. .r. «v: .fi .V.c.;nam..1w . fit. 17...: duta‘ :9 3:- 11:13. I ‘ W mafia 26061 This is to certify that the thesis entitled ANTIBODY RESPONSE INDUCED AFTER INTRAOCULAR VIRAL GENE ADDITION THERAPY USING ADENO—ASSOCIATED VIRUS. LENTIVIRUS, AND ADENOVIRUS VECTORS WITH THE GFP TRANSGENE IN DOGS presented by Jennifer Vander Kooi Den Houter has been accepted towards fulfillment of the requirements for the MS. degree in Comparative Medicine and Integrative Biology 7W3 - _ Major Professor’s Signature “9/670 2? Date MSU is an Affirmative Action/Equal Opportunity Employer .--n-n-I-o-l-I-I-l-l-D‘I—I-t-O-I—-...— ._ .- on-n-o-.-o--u-u-e-a-a-n-.--o-u--g--.-.— — PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATEDUE DAIEDUE DAIEDUE 0911 15 5/08 K:/PrqlAcc&Pres/ClRC/DateDue indd ANTIBODY RESPONSE INDUCED AFTER INTRAOOULAR VIRAL GENE ADDITION THERAPY USING ADENO-ASSOCIATED VIRUS, LENTIVIRUS, AND ADENOVIRUS VECTORS WITH THE GFP TRANSGENE IN DOGS By Jennifer Vander Kooi Den Houter A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE Comparative Medicine and Integrative Biology 2008 ABSTRACT ANTIBODY RESPONSE INDUCED AFTER INTRAOCULAR VIRAL GENE ADDITION THERAPY USING ADENO-ASSOCIATED VIRUS, LENTIVIRUS, AND ADENOVIRUS VECTORS WITH THE GFP TRANSGENE IN DOGS By Jennifer Vander Kooi Den Houter A humoral immune response induced after ocular gene addition therapy using viral vectors, to either the transgene or the viral vector, can impact therapy safety and efficacy. Three vectors with the green fluorescent protein transgene (GFP): adenO-associated virus (AAV), lentivirus, and adenovirus (Av-1), were studied in order to monitor the Circulatory antibody response after intraocular treatment in dogs. Aqueous samples were taken both pre- and post-treatment from AAV treated dogs to monitor the local humoral response. In order to monitor the antibody response, two dog-specific enzyme-linked immunosorbent assays (ELISAS) were designed: a GFP ELISA and an AAV ELISA. In dogs treated with AAV-GFP, a high level of Circulating anti-GFP antibodies were detected in 4 out of 22 dogs. The anti-GFP antibody response was observed to be influenced by using the chicken-beta actin promoter (CBA) and treatment age of 3 weeks. Anti-GF P antibodies were also found in aqueous samples up to 96 weeks post- treatment, and a significant anti-AAV antibody response was also detected. Anti- GFP antibodies were also found for lentivirus-GFP and Av-1-GFP treated dogs. These results Show that despite the immune—privileged nature of the eye, an antibody response to both the transgene protein and the viral vector occurs and should remain a concern in future intraocular gene therapy studies. Dedicated to my father, Leonard H. Den Houter ACKNOWLEDGEMENTS I would like to thank my committee: Dr. Simon Petersen-Jones, Dr. Vilma Yuzbasiyan-Gurkan, Dr. Jeanne Burton, Dr. Art Weber, Dr. Patrick Venta, and Dr. Matti Kiupel for your continued support, empathy, and flexibility. Friends from the Comparative Ophthalmology Laboratory, Dr. Nalinee Tuntivanich, Michelle Curcio, Gillian Shaw, Dr. Simon Petersen-Jones, and Janice Forcier, thank you for being such a blessing over these past two years. Thank you to the members of the Immunogenetics Laboratory for your patience and support. Dr. Jeanne Burton, Dr. Patricia Weber, and Dr. Patricia Almeida, thank you for helping me through the ELISA development and statistical analysis processes. My family and friends, thank you for your support as well as a listening ear through this experience. Mom, thank you for your continued persistence. Jon, Jessica, and Ben, thank you for being the greatest siblings anyone could ever have. Last but not least, thank you to my father, who always believed in me and Challenged me until the end. TABLE OF CONTENTS LIST OF TABLES ........................................................................................ viii LIST OF FIGURES ...................................................................................... ix LIST OF ABBREVIATIONS ........................................................................ xii CHAPTER 1. INTRODUCTION 1.1. Somatic Cell Gene Addition Therapy for Inherited Retinal Degenerations ............................................................................ 1 1.2. Literature Review ........................................................................ 3 1.2.1. Overview Of Canine Ocular Anatomy .................................. 3 1.2.2. The Retina ........................................................................... 5 1.2.3. Phototransduction and the Visual Cycle .............................. 9 1.2.4 The Central Visual Pathway ................................................ 13 1.2.5. Inherited Retinal Dystrophies .............................................. 14 1.2.5.a. Small Animal Models of Retinal Degenerations ............. 16 1.2.5.b. Large Animal Models of Retinal Degenerations ............. 18 1.2.6. Gene Therapy Viral Vectors ................................................ 19 1.2.6.a. Recombinant Adeno-Associated Virus (AAV) ............... 19 1.2.6.b. Recombinant Lentivirus ................................................ 22 1.2.6.C. Recombinant Adenoviruses (Av-1) ................................ 25 1.2.7. Summary of Successful Retinal Gene Therapy Treatments. 28 1.2.7.a. AAV Preliminary Studies in the Eye ............................. 28 1.2.7.b. Successful AAV Gene Addition Therapy for Retinal Degenerations .............................................................. 29 1.2.7.c. Lentivirus Preliminary Studies in the Eye ..................... 29 1.2.7.d. Successful Lentivirus Gene Addition Therapy for Retinal Degenerations ................................................. 30 1.2.7.e. Adenovirus Preliminary Studies in the Eye .................. 31 1.2.7.f. Successful Adenovirus Gene Addition Therapy for Retinal Degenerations ................................................. 32 1.2.8. Safety Concerns that Arise from Viral Gene Therapy .......... 33 1.2.8.a. Host Immune Response .............................................. 34 1.2.8.b. Adeno-Associated Vectors- Safety and Host Immune Response ..................................................................... 38 1.2.8.c. Safety of Lentivirus Vectors .......................................... 41 1.2.8.d. Adenovirus Vectors in the Eye- Safety and Host Immune Response ....................................................... 42 1.2.8.e. Host Immune Response to Expressed Transgene in the Eye .......................................................................... 44 1.2.8.f. Biodistribution of the Viral Vector within the Host After Intraocular Treatment .......................................... 44 1.2.8.9. Genotoxicity ................................................................. 46 1.3. Objectives, Hypotheses, and Experimental Design ...................... 48 1.3.1. Objectives ............................................................................ 48 1.3.2. Hypotheses .......................................................................... 48 1.3.3. Experimental Design ............................................................ 49 CHAPTER 2. DEVELOPMENT OF ELISA ASSAYS 2.1. Introduction .................................................................................... 52 2.2. Materials and Methods ................................................................... 52 2.2.1. Immunization of Dogs ........................................................... 52 2.2.2. Development Of GFP ELISA .................................................. 54 2.2.2.a. Reagents ....................................................................... 55 2.2.2.b. Detailed GFP ELISA Protocol ....................................... 56 2.2.2.c. Results of GFP ELISA Development ............................. 58 2.2.3. Development of AAV ELISA .................................................. 59 2.2.3.a. Reagents ....................................................................... 60 2.2.3.b. Detailed AAV ELISA Protocol ....................................... 60 2.2.3.c. Results of AAV ELISA Development ............................. 61 CHAPTER 3. HUMORAL IMMUNE RESPONSES DETECTED IN DOGS GIVEN INTRAOCULAR TREATMENT OF VIRAL VECTOR 3.1. Introduction .................................................................................... 67 3.1.1. Intraocular Injections .............................................................. 67 3.1.2. Anesthesia ............................................................................ 68 3.1.3. rAAV2/5, Lentivirus, and Av-1 ............................................... 68 3.1.4. Injection Technique Used in Puppies Where the Eyelids Have Opened ........................................................................ 69 3.1.5. Injection Technique Used in Neonates .................................. 71 3.1.6. Dogs Used in Study and Sample Collection .......................... 