; Wilson et al., 2002; Jester et al., 1999). Scores of
clinical and animal studies have found distinctly different wound
healing responses in the cornea after surgical, mechanical, or
Fig. 8. Representative image showing various minimally invasive defined vector-delivery techniques developed using cloning cylinder (A), soaked circular sponge (B), and glasscapillary (C) for targeted gene therapy approaches for the cornea.
54
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
chemical injury. Many investigators including us have reported that
injury to the corneal epithelium causes keratocyte apoptosis in the
anterior stroma. However, no studies were performed to examine
whether the intensity of mechanical injury dictates the amount of
keratocyte death in the anterior stroma. Recently, we hypothesized
that careful, gentle removal of the corneal epithelium with a #64
surgical blade under controlled conditions induces clinically
insignificant keratocyte apoptosis and wound healing in the cornea
in vivo contrary to rough corneal epithelium removal with a #64
blade under undefined conditions. To test the hypothesis we performed a very simple experiment using a rabbit model. In the same
rabbit eye, 5 mm central corneal epthelium was gently removed
following defined condition and 4 mm peripheral corneal epithelium was removed in a rough manner with no defined conditions
with a #64 surgical blade. Fig. 9 shows the TUNEL assay of the
rabbit cornea collected 24 h after euthanasia. The detection of a few
TUNELþ cells in the central cornea where the epithelium was
removed by defined gentle technique and several TUNELþ cells in
the peripheral cornea where the epithelium was removed by
undefined rough technique strongly support our hypothesis.
Additional support to our hypothesis comes from our recent hair
dryer study in which we observed almost no inflammatory
response in corneas subjected to up to 3 rounds of 10 s dehydration
as a comparable quantity of inflammatory cells were seen in murine
corneas that did not experience hair dryer-assisted dehydration
(Fig. 4, Mohan et al., 2010b). Thus we hold that the administration
of an efficient vector via defined vector-delivery technique can
successfully deliver therapeutic genes in the corneal stroma in vivo
without compromising corneal integrity or function. More research
is required to fully define parameters and techniques that minimize
side effects, improve safety, and augment targeted delivery of
therapeutic genes into keratocytes or the stroma in vivo with AAV
vectors.
Delivering genes precisely into the corneal endothelium is
a huge challenge and a major limitation for developing gene
therapy for corneal dystrophies such as Fuchs’ dystrophy, a leading
cause of corneal blindness with no effective treatments available.
To address this challenge we developed minimally invasive
microsurgical techniques based on the principle of air pressure
force like a water gun as shown in Fig. 10. In our studies we found
these techniques improved target-specificity significantly potentiating transgene delivery into mouse corneal endothelium in vivo.
The newly optimized microsurgical procedures derived from
microinjection technique are simple and may be used routinely in
ophthalmology but may require practice to attain proficiency like
all other surgical techniques.
4. Gene therapy updates in various corneal disorders
Trauma, injury and/or infections to the eye lead to corneal
dysfunction and vision impairment. Corneal disorders are the 3rd
leading cause of blindness in the world according to the World
Health Organization (World Health Organization, 2011; Whitcher
et al., 2001). Eight million people in the world, including 1.5
million children, are blind due to corneal abnormalities (Whitcher
et al., 2001). Corneal disease and disorders have a broad range of
pathology leading to diverse outcomes. Haze, scarring, abnormal
wound healing, new blood vessel ingrowth, conjunctivitis, dry eye,
graft failure, endothelial defects, and Fuchs’ dystrophy are prevalent corneal problems among many acquired and genetic corneal
diseases. Gene therapy holds great promise for treating as well as
preventing corneal diseases and disorders. In the field of corneal
gene therapy emphasis is more on treating common corneal
disorders instead of curing genetic defects. Here we provide an
overview of the progress made in the field of corneal gene therapy
with a brief description about the disease condition/cause.
4.1. Corneal graft rejection
Keratoplasty is currently used for treating many corneal
diseases. According to the Eye Bank Association of America more
than 42,600 corneal transplants were performed in the United
States in 2010 alone (Eye Bank Association of America, 2010).
Although perhaps the most successful transplant procedure, it is
not without blemish. Corneal graft rejection due to immunological
reaction is the chief cause of failure despite the cornea enjoying an
immune-privilege status (Williams and Coster, 2010). Postoperative complications and lack of good quality donor corneas
are the other concerns. Gene therapy approaches have been tested
to improve allograft survival by delivering various therapeutic
genes to modulate cellular transport, apoptosis, angiogenesis, and
wound healing in vivo (Williams and Coster, 2010). In 2006, the
Larkin group reported the delivery of indoleamine 2,3dioxygenase (IDO) in murine corneal endothelium with lentivirus vector (Beutelspacher et al., 2006). An increase in IDO mRNA
and protein levels was detected and IDO expression was found to
be intracellular and restricted to the endothelial layer of the
cornea. Ironically corneal endothelial pathology accounts for
almost 48% of penetrating keratoplasty. Once activated, IDO, an
intracellular enzyme, induced by pro-inflammatory cytokines like
interferon (IFN) and TNF, leads to the immunoregulatory catabolism of tryptophan via opening of the essential amino acid’s indole
ring forming N-formylkynurenine and subsequently L-kynurenine
Fig. 9. Representative TUNEL assay images showing effects of epithelial removal on keratocyte death by custom (A) and non-custom (B) epithelial scrape techniques in the rabbit
corneas. Few TUNELþ cells were noted in customized-gentle scrape technique (A) compared to non-optimized epithelial removal technique (B). Nuclei are stained blue with DAPI
and TUNELþ cells in red.
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
55
Fig. 10. Representative images showing technique utilized for achieving preferential gene delivery into mouse corneal endothelium in vivo.
(Higuchi and Hayaishi, 1967). The IDO-mediated diminution of
tryptophan, in addition to the extracellular release of tryptophan
catabolites, is believed to halt activated T cells in the cell cycle’s G1
phase encouraging immune tolerance (Munn et al., 1999) and
apoptosis essential to the survival of transplanted corneas. Fullthickness corneal grafts were also used by the Larkin group to
study vector-mediated IDO over-expression (Beutelspacher et al.,
2006). The study demonstrated significant prolongation of
corneal allograft survival after IDO transduction of excised donor
cornea ex vivo prior to transplantation compared to GFP and nonIDO-transduced control corneas (Beutelspacher et al., 2006). This
demonstrates the potential of IDO gene therapy to thwart corneal
allograft rejection. A year later in 2007, the Dana group reported
the use of a lentivirus vector to over-express four anti-apoptotic
genes (bcl-xL, bcl-2, survivin and p35) in a rodent corneal transplantation model. They found that lentivirus-mediated transfer of
bcl-xL gene was not only effective at inhibiting apoptosis in the
corneal endothelium in vitro, but also significantly enhanced
corneal graft survival in vivo as a 90% success rate was observed at
8 weeks post-transplantation compared to 40% in uninfected
controls and 30% in reporter gene controls (Barcia et al., 2007).
More recently, the same group studied lentivirus-driven bcl-xL
and p35 gene delivery in both primary human endothelial cells
and human corneas, and found both genes displayed antiapoptotic properties in the corneal endothelium although p35
had greater anti-apoptotic efficiency than bcl-xL (Fuchsluger et al.,
2011). It is also believed that gene therapy at earlier steps like the
sensitization of host to foreign antigen may be more efficacious
(Williams and Coster, 2010). Other potential gene therapy
approaches include utilizing IL4, IL10, CTLA4-Ig, p40-IL12, viral
MIPII, and nerve growth factor (Williams and Coster 2010).
Another avenue for the utilization of gene therapy with regard
to corneal transplantation involves corneal endothelial cell density
maintenance in corneal tissue procured and stored at eye banks.
Recombinant adenovirus-driven over-expression of transcription
factor E2F2 ex vivo demonstrated increased endothelial cell count
in rabbit and human corneas (Joyce et al., 2004; McAlister et al.,
2005). Endothelial cells, normally amitotic, progressed through
the cell cycle from G1 to S phase in significant quantities accounting
for the boost in the monolayer cell density. E2F2 expression was
short lived with expression dropping off 7 d following a 2 h vector
exposure (McAlister et al., 2005). This has great potential in not
only the long-term preservation of corneas but also the donation of
corneas with low endothelial density previously considered
unsuitable for keratoplasty involving endothelial replacement. In
addition, the benefits of the vector-mediated transfer of E2F2 may
prove beneficial in the treatment of corneal endotheliopathies.
4.2. Corneal scarring and wound healing
Wound healing plays a central part in the maintenance of
corneal transparency and thus normal vision. Corneal injury,
regardless of cause, may ignite a dysregulated wound healing
response that commonly leads to corneal fibrosis and loss of visual
function. Corneal wound healing is an extremely intricate process
under the regulation of several cytokines and growth factors.
Among several cytokines transforming growth factor beta (TGFb)
has been identified as a chief player in the formation of a myofibroblasts and opacity in the cornea. Thus, it has become a prime
target for gene therapy aimed at preventing corneal scarring and
other corneal disorders caused by TGFb. Many investigators share
the thought that impeding TGFb or its signal transduction represents a powerful strategy to modulate uncontrolled corneal healing
and prevent or even cure corneal scarring. Recently, using an
in vitro model our laboratory demonstrated that decorin overexpression in human corneal fibroblasts significantly inhibits
TGFb-driven keratocyte differentiation to myofibroblasts without
compromising cellular viability (Mohan et al., 2010a). Decorin,
a member of the small leucine-rich proteoglycan family, is
expressed in the corneal stroma and plays an important role in
wound healing and structural support in the cornea (Mohan et al.,
2011a). Using laser capture technique, we found that photorefractive keratectomy-induced laser injury to the cornea does not
alter decorin significantly but dramatically increases TGFb levels in
the rabbit cornea, in vivo (Tandon et al., 2010). This observation
concurs with our hypothesis that AAV-mediated decorin gene
therapy can effectively reduce corneal scarring in vivo. Indeed, in
a very recent study we detected significant inhibition of corneal
scarring in the rabbit eye (59e73%; p < 0.001 or <0.01) with no
adverse effects at one-month time point with AAV5-mediated
decorin gene therapy (Mohan et al., 2011c). This study was the
first to demonstrate the therapeutic potential of decorin for treating corneal diseases via targeted gene therapy (Fig. 11). Sensing the
translational potential of AAV-mediated decorin gene therapy for
corneal scarring we are studying whether long-term targeted overexpression of decorin causes any complication to the eye or changes
in corneal function or transparency. The clinical and slit-lamp
examination in the eyes of live rabbit at up to 4 months showed no
sign of adverse effects indicating that decorin gene therapy is safe
for patients (Mohan et al., unpublished data). The pending histological, immunochemical, molecular and transmission electron
microscopy studies will validate this notion.
Venturing to block specific targets in TGFb signaling is another
approach used by scientists to develop corneal fibrosis treatment.
Numerous studies have shown that TGFb uses the Smad pathway
56
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
to relay its signal to the nucleus leading to fibrosis (Tandon et al.,
2010). The translocation of Smad2 and Smad4 proteins during
corneal wound healing in vivo has been demonstrated by performing keratectomy and epithelial debridement wounds in
SpragueeDawley rats (Hutcheon et al., 2005). This study motivated us to perform in vitro studies with Smad2, Smad3, Smad4,
and Smad7 siRNA to examine whether TGFb in human corneal
fibroblast cells relays its signal via the Smad pathway. The results
of our study showed that TGFb-mediated keratocyte transdifferentiation to myofibroblasts occurs via Smad signaling and
that Smad7 is an attractive target to prevent myofibroblast
formation. Our findings are in accord with many literature reports
as Smad7 gene therapy has been reported to inhibit fibrosis in
many non-ocular tissues such as kidneys, peritoneum, liver, etc.
(Chen et al., 2011; Dooley et al., 2003). Recently, Galiacy et al.
(2011) used gene therapy strategies to hinder excessive collagen
deposition in corneal scar development. Using a recombinant
AAV-based vector (AAV2/8) the researchers over-expressed
a specific fibril collagenase, matrix metalloproteinase (MMP) 14,
to prevent collagen deposition and subsequent scarring following
incisional injury to the cornea in mice. After microdissecting
a 0.75 mm full-thickness button from the central corneal region,
a single injection of 10 ml PBS into the stroma in control mice was
performed (vector was injected 10 h prior to incisional injury in
the case of AAV2/8-GFP and AAV2/8-MMP14). This previously
outlined model encourages the fibroproliferative response during
repair including the development of alpha smooth muscle actin
(aSMA)-expressing myofibroblasts from stromal keratocytes
(Stramer et al., 2003). The investigators demonstrated that a single
intrastromal injection with recombinant AAVeMMP14 mediated
murine stromal keratocyte gene expression. This was evident with
decreased mRNA expression of aSMA and type III collagen, two
key gene markers of corneal fibrosis (Galiacy et al., 2011). MMP,
like TGFb, also has important implications in other processes such
as neovessel formation.
4.3. Corneal neovascularization
Neovascularization may occur in any of the layers of the cornea
following ocular trauma, infection, injury, etc. and leads to corneal
opacity. The sprouting of new blood vessels in the cornea from the
limbus is closely linked to the inflammatory response and poses
a major risk for corneal allograft rejection (Klausner et al., 2007).
The mechanism of neovascular formation and regression is
tremendously complex involving a slew of cytokines, growth
factors, and cell types. However, it is widely accepted that vascular
endothelial growth factor (VEGF) plays a pivotal role in the development of new blood vessels. A well-designed study performed by
the Ambati group reported that the avascular phenotype of the
cornea is due, at least in part, to soluble VEGF receptor-1(sVEGFR-1)
or sflt-1, an endogenous trap for VEGF (Ambati et al., 2006). Thus
patients with aniridia and mutations in Paired box gene 6 (PAX6)
lack sflt-1 and display spontaneous corneal neovascularization
(CNV). Gene therapy with sflt-1 may greatly benefit patients with
these types of corneal disturbances. Five years earlier in 2001, Lai
and cohorts conducted a study in a rat model showing the inhibition of CNV with recombinant adenovirus-mediated delivery of
sflt-1 (Lai et al., 2001). Gene therapy utilizing sflt-1 may certainly
prove beneficial in averting VEGF-induced neovascularization in
the setting of corneal graft rejection and other corneal disorders. A
variety of gene therapy methodologies have been probed to stem
corneal angiogenesis using experimental models and different
transgenes that target VEGF such as decorin, angiostatin, PEDF,
vasohibin-1 and VEGF receptors Flt-1 and Flk-1 (Cheng et al., 2007;
Jani et al., 2007; Lai et al., 2007; Oh et al., 2010; Kuo et al., 2009; Yu
et al., 2007; Zhou et al., 2010; Mohan et al., 2011b). Adenovirusmediated gene transfer of soluble VEGF receptor-2, sFlk-1 was
shown to inhibit VEGF-stimulated proliferation of murine and
human umbilical vein endothelial cells (Yu et al., 2007). The same
study demonstrated that CNV development in rats in vivo following
cauterization is suppressed by anterior chamber injection of
Fig. 11. Representative stereomicroscopy (A, B) and a-smooth muscle actin immunostaining (C, D) images showing corneal haze inhibition by targeted AAV5-decorin gene therapy
in rabbits in vivo. AAV5 viral titer (6.5 1012 mg/ml) was topically applied onto the cornea once for 2 min after PRK.
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
adenovirus delivery of sFlk-1 (Yu et al., 2007). Immunohistochemistry studies of corneas from these animals showed localization of protein production in the stroma and endothelium 14 days
following cauterization compared to absent sFlk-1 expression in
controls (adenovirus-null and adenovirus-GFP). This strongly
suggests that adenovirus gene therapy has the ability to transduce
corneal cells of the endothelial and stromal layers with sFlk-1,
a vital element in the development of angiogenesis (Yu et al., 2007).
Gene therapy strategies using endostatin have also shown
promise in treating CNV. Endostatin, a naturally occurring fragment
of collagen type XVIII, blocks endothelial cell adhesion, migration
and proliferation, and inhibits apoptosis (O’Reilly et al., 1997). Mice
eyes treated via single subconjunctival injection with recombinant
AAV encoding endostatin following silver nitrate cauterization to
induce CNV showed significant inhibition of corneal angiogenesis.
Endostatin released from the conjunctiva suppressed the migrating
limbal vasculature, was detected at 4 days post-injection, and
retained stable levels for more than 8 months. Interestingly no
difference in immune response (measured by CD4 and CD8
lymphocyte staining) was noticed in mice treated with recombinant
AAV-GFP injection, PBS injection, and no injection (Lai et al., 2007).
Another gene therapy approach to obstruct VEGF-induced CNV
development utilized intrastromal injection of intraceptors linked
to the endoplasmic reticulum retaining peptide KDEL. Plasmids
encoding for specific VEGF binding domains of sFlt-1, Flt23K and
Flt24K, coupled to KDEL were delivered via pCMV vector and were
shown to interrupt VEGF signaling in vitro and in vivo. These
peptides offset intracellular VEGF as they are not liberated from the
cell into the extracellular milieu. Interestingly a variation in VEGF
downregulation was seen between the two intraceptors with
Flt23K suppressing injury induced CNV by 66.8% and Flt24K by
almost 50%, possibly due to differences in protein configuration
(Singh et al., 2005). The same group then tested the ability of Flt23K
to impede corneal neovessel growth using albumin nanoparticles
and showed CNV regression up to 40% at 5 weeks post intrastromal
injection in a murine model (Jani et al., 2007).
Recently Chen and cohorts evaluated gene therapy with
multiple genes known to have antivasculogenic activity in a murine
model of CNV (Chen et al., 2010). Retrovirus encoding murine sFlk1, sTie2, and endostatin genes was used in cell cultures of human
umbilical vein endothelial cells (HUVEC) as well as in vivo studies
involving subconjunctival injection of vector into corneas of C57B1/
6 mice with chemically induced CNV. Studies in vitro showed
decreased HUVEC proliferation with obvious additive effects when
two or three genes were combined (i.e. sFlk-1/sTie2, msFlk-1/
endostatin, sTie2/endostatin, sFlk-1/sTie2/endostatin). However,
although HUVEC migration was also decreased by these genes
individually, the additive effect seen in proliferation was absent
highlighting the various pathways involved in these intricate
processes of CNV. For the in vivo segment of the study, gene
combination showed efficient CNV inhibition specifically downregulating levels of IL-1b, MMP-9, and VEGF as measured by
western blotting and RT-PCR. In addition, the combinations of sFlk1/endostatin and sFlk-1/sTie2/endostatin were found to significantly decrease nuclear factor (NF)-kB phosphorylation and super
repressor inhibitor kappa Ba (IkBa) expression without regard to
total NF-kB quantity (Chen et al., 2010). This has direct implications
to CNV as NF-kB leads to VEGF expression while IkBa inhibits NF-kB
by maintaining it in an inactive complex in the cytoplasm (Josko
and Mazurek, 2004; Karin, 1999).
Combining genes to thwart CNV is not a new concept. For
example, an almost decade old report from Casey Eye Institute
evaluated lentivirus transfer of a fusion protein in an allogenic
transplantation model of CNV. Lentivirus was employed to transfect
rabbit corneal buttons ex vivo with a fusion gene made up of
57
endostatin and the kringle 5 domain of plasminogen. This small
specific plasminogen fragment is known to display robust inhibition of angiogenesis. Corneas transduced with the endostatin and
kringle 5 fusion protein showed decreased proliferation and
migration of endothelial cells and subsequently less post-operative
neovascularization and inflammation in comparison to controls
(Murthy et al., 2003). Other kringle elements have also been
assessed for their antiangiogenic properties. Angiostatin is
a product of proteolytic cleavage that comprises the first four
kringle domains of plasminogen. One group examined the role of
subconjunctival injection of AAV carrying angiostatin in a rat model
of alkali-induced CNV. The researchers illustrated corneal gene
transduction of angiostatin and a post-operative day 7 mean
CNV area of 15.83 0.55 mm2 in untreated controls and
6.87 2.23 mm2 in angiostatin treated animals reflecting 56.6%
abatement of CNV due to angiostatin (Cheng et al., 2007).
In another study, mouse corneas were transfected with DNA
plasmid encoding IL18 using intrastromal injection. The angiostatic
property of IL18 is well known making it potentially useful in the
treatment of ocular disorders associated with CNV such as herpes
keratitis. The investigators showed a decrease in VEGF production
in IL18 treated eyes and subsequently a weakened immunoinflammatory response compared to controls. Furthermore, bFGF
induced MBE cell lines treated with IL18 demonstrated diminished
VEGF production and gene expression in contrast to cells incubated
with OVA (Kim et al., 2005).
Vasohibin-1, an endothelium-derived negative feedback
modulator of neovessel formation characterized by Watanabe, has
also been studied in gene therapy studies as a means to inhibit CNV
(Watanabe et al., 2004; Zhou et al., 2010). A 2010 report by Zhou
and colleagues described the subconjunctival injection of adenovirus encoding vasohibin-1 that resulted in significant CNV
reduction. It was speculated that the antiangiogenic nature of
vasohibin-1 may be related to the downregulation of VEGF type 2
receptor (Zhou et al., 2010).
Other gene transfer studies have assessed the utility of CD36
(Mwaikambo et al., 2006), pigment epithelium derived factor (Kuo
et al., 2009), and GA binding protein (Yoon et al., 2009) among
other approaches to treating CNV. Future experiments may include
vector-mediated delivery of inhibitors of angiogenesis derived from
basement membrane such as neostatin, restin, arresten, canstatin,
and tumstatin (Ellenberg et al., 2010). Recently, our laboratory
detected significant CNV reduction in rabbit eyes after targeted
decorin gene therapy using AAV5 vector (Mohan et al., 2011b). CNV
in rabbit eyes was induced by VEGF implantation and AAV
expressing decorin or GFP gene was topically applied for two
minutes on the rabbit cornea one day after VEGF implantation. As
shown in Fig. 12, targeted decorin delivery in the stroma showed
(52e66% p < 0.05, p < 0.001 or p < 0.01) reduction at various stages
of CNV in rabbit model (Mohan et al., 2011b).
4.4. Corneal alkali burn
Alkali burn injury to the cornea is a major clinical entity that leads
to sight-threatening sequelae such as corneal scarring, neovessel
formation, and ulceration. Scores of preclinical studies have been
dedicated to finding ways to halt the progression of these negative
after effects of alkali burn. In 2007 the Saika group outlined the use of
adenovirus to introduce peroxisome proliferator-activated receptorg (PPARg) to keratocytes in a mouse alkali burn model (Saika et al.,
2007). Gene therapy employing PPARg, a nuclear hormone
receptor with immunomodulatory functions, has been evaluated
extensively for non-ocular tissues (Lewis et al., 2008; Klotz et al.,
2009; Kulkarni et al., 2011). In the cornea, a faint expression of
PPARg was noted via immunohistochemistry in the undamaged
58
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
corneal epithelium as well as corneas subjected to NaOH and treated
with Cre-adenovirus. Conversely PPARg in burned corneas treated
with adenovirus-PPARg showed strong protein expression in
stromal and newly generated epithelial layers (Saika et al., 2007).
PPARg inhibited TGFb1-induced fibroblast to myofibroblast transdifferentiation as well as the production of profibrogenic factors
TGFb1, TGFb2, and connective tissue growth factor (CTGF) in addition to collagen type Ia2 and MMP2/9. Corneal epithelium recovery
and reconstruction of the basement membrane were also encouraged by PPARg gene delivery in healing mice corneas in vivo. This
study also showed that in the cornea PPARg blocks a critical step in
Smad signaling, the nuclear entry of phosphorylated Smad2/3 (Saika
et al., 2007). Another study from same authors reported that
adenovirus-mediated Smad7 over-expression in a murine eye
accelerated corneal wound healing and restricted corneal opacity
(Saika et al., 2005a). Furthermore, adenovirus introduction of bone
morphogenic protein 7 (BMP7) was found to promote corneal reepithelialization, inhibit myofibroblast production and monocyte/
macrophage infiltration, as well as downregulate TGFb, MCP-1, and
collagen expression in the stroma (Saika et al., 2005b). BMP7 overexpression activated Smad1, 5, and 8 and partly inhibited phosphorylated Smad2. However, it was unsuccessful at suppressing
levels of VEGF and the development of NaOH-induced CNV (Saika
et al., 2005b). Apparently, none of the previously discussed studies
investigated the downside of PPARg, SMAD7, or BMP7 gene transfer
in the cornea as no information regarding side effects or adverse
reactions has been reported.