72 3.1.7. Monitoring GFP Expression .................................................. 75 3.1.8. Data Analysis using SAS ....................................................... 76 3.2. Results ......................................................................................... 79 3.2.1. Presence of Circulating Anti-GFP Antibodies after Intraocular Treatment with AAV-GFP .................................... 79 3.2.2. Presence of Anti-GFP Antibodies in Aqueous after Intraocular Treatment with AAV-GFP ..................................... 81 3.2.3 Correlation between Anti-GFP Antibody Responses in Serum and Aqueous Samples .............................................. 90 3.2.4. Presence of Circulating Anti-AAV Antibodies after Intraocular Treatment with AAV-GFP .................................... 90 3.2.5. Correlation between Anti-AAV and Anti-GFP Antibodies in the Serum .......................................................................... 91 3.2.6. Presence of Circulating Anti-GFP Antibodies after Intraocular Treatment with Lentivirus .................................... 91 3.2.7. Presence of Circulating Anti—GFP Antibodies after Intraocular Treatment with Av-1 ............................................ 92 vi 3.3. Discussion and Future Work .......................................................... 100 3.3.1. Discussion and Summary of Results ..................................... 100 3.3.1 .a. Anti-GFP Antibody Response after Treatment with AAV-GFP ....................................................................... 1 00 3.3.1.b. Anti-AAV Antibody Response after Treatment with AAV-GFP ...................................................................... 1 00 3.3.1.0. Discussion of Results from AAV Treated Dogs ............ 101 3.3.1.d. Correlation Analyses .................................................... 103 3.3.1.e. Anti-GFP Antibody Response after Treatment with Lentivirus ...................................................................... 1 03 3.3.1.f. Anti-GFP Antibody Response after Treatment with Av-1 ............................................................................. 1 04 3.3.1.9. AAV-GFP, Lentivirus-GFP, and Av-1 Antibody Responses Compared ................................................. 104 3.3.2. Future Research .................................................................... 105 3.3.3. Conclusion ............................................................................ 107 APPENDIX Appendix A. ELISA GFP Laboratory Protocol .......................................... 109 Appendix B. Anti-AAV ELISA Protocol and Summary of References ........ 113 Appendix C. AAV2/5-GFP Vector Construction ......................................... 116 Bibliography ................................................................................................ 124 vii LIST OF TABLES Table 2.1. lmmunized dogs ........................................................................ 54 Table 3.1. Colony dogs injected with AAV2/5-CBA-GFP or AAV2/5-mOPS-GFP vectors for the GFP ELISA protocol ........ 73 Table 3.2. Colony dogs injected with AAV2/5 vectors that were used for the Anti-AAV ELISA protocol ............................................... 74 Table 3.3. Least Squares Means of GFP ELISA OD Values from Dog Serum Samples (1 :5) for Independent Variables ...................... 86 Table 3.4. Least Squares Means of GFP ELISA OD Values from Dog Aqueous Samples (1 :20) for Independent Variables ................. 89 Table 3.5. Least Squares Means of AAV ELISA OD Values for Dog Serum Samples (1:5) for Independent Variables ...................... 96 viii Figure 1.1. Figure 1.2. Figure 1.3. Figure 1.4. Figure 1.5. Figure 1.6. Figure 1.7. Figure 1.8. Figure 1.9. Figure 1.10. Figure 1.11. Figure 1.12. Figure 1.13. Figure 1.14. Figure 1.15. Figure 1.16. Figure 2.1. Figure 2.2. LIST OF FIGURES Images in this thesis/dissertation are presented in color. Image of the vertebrate eye with an expanded view of the retina ....................................................................................... 4 Schematic of the retinal layers ................................................ 6 Diagram of a rod photoreceptor from the vertebrate retina ..... 7 Phototransduction within a rod photoreceptor cycle ................ 10 Diagram of the visual cycle ..................................................... 12 An illustration of the central visual pathway ............................ 14 Fundus pictures from two different human patients ................ 15 A representation of the adeno-associated virus genome ........ 20 A diagram of the infection pathway of adeno-associated virus ....................................................................................... 22 Diagram of the HIV provirus .................................................... 23 A representation of an HIV particle ......................................... 24 A representation of the adenovirus genome ........................... 26 Immunohistochemistry of the dog retina transduced by an Av-1 vector after subretinal injection ...................................... 33 An overview of the host immune response to a foreign invader ................................................................................... 37 The typical host immune response to recombinant adenovirus vectors ................................................................. 43 Flow Chart of experimental design for the AAV-treated dogs. 50 GFP ELISA Protocol Flow Chart ............................................. 56 GFP ELISA Development: Anti-GFP Rabbit Serum Used to Determine the Optimal GF P Concentration Range ................ 63 Figure 2.3. Figure 2.4. Figure 2.5. Figure 3.1. Figure 3.2. Figure 3.3. Figure 3.4. Figure 3.5. Figure 3.6. Figure 3.7. Figure 3.8. Figure 3.9. Figure 3.10. Figure 3.11. Figure 3.12. GFP ELISA Development: Anti-GFP Dog Serum Used to Optimize the Dog-Specific GFP ELISA .................................. 64 GFP ELISA Development: Variation in Control Dog Serum Dilutions Using 1:500 GFP in Order to Determine the Positive and Negative Control Sera for the GFP ELISA ......... 65 AAV2/5 ELISA Development: Variations in Dog Sera and AAV to Determine the Optimal Anti-AAV Assay .............................. 66 Path taken by the injector for the subretinal injection procedure ................................................................................ 70 Subretinal injector (RetinaJect Injector, Surrnodics, Irvine, CA) used to perform the subretinal injections ................................. 70 Before and after subretinal injection in a dog’s eye ................. 71 Injection site in neonatal puppies ............................................ 72 Serum and Aqueous Sampling Time Line ............................... 75 GFP expression was monitored using a RetCam ................... 76 Least Squares Mean ODS for Anti-GFP Antibody Levels in Serum for dilutions 1:5, 1:10, and 1:20 compared after AAV-GFP Intraocular Injection Collected on Weeks 0, 2, 4, 6, 8, and >8 .............................................................................. 82 Individual Dog ODS for Anti-GFP Antibody Levels in Semm Pre- and Post-Treatment (1:5 dilution shown) ......................... 83 Least Squares Mean (+/- SEM) ODS for Anti-GFP Antibody Levels in Serum after AAV-GFP Intraocular Injection Collected Pre and Post Treatment (1 :5 serum shown) ........... 84 Least Squares Mean (+/- SEM) ODS for Anti-GFP Antibody Levels in Serum after AAV-GFP Intraocular Injection Collected on Weeks 0, 2, 4, 6, 8, and >8 (1 :5 serum Shown) . 85 Least Squares Mean (+/-SEM) ODS for Anti-GFP Antibody Levels in Aqueous Collected Pre- and Post-Injection (1 :20 dilution of aqueous) ....................................................... 87 Individual Dog Aqueous GFP ELISA OD Values at Sacrifice .. 88 Figure 3.13. Correlation of Anti-GFP ELISA OD Values in Aqueous and Serum Samples ...................................................................... 93 Figure 3.14. Least Squares Mean (+/-SEM) ODS for Anti-AAV Antibody Levels in Serum Collected Pre- and Post-Injection after Intraocular Treatment with AAV-GFP (1 :5 dilution of serum shown) ........................................................................ 94 Figure 3.15. Least Squares Mean (+/-SEM) ODS for Anti-AAV Antibody Levels in Serum Collected 0, 2, and 4 Weeks Post- Treatment with AAV-GFP (1 :5 dilution of serum shown) ........ 95 Figure 3.16. Correlation of Anti-AAV and Anti-GFP ELISA OD Values from Serum Samples ............................................................. 97 Figure 3.17. Least Squares Mean (+/-SEM) ODS for Anti-GFP Antibody Levels in Serum Collected Pre and Post Treatment with Lentivirus-GFP (1:5 dilution Of serum shown) ........................ 98 Figure 3.18. Least Squares Mean (+/-SEM) ODS for Anti-GFP Antibody Levels in Serum Collected on Weeks 0, 2, 4, 6, and 8 Post Treatment with Lentivirus-GFP (1 :5 dilution Of serum shown) ........................................................................ 