4.5. Other corneal disorders
Corneal inflammation due to ocular herpes simplex virus (HSV)
infection, or herpes stromal keratitis (HSK), is a leading cause of
infectious blindness worldwide particularly in more developed
nations including the U.S. Recurrent infections often lead to corneal
scarring, neovascularization, and thinning resulting in visual
abnormalities and blindness (Kaye and Choudhary, 2006). Many
studies have shared the common goal of developing a vaccine using
gene transfer technology to enhance protection against ocular HSV.
These studies have used individual HSV-1 glycoproteins (i.e. gD) as
well as a cocktail of glycoproteins and have shown variable success
in controlling HSV keratitis (Sharma et al., 2010a). The recent
development of a humanized HLA-Tg rabbit model will hopefully
assist in the design of future vaccines for HSV-1 keratitis as it
demonstrates spontaneous HSV reactivation leading to recurrent
HSK comparable to what is seen clinically. Moreover, this rabbit
model expresses human, rather than rabbit, HLA class I proteins
(Hu et al., 2006; Dasgupta et al., 2009).
In the past siRNAs and RNA enzymes have shown promise in cell
culture experiments but their instability in vivo has been a barrier
to their translation into clinical use. A group of researchers from
different medical disciplines at the University of Florida converged
to engineer a hammerhead ribozyme with a specific target, the
mRNA of an essential late HSV-1 gene, UL20. Ribozymes are RNA
molecules that possess the ability to catalyze the cleavage and
formation of covalent bonds in RNA strands at specific sites. The
“hammerhead” motif is the smallest (approximately 30nucleotides long) and best characterized small catalytic ribozyme
(Citti and Rainaldi, 2005; Mulhbacher et al., 2010). Gene-tailored
ribozymes have been designed, produced, and given to cells to
knock down the expression of specific genes (Citti and Rainaldi,
2005). The investigators in the aforementioned study used iontophoresis and an adenovirus vector to deliver the ribozyme in rabbit
skin cells in addition to rabbit eyes in vivo and discovered significant ribozyme-induced UL20 inhibition leading to a subsequent
decrease in viral load in vitro and decreased ocular HSV-1 severity
in vivo (Liu J, et al. IOVS 2006;47:ARVO E-Abstract 3080). A follow
up study by the same group used the ribozyme to decrease HSV-1
severity in a murine footpad model (Liu et al., 2008b). These findings give credence to the value of RNA gene therapy for HSV-1 by
targeting late genes required for HSV-1 survival and may find
a niche in the treatment of ocular herpes including HSK in the
future. Another group used gene gun method to deliver gold
particles carrying plasmid DNA encoding specific interleukins to
determine effect in HSK. A one time low dose (1 mg/shot) of IL4 or
Fig. 12. Representative stereomicroscopy (A, B) and H & E (C, D) images showing VEGF-induced corneal neovascularization inhibition in rabbits in vivo by AAV5-decorin gene
therapy. Decorin gene was delivered in the rabbit stroma by a single topical application of AAV5 titer (5 1012 mg/ml) on the cornea for 2 min.
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
IL10 plasmid DNA was found to significantly abrogate the clinical
course of HSK when administered at 300 psi helium pressure with
minimal alterations in immune response systemically. IL6, shown
to play a role in HSK development (Banerjee et al., 2004), was also
downregulated. In addition, gene gun utilization did not lead to
significant corneal damage as ascertained by histology and clinical
examination (Bauer et al., 2006).
Diabetic disease of the cornea is a common occurrence in
patients with type I and II diabetes mellitus and can lead to sightthreatening corneal neuropathy and epitheliopathy. Currently, no
treatment exists to thwart the progression or cure the disease and
treatment regimens are largely symptomatic. Saghizadeh et al.
(2010) recently used recombinant AV to over-express the hepatocyte growth factor (HGF) receptor c-met proto-oncogene in corneal
epithelium of diabetic human corneas in vitro. Their past studies
outlined disturbances in HGF/c-met signaling in diabetic corneas as
c-met expression was reduced in the face of upregulated HGF
expression (Saghizadeh et al., 2005). With the introduction of
exogenous c-met via recombinant AV vector they reported
improved reestablishment of HGF signaling and subsequent
restoration of marker protein patterns and enhanced wound
healing (Saghizadeh et al., 2010).
Mucopolysaccharidosis (MPS) is a set of metabolic disorders
characterized by the inability to breakdown glycosaminoglycans
(GAG) due to lysosomal enzyme deficiency. GAGs thus accumulate
in lysosomes systemically as well as in the eyes (Ashworth et al.,
2006). MPS VII, or Sly disease, is due to b-glucuronidase (GUSB)
insufficiency leading to the stockpiling of GAGs (dermatan, heparan, and chondroitin sulfate) while MPS VI, or MaroteauxeLamy
syndrome, results from aryl N-acetylgalactosamine-4-sulfatase
deficiency leading to accumulation of dermatan sulfate
(Ashworth et al., 2006). Pathology is common in the ocular system
and results in visual impairment due to opacification of the cornea
and disorders of other ocular tissues (Ashworth et al., 2006). While
lysosomal accumulation occurs in the corneal endothelium, it is
their storage in keratocytes that causes corneal opacity (Williams
and Coster, 2010). Systemic gene therapy using a retroviral vector
has been shown to avert GAG accrual and ensuing corneal clouding
due to MPS using neonatal canines (Wang et al., 2006; Traas et al.,
2007; Ponder and Auricchio, 2010). Even 6.5e8 years following
therapy, little or no corneal opacity was detected in dogs with MPS
VII (Ponder and Auricchio, 2010). Gene therapy may have a role as
an alternative therapeutic strategy for MPS disorders by introducing genes that encode for specific enzymes, such as GUSB,
clearing lysosome build up in the eye and other tissues.
5. Lessons learned from corneal genetic studies: humans and
animals
Advances in molecular biology have led to the identification of
numerous genes and gene mutations associated with dystrophic
diseases of the cornea. Different modes of Mendelian inheritance are
evident in corneal dystrophies likely due to alterations in several
genes including CHST6, KRT3, TGFBI, and UBIAD1 (Klintworth, 2009).
The TGFb induced gene (TGFBI), commonly known as big-h3, encodes
the TGFBI protein (TGFBIp or keratoepithelin). Changes in the
genomic sequence of TGFBI have been linked to several dystrophies
of the corneal stroma such as Lattice dystrophy type I, Reis-Buckler
dystrophy, Thiel-Behnke corneal dystrophy, granular corneal
dystrophy type I, and granular corneal dystrophy II (Avellino corneal
dystrophy) (Runager et al., 2008; Klintworth, 2009). Indeed 38
different mutations in the TGFBI gene are responsible for various
corneal dystrophies, the majority being missense mutations
(Runager et al., 2008). TGFBIp functions as a cell adhesion molecule
acting by way of integrins (a1b1, a3b1, avb3, etc) and elements of the
59
ECM including proteoglycans (i.e. decorin), fibronectin, and collagens
(Runager et al., 2008). The precise function of these interactions
remains to be identified although TGFBIp may play a role as a regulatory modulator between the ECM and cells found in its supporting
meshwork (Runager et al., 2008). Dystrophic corneas contain
aggregates of unknown makeup although increased TGFBIp has been
detected in addition to clusterin, a proaggregation protein, in Fuchs
endothelial corneal dystrophy (Runager et al., 2008; Jurkunas et al.,
2009). A recent report identified the two corneal dystrophies,
gelatinous drop-like corneal dystrophy and spheroidal degeneration,
in the same patient (Zhang and Yao, 2010). Upon molecular genetic
examination a new missense mutation in the tumor associated
calcium signal transducer 2 (TACSTD2) gene was unearthed (Zhang
and Yao, 2010). Earlier it was found that alterations in the UbiA prenyltransferase domain containing 1 gene are responsible for
Schnyder corneal dystrophy (Weiss et al., 2007). The corneal endothelial defect was found to be caused, at least in part, by variations in
the transcription factor 4 (TCF4) gene (Baratz et al., 2010). The
knowledge of gene localization is paramount in creating an animal
model of disease and treatment approaches via gene therapy.
Previously it was thought that type I and II macular corneal dystrophy
were due to genetic aberrations of carbohydrate sulfotransferase 6
(CHST6) in the epithelial layer (Abbruzzese et al., 2004) and that
limbal stem-cell therapy might possibly offer benefits to patients in
terms of therapy. It was recently shown however, that CHST6 along
with keratan sulfate are chiefly the stromal products of keratocytes
underscoring the need for therapeutic modalities, including gene
therapy, to target stromal keratocytes (Di Iorio et al., 2010).
Nonetheless, the lack of in vivo animal models has sharply
hindered progress in the development of gene therapy options for
patients suffering from corneal dystrophies (Weiss, 2010).
Furthermore, the pathogenesis and localization of genes responsible for many of these disorders has yet to be elucidated. Recently,
an animal model mimicking human corneal dystrophy was reported by Chen et al. (2010). A mouse model of congenital stromal
corneal dystrophy, caused by a faulty decorin gene leading to
corneal clouding, was created utilizing a Cre-on approach. It was
also surmised that mutations in decorin accounting for the disorganized stromal architecture may be influenced by class II SLRPs
lumican and keratocan (Chen S, et al. IOVS 2010;51:ARVO EAbstract 3832). It is conceivable that keratocytes may induce an
increase in GAG chain and SLRP production to offset deficiencies in
stromal ECM GAGs or SLRPs brought about by genetic causes. The
tissue-selective targeted gene-based therapeutic approaches we
are attempting to define for the cornea have great potential for
corneal genetic dystrophies.
6. Ocular gene therapy clinical trials: current status
Currently 20 gene therapy trials for eye disease are listed on The
Journal of Gene Medicine Clinical Trial Website. Out of 20 gene
therapy clinical trials, 18 are directed toward retinal disorders, 1 for
glaucoma, and 1 for the cornea. The lone corneal gene therapy
Phase I/II clinical trial is focused on evaluating the safety and efficacy of a matrix-targeted retroviral vector bearing a dominant
negative cyclin G1 construct (Mx-dnG1) as adjunctive intervention
for superficial corneal opacity/corneal scarring. The findings of the
clinical trial assessing gene therapy for diseases of the cornea have
not yet been published.
7. Future directions: regenerative medicine, nanomedicine
and other approaches
The future of gene therapy for the cornea is very promising and
involves different branches of medicine including regenerative
60
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
medicine and nanotechnology. Indeed, nanotechnology will lead to
the development of regenerative medicine, microsensors and
feedback apparatus, improved imaging in vivo, and artificial vision
(Zarbin et al., 2010a). Regenerative nanomedicine is a new field in
medicine gaining widespread attention. It entails the use of nanoparticles containing regulatory molecules such as gene transcription factors that allow for cellular reprogramming in vivo (Zarbin
et al., 2010a). Nanoparticles may not only serve as vectors for
therapeutic gene delivery but may also act as biocarriers of genes
which may assist in the diagnosis and monitoring of corneal
disease. This process of coupling disease diagnosis with targeted
treatment is termed theragnostics and has enormous potential in
the ocular system including the cornea. For example, oxidative
stress has been implicated in numerous inflammatory, metabolic,
traumatic, and iatrogenic corneal diseases (Shoham et al., 2008). By
coupling a therapeutic gene such as peroxidase, superoxide dismutase, or catalase (as well as a marker gene like GFP) to nanoparticles capable of activating antioxidant response element, one
may produce a nano-device with dual diagnosticetherapeutic
effects enabling corneal cells to treat themselves following oxidative damage (Zarbin et al., 2010b).
Fagerholm and colleagues recently reported the results of
a clinical study in Sweden involving corneal regeneration with
recombinant human collagen. Recombinantly produced human
collagen type III was cross-linked with 1-ethyl-3-(3dimethylaminopropyl) carbodiimide (EDC) and N-hydroxysuccinimide (NHS) and fashioned into substitutes with human
corneal dimensions. This biomimetic substitute was then transplanted into patients following anterior lamellar keratoplasty. The
investigators found that a cell-free corneal substitute imitating the
cornea’s unique ECM can successfully provide support for endogenous restoration of corneal epithelium, stroma, and nerves. Of
note was the absence of inflammation, neovascularization, or
rejection that plagues recipients of human donor corneas
(Fagerholm et al., 2009). Although the study had a small patient
population (n ¼ 10), it clearly highlights the safety profile of
recombinant human collagen and cross-linking methods
(Fagerholm et al., 2009). Preliminary results from the study
demonstrate improved visual acuity, ocular surface quality, and
corneal sensitivity in patients with a biomimetic corneal substitute.
These corneal substitutes may prove beneficial in circumstances
necessitating short-term or emergency corneal replacements,
where availability of human donor corneas is poor or nonexistent
and where prostheses are unfavorable (Fagerholm et al., 2009). In
the future these biomimetic corneas may serve as efficient
permanent corneal replacements and a viable option to human
corneal tissue in transplantation. These substitutes in combination
with single or multiple gene therapy may find a niche in the
treatment of recurrent CNV and other disorders of the ocular
surface.
Cell-based therapy for corneal diseases has also been examined.
A study from the Funderburgh group (Du et al., 2009) used stem
cells isolated from adult human corneal stroma to rectify stromal
opacity due to SLRP lumican deficiency in mice via intrastromal
injection of cells. Stem cells subsisted in wild-type mice for months
without host-cell fusion or provoking an immune T-cell response.
Human stromal stem cells introduced into the stroma of lumicannull mice with corneal opacity (similar in nature to that of scar
tissue due to disorganized stromal collagen) led to rectification of
fibrillar collagen defects and stromal thickness and restoration of
corneal transparency (Du et al., 2009). Thus stem-cell therapy
shows proof of principle in treating corneal disease through the
regeneration of corneal tissue in a safe and efficacious manner
similar to organogenesis. The results of a remarkable clinical study
in Italy involving autologous limbal cell treatment were recently
reported (Rama et al., 2010). Currently limbal stem cells, a group of
cells responsible for corneal epithelial renewal and regeneration,
are transplanted as a treatment for limbal stem-cell deficiency
(Pellegrini et al., 2009). 112 patients with burn-related corneal
destruction associated with limbal stem-cell deficiency were
treated with autologous limbal stem cells cultivated on fibrin.
Corneal transparency was restored in 76.6% of eyes up to 10 years
after corneal transplantation with limbal stem-cell grafts identifying limbal stem-cell cultures as a source of transplantable cells for
treating corneal burns (Rama et al., 2010).
Future studies involving gene therapy for corneal diseases may
include non-coding RNA (ncRNA). ncRNA includes several types of
functional RNA molecules not translated into proteins that play key
roles in several cellular processes such as transfer RNA, ribosomal
RNA, small interfering RNAs, microRNAs, PIWI-associated RNAs,
small nucleolar RNAs, promoter-associated RNAs, and telomere
specific small RNAs. It has been shown that ncRNAs play a part in
oncogenesis and may be a factor in molecular aberrations of other
diseases (Galasso et al., 2010).
In the recent past, toll like receptors (TLRs) have gained attention in the cornea as their activation sets off a multifaceted
signaling pathway instigating innate and adaptive immunity
generating inflammatory cytokines and co-stimulatory molecules
(Redfern and McDermott, 2010). The production of chemokines
encourages corneal infiltration of macrophages and neutrophils
resulting in the functional loss of corneal epithelium, stroma, and
endothelium leading to the loss of corneal transparency and visual
activity (Kanwar et al., 2010). The modulation of TLR signaling via
gene therapy methods may play a vital role in blocking immunemediated corneal graft rejection as well as aberrant wound healing following corneal infection.
The future will reveal animal models of human corneal disease
in which to test highly efficient viral or nonviral vectors with lowimmunogenicity and sustained transgene expression in combination with cell-based therapy and regenerative approaches. Presently the cornea lags behind the retina in gene therapy for various
reasons including lack of animal models of inherited disease for
experimentation. Currently available technologies are likely
nontoxic and effective, at least in the short term, with little further
development (Weiss, 2010).
8. Conclusions
The last five to six years have brought about great progress and
advancement in the field of corneal gene therapy. The increased
understanding of the molecular mechanisms and course of
inherited and acquired corneal diseases have allowed for the
emergence of targets for gene-based therapy. The testing of various
viral and nonviral vectors and multiple delivery systems has been
the key to the success observed in various models of corneal
disease. We along with other investigators have examined the
efficacy of gene therapy using in vitro, ex vivo, and in vivo animal
(equine, murine, canine, etc) and human corneal tissues, identified
several efficient AAV serotypes and nanoparticles, and defined
simple minimally invasive vector-delivery techniques for delivering
genes precisely into targeted cells (keratocytes and endothelium) of
the cornea in vivo. Furthermore, using a combination of vector and
delivery techniques, we have defined tissue-selected targeted gene
therapy approaches for the cornea. Although much of the corneal
gene therapy investigations have been accomplished in animals,
there is a dearth of research that has been translated fully from
bench to bedside including the clinic. The progress in establishing
corneal gene therapy clinical trials has been meager compared to
gene therapy trials conducted in other ocular tissues. Even though
the cornea is easily accessible for gene transfer, it is lagging far
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
behind the retina in terms of clinical application of gene therapy for
human patients. The future is bright however, as novel innovations
and treatment approaches have proved effective in animal models
(i.e. rabbits) that closely resemble the human cornea in terms of
anatomy, wound healing response, and function.
Over the past few years, much effort has been made to develop
strategies for effective delivery of DNA to the nucleus of target cells.
The most pertinent question concerning the success of gene
therapy is “how to introduce therapeutic genes into target cells/
tissues?” Vectors are important tools for introducing foreign genes
into cells and are one of the two major determinants for the success
of gene therapy, the other being gene delivery technique. Several
viral and nonviral vectors have been developed and each has its
own advantages as well as limitations. In addition, many vectordelivery techniques have been developed to enhance vector efficiency and achieve target-specificity. With the discovery of animal
models of human corneal disease and the combination of gene
therapy approaches with stem-cell therapy and regenerative
medicine, the cure for corneal blindness may well become a reality
within the next decade.
Acknowledgments
The work was supported from the RO1EY17294 (R.R.M.)
National Eye Institute, NIH, Bethesda, MD, NEI/NIH RO1EY17294S2
Diversity Supplement (J.C.K.T.), 1I01BX00035701 (R.R.M.) Veteran
Health Affairs, Washington, DC, and an Unrestricted Research to
Prevent Blindness, New York, NY grants.
References
Abbruzzese, C., Kuhn, U., Molina, F., Rama, P., De Luca, M., 2004. Novel mutations in
the CHST6 gene causing macular corneal dystrophy. Clin. Genet. 65, 120e125.
Adriaansen, J., Vervoordeldonk, M.J., Tak, P.P., 2006. Gene therapy as a therapeutic
approach for the treatment of rheumatoid arthritis: innovative vectors and
therapeutic genes. Rheumatology (Oxford) 45, 656e668.
Akache, B., Grimm, D., Pandey, K., Yant, S.R., Xu, H., Kay, M.A., 2006. The 37/67kilodalton laminin receptor is a receptor for adeno-associated virus serotypes
8, 2, 3, and 9. J. Virol. 80, 9831e9836.
Alexander, J.J., Hauswirth, W.W., 2008. Adeno-associated viral vectors and the
retina. Adv. Exp. Med. Biol. 613, 121e128.
Ambati, B.K., Nozaki, M., Singh, N., Takeda, A., Jani, P.D., Suthar, T., Albuquerque, R.J.,
Richter, E., Sakurai, E., Newcomb, M.T., Kleinman, M.E., Caldwell, R.B., Lin, Q.,
Ogura, Y., Orecchia, A., Samuelson, D.A., Agnew, D.W., St Leger, J., Green, W.R.,
Mahasreshti, P.J., Curiel, D.T., Kwan, D., Marsh, H., Ikeda, S., Leiper, L.J.,
Collinson, J.M., Bogdanovich, S., Khurana, T.S., Shibuya, M., Baldwin, M.E.,
Ferrara, N., Gerber, H.P., De Falco, S., Witta, J., Baffi, J.Z., Raisler, B.J., Ambati, J.,
2006. Corneal avascularity is due to soluble VEGF receptor-1. Nature 443,
993e997.
Ashworth, J.L., Biswas, S., Wraith, E., Lloyd, I.C., 2006. Mucopolysaccharidoses and
the eye. Surv. Ophthalmol. 51, 1e17.
Bainbridge, J.W., Tan, M.H., Ali, R.R., 2006. Gene therapy progress and prospects: the
eye. Gene Ther. 13, 1191e1197.
Bainbridge, J.W., Smith, A.J., Barker, S.S., Robbie, S., Henderson, R., Balaggan, K.,
Viswanathan, A., Holder, G.E., Stockman, A., Tyler, N., Petersen-Jones, S.,
Bhattacharya, S.S., Thrasher, A.J., Fitzke, F.W., Carter, B.J., Rubin, G.S., Moore, A.T.,
Ali, R.R., 2008. Effect of gene therapy on visual function in Leber’s congenital
amaurosis. N. Engl. J. Med. 358, 2231e2239.
Banerjee, K., Biswas, P.S., Kim, B., Lee, S., Rouse, B.T., 2004. CXCR2 / mice show
enhanced susceptibility to herpetic stromal keratitis: a role for IL-6-induced
neovascularization. J. Immunol. 172, 1237e1245.
Baratz, K.H., Tosakulwong, N., Ryu, E., Brown, W.L., Branham, K., Chen, W., Tran, K.D.,
Schmid-Kubista, K.E., Heckenlively, J.R., Swaroop, A., Abecasis, G., Bailey, K.R.,
Edwards, A.O., 2010. E2-2 protein and Fuchs’s corneal dystrophy. N. Engl. J. Med.
363, 1016e1024.
Barcia, R.N., Dana, M.R., Kazlauskas, A., 2007. Corneal graft rejection is accompanied
by apoptosis of the endothelium and is prevented by gene therapy with bcl-xL.
Am. J. Transplant. 7, 2082e2089.
Bauer, D., Lu, M., Wasmuth, S., Li, H., Yang, Y., Roggendorf, M., Steuhl, K.P.,
Heiligenhaus, A., 2006. Immunomodulation by topical particle-mediated
administration of cytokine plasmid DNA suppresses herpetic stromal keratitis
without impairment of antiviral defense. Graefes Arch. Clin. Exp. Ophthalmol.
244, 216e225.
Bemelmans, A.P., Arsenijevic, Y., Majo, F., 2009. Efficient lentiviral gene transfer into
corneal stroma cells using a femtosecond laser. Gene Ther. 16, 933e938.
61
Berdahl, J.P., Johnson, C.S., Proia, A.D., Grinstaff, M.W., Kim, T., 2009. Comparison of
sutures and dendritic polymer adhesives for corneal laceration repair in an
in vivo chicken model. Arch. Ophthalmol. 127, 442e447.
Beutelspacher, S.C., Ardjomand, N., Tan, P.H., Patton, G.S., Larkin, D.F., George, A.J.,
McClure, M.O., 2005. Comparison of HIV-1 and EIAV-based lentiviral vectors in
corneal transduction. Exp. Eye Res. 80, 787e794.
Beutelspacher, S.C., Pillai, R., Watson, M.P., Tan, P.H., Tsang, J., McClure, M.O.,
George, A.J., Larkin, D.F., 2006. Function of indoleamine 2,3-dioxygenase in
corneal allograft rejection and prolongation of allograft survival by overexpression. Eur. J. Immunol. 36, 690e700.