99 Figure 3.19. AAV-GFP, Av-1, and Lentivirus-GFP ODS for Anti-GFP Antibody Levels in Serum after Intraocular Treatment (1 :5 serum shown for AAV and Lentivirus-GFP, 1:20 Shown for Av-1 ) ...................................................................................... 108 Figure A. The CBA promoter sequence ..................................................... 117 Figure B. The mOPSSOO promoter sequence ............................................ 117 Figure C. The GFP CDNA sequence used in the AAV viral vector Constructs .................................................................................. 1 18 Figure D. The pTR-UF 11 plasmid with CMV enhancer, CBA promoter, and GFP transgene ................................................................... 119 xi LIST OF ABBREVIATIONS AAV = adeno-associated virus AAV2= adeno-associated virus serotype 2 AAV5 = adeno-associated virus serotype 5 AAV-CBA-GFP = adeno-assoaciated2/5 vector with CBA and GFP AAV-MOPS-GFP = adeno-assoaciated2/5 vector with mOPS and GFP APC = antigen presenting cells Av—1 = adenovirus CAR = coxsackie-adenovirus receptor CBA = Chicken beta-actin promoter cGMP-PDE = cyclic GMP phosphodiesterase holoenzyme CMV = cytomegalovirus dsDNA = double-stranded DNA EAM = encapsidated adenovirus mini-chromosomes ELISA = enzyme-linked immunosorbent assay ERG = electroretinography GC = guanylate cyclase GFP = green fluorescent protein HIV = human immunodeficiency virus IU = infectious unit LacZ = B-galactosidase LCA = leber congenital amaurosis xii LGN = lateral geniculate nucleus LSM = least squares mean LTR = long terminal repeats mOPS = mouse opsin promoter NK cells = natural killer cells OD = optical density OTC = omithine transcarboxylase PBS = phosphate-buffered saline PDE= phosphodiesterase qPCR = quantitative PCR rAAV2/5 = recombinant AAV serotype 2/5 rAAV = recombinant AAV RAC = NIH Recombinant DNA Advisory Committee Rh= rhodopsin rHIV = recombinant HIV RK= rhodopsin kinase RP = retinitis pigmentosa RPE = retinal pigment epithelium SAS = Statistical Analysis Software SClD-X1 = severe combined immunodeficiency disease SEM= standard error of the mean T= transducing VSV-G = vesicular stomatitis virus xiii Chapter 1 INTRODUCTION 1.1. Somatic Cell Gene Addition Therapy for Inherited Retinal Degenerations Gene therapy has been at the forefront of research since the 1980’s and continues to draw considerable attention as a novel therapy. Currently, treatments are progressing for diseases that are difficult or even impossible to treat with contemporary medicine, such as hemophilia B (Chao and Walsh, 2004), severe combined immune deficiency (Cavazzana-Calvo et al., 2000; Cavazzana-Calvo et al., 2005), and many forms of cancer. Gene therapy has been considered only for fatal, Chronic, or severely debilitating diseases in the past. Recently though, diseases of the special sense organs, such as the ear and eye, have been considered for gene therapy. The eye is a particularly interesting organ for gene therapy research since it is a relatively isolated part of the body and also an immune-privileged Site and thus gene therapies would theoretically allow for long-term treatment success with leSS risk of adverse immune responses. Examples of gene therapy in the eye include somatic cell gene addition therapies for autosomal recessive diseases in which the disease results from a lack of gene product (Auricchio and Rolling, 2005); antisense RNA and ribozyme treatments for autosomal dominant diseases in which the disease results from accumulation of the mutated gene product (Lewin et al., 1998); and generic treatments such as introduction of neurotrophic factors to protect and sustain the photoreceptors as long as possible (Bessant et al., 2001 ). Unfortunately, each approach has its own challenges. For instance, when using gene addition therapy in an attempt to produce the missing protein for an individual with a null mutation, the vector may be expressing a protein for which the body has not had previous exposure, thus increasing the potential for a host immune response against the therapeutic gene’s protein (Tripathy et al., 1996). Since gene therapy is a relatively new field, much research needs to be done to determine the most efficacious and safest methods for treatment of a variety of inherited, debilitating eye diseases. Currently, little research has been performed to investigate in detail the immune response, either within the eye or systemically, after intraocular injection with a vector that expresses a foreign transgene. With the potential for gene therapy currently under scrutiny, the need to learn more about the immune response after intraocular gene therapy is necessary to determine the safety and efficacy of this form of treatment for future research and Clinical trials. 1.2. Literature Review 1.2.1. Overview of Canine Ocular Anatomy The anatomy of the eye is quite complex with each region or tissue having its own specific function. Sequentially, from the cornea to the optic nerve, a brief description will be given. Overall, there are three important layers of the eye as shown in Figure 1.1: 1) the fibrous, outermost layer consists of the cornea and SClera, deemed the comeoscleral layer with the junction between cornea and sclera termed the limbus, 2) the middle layer consists of the uveal tract which is composed of the iris, the Ciliary body, and the choroid, and 3) the innermost retinal layer which consists of the retinal pigment epithelium (RPE) and neurosensory retina which are continuous with the Ciliary epithelium anteriorly. The cornea is the roughly circular, transparent area at the front of the eye that allows light to enter the globe. The majority of light refraction occurs at the corneal surface. Light then passes through the anterior chamber, which is filled with aqueous, and then continues through the lens where it is further refracted. The lens is a transparent, crystalin structure that is held in place by the lens zonules and its shape is modified by the action of the Ciliary body musculature. Just in front of the lens is the iris which acts aS a diaphragm to control the amount of light that enters the lens. Along the periphery Of the iris lies the iridocomeal drainage apparatus that allows fluid drainage from the anterior chamber to the blood stream and thus controls the pressure in the globe. Sclera _ __ Choroid Optic nerve Ciliary body Figure 1.1. Image of the vertebrate eye with an expanded view of the retina. (http://www.webvision.med.utah.edu, 2-24-07). Light continues through the lens and through the vitreous to be focused onto the retina. The retina, which lines the back of the eye, is composed of several layers, and acts to convert light into an electric signal which is modified within the inner retina and then travels to the brain initially via the optic nerve and allows the organism to visualize its environment. 1.2.2. The Retina The retina lines the back of the eye globe as shown in Figure 1.2. It ends anteriorly at the ora Ciliaris retinae which is positioned just posterior of the Ciliary body. The outer layer of the retina is known as the retinal pigment epithelium (RPE) and is a monolayer of cells that is in intimate contact with the photoreceptors of the neurosensory retina and plays a vital role in their nutrition and maintenance. The RPE lies adjacent to BrUCh’s membrane posteriorly. Tight junctions in the RPE and the retinal vessel endothelium create the blood- retinal barrier which limits access of molecules to the neurosensory retina. The sub-retinal space between the RPE and the photoreceptor outer segments, a potential space into which gene therapy injections are given, has been Shown specifically to be immune-privileged due to the action of the blood-retinal barrier (Hoffman et al., 1997; Wenkel and Streilein, 1998; Anand et al., 2002; Isenmann et al., 2001). With this in mind, gene therapy of the eye may be influenced by this deviant immune response due to this immune-plivileged environment. The three main neuronal cells of the retina are the photoreceptors, the bipolar cells, and the ganglion cells. Photoreceptors are slender cells responsible for the conversion of light into an electrical signal. There are two main types of photoreceptors: cones and rods. Approximately 95% of photoreceptors are rods in the canine eye which is comparable to the human eye in which approximately 95% are rods and 5% are cones (Koch and Rubin, 1972; Forrester et al., 1996). In contrast, the distribution of rod and cones across the retina varies between the human and canine eye. In the human eye, the macula, which is superotemporal to the optic disc, contains a clustering of cone photoreceptors while in the canine there is a region of high cone density in the area centralis which forms a streak above