Broekman, M.L., Comer, L.A., Hyman, B.T., Sena-Esteves, M., 2006. Adeno-associated
virus vectors serotyped with AAV8 capsid are more efficient than AAV-1 or -2
serotypes for widespread gene delivery to the neonatal mouse brain. Neuroscience 138, 501e510.
Cai, X., Conley, S., Naash, M., 2008. Nanoparticle applications in ocular gene therapy.
Vision Res. 48, 319e324.
Calabretta, M.K., Kumar, A., McDermott, A.M., Cai, C., 2007. Antibacterial activities of
poly(amidoamine) dendrimers terminated with amino and poly(ethylene
glycol) groups. Biomacromolecules 8, 1807e1811.
Carlson, E.C., Liu, C.Y., Yang, X., Gregory, M., Ksander, B., Drazba, J., Perez, V.L., 2004.
In vivo gene delivery and visualization of corneal stromal cells using an
adenoviral vector and keratocyte-specific promoter. Invest. Ophthalmol. Vis. Sci.
45, 2194e2200.
Challa, P., Luna, C., Liton, P.B., Chamblin, B., Wakefield, J., Ramabhadran, R.,
Epstein, D.L., Gonzalez, P., 2005. Lentiviral mediated gene delivery to the
anterior chamber of rodent eyes. Mol. Vis. 11, 425e430.
Chen, H.Y., Huang, X.R., Wang, W., Li, J.H., Heuchel, R.L., Chung, A.C., Lan, H.Y., 2011.
The protective role of Smad7 in diabetic kidney disease: mechanism and
therapeutic potential. Diabetes 60, 590e601.
Chen, P., Yin, H., Wang, Y., Mi, J., He, W., Xie, L., Wang, Y., 2010. Multi-gene targeted
antiangiogenic therapies for experimental corneal neovascularization. Mol. Vis.
16, 310e319.
Cheng, H.C., Yeh, S.I., Tsao, Y.P., Kuo, P.C., 2007. Subconjunctival injection of
recombinant AAV-angiostatin ameliorates alkali burn induced corneal angiogenesis. Mol. Vis. 13, 2344e2352.
Chikama, T., Hayashi, Y., Liu, C.Y., Terai, N., Terai, K., Kao, C.W., Wang, L., Hayashi, M.,
Nishida, T., Sanford, P., Doestchman, T., Kao, W.W., 2005. Characterization of
tetracycline-inducible bitransgenic Krt12rtTA/þ/tet-O-LacZ mice. Invest. Ophthalmol. Vis. Sci. 46, 1966e1972.
Citti, L., Rainaldi, G., 2005. Synthetic hammerhead ribozymes as therapeutic tools to
control disease genes. Curr. Gene Ther. 5, 11e24.
Dannowski, H., Bednarz, J., Reszka, R., Engelmann, K., Pleyer, U., 2005. Lipid-mediated gene transfer of acidic fibroblast growth factor into human corneal
endothelial cells. Exp. Eye Res. 80, 93e101.
Dasgupta, G., Chentoufi, A.A., Nesburn, A.B., Wechsler, S.L., BenMohamed, L., 2009.
New concepts in herpes simplex virus vaccine development: notes from the
battlefield. Expert Rev. Vaccines 8, 1023e1035.
De Jong, W.H., Hagens, W.I., Krystek, P., Burger, M.C., Sips, A.J., Geertsma, R.E., 2008.
Particle size-dependent organ distribution of gold nanoparticles after intravenous administration. Biomaterials 29, 1912e1919.
de la Fuente, M., Seijo, B., Alonso, M.J., 2008a. Bioadhesive hyaluronanechitosan
nanoparticles can transport genes across the ocular mucosa and transfect
ocular tissue. Gene Ther. 15, 668e676.
de la Fuente, M., Seijo, B., Alonso, M.J., 2008b. Novel hyaluronic acid-chitosan nanoparticles for ocular gene therapy. Invest. Ophthalmol. Vis. Sci. 49, 2016e2024.
Degoricija, L., Johnson, C.S., Wathier, M., Kim, T., Grinstaff, M.W., 2007. Photo crosslinkable biodendrimers as ophthalmic adhesives for central lacerations and
penetrating keratoplasties. Invest. Ophthalmol. Vis. Sci. 48, 2037e2042.
Di Iorio, E., Barbaro, V., Volpi, N., Bertolin, M., Ferrari, B., Fasolo, A., Arnaldi, R.,
Brusini, P., Prosdocimo, G., Ponzin, D., Ferrari, S., 2010. Localization and
expression of CHST6 and keratan sulfate proteoglycans in the human cornea.
Exp. Eye Res. 91, 293e299.
Dooley, S., Hamzavi, J., Breitkopf, K., Wiercinska, E., Said, H.M., Lorenzen, J., Ten
Dijke, P., Gressner, A.M., 2003. Smad7 prevents activation of hepatic stellate
cells and liver fibrosis in rats. Gastroenterology 125, 178e191.
Douar, A.M., Poulard, K., Stockholm, D., Danos, O., 2001. Intracellular trafficking of
adenoassociated virus vectors: routing to the late endosomal compartment and
proteasome degradation. J. Virol. 75, 1824e1833.
Dratviman-Storobinsky, O., Lubin, B.C., Hasanreisoglu, M., Goldenberg-Cohen, N.,
2009. Effect of subconjuctival and intraocular bevacizumab injection on
angiogenic gene expression levels in a mouse model of corneal neovascularization. Mol. Vis. 15, 2326e2338.
Du, Y., Carlson, E.C., Funderburgh, M.L., Birk, D.E., Pearlman, E., Guo, N., Kao, W.W.,
Funderburgh, J.L., 2009. Stem cell therapy restores transparency to defective
murine corneas. Stem Cells 27, 1635e1642.
Durairaj, C., Kadam, R.S., Chandler, J.W., Hutcherson, S.L., Kompella, U.B., 2010.
Nanosized dendritic polyguanidilyated translocators for enhanced solubility,
permeability, and delivery of gatifloxacin. Invest. Ophthalmol. Vis. Sci. 51,
5804e5816.
Ellenberg, D., Azar, D.T., Hallak, J.A., Tobaigy, F., Han, K.Y., Jain, S., Zhou, Z.,
Chang, J.H., 2010. Novel aspects of corneal angiogenic and lymphangiogenic
privilege. Prog. Retin. Eye Res. 29, 208e248.
Eye Bank Association of America 2010 Statistical Report. URL: http://www.restoresight.
org/about-us/understanding-donation/cornea-donation-transplantation-statistics/
(accessed on 23.09.11).
62
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
Fagerholm, P., Lagali, N.S., Carlsson, D.J., Merrett, K., Griffith, M., 2009. Corneal
regeneration following implantation of a biomimetic tissue-engineered
substitute. Clin. Transl. Sci. 2, 162e164.
Fuchsluger, T.A., Jurkunas, U., Kazlauskas, A., Dana, R., 2011. Corneal endothelial cells
are protected from apoptosis by gene therapy. Hum. Gene Ther. 22, 549e558.
Galasso, M., Elena Sana, M., Volinia, S., 2010. Non-coding RNAs: a key to future
personalized molecular therapy? Genome Med. 2, 12.
Galiacy, S.D., Fournié, P., Massoudi, D., Ancèle, E., Quintyn, J.C., Erraud, A., RaymondLetron, I., Rolling, F., Malecaze, F., 2011. Matrix metalloproteinase 14 overexpression reduces corneal scarring. Gene Ther. 18, 462e468.
Gao, G., Vandenberghe, L.H., Wilson, J.M., 2005. New recombinant serotypes of AAV
vectors. Curr. Gene Ther. 5, 285e297.
Gao, G., Lu, Y., Calcedo, R., Grant, R.L., Bell, P., Wang, L., Figueredo, J., Lock, M.,
Wilson, J.M., 2006. Biology of AAV serotype vectors in liver-directed gene
transfer to nonhuman primates. Mol. Ther. 13, 77e87.
Gao, X., Kim, K.S., Liu, D., 2007. Nonviral gene delivery: what we know and what is
next. AAPS J. 9, E92e104.
Ghosh, A., Yue, Y., Long, C., Bostick, B., Duan, D., 2007. Efficient whole-body transduction with trans-splicing adeno-associated viral vectors. Mol. Ther. 15,
750e755.
Ghosh, P.S., Kim, C.K., Han, G., Forbes, N.S., Rotello, V.M., 2008. Efficient gene
delivery vectors by tuning the surface charge density of amino acidfunctionalized gold nanoparticles. ACS Nano. 2, 2213e2218.
Gong, N., Pleyer, U., Vogt, K., Anegon, I., Flügel, A., Volk, H.D., Ritter, T., 2007a. Local
overexpression of nerve growth factor in rat corneal transplants improves
allograft survival. Invest. Ophthalmol. Vis. Sci. 48, 1043e1052.
Gong, N., Pleyer, U., Volk, H.D., Ritter, T., 2007b. Effects of local and systemic viral
interleukin-10 gene transfer on corneal allograft survival. Gene Ther. 14,
484e490.
Hacein-Bey-Abina, S., Garrigue, A., Wang, G.P., Soulier, J., Lim, A., Morillon, E.,
Clappier, E., Caccavelli, L., Delabesse, E., Beldjord, K., Asnafi, V., MacIntyre, E., Dal
Cortivo, L., Radford, I., Brousse, N., Sigaux, F., Moshous, D., Hauer, J.,
Borkhardt, A., Belohradsky, B.H., Wintergerst, U., Velez, M.C., Leiva, L.,
Sorensen, R., Wulffraat, N., Blanche, S., Bushman, F.D., Fischer, A., CavazzanaCalvo, M., 2008. Insertional oncogenesis in 4 patients after retrovirus-mediated
gene therapy of SCID-X1. J. Clin. Invest. 118, 3132e3142.
Hainfeld, J.F., Slatkin, D.N., Focella, T.M., Smilowitz, H.M., 2006. Gold nanoparticles:
a new X-ray contrast agent. Br. J. Radiol. 79, 248e253.
Hao, J., Li, S.K., Liu, C.Y., Kao, W.W., 2009. Electrically assisted delivery of macromolecules into the corneal epithelium. Exp. Eye Res. 89, 934e941.
Hao, J., Li, S.K., Kao, W.W., Liu, C.Y., 2010. Gene delivery to cornea. Brain Res. Bull. 81,
256e261.
Hauswirth, W.W., Aleman, T.S., Kaushal, S., Cideciyan, A.V., Schwartz, S.B., Wang, L.,
Conlon, T.J., Boye, S.L., Flotte, T.R., Byrne, B.J., Jacobson, S.G., 2008. Treatment of
leber congenital amaurosis due to RPE65 mutations by ocular subretinal
injection of adeno-associated virus gene vector: short-term results of a phase I
trial. Hum. Gene Ther. 19, 979e990.
He, Z., Pipparelli, A., Manissolle, C., Acquart, S., Garraud, O., Gain, P., Thuret, G., 2010.
Ex vivo gene electrotransfer to the endothelium of organ cultured human
corneas. Ophthalmic Res. 43, 43e55.
Herzog, R.W., 2010. Gene therapy for SCID-X1: round 2. Mol. Ther. 18, 1891.
Higuchi, K., Hayaishi, O., 1967. Enzymic formation of d-kynurenine from dtryptophan. Arch. Biochem. Biophys. 120, 397e403.
Hillaireau, H., Couvreur, P., 2009. Nanocarriers’ entry into the cell: relevance to drug
delivery. Cell. Mol. Life Sci. 66, 2873e2896.
Hornof, M., de la Fuente, M., Hallikainen, M., Tammi, R.H., Urtti, A., 2008. Low
molecular weight hyaluronan shielding of DNA/PEI polyplexes facilitates CD44
receptor mediated uptake in human corneal epithelial cells. J. Gene Med. 10,
70e80.
Hu, J., Peng, X., Schell, T.D., Budgeon, L.R., Cladel, N.M., Christensen, N.D., 2006. An
HLA-A2.1-transgenic rabbit model to study immunity to papillomavirus infection. J. Immunol. 177, 8037e8045.
Hudde, T., Rayner, S.A., Comer, R.M., Weber, M., Isaacs, J.D., Waldmann, H.,
Larkin, D.F., George, A.J., 1999. Activated polyamidoamine dendrimers, a nonviral vector for gene transfer to the corneal endothelium. Gene Ther. 6,
939e943.
Hutcheon, A.E., Guo, X.Q., Stepp, M.A., Simon, K.J., Weinreb, P.H., Violette, S.M.,
Zieske, J.D., 2005. Effect of wound type on Smad 2 and 4 translocation. Invest.
Ophthalmol. Vis. Sci. 46, 2362e2368.
Jani, P.D., Singh, N., Jenkins, C., Raghava, S., Mo, Y., Amin, S., Kompella, U.B.,
Ambati, B.K., 2007. Nanoparticles sustain expression of Flt intraceptors in the
cornea and inhibit injury-induced corneal angiogenesis. Invest. Ophthalmol.
Vis. Sci. 48, 2030e2036.
Jester, J.V., Petroll, W.M., Cavanagh, H.D., 1999. Corneal stromal wound healing in
refractive surgery: the role of myofibroblasts. Prog. Retin. Eye Res. 18,
311e356.
Josko, J., Mazurek, M., 2004. Transcription factors having impact on vascular
endothelial growth factor (VEGF) gene expression in angiogenesis. Med. Sci.
Monit. 10, RA89eRA98.
Joyce, N.C., Harris, D.L., McAlister, J.C., Ali, R.R., Larkin, D.F.P., 2004. Overexpression
of the transcription factor E2F2 induces cell cycle progression in rabbit corneal
endothelial cells. Invest. Ophthalmol. Vis. Sci. 45, 1340e1348.
Jurkunas, U.V., Bitar, M., Rawe, I., 2009. Colocalization of increased transforming
growth factor-beta-induced protein (TGFBIp) and Clusterin in Fuchs endothelial
corneal dystrophy. Invest. Ophthalmol. Vis. Sci. 50, 1129e1136.
Kanwar, J.R., Mohan, R.R., Kanwar, R.K., Roy, K., Bawa, R., 2010. Applications of
aptamers in nanodelivery systems in cancer, eye and inflammatory diseases.
Nanomedicine (Lond.) 5, 1435e1445.
Karin, M., 1999. How NF-kappaB is activated: the role of the IkappaB kinase (IKK)
complex. Oncogene 18, 6867e6874.
Kaye, S., Choudhary, A., 2006. Herpes simplex keratitis. Prog. Retin. Eye Res. 25,
355e380.
Kim, B., Lee, S., Suvas, S., Rouse, B.T., 2005. Application of plasmid DNA encoding IL18 diminishes development of herpetic stromal keratitis by antiangiogenic
effects. J. Immunol. 175, 509e516.
Kim, T.H., Jiang, H.L., Jere, D., Park, I.K., Cho, M.H., Nah, J.W., Choi, U.J., Akaike, T.,
Cho, C.S., 2007. Chemical modification of chitosan as a gene carrier in vitro and
in vivo. Prog. Polym. Sci. 32, 726e753.
Klausner, E.A., Peer, D., Chapman, R.L., Multack, R.F., Andurkar, S.V., 2007. Corneal
gene therapy. J. Control Release 124, 107e133.
Klausner, E.A., Zhang, Z., Chapman, R.L., Multack, R.F., Volin, M.V., 2010. Ultrapure chitosan oligomers as carriers for corneal gene transfer. Biomaterials 31, 1814e1820.
Klibanov, A.L., 2006. Microbubble contrast agents: targeted ultrasound imaging and
ultrasound-assisted drug-delivery applications. Invest. Radiol. 41, 354e362.
Klintworth, G.K., 2009. Corneal dystrophies. Orphanet. J. Rare Dis. 4, 7.
Klotz, L., Hucke, S., Thimm, D., Classen, S., Gaarz, A., Schultze, J., Edenhofer, F.,
Kurts, C., Klockgether, T., Limmer, A., Knolle, P., Burgdorf, S., 2009. Increased
antigen cross-presentation but impaired cross-priming after activation of
peroxisome proliferator-activated receptor gamma is mediated by upregulation of B7H1. J. Immunol. 183, 129e136.
Kulkarni, A.A., Thatcher, T.H., Olsen, K.C., Maggirwar, S.B., Phipps, R.P., Sime, P.J.,
2011. PPAR-g ligands repress TGFb-induced myofibroblast differentiation by
targeting the PI3K/Akt pathway: implications for therapy of fibrosis. PLoS One
6, e15909.
Kuo, C.N., Yang, L.C., Yang, C.T., Chen, M.F., Lai, C.H., Chen, Y.H., Chen, C.H., Chen, C.H.,
Wu, P.C., Kou, H.K., Tsai, J.C., Hung, C.H., 2008. A novel vector system for gene
transfer into the cornea using a partially dried plasmid expressing 18 basic
fibroblast growth factor-synthetic amphiphile INTeraction-18 (SAINT-18)
complex. Curr. Eye Res. 33, 839e848.
Kuo, C.N., Yang, L.C., Yang, C.T., Lai, C.H., Chen, M.F., Chen, C.Y., Chen, C.H., Wu, P.C.,
Kou, H.K., Chen, Y.J., Hung, C.H., Tsai, C.B., 2009. Inhibition of corneal neovascularization with plasmid pigment epithelium-derived factor (p-PEDF)
delivered by synthetic amphiphile Interaction-18 (SAINT-18) vector in an
experimental model of rat corneal angiogenesis. Exp. Eye Res. 89, 678e685.
Lai, C.M., Brankov, M., Zaknich, T., Lai, Y.K., Shen, W.Y., Constable, I.J., Kovesdi, I.,
Rakoczy, P.E., 2001. Inhibition of angiogenesis by adenovirus-mediated sFlt-1
expression in a rat model of corneal neovascularization. Hum. Gene Ther. 12,
1299e1310.
Lai, L.J., Xiao, X., Wu, J.H., 2007. Inhibition of corneal neovascularization with
endostatin delivered by adeno-associated viral (AAV) vector in a mouse corneal
injury model. J. Biomed. Sci. 14, 313e322.
Lebherz, C., Maguire, A., Tang, W., Bennett, J., Wilson, J.M., 2008. Novel AAV serotypes for improved ocular gene transfer. J. Gene Med. 10, 375e382.
Lech, P., Somaia, N.V., 2008. Retrovirus vectors. In: Benigni, A., Remuzzi, G. (Eds.),
Gene Therapy for Renal Diseases and Transplantation. Karger, Basel, pp. 30e46.
Lewis, J.D., Lichtenstein, G.R., Deren, J.J., Sands, B.E., Hanauer, S.B., Katz, J.A.,
Lashner, B., Present, D.H., Chuai, S., Ellenberg, J.H., Nessel, L., Wu, G.D., Rosiglitazone for Ulcerative Colitis Study Group, 2008. Rosiglitazone for active
ulcerative colitis: a randomized placebo-controlled trial. Gastroenterology 134,
688e695.
Li, D., Li, P., Li, G., Wang, J., Wang, E., 2009a. The effect of nocodazole on the
transfection efficiency of lipid-bilayer coated gold nanoparticles. Biomaterials
30, 1382e1388.
Li, H.L., Zheng, X.Z., Wang, H.P., Li, F., Wu, Y., Du, L.F., 2009b. Ultrasound-targeted
microbubble destruction enhances AAV-mediated gene transfection in human
RPE cells in vitro and rat retina in vivo. Gene Ther. 16, 1146e1153.
Li, M., Jayandharan, G.R., Li, B., Ling, C., Ma, W., Srivastava, A., Zhong, L., 2010. Highefficiency transduction of fibroblasts and mesenchymal stem cells by tyrosinemutant AAV2 vectors for their potential use in cellular therapy. Hum. Gene
Ther. 21, 1527e1543.
Limberis, M.P., Wilson, J.M., 2006. Adeno-associated virus serotype 9 vectors
transduce murine alveolar and nasal epithelia and can be readministered. Proc.
Natl. Acad. Sci. U.S.A. 103, 12993e12998.
Lindner, J.R., 2004. Microbubbles in medical imaging: current applications and
future directions. Nat Rev Drug Discov 3, 527e532.
Liu, J., Saghizadeh, M., Tuli, S.S., Kramerov, A.A., Lewin, A.S., Bloom, D.C.,
Hauswirth, W.W., Castro, M.G., Schultz, G.S., Ljubimov, A.V., 2008a. Different
tropism of adenoviruses and adeno-associated viruses to corneal cells: implications for corneal gene therapy. Mol. Vis. 14, 2087e2096.
Liu, J., Lewin, A.S., Tuli, S.S., Ghivizzani, S.C., Schultz, G.S., Bloom, D.C., 2008b.
Reduction in severity of a herpes simplex virus type 1 murine infection by
treatment with a ribozyme targeting the UL20 gene RNA. J. Virol. 82, 7467e7474.
Lipkowitz, M.S., Hanss, B., Tulchin, N., Wilson, P.D., Langer, J.C., Ross, M.D.,
Kurtzman, G.J., Klotman, P.E., Klotman, M.E., 1999. Transduction of renal cells
in vitro and in vivo by adeno-associated virus gene therapy vectors. J. Am. Soc.
Nephrol. 10, 1908e1915.
Liu, Y., Yang, H., Sakanishi, A., 2006. Ultrasound: mechanical gene transfer into plant
cells by sonoporation. Biotechnol. Adv. 24, 1e16.
Lungwitz, U., Breunig, M., Blunk, T., Gopferich, A., 2005. Polyethylenimine based
non-viral gene delivery systems. Eur. J. Pharm. Biopharm. 60, 247e266.
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
McAlister, J.C., Joyce, N.C., Harris, D.L., Ali, R.R., Larkin, D.F., 2005. Induction of
replication in human corneal endothelial cells by E2F2 transcription factor
cDNA transfer. Invest. Ophthalmol. Vis. Sci. 46, 3597e3603.
Maguire, A.M., Simonelli, F., Pierce, E.A., Pugh Jr., E.N., Mingozzi, F., Bennicelli, J.,
Banfi, S., Marshall, K.A., Testa, F., Surace, E.M., Rossi, S., Lyubarsky, A.,
Arruda, V.R., Konkle, B., Stone, E., Sun, J., Jacobs, J., Dell’Osso, L., Hertle, R.,
Ma, J.X., Redmond, T.M., Zhu, X., Hauck, B., Zelenaia, O., Shindler, K.S.,
Maguire, M.G., Wright, J.F., Volpe, N.J., McDonnell, J.W., Auricchio, A., High, K.A.,
Bennett, J., 2008. Safety and efficacy of gene transfer for Leber’s congenital
amaurosis. N. Engl. J. Med. 358, 2240e2248.
Maguire, A.M., High, K.A., Auricchio, A., Wright, J.F., Pierce, E.A., Testa, F., Mingozzi, F.,
Bennicelli, J.L., Ying, G.S., Rossi, S., Fulton, A., Marshall, K.A., Banfi, S., Chung, D.C.,
Morgan, J.I., Hauck, B., Zelenaia, O., Zhu, X., Raffini, L., Coppieters, F., De Baere, E.,
Shindler, K.S., Volpe, N.J., Surace, E.M., Acerra, C., Lyubarsky, A., Redmond, T.M.,
Stone, E., Sun, J., McDonnell, J.W., Leroy, B.P., Simonelli, F., Bennett, J., 2009. Agedependent effects of RPE65 gene therapy for Leber’s congenital amaurosis:
a phase 1 dose-escalation trial. Lancet 374, 1597e1605.
Marano, R.J., Toth, I., Wimmer, N., Brankov, M., Rakoczy, P.E., 2005. Dendrimer
delivery of an anti-VEGF oligonucleotide into the eye: a long-term study into
inhibition of laser-induced CNV, distribution, uptake and toxicity. Gene Ther. 12,
1544e1550.
Maurice, D.M., 1984. Cornea and sclera. In: Davson, D. (Ed.), The Eye. Academic
Press, London, pp. 1e158.