the optic disc. Rod photoreceptors as Shown in Figure 1.3 are responsible for vision in dim light while cone photoreceptors provide for color, bright light vision. The number of cone types vary between species in mammals. Two or three types of cones are typically present: long and short wavelength or blue, green, and red (short, medium, and long wavelength), respectively. Choroid Retinal Pigment Epithelium (RPE) Photoreceptor Layer External Limiting Membrane Outer Nuclear Layer(ONL) 194" :0. i "' ' {a "ij; Inner Nuclear Layer(lNL) paws} . _,' :31"? ’1 . , C ’ i _ 1 £13“ r L ‘ Ganglion Cell Layer(GCL) __,,.Optic Fiber Layer Vitreous Humor \ Inner Limiting Membrane(lLM) Light Figure 1.2. Schematic of the retinal layers. (http://webvision.med.utah.edu/20030lb.pdf#How°/o20the°/o20Retina%20Works, 2-8-07) Discs Outer sfigment \ - CytoplaSm Plasma membrane ”iii” I [iiiilflljmig Cilium Mitochondria Inner segment 05 :1 Golgi Ciomplex cell body Cell nUCIeuS Synaptic . terminal Slv’naptlc \ Vesicles Figure 1.3. Diagram of a rod photoreceptor from the vertebrate retina. The rod photoreceptor consists of four main regions: the outer segment complete with discs where phototransduction occurs, the inner segment which is connected to the outer segment via the Cilium and creates the energy for the cell, the cell body which holds the nucleus of the cell, and finally the synaptic terminal which contains synaptic vesicles that transmit the message to the next neuron MEN/www.csulb.edu/~cwallis/482/visualsvstem/cwrod.ipg , 7-20-06). The photoreceptor layer is made up of the inner and outer segments of the photoreceptors, the outer nuclear layer is made up of the nuclei of the photoreceptors, and the outer plexiform layer is made up of the photoreceptor termini. The outer segment of the photoreceptor, made up of stacks of discs for the rods and a comb-like structure for the cones, is connected to the inner segment via a Cilium. Within the inner segments of the photoreceptor, metabolic processes occur to keep the cell alive (Baldridge and Kurennvi., 1998). The outer and inner segments are made distinct by the thin outer-limiting membrane which creates a layer across the retina at this level (Buntmilam et al., 1985). The termini of the photoreceptors contain synaptic vesicles with the neurotransmitter glutamate and form synapses with both bipolar and horizontal cells of the inner nuclear layer. Bipolar cells are second order neurons that bridge between 20-60 photoreceptors to the ganglion cells. Rod ON-bipolar cells connect to the ganglion cells via amacrine cells but may also contact them via cone OFF-bipolar cells (Soucy et al., 1998; Hack et al., 1999). Horizontal cells, which are responsible for horizontal Signaling across the retina, also reside in the inner nuclear layer and send dendrites out to synapse with the termini of the photoreceptors. The inner plexiform layer is formed by the connections of the inner nuclear layer cells, such as amacrine cells and bipolar cells, with ganglion cells and others. Amacrine cells help the communication between the inner and outer retina through dopaminergic pathways between the bipolar cells and ganglion cells. The ganglion cell layer contains the cell bodies of the ganglion cells as well as those of some amacrine and astroglial cells. Ganglion cells receive input from many photoreceptors within their receptive field and then send this electrical Signal in the form of action potentials through the unmyelinated nerve fiber layer to the myelinated optic nerve. There are two classes of ganglion cells: 1) ON- center which depolarize and 2) OFF-center which hyperpolarize in response to light stimulation. The innermost layer Of the retina is the inner limiting membrane that is composed of Miller cell end feet and astrocytes. Miiller cells are retinal glial cells that originate from hemopoietic cells. Their cell body is in the inner nuclear layer while their cell processes spread out to form the outer and inner limiting membranes. 1.2.3. Phototransduction and the Visual Cycle Phototransduction (Figure 1.4.) is the process by which the photoreceptors convert photons of light into an electrical Signal which is then transmitted through inner retina to the ganglion cell and via ganglion cell axons to the brain. Rod , Light ROD [ ' Pf Plasma membrane : i _isk membrane If: Na+ 'T‘T‘i cGMP . . .. i . Mg 1-... InthIts ‘ == CNG . Channel Rh*~P inhibits Mg2 . y ' GTP ; 9 i? ll ........ 1 K+ 4, Ca2 Na/Ca-K Exchanger To Na+ pump in inner ‘. segment Figure 1.4. Phototransduction within a rod photoreceptor. htt ://www.biol.tsukuba.ac.' /~nakatani/ hototransduction-scheme-1.' 2-7-07) light to be converted to an electrical message through the Stimulation of the photopigment rhodopsin. Fi= rhodopsin, T= transducin, PDE= phosphodiesterase, Gc= guanylate cyclase, and BK: rhodopsin kinase. phototransduction has been studied extensively, and it initiates with the activation of the photopigment rhodopsin (see Pugh and Lamb, 2005 for a review). Rhodopsin molecules are formed by the combination of 11-ciS retinal with the transmembrane protein rod opsin. Rhodopsin is activated by the absorption-of photons within the outer segment of the photoreceptor. The 11-cis retinal of the rhodopsin molecule photoisomerizes to all-trans retinal and is released from the rhodopsin complex. This all-trans retinal is cycled back into all-trans retinol which is then transported to the RPE where it is converted back to 11-ciS retinal via the visual cycle involving rpe65 isomerase (Figure 1.5) and returned to the outer segments to once again recombine with opsin to form rhodopsin (Redmond et al., 1998; Mata et al., 2004). Following light-activation, activated-rhodopsin then forms a complex with transducin, the rhodopsin-transducin binding complex. Transducin is a trimeric G-protein consisting of: To which is the active subunit, and TB and Ty which are the inhibitory subunits. The light-activated rhodopsin changes the GDP of the transducin complex to a GTP and TBy then dissociate to release To and GTP. The active transducin then stimulates the cyclic GMP phosphodiesterase holoenzyme (cGMP-PDE). cGMP-PDE iS a heterotrimeric complex with two catalytic subunits, PDEa and PDEB that are inhibited by the two PDEv subunits (see Stryer, 1991 for a review). Activated Ta removes the inhibitory gamma subunits from this complex leaving the active PDEq/B complex. PDEa/B hydrolyzes cGMP to GMP resulting in closure of the cGMP-gated Channels due to the decreased concentration of cytosolic cGMP. The Closure of 11 these channels results in the hyperpolarization of the photoreceptor cell membrane. ' . all-trans retinyl ester l 1 all-trans retinol all-trans retinol Figure 1.5. Diagram of the visual cycle. The cycling of the compounds all- trans retinal to 11-cis retinol occurs through a series of steps with various isomerases and through transport via lRBP (interphotoreceptor binding protein) between the RPE and photoreceptor layers. Rh is the molecule rhodopsin which is the photopigment involved in phototransduction (http://www.bumc.bu.edu/www/busm/by/images/carter.jpg, 2-7-07) The closed cGMP-gated Channels consist of two subunits and one glutamate residue which block the cation flow into the cell when cytosolic cGMP is in low concentrations. Consequently, cations such as calcium are not able to flow into the cell and at the same time the Na/Ca-K ion pumps are continuously pumping Ca2+ out of the cell which then results in hyperpolarization of the cell (Haase et al., 1990). This results in a discontinuation of the ‘dark current’ and consequently inhibits glutamate release at the photoreceptor synaptic terminus. 1.2.4. The Central Visual Pathway When the layers of the retina are working in harmony, the retina turns light into an electric signal which is then sent to the brain for perception of the image (Figure 1.6.). This Signal is sent through the ganglion cell axons via the optic nerve. The two optic nerves, from the left and right eyes, meet at the optic chiasm and fibers either continue to the ipsilateral optic tract or decussate to the contralateral optic tract. If the impulse originated from the nasal portion of the retina, then the Signal travels to the opposite side of the brain. Those from the temporal side of the retina project to the ipsilateral Optic tract. Fibers travel to the lateral geniculate nucleus (LGN) for conscious perception of vision, the pretectum, and the superior colliculus for reflex pathways. Ganglion cell axons synapse in the LGN, and then axons pass via the optic radiation to the visual cortex. The visual cortex is divided into regions specific, also known as retinotopic, to the many areas of the visual field (Vanduffel et al., 2002). 13 i Optic nerves l Optic chiasm i Optic tract l ./ synaps {BK RE?) i Lateral geniculate nuclei i Visual cortex l Figure 1.6. An illustration of the central visual pathway. This picture shows the visual field in relationship to the path that the information portraying the image follows along the Optic nerve, through the optic chiasm, and via a synapse in the lateral geniculate nucleus to the visual cortex (Forrester et al., 1996). 1.2.5. Inherited Retinal Dystrophies At the time of writing, 185 different genes have been linked to retinal dystrophies (summarized in RetNet, http://www.sph.uth. tmc.edu/Retnet/sum-dis.htm#A- genes, 2007). Retinal dystrophies are divided into many forms including Leber Congenital Amaurosis and retinitis pigmentosa (RP). RP, which was first described in 1857, is a subset of these blinding hetergenous, inherited diseases that is estimated to affect 1 in 3,000 humans (Bundey and Crews, 1984). This bilateral disease is described as a rod-cone dystrophy since the rods degenerate initially and are often followed by cone degeneration (Zeiss et al., 2004; Huang et al., 1995). The inheritance of the disease can be autosomal dominant, autosomal recessive, or X-linked. RP can present either as a slow or fast onset disease with the most common symptoms being tunnel vision, night blindness, headaches, and unexplained light flashes (Heckenlively et al., 1988). Secondary to these common symptoms are cataract formation, pigment changes in the retinal layers, a decreased ERG rod response initially followed by a decreased cone response, and retinal thinning (Figure 1.7). Figure 1.7. Fundus pictures from two different human patients. The retina on the left is normal while the retina on the right is affected with retinitis pigmentosa. The diseased fundus has bone-spicule pigmentation as well as vascular attenuation. There are four distinct categories into which most of the RP mutations fall: 1) phototransduction cascade proteins, 2) photoreceptor structural proteins, 3) proteins involved in photoreceptor and retinal pigment epithelial metabolism, and 4) proteins that regulate gene expression (Bessant et al., 2001). Leber Congenital Amaurosis (LCA) is a severe early-onset condition that causes blindness in Children (Morimura et al., 1998). Causal mutations have been identified in several genes that are either expressed in the RPE or retinal cells. One example is the RPE65 gene which codes for an isomerase in the RPE and plays an important role in the visual cycle; a mutation in this gene results in a severe vision loss but only slow degeneration of the retina and has shown promise for gene therapy treatments (Cremers, et al., 2002). LCA is Characterized by early onset blindness and typically include a decreased electroretinographic (ERG) rod response initially followed by a decreased cone response. Nystagmus, cataract formation, and impairment of the pupillary light reflex are also features of the condition. 1.2.5.a. Small Animal Models of Retinal Degenerations There are numerous small animal models of retinal degenerations in a variety Of species including Drosophila, zebra fish, and many rodent models. Most of the models are autosomal recessive while a few are inherited in an autosomal dominant or X-linked manner. The most widely used species are the laboratory rodents: rats and mice. Rat models include the Royal College of Surgeons rat (RCS rat), the Rho knockout rat, and the P23H transgenic rat while the mouse models include the spontaneously occurring rd1 mouse, the peripherin 2 (Prph2) mouse (also known as rd2), and a knockout RPE65 mouse. 16 The RCS rat is an inbred line with an autosomal recessive mutation in the Mertk gene which normally serves a role in RPE phagocytosis of the outer segments of photoreceptors. In the RC8 rat, there is a build up of photoreceptor outer segment debris in the subretinal Space and the photoreceptors eventually degenerate as a consequence (for a review see Dejneka et al., 2003). This rat serves as a model for the humans with Mertk gene mutation (Gal et al., 2000; D’Cruz et al., 2000). The P23H rat is a transgenic rat that was created to have an autosomal dominant mutation in the rhodopsin gene due to the alteration of proline-23 to histidine (RhoP23H) in one allele. This mutation leads to eventual blindness, and is one of the most common rhodopsin mutations of humans (for a review see Dejneka et al., 2003; Chader, 2002). The rd1 mouse has an autosomal recessive mutation in the phoshodiesterase beta subunit (PDEB) gene which normally functions as a component of the rod phototransduction cascade. This mutation leads to a fast retinal degeneration and results in blindness. This was the first mouse model of retinal degeneration discovered and has been widely used as a model to mimic phototransduction- related disease in humans (Chader, 2002; Pittler et al., 1991). The knockout RPE65 mouse was developed as a model for LCA. This mouse has been used extensively for gene therapy trials Since it has a similar disease phenotype to the human LCA patient (for a review see Dejneka et al., 2003). 17 1.2.5.b. Large Animal Models of Retinal Degenerations Progressive Retinal Atrophy is the canine equivalent of RP and LCA and Similarly shows genetic heterogeneity. These Spontaneous canine models are useful because the canine eye is Similar in Size to the human eye. Rod cone dysplasia type 1 (rcd1) affecting the Irish Setter breed is a model of early onset autosomal recessive RP and has a nonsense mutation in the PDE6B gene in exon 21 (Suber et al., 1993). Rcd1 has a fast onset with night blindness occurring within 25 days after birth. The rcd1 Irish Setter is used as a model for fast onset, phototransduction-related RP. The Cardigan Welsh Corgi has an autosomal recessive PRA due to a 1 base pair deletion mutation in exon 16 of the phosphodiesterase Six alpha subunit (PDE6A) gene which results in a frameshift and downstream premature stop codon (Petersen-Jones et al., 1999). The disease onset is comparable to the rcd1lrish Setter and has been named ‘rod-cone dysplasia type 3’ (rcd3). This disease was described in detail by Tuntivanich (2006). The Briard, with a 4-bp deletion mutation in the RPE65 gene that leads to a frameshift and downstream premature stop codon, is a canine model of LCA (Veske et al., 1999; Aguirre et al., 1998; Wrigstad, 1994). The absence of the rpe65 protein in the Briard results in a severe loss of vision and markedly reduced ERG responses. However despite the early onset of vision loss, 18 structural deterioration of the photoreceptors is Slow which provides a wide opportunity to test possible gene therapy treatments. 1.2.6. Gene Therapy Viral Vectors Viral vectors are currently used extensively for the transport of a transgene into a target cell. This method has been the basis for many gene addition therapy trials, and has shown success in many studies. Three commonly used viral vectors are adeno-associated virus, lentivirus, and adenovirus vectors. These vectors have demonstrated beneficial treatment effects but also unexpected Side effects. Most viral vectors are Chosen based on the disease that is to be treated. Disease factors such as degree of transgene expression required, duration of treatment, and target cell determine the viral vector that would be optimal. The ultimate goals for a gene therapy vector include, but are not limited to: 1) low immunogenicity, 2) regulated vs. unregulated expression, 3) duration of transgene expression, 4) amount of vector needed for transduction, 5) integration vs. no integration of the viral genome, and 6) tissue specificity (Kay et al., 2001). 1.2.6.a. Recombinant Adena-Associated Virus (AAV) One of the first successful gene therapy trials utilized a recombinant adeno- associated virus vector to treat hemophilia. This disease was treated using a serotype 2 recombinant AAV vector that contained the CDNA sequence for the coagulation factor IX gene. Not only has this vector been shown to be safe as a gene therapy vector in a wide variety Of studies, it has also been shown to be 19 efficacious in gene delivery and long-term transgene expression (Chao and Walsh, 2004; Flannery et al., 2005). Adeno-associated viruses are in the family Parvoviridae and are “helper- dependent’ viruses. Wild type AAV are typically associated with subclinical or latent infections, and typical viral gene expression in the cell occurs during the ‘S’ phase of the cell cycle which can be activated by an adenovirus or herpesvirus infection, for example. Adeno-associated viruses are small particles 26 nm in diameter with an icosahedral capsid made up of viral particles VP1 and VP2. The genome is Single-stranded DNA with flanking terminal repeat regions, the normal genes of the virus being rep and cap, and is approximately 5,000 bases in size which Significantly limits the space available, to approximately 2500bp, for the transgene of interest (Figure 1.8) (Vihinen-Ranta et al., 2004). ITR _ REP CAP — ITR Figure 1.8. A representation of the adeno—associated virus genome. The viral genome consists of the Rep and Cap genes with flanking inverted