Mintzer, M.A., Simanek, E.E., 2009. Nonviral vectors for gene delivery. Chem. Rev.
109, 259e302.
Mitragotri, S., 2005. Healing sound: the use of ultrasound in drug delivery and other
therapeutic applications. Nat. Rev. Drug Discov. 4, 255e260.
Mo, Y., Barnett, M.E., Takemoto, D., Davidson, H., Kompella, U.B., 2007. Human
serum albumin nanoparticles for efficient delivery of Cu, Zn superoxide dismutase gene. Mol. Vis. 13, 746e757.
Mohan, R.R., Hutcheon, A.E., Choi, R., Hong, J., Lee, J., Mohan, R.R., Ambrósio Jr., R.,
Zieske, J.D., Wilson, S.E., 2003. Apoptosis, necrosis, proliferation, and myofibroblast generation in the stroma following LASIK and PRK. Exp. Eye Res. 76,
71e87.
Mohan, R.R., Sharma, A., Netto, M.V., Sinha, S., Wilson, S.E., 2005. Gene therapy in
the cornea. Prog. Retin. Eye Res. 24, 537e559.
Mohan, R.R., Gupta, R., Mehan, M.K., Cowden, J.W., Sinha, S., 2010a. Decorin
transfection suppresses profibrogenic genes and myofibroblast formation in
human corneal fibroblasts. Exp. Eye Res. 91, 238e245.
Mohan, R.R., Sharma, A., Cebulko, T.C., Tandon, A., 2010b. Vector delivery technique
affects gene transfer in the cornea in vivo. Mol. Vis. 16, 2494e2501.
Mohan, R.R., Tovey, J.C., Gupta, R., Sharma, A., Tandon, A., 2011a. Decorin
biology, expression, function and therapy in the cornea. Curr. Mol. Med. 11,
110e128.
Mohan, R.R., Tovey, J.C., Sharma, A., Schultz, G., Cowden, J., Tandon, A., 2011b.
Targeted decorin gene therapy delivered with adeno-associated virus
effectively retards corneal neovascularization in vivo. PLoS One, in press.
Mohan, R.R., Tandon, A., Sharma, A., Cowden, J.W., Tovey, J.C., 2011c. Significant
inhibition of corneal scarring in vivo with tissue-selective, targeted AAV5
decorin gene therapy. Invest Ophthalmol Vis Sci. 52, 4833e4841.
Montini, E., Cesana, D., Schmidt, M., Sanvito, F., Ponzoni, M., Bartholomae, C., Sergi
Sergi, L., Benedicenti, F., Ambrosi, A., Di Serio, C., Doglioni, C., von Kalle, C.,
Naldini, L., 2006. Hematopoietic stem cell gene transfer in a tumor-prone
mouse model uncovers low genotoxicity of lentiviral vector integration. Nat.
Biotechnol. 24, 687e696.
Montini, E., Cesana, D., Schmidt, M., Sanvito, F., Bartholomae, C.C., Ranzani, M.,
Benedicenti, F., Sergi, L.S., Ambrosi, A., Ponzoni, M., Doglioni, C., Di Serio, C., von
Kalle, C., Naldini, L., 2009. The genotoxic potential of retroviral vectors is
strongly modulated by vector design and integration site selection in a mouse
model of HSC gene therapy. J. Clin. Invest. 119, 964e975.
Mulhbacher, J., St-Pierre, P., Lafontaine, D.A., 2010. Therapeutic applications of
ribozymes and riboswitches. Curr. Opin. Pharmacol. 10, 551e556.
Munn, D.H., Shafizadeh, E., Attwood, J.T., Bondarev, I., Pashine, A., Mellor, A.L., 1999.
Inhibition of T cell proliferation by macrophage tryptophan catabolism. J. Exp.
Med. 189, 1363e1372.
Murthy, R.C., McFarland, T.J., Yoken, J., Chen, S., Barone, C., Burke, D., Zhang, Y.,
Appukuttan, B., Stout, J.T., 2003. Corneal transduction to inhibit angiogenesis
and graft failure. Invest. Ophthalmol. Vis. Sci. 44, 1837e1842.
Mwaikambo, B.R., Sennlaub, F., Ong, H., Chemtob, S., Hardy, P., 2006. Activation of
CD36 inhibits and induces regression of inflammatory corneal neovascularization. Invest. Ophthalmol. Vis. Sci. 47, 4356e4364.
Netto, M.V., Mohan, R.R., Medeiros, F.W., Dupps Jr., W.J., Sinha, S., Krueger, R.R.,
Stapleton, W.M., Rayborn, M., Suto, C., Wilson, S.E., 2007. Femtosecond laser and
microkeratome corneal flaps: comparison of stromal wound healing and
inflammation. J. Refract. Surg. 23, 667e676.
O’Reilly, M.S., Boehm, T., Shing, Y., Fukai, N., Vasios, G., Lane, W.S., Flynn, E.,
Birkhead, J.R., Olsen, B.R., Folkman, J., 1997. Endostatin: an endogenous inhibitor
of angiogenesis and tumor growth. Cell 88, 277e285.
Oh, J.Y., Roddy, G.W., Choi, H., Lee, R.H., Ylöstalo, J.H., Rosa Jr., R.H., Prockop, D.J.,
2010. Anti-inflammatory protein TSG-6 reduces inflammatory damage to the
cornea following chemical and mechanical injury. Proc. Natl. Acad. Sci. U.S.A.
107, 16875e16880.
Pacak, C.A., Mah, C.S., Thattaliyath, B.D., Conlon, T.J., Lewis, M.A., Cloutier, D.E.,
Zolotukhin, I., Tarantal, A.F., Byrne, B.J., 2006. Recombinant adeno-associated
63
virus serotype 9 leads to preferential cardiac transduction in vivo. Circ. Res.
99, e3e9.
Palmer, D.J., Ng, P., 2005. Helper-dependent adenoviral vectors for gene therapy.
Hum. Gene Ther. 16, 1e16.
Parker, D.G., Kaufmann, C., Brereton, H.M., Anson, D.S., Francis-Staite, L., Jessup, C.F.,
Marshall, K., Tan, C., Koldej, R., Coster, D.J., Williams, K.A., 2007. Lentivirusmediated gene transfer to the rat, ovine and human cornea. Gene Ther. 14,
760e767.
Parker, D.G., Brereton, H.M., Klebe, S., Coster, D.J., Williams, K.A., 2009. A steroidinducible promoter for the cornea. Br. J. Ophthalmol. 93, 1255e1259.
Pellegrini, G., Rama, P., Mavilio, F., De Luca, M., 2009. Epithelial stem cells in corneal
regeneration and epidermal gene therapy. J. Pathol. 217, 217e228.
Petrs-Silva, H., Dinculescu, A., Li, Q., Min, S.H., Chiodo, V., Pang, J.J., Zhong, L.,
Zolotukhin, S., Srivastava, A., Lewin, A.S., Hauswirth, W.W., 2009. High-efficiency transduction of the mouse retina by tyrosine-mutant AAV serotype
vectors. Mol. Ther. 17, 463e471.
Petry, H., Brooks, A., Orme, A., Wang, P., Liu, P., Xie, J., Kretschmer, P., Qian, H.S.,
Hermiston, T.W., Harkins, R.N., 2008. Effect of viral dose on neutralizing antibody response and transgene expression after AAV1 vector re-administration in
mice. Gene Ther. 15, 54e60.
Pillai, R.G., Beutelspacher, S.C., Larkin, D.F., George, A.J., 2008. Expression of the
chemokine antagonist vMIP II using a non-viral vector can prolong corneal
allograft survival. Transplantation 85, 1640e1647.
Pissuwan, D., Niidome, T., Cortie, M.B., 2011. The forthcoming applications of gold
nanoparticles in drug and gene delivery systems. J. Control. Release 149, 65e71.
Ponder, K.P., Auricchio, A., 2010. Gene therapy for ocular problems in mucopolysaccharidosis: an experimental and promising approach with benefits in animal
models e a review. Clin. Exp. Ophthalmol. 38, 43e51.
Qaddoumi, M.G., Ueda, H., Yang, J., Davda, J., Labhasetwar, V., Lee, V.H., 2004. The
characteristics and mechanisms of uptake of PLGA nanoparticles in rabbit
conjunctival epithelial cell layers. Pharm. Res. 21, 641e648.
Ragusa, A., García, I., Penadés, S., 2007. Nanoparticles as nonviral gene delivery
vectors. IEEE Trans. Nanobiosci. 6, 319e330.
Rama, P., Matuska, S., Paganoni, G., Spinelli, A., De Luca, M., Pellegrini, G., 2010.
Limbal stem-cell therapy and long-term corneal regeneration. N. Engl. J. Med.
363, 147e155.
Redfern, R.L., McDermott, A.M., 2010. Toll-like receptors in ocular surface disease.
Exp. Eye Res. 90, 679e687.
Richter, M., Iwata, A., Nyhuis, J., Nitta, Y., Miller, A.D., Halbert, C.L., Allen, M.D., 2000.
Adeno-associated virus vector transduction of vascular smooth muscle cells
in vivo. Physiol. Genom. 2, 117e127.
Ritter, T., Yang, J., Dannowski, H., Vogt, K., Volk, H.D., Pleyer, U., 2007. Effects of
interleukin-12p40 gene transfer on rat corneal allograft survival. Transpl.
Immunol. 18, 101e107.
Runager, K., Enghild, J.J., Klintworth, G.K., 2008. Focus on molecules: transforming
growth factor beta induced protein (TGFBIp). Exp. Eye Res. 87, 298e299.
Saghizadeh, M., Kramerov, A.A., Tajbakhsh, J., Aoki, A.M., Wang, C., Chai, N.N.,
Ljubimova, J.Y., Sasaki, T., Sosne, G., Carlson, M.R., Nelson, S.F., Ljubimov, A.V.,
2005. Proteinase and growth factor alterations revealed by gene microarray
analysis of human diabetic corneas. Invest. Ophthalmol. Vis. Sci. 46, 3604e3615.
Saghizadeh, M., Kramerov, A.A., Yu, F.S., Castro, M.G., Ljubimov, A.V., 2010.
Normalization of wound healing and diabetic markers in organ cultured human
diabetic corneas by adenoviral delivery of c-Met gene. Invest. Ophthalmol. Vis.
Sci. 51, 1970e1980.
Saika, S., Ikeda, K., Yamanaka, O., Miyamoto, T., Ohnishi, Y., Sato, M., Muragaki, Y.,
Ooshima, A., Nakajima, Y., Kao, W.W., Flanders, K.C., Roberts, A.B., 2005a.
Expression of Smad7 in mouse eyes accelerates healing of corneal tissue after
exposure to alkali. Am. J. Pathol. 166, 1405e1418.
Saika, S., Ikeda, K., Yamanaka, O., Flanders, K.C., Nakajima, Y., Miyamoto, T.,
Ohnishi, Y., Kao, W.W., Muragaki, Y., Ooshima, A., 2005b. Therapeutic effects of
adenoviral gene transfer of bone morphogenic protein-7 on a corneal alkali
injury model in mice. Lab. Invest. 85, 474e486.
Saika, S., Yamanaka, O., Okada, Y., Miyamoto, T., Kitano, A., Flanders, K.C., Ohnishi, Y.,
Nakajima, Y., Kao, W.W., Ikeda, K., 2007. Effect of overexpression of PPARgamma
on the healing process of corneal alkali burn in mice. Am. J. Physiol. Cell.
Physiol. 293, C75eC86.
Sharma, A., Ghosh, A., Siddappa, C., Mohan, R.R., 2010a. Ocular surface: gene
therapy. In: Besharse, J., Dana, R., Dartt, D.A. (Eds.), Encyclopedia of the Eye.
Elsevier, pp. 185e194.
Sharma, A., Ghosh, A., Hansen, E.T., Newman, J.M., Mohan, R.R., 2010b. Transduction
efficiency of AAV 2/6, 2/8 and 2/9 vectors for delivering genes in human corneal
fibroblasts. Brain Res. Bull. 81, 273e278.
Sharma, A., Tovey, J.C., Ghosh, A., Mohan, R.R., 2010c. AAV serotype influences gene
transfer in corneal stroma in vivo. Exp. Eye Res. 91, 440e448.