terminal repeat regions (ITR). Two of the many AAV serotypes are more commonly used for gene therapy trials: serotype 2 (AAV2) and serotype 5 (AAV5). The major difference between the two serotypes is mainly the viruses’ method of cell entry. AAV2 binds the heparin-sulfate proteoglycan on the cell surface while AAV5 binds 02,3 or (12,6 sialic acid on the cell surface for entry through receptor-mediated endocytosis (Vihinen-Ranta et al., 2004). Upon entry into the cell, the endosome undergoes 20 acidification which results in capsid degradation and viral escape into the cytoplasm. Once in the cytoplasm, the virus is shuttled on microtubules to the nuclear pore. When the virus contacts the nuclear pore at the 5—fold axis of the virus capsid, the DNA is made into double-stranded DNA (dsDNA) by a DNA polymerase that lies at this axis. The dsDNA is then transported into the nucleus, through the pore at the 5-fold axis, where it is free to be transcribed into RNA through the host cell transcription process (Figure 1.9). AAV2 has been the primary rAAV for gene therapy studies while AAV5 also has been studied in retinal gene therapy research in recent years. AAV2 was efficacious in retinal gene therapy studies; it had the ability to infect ganglion cells, photoreceptors, and the RPE of dogs (Bainbridge et al., 2003), mice (Grant et al., 1997; Ali et al. 1998), and non-human primates (Bennett et al., 1999). AAV5 has been Shown to be up to 1,000 times more efficient than AAV2 when infecting retinal cells, such as photoreceptors, in the mouse and non-human primate eye, with the average transgene activation time being between 2-4 weeks (Yang et al., 2002; Lotery et al., 2003). A further study pseudotyped AAV2 with the capsid of AAV5 (AAV2/5) and thiS vector was found to be just as efficient at infecting photoreceptors as AAV5 (Auricchio et al., 2001). 21 AAV Host Cell Entry Cytoplasm Nucleus Clathrin- Early endosome coated Endosome vessicle X Perinuclear recycling endosome Figure 1.9. A diagram of the infection pathway of adeno-associated virus (Modified from Vihinen-Ranta et al., 2004). The AAV enters the host cell via receptor-mediated endocytosis. After escape from the endosome, the virus makes a dsDNA version of its SSDNA genome at the 5-fold axis of the capsid. The dsDNA then travels through the nuclear pore into the nucleus where it is then transcribed into RNA. 1.2.6.b. Recombinant Lentivirus Lentiviruses, such as the human immunodeficiency virus (HIV) are a kind of retrovirus. Retroviruses, which were some of the first vectors used to transfer cellular DNA, consist of a single stranded RNA genome, reverse transcriptase, a viral envelope, and a complex genome (Figure 1.10.). 22 LTR Figure 1.10. Diagram of the HIV provirus. The provirus is the dsDNA genome that has been altered from a dsRNA genome via reverse transcriptase within the host cell and is ready to be inserted into the host cell genome (modified from Naldini, 1996). All genes of the provirus are shown in this diagram including the accessory viral proteins: Env=vira| envelope, Rev=reverse transcriptase, Pol=viral polymerase, SD=sp|ice donor site, Lp=packaging signal, Gag=structural polyprotein, Vif, Tat, Nef=regu|atory proteins, and Pro and R=accessory proteins. The two essential components needed to make a recombinant HIV (rHIV) vector in cell culture are: 1) a modified genome with packaging signals (W), a primer- binding site, a polyurine tract, the flanking LTRs, and the transgene in cis-acting sequence and 2) a helper virus that can provide all the packaging materials to create the vector (Figure 1.11.) (Lever, 1999). Wild-type HIV are able to infect T-cells by binding helper T cell surface receptors CD4 and CXCr4. Recombinant viruses have been pseudotyped with the Vesicular Stomatitis Virus Glycoprotein (VSV-G) which is related to the rabies vims and consequently allows the recombinant virus to infect neuronal cells (Cronin et al., 2005). Once the virus has bound to the cellular receptor, the viral envelope fuses with the host cell membrane. The viral core then travels into the cell’s cytoplasm where reverse transcription takes place. The now double- stranded DNA genome enters the nucleus where it integrates with the host cell 23 genome; this dsDNA genome is termed the ‘provirus’. Transcription using the host cell machinery occurs and viral proteins are produced. gp120 9P41 \ lipid layer host cell proteins p17 matrix antioen integrase p24 core antigen HIV-RNA reverse D7 gag transcriptase P9 939 Figure 1.11. A representation of an HIV particle. (http://wwwhivmedicine.com/textbook/images/image65.ipg, 2/8/07). The HIV particle has both a protein capsid as well as a lipid outer membrane. Within the viral capsid, the reverse transcriptase, integrase, and dsRNA genome reside. One desirable trait of rHIV with respect to gene therapy applications is its potential to infect non-dividing cells. Retinal cells do not divide after differentiation, thus rHIV may prove beneficial as a vector for retinal gene therapy. In retinal research, it has been found that the rHIV pseudotyped with VSV-G is able to enter and integrate into maturing photoreceptors within the first 15 days of a mouse’s life (Pang et al., 2006a). rHIV was shown to be able to transfect both photoreceptors and RPE with the most efficient transfection occurring from postnatal day 1 to 3. Use of enzymes has been Shown to 24 increase photoreceptor transduction in the eye, as access of lentivirus vectors to photoreceptors may be limited by a physical barrier within the adult retina (Gruter et al., 2005). 1.2.6.C. Recombinant Adenoviruses (Av-1) Recombinant adenoviruses have been used successfully as vectors for gene therapy ranging from management of cancer to treatment of inherited systemic diseases. Adenovimses are non-enveloped viruses that consist of an icosahedral, Spiked capsid that is 70-90nm in diameter. The viral genome is linear, double-stranded DNA and iS approximately 36KB in size (Figure 1.12) (Connelly, 1999). This virus is in the family Adenoviridae and consists of at least 50 distinct serotypes of which group ‘C’ serotype 5 iS the most commonly used for gene therapy purposes. Adenoviruses infect many different cell types including liver, intestine, and even cells of the retina. Wild-type serotype 5 adenovirus can cause respiratory infections, gastroenteritis, and even conjunctivitis, but typically little inflammation is induced from the virus alone (Connelly, 1999). Three main recombinant adenoviruses are available: Av-1, Av-2. and Av-3. Av-1 is the first generation recombinant adenovirus that was created by deleting the entire region for the E1A gene and 60% of the E1 B gene which was then replaced with the transgene of interest. Av-1 uses the cytomegalovirus (CMV) 25 promoter, a strong viral promoter that has been known to induce a strong inflammatory response (Connelly, 1999). Av-2 vectors have one or more genes deleted in any combination of the following genes: E1, E2, E3, and E4. The transgene insert site is still located in the E1 gene deleted region, but these vectors use either the CMV promoter or can use another promoter of choice (Connelly, 1999). Av-3, also known as ‘Helper—Dependent Ads’, are the most recent adenovirus vectors produced. In Av-3 vectors, the entire viral genome has been deleted and therefore the vector must be grown in the presence of ‘helper’ viruses. The DNA within the virus is the promoter and transgene DNA along with more ‘filler’ DNA to stabilize the structural integrity of the virus (Kumar—Singh et al., 1998; Connelly, 1999). E1A E1B L1 L2 L3 L4 E3 L5 ——> —> > >——> ——> —> —> 4 4— ‘— EZB E2A E4 Figure 1.12. A representation of the adenovirus genome (Modified from http://www-ermm.cbcu.cam.ac.uk/flg005ifo.qif, 2-24-07). The adenovirus genome is a linear dsDNA genome that is transcribed from both 5’ ends since viral genes can be oriented in both directions. ‘E’ genes represent the early transcribed genes whereas the ‘L’ genes represent the later transcribed genes. Adenoviruses enter the host cell by binding a cell surface receptor, of which two are of importance to retinal gene therapy: CAR (Coxsackie-adenovirus receptor), which are present on most cells but not photoreceptors, and CD46, which has 26 been found on photoreceptors and binds to a fiber, F35, on the adenovirus capsid (Mallam et al., 2004; Von Seggem et al., 2003). Integrins are also involved in the entry of the virus and aid the cell in the process of endocytosis. The virus can tolerate the acidic conditions within the vesicle and escapes into the cytoplasm with the viral capsid fully intact. Once in the cytoplasm, the virus is transported along microtubules and directed toward the nuclear pores. The adenovirus attaches to the nuclear pore and the viral genome is transported into the nucleus where it is then transcribed (Connelly, 1999). When using recombinant adenoviruses for gene therapy, it has been found that there is typically a waning of transgene expression approximately 2 weeks post transfection. There are many theories to explain this phenomenon. One theory suggests that antibodies generated by the humoral immune system eliminate the transgene and virus. Another theory suggests that there is CMV-promoter shutdown. This was eloquently demonstrated by Everett in a study which compared the same vector in both normal mice with an intact immune system and nude mice which lack an intact immune system (Everett et al., 2004). Both of the mice, despite the lack of an immune response in the nude mice, Showed a decrease in transgene expression over time, suggesting promoter shutdown rather than immune response was the cause of loss of transgene expression. 27 1.2.7. Summary of Successful Retinal Gene Therapy Treatments 1.2.7.a. AAV Preliminary Studies in the Eye Recombinant AAV vectors have been studied extensively for gene therapy purposes. AAV vectors are easy to create in large quantities, have low immunogenicity, and can transduce many different cell types. Several studies have looked at the ability and efficiency of these vectors to infect retinal cells in various species. Initially mouse models were studied followed later by dog and primate models. AAV vectors have been shown to be effective and safe with little inflammation or toxicity and to produce stable long term transgene expression (Auricchio and Rolling, 2005; Ali et al., 1998; Bainbridge et al., 2003; Bennett et al., 1999). The kinetics of the AAV5 vectors was studied in the mouse eye in order to understand the transduction and gene expression variables. Mice were given injections of between 2x106 and 2x1010 lU/mL into the subretinal space where the vector transduced retinal cells as soon as 3 days after injection. The time lag between transduction of retinal cells and gene expression is believed to be due to the requirement for dsDNA synthesis from the SSDNA viral genome. The retinal cellS that were the main targets of the vector were located at the injection site, but some vector diffused past the borders of the injection site for unknown reasons. It was also determined that increasingly higher titers were able to transduce cells more effectively and were able to yield higher gene expression rates in retinal cells (Sarra et al., 2002). 28 1.2.7.b. Successful AAV Gene Addition Therapy for Retinal Degenerations Attempts to treat mouse models of RP and LCA using AAV vectors have been at least partially successful in several mouse models. Both the RPE65 mutant mouse and the rd1 mouse have shown improvements after AAV gene addition therapy (Pang et al., 2006b; Lai et al., 2004; Jomary et al., 1997). Further work into the treatment of the RPE65 null mutation model of LCA has utilized a large animal model, the Briard dog. Using an AAV vector transporting the normal RPE65 transgene, subretinal injection was performed in an attempt to treat a portion of the retina by gene addition therapy. These treated dogs showed significant improvement in vision with dose—dependent, long-term results for several years (Acland et al., 2001; Narfstrom et al., 2003a; Ford et al., 2003; Narfstrom et al., 2003b; ACland et al., 2005; Jacobson et al., 2006). There have also been other successful retinal treatments using an AAV vector; the Prph2 mutant mouse showed rescue when an AAV vector with a rhodopsin promoter driving expression of peripherin was used (Ali et al., 2000), and the ROS rat with a mutation in the Mertk gene was rescued using an AAV vector to deliver a normal copy of the Mertk gene as well (Smith et al., 2003). 1.2.7.e. Lentivirus Preliminary Studies in the Eye HIV vectors pseudotyped with the VSV glycoprotein were created with the aim of transducing differentiated, non-dividing cells such as retinal cells (Naldini et al., 1996; Bemelmans et al., 2005). Work using these vectors in rodent models 29 Showed some successful results but with limitations. In one study, subretinal injections of the vector in rat eyes showed sustained transgene expression 12 weeks after injection with no Signs of decrease in expression (Miyoshi et al., 1997) 1.2.7.d. Successful Lentivirus Gene Addition Therapy for Retinal Degenerations Continued efforts have shown the efficiency and promise of recombinant lentivirus vectors in delivering the missing gene responsible for fast onset retinal degenerations. The rd1 mouse, the RPE65 knockout mouse, and the ROS rat have been treated using a rHIV vector. Early treatment of the rd1 mouse has Shown promise in slowing photoreceptor degeneration for up to 24 weeks (Takahashi et al., 1999). In the RPE65 knockout mouse, early treatment of the retina led to increased cone photoreceptor survival for up to 4 months while adults treated with the same vector did not Show increased cone photoreceptor survival (Bemelmans et al., 2006). Finally, lentivirus vector treatment of the RC8 rat by subretinal injection at 10 days of age resulted in survival of photoreceptors up to 7 months (TSChemutter et al., 2005;). Unfortunately, for unknown reasons the rescue was only temporary. 30 1.2.7.e. Adenovirus Preliminary Studies in the Eye Adenoviruses were among the first viral vectors that were studied for gene addition therapy in the retina. Today, this vector is still being researched as a potential vector for treatment of many types of ocular disease. As stated earlier, adenoviruses enter the host cell by binding a cell surface receptor, either CAR or CD46, depending on the viral capsid (Von Seggem et al., 2003). Importantly, CAR receptors do not exist in large numbers on the surface of photoreceptors and therefore these cells are not well transduced by the commonly used AdV5 vectors for gene therapy research (Mallam et al., 2004). The first study using adenoviruses for retinal gene therapy were done using Av-1 vectors with the beta-galactosidase (LacZ) transgene in mice. Within 24-48 hours after subretinal injection, transgene expression was evident in the retinal layer (Bennett et al., 1994). Transduction by the recombinant virus and also transgene expression was found to be dose-dependent in that study. Two weeks after the injection, the transgene expression had waned completely. Another study looking to determine the length of transgene expression used the same vector type as the above experiment and found that the adenovirus- delivered transgene could be expressed for almost 16 months in rat retinas, with most of the infected cells being RPE with only a few cells of the neurosensory retina infected (Loewen et al., 2004). 31 1 .2.7.f. Successful Adenovirus Gene Addition Therapy for Retinal Degenerations One of the first successes using Av-1 for treatment of a retinal disease involved treatment of rd1 mice. This study demonstrated photoreceptor survival up to six weeks post injection. The reporter transgene slowly waned for a period of two weeks and then became completely undetectable thereafter (Bennett et al., 1996). This study established the potential usefulness of this vector, so other researchers have attempted to use this vector for other models Of retinal degeneration. (See Figure 1.13. for an image of a dog retina transduced by an Av-2 vector.) Since the RPE is transduced well by Av-1 vectors, several retinal degeneration models were suitable for treatment using this vector. For instance, the ROS rat showed retinal saving after subretinal injection of the vector carrying the Mertk transgene (Vollrath et al., 2001). Treatment of the RPE65 knockout mouse resulted in the presence of functional rpe65 and preservation of photoreceptors (Chen et al., 2006). Av-3 vectors have also been investigated for retinal degeneration treatments. An EAM with a B-pde transgene and B-pde promoter was injected into the subretinal space of the rd1 mouse eye. This vector resulted in improved photoreceptor 32 Figure 1.13. ' L' ‘ ‘ ' ‘ y of the dog retina transduced by an Av-1 vector after subretinal injection. The red fluorescence indicates the green fluorescent protein (GFP) within the RPE layer of the retina and the blue fluorescence indicates the nuclei of individual cells. survival compared with the results using an Av-1 vector (Kumar-Singh and Farber., 1998). 