Sharma, A., Tandon, A., Tovey, J.C., Gupta, R., Robertson, J.D., Fortune, J.A.,
Klibanov, A.M., Cowden, J.W., Rieger, F.G., Mohan, R.R., 2011. Polyethylenimineconjugated gold nanoparticles: gene transfer potential and low toxicity in the
cornea. Nanomedicine PMID: 21272669.
Shiraishi, A., Converse, R.L., Liu, C.Y., Zhou, F., Kao, C.W., Kao, W.W., 1998. Identification of the cornea-specific keratin 12 promoter by in vivo particle-mediated
gene transfer. Invest. Ophthalmol. Vis. Sci. 39, 2554e2561.
Shoham, A., Hadziahmetovic, M., Dunaief, J.L., Mydlarski, M.B., Schipper, H.M., 2008.
Oxidative stress in diseases of the human cornea. Free Radic. Biol. Med. 45,
1047e1055.
64
R.R. Mohan et al. / Progress in Retinal and Eye Research 31 (2012) 43e64
Simonelli, F., Maguire, A.M., Testa, F., Pierce, E.A., Mingozzi, F., Bennicelli, J.L.,
Rossi, S., Marshall, K., Banfi, S., Surace, E.M., Sun, J., Redmond, T.M., Zhu, X.,
Shindler, K.S., Ying, G.S., Ziviello, C., Acerra, C., Wright, J.F., McDonnell, J.W.,
High, K.A., Bennett, J., Auricchio, A., 2010. Gene therapy for Leber’s congenital
amaurosis is safe and effective through 1.5 years after vector administration.
Mol. Ther. 18, 643e650.
Singh, N., Amin, S., Richter, E., Rashid, S., Scoglietti, V., Jani, P.D., Wang, J., Kaur, R.,
Ambati, J., Dong, Z., Ambati, B.K., 2005. Flt-1 intraceptors inhibit hypoxiainduced VEGF expression in vitro and corneal neovascularization in vivo.
Invest. Ophthalmol. Vis. Sci. 46, 1647e1652.
Sonoda, S., Tachibana, K., Uchino, E., Okubo, A., Yamamoto, M., Sakoda, K.,
Hisatomi, T., Sonoda, K.H., Negishi, Y., Izumi, Y., Takao, S., Sakamoto, T., 2006.
Gene transfer to corneal epithelium and keratocytes mediated by ultrasound
with microbubbles. Invest. Ophthalmol. Vis. Sci. 47, 558e564.
Stramer, B.M., Zieske, J.D., Jung, J.C., Austin, J.S., Fini, M.E., 2003. Molecular mechanisms controlling the fibrotic repair phenotype in cornea: implications for
surgical outcomes. Invest. Ophthalmol. Vis. Sci. 44, 4237e4246.
Surace, E.M., Auricchio, A., 2008. Versatility of AAV vectors for retinal gene transfer.
Vision Res. 48, 353e359.
Tandon, A., Tovey, J.C., Sharma, A., Gupta, R., Mohan, R.R., 2010. Role of transforming
growth factor Beta in corneal function, biology and pathology. Curr. Mol. Med.
10, 565e578.
The Journal of Gene Medicine Clinical Trial Website. update June 2010 Available at:
http://www.abedia.com/wiley/index.html (accessed 23.05.11).
Thomas, M., Klibanov, A.M., 2003. Conjugation to gold nanoparticles enhances
polyethylenimine’s transfer of plasmid DNA into mammalian cells. Proc. Natl.
Acad. Sci. U.S.A. 100, 9138e9143.
Thomas, M., Ge, Q., Lu, J.J., Chen, J., Klibanov, A.M., 2005. Cross-linked small polyethylenimines: while still nontoxic, deliver DNA efficiently to mammalian cells
in vitro and in vivo. Pharm. Res. 22, 373e380.
Tiyaboonchai, W., Woiszwillo, J., Middaugh, C.R., 2003. Formulation and characterization of DNAepolyethylenimineedextran sulfate nanoparticles. Eur. J.
Pharm. Sci. 19, 191e202.
Tong, Y.C., Chang, S.F., Liu, C.Y., Kao, W.W., Huang, C.H., Liaw, J., 2007. Eye drop
delivery of nano-polymeric micelle formulated genes with cornea-specific
promoters. J. Gene Med. 9, 956e966.
Toropainen, E., Hornof, M., Kaarniranta, K., Johansson, P., Urtti, A., 2007. Corneal
epithelium as a platform for secretion of transgene products after transfection
with liposomal gene eyedrops. J. Gene Med. 9, 208e216.
Traas, A.M., Wang, P., Ma, X., Tittiger, M., Schaller, L., O’donnell, P., Sleeper, M.M.,
Vite, C., Herati, R., Aguirre, G.D., Haskins, M., Ponder, K.P., 2007. Correction of
clinical manifestations of canine mucopolysaccharidosis I with neonatal retroviral vector gene therapy. Mol. Ther. 15, 1423e1431.
Van Vliet, K.M., Blouin, V., Brument, N., Agbandje-McKenna, M., Snyder, R.O., 2008.
The role of the adeno-associated virus capsid in gene transfer. Methods Mol.
Biol. 437, 51e91.
Wang, B., O’Malley, T.M., Xu, L., Vite, C., Wang, P., O’Donnell, P.A., Ellinwood, N.M.,
Haskins, M.E., Ponder, K.P., 2006. Expression in blood cells may contribute to
biochemical and pathological improvements after neonatal intravenous gene
therapy for mucopolysaccharidosis VII in dogs. Mol. Genet. Metab. 87, 8e21.
Wang, Z., Zhu, T., Qiao, C., Zhou, L., Wang, B., Zhang, J., Chen, C., Li, J., Xiao, X., 2005.
Adeno-associated virus serotype 8 efficiently delivers genes to muscle and
heart. Nat. Biotechnol. 23, 321e328.
Watanabe, K., Hasegawa, Y., Yamashita, H., Shimizu, K., Ding, Y., Abe, M., Ohta, H.,
Imagawa, K., Hojo, K., Maki, H., Sonoda, H., Sato, Y., 2004. Vasohibin as an
endothelium-derived negative feedback regulator of angiogenesis. J. Clin.
Invest. 114, 898e907.
Weiss, J.S., Kruth, H.S., Kuivaniemi, H., Tromp, G., White, P.S., Winters, R.S., Lisch, W.,
Henn, W., Denninger, E., Krause, M., Wasson, P., Ebenezer, N., Mahurkar, S.,
Nickerson, M.L., 2007. Mutations in the UBIAD1 gene on chromosome short arm
1, region 36, cause Schnyder crystalline corneal dystrophy. Invest. Ophthalmol.
Vis. Sci. 48, 5007e5012.
Weiss, J.S., Fifth ARVO/Pfizer Ophthalmics Research Institute Conference Working
Group, 2010. Corneal dystrophies: molecular genetics to therapeutic interventiondFifth ARVO/Pfizer Ophthalmics Research Institute Conference. Invest.
Ophthalmol. Vis. Sci. 51, 5391e5402.
Whitcher, J.P., Srinivasan, M., Upadhyay, M.P., 2001. Corneal blindness: a global
perspective. Bull. World Health Org. 79, 214e221.
Williams, K.A., Coster, D.J., 2010. Gene therapy for diseases of the cornea e a review.
Clin. Exp. Ophthalmol. 38, 93e103.
Wilson, S.E., Mohan, R.R., Hong, J., Lee, J., Choi, R., Liu, J.J., Mohan, R.R., 2002.
Apoptosis in the cornea in response to epithelial injury: significance to wound
healing and dry eye. Adv. Exp. Med. Biol. 506, 821e826.
World Health Organization Report, fact sheet 282, April 2011. Visual Impairment
and Blindness. URL: http://www.who.int/mediacentre/factsheets/fs282/en/
index.html (accessed 23.09.11).
Wu, Z., Miller, E., Agbandje-McKenna, M., Samulski, R.J., 2006. Alpha2,3 and alpha2,
6 N-linked sialic acids facilitate binding and transduction by adenoassociated
virus types 1 and 6. J. Virol. 80, 9093e9103.
Xu, Q., Wang, C.H., Pack, D.W., 2010. Polymeric carriers for gene delivery: chitosan
and poly(amidoamine) dendrimers. Curr. Pharm. Des. 16, 2350e2368.
Yamashita, T., Sonoda, S., Suzuki, R., Arimura, N., Tachibana, K., Maruyama, K.,
Sakamoto, T., 2007. A novel bubble liposome and ultrasound-mediated gene
transfer to ocular surface: RC-1 cells in vitro and conjunctiva in vivo. Exp. Eye
Res. 85, 741e748.
Yoon, K.C., Bae, J.A., Park, H.J., Im, S.K., Oh, H.J., Lin, X.H., Kim, M.Y., Lee, J.H., Lee, S.E.,
Ahn, K.Y., Kim, K.K., 2009. Subconjunctival gene delivery of the transcription
factor GA-binding protein delays corneal neovascularization in a mouse model.
Gene Ther. 16, 973e981.
Young, L.S., Searle, P.F., Onion, D., Mautner, V., 2006. Viral gene therapy strategies:
from basic science to clinical application. J. Pathol. 208, 299e318.
Yu, H., Wu, J., Li, H., Wang, Z., Chen, X., Tian, Y., Yi, M., Ji, X., Ma, J., Huang, Q., 2007.
Inhibition of corneal neovascularization by recombinant adenovirus-mediated
sFlk-1 expression. Biochem. Biophys. Res. Commun. 361, 946e952.
Yuan, J., Chen, J.Q., Zhou, S.Y., Liu, Z.G., Wang, Z.C., Gu, J.J., 2006. The transfection and
expression of IL-1ra gene to the rabbit cornea in situ via cation polymer
mediation. Zhonghua Yan Ke Za Zhi 42, 686e693.
Zagon, I.S., Sassani, J.W., Malefyt, K.J., McLaughlin, P.J., 2006. Regulation of corneal
repair by particle-mediated gene transfer of opioid growth factor receptor
complementary DNA. Arch. Ophthalmol. 124, 1620e1624.
Zarbin, M.A., Montemagno, C., Leary, J.F., Ritch, R., 2010a. Nanomedicine in
ophthalmology: the new frontier. Am. J. Ophthalmol. 150, 144e162.
Zarbin, M.A., Montemagno, C., Leary, J.F., Ritch, R., 2010b. Nanotechnology in
ophthalmology. Can. J. Ophthalmol. 45, 457e476.
Zhang, B., Yao, Y.F., 2010. Gelatinous drop-like corneal dystrophy with a novel
mutation of TACSTD2 manifested in combination with spheroidal degeneration
in a Chinese patient. Mol. Vis. 16, 1570e1575.
Zhong, L., Li, B., Jayandharan, G., Mah, C.S., Govindasamy, L., Agbandje-McKenna, M.,
Herzog, R.W., Weigel-Van Aken, K.A., Hobbs, J.A., Zolotukhin, S., Muzyczka, N.,
Srivastava, A., 2008. Tyrosine-phosphorylation of AAV2 vectors and its consequences
on viral intracellular trafficking and transgene expression. Virology 381, 194e202.
Zhou, R., Dean, D.A., 2007. Gene transfer of interleukin 10 to the murine cornea
using electroporation. Exp. Biol. Med. (Maywood) 232, 362e369.
Zhou, S.Y., Xie, Z.L., Xiao, O., Yang, X.R., Heng, B.C., Sato, Y., 2010. Inhibition of mouse
alkali burn induced-corneal neovascularization by recombinant adenovirus
encoding human vasohibin-1. Mol. Vis. 16, 1389e1398.
Zhou, X., Zhang, X., Yu, X., Zha, X., Fu, Q., Liu, B., Wang, X., Chen, Y., Chen, Y., Shan, Y.,
Jin, Y., Wu, Y., Liu, J., Kong, W., Shen, J., 2008. The effect of conjugation to gold
nanoparticles on the ability of low molecular weight chitosan to transfer DNA
vaccine. Biomaterials 29, 111e117.