1.2.8. Safety Concerns that Arise from Viral Gene Therapy On September 17th, 1999, Jesse Gelsinger, an 18 year old omithine transcarboxylase (OTC) deficient patient who was a voluntary participant in a gene therapy trial died 4 days after the treatment due to multi-organ-system failure (Stolberg, 1999; Hartogs, 1999; Recombinant DNA Advisory Committee, 2002). Adenovirus vectors were used in the trial in an attempt to treat the OTC deficiency. Gelsinger received the highest dose used in the trial of 3.8 x 1013 viral particles in an effort to determine the dose-dependent nature of the treatment. It was determined on autopsy that the extreme reaction to the vector was to the viral capsid. In response to this unfortunate event, the NIH Recombinant DNA Advisory Committee (RAC) made recommendations for the 33 protocol and monitoring of human Clinical trials in order to lessen the chance of future adverse events (RAC, 2002). Another serious adverse event occurred during Clinical trials using gene therapy to treat X-Iinked severe combined immunodeficiency disease (SCID—X1). In this study, retroviruses were used to infect T cells ex vivo that were then injected into the blood stream. Two of the youngest patients contracted leukemia due to insertional oncogenesis of the retrovirus near the LMO2 promoter, which is a proto-oncogene (Hacein-Bey—Abina et al., 2003; ASGT, 2002). The NIH RAC has produced statements in regards to these concerns and has warned researchers of this rare but serious consequence that is possible when using retroviral vectors for gene therapy (Sadelain, 2004). The safety and efficacy of gene addition therapy is a serious and legitimate concern. Issues encompass several areas including: the concern of viral biodistribution, insertional oncogenesis, host immune responses to both the transgene and viral vector, and the potential for genotoxicity of the transduced cells. 1.2.8.a. Host Immune Response The host immune system is stimulated to eradicate foreign invaders as quickly and as efficiently as possible (Figure 1.14). Initially, the host tries to eradicate the invader through the innate immune response such as through phagocytosis, 34 neutralization, and complement (Janeway et al., 2005). If the innate immune system is unable to inhibit or destroy the Wars, host cells are then infected. This is where the life cycle of the vian begins and the eventual death of the cell ensues. The host’s immune cells, such as antigen presenting cells (APCS); macrophages, neutrophils, and dendritiC cells, and also natural killer cells (NK cells), recognize the infected cells and thus the antigen. The host also mounts a second attack against the antigen known as the adaptive immune response. This response takes 4-7 days for elimination of the antigen after infection, which involves both the cell-mediated and humoral pathways, separately and collaboratively (Janeway et al., 2005). The cell-mediated immune response is through activation of T cells by antigen presentation by APCS within lymph nodes. Cytotoxic T cells, also known as CD8+ T cells, and helper T cells, CD4+ Th1 and CD4+ Th2 cells, are activated by antigens presented by APCS. CD8+ T cells, are the effector cells that destroy infected cells by way of a pathway that induces apoptosis (Janeway et al., 2005). CD4+ Th1 and Th2 cells each have their own unique role. Th1 cells leave the lymph node after activation to stimulate cells, such as macrophages, into action. Th2 cells activate B cells within the lymph node (Janeway et al., 2005). The humoral immune response is activated immediately to any antigen upon entry into the host. The humoral immune response is initiated by Th2 cell activation of B cells and also B cell presentation of antigens to Th2 cells through 35 the MHCll receptor. These immature B cells produce antibodies and they are dispersed into the blood. lgM antibodies are the first antibodies produced by immature B cells within the first few days of an infection (Janeway et al., 2005). Some of the B cells travel to the germinal centers within the lymph nodes where they mature through isotype switching, which allows for a variety of antibodies to be produced such as lgG, thus creating plasma cells. The peak lgG production occurs between 7-10 days post-infection. The antibodies bind to the antigens and allow for neutralization of the antigen, induction of complement, or promotion of phagocytosis (Janeway et al., 2005). Memory T and B cells are formed after an initial exposure to an antigen. Both memory T and B cells are thought to be sustained through constant cytokine stimulation. Upon a repeat exposure to the same antigen, such as a viral vector, B cells undergo clonal expansion and are able to produce high levels of IgG within a few days after infection. T cells also undergo proliferation after encountering the same antigen and create effector cells that travel throughout the host killing infected cells. For the viruses that some of the recombinant vectors are derived from, the details of the host response have not been elucidated, but for others, such as adenoviruses, extensive work has been done in order to understand the immune response (for a review see Schaack, 2005). 36 Virus enters host Host Immediate Immune Responses Pre- -existing neutralizing body Inflammatory response if at *T— 00 Macrophages and neutrophils0 phagocytize virus and then alert host *v , / I / /’ I I Cell- mediated Immune Response ----- * Humoral Immune Response Secondary Host Immune Response Flgure 1 14 An overVIew of the host immune response to a forelgn invader. NK_ natural killer cells, T: T cell and B_ B cell 37 1.2.8.b. Adena-Associated Vectors- Safety and Host Immune Response Wild-type adeno-associated viruses are known for their low immunogenicity during infections. Because of this trait, they have proven useful as gene therapy vectors with the potential for repeated injection without adverse effects (Anand et al. 2002, Anand et al., 2000). Studies to determine the safety of AAV vectors have included the systemic safety, the possibility of gerrnline transmission, the immune response to the viral capsid after intraocular injection, and the effect of the immune response on the repeated administration of the vector. AAV vectors have been found to be safe and efficacious for systemic use (Chao and Walsh, 2004). While this may not be a major concern for ocular therapy, the potential for spread of the vector from the eye to other parts of the body does remain a concern. In order to determine the overall safety of the AAV vector systemically, intramuscular injections of an AAV vector were performed on nonhuman primates (Favre et al., 2001). Within 48-72 hours after the injection, the viral genome was detected in the serum and up to 6 days post-injection the vector was detected in various body fluids. No tissue abnormalities were found at time of necropsy performed 8-18 months post-injection, but the vector was found in the lymph nodes and liver (Favre et al., 2001). Gerrnline transmission has been a concern after an AAV vector was used to treat hemophilia in human patients and the AAV vector DNA was found in the semen of treated males (Manno et al., 2003; Arruda et al., 2001). Schuettrumpf et al. 38 designed a study to determine if the germ cells were infected or if the viral vector was only in the semen transiently. Using rabbits to study the gerrnline transmission after a systemic injection of AAV, vector sequences were found in the semen in a dose-dependent manner and disappeared also in a dose- dependent manner. No infection of the germ cells was evident when monitored over a period of 18 months when analyzed using a quantitative real-time polymerase Chain reaction (qPCR) (Schuettrumpf et al., 2006). One issue of remaining controversy over this vector is whether or not rAAV has the ability to insert itself into the host cell’s genome. Random insertion into the genome could potentially create insertional oncogenesis. In humans, it has been Shown that the wild-type virus has the ability to integrate into the genome at a specific place on chromosome 19 (Kotin et al., 1990). Others contest that the viral DNA, once inside the nucleus, does not integrate into the host genome (Song et al., 2004). AS this continues to be debated, the possibility of integration remains a concern for the safety of gene therapy trials using AAV vectors. Immune responses to the AAV capsid have been previously studied. After intraocular injection of the vector, mild to moderate inflammation has been Observed in Briard dogs, but this resolved over a period of 3 months post- injection (Jacobson et al., 2006). An adaptive immune response has been detected after intraocular injection of the vector. A slight qualitative anti-AAV humoral response was detected in mice and dogs after intravitreal and subretinal 39 injection in both pre- and post-injection (time not specified) serum and aqueous samples (Dudus et al., 1999). Further work into the adaptive immune response was studied in mice; subretinal injections were performed in mice and a deviant immune response was found in the eye. A footpad thickness test performed after intraocular injection (which demonstrates the cell-mediated response) the results of which indicated there was a minimal response against the viral capsid (Anand et al., 2002). Bennett et al. detected a Th2-dependent humoral response to the viral capsid (by ELISA) and found that individual primates had up to a 16-fold increase in IgG and Dudus et al. showed a statistically Significant increase in mice and dogs of lgG antibodies to the viral capsid (Bennett et al, 1999; Dudus et aL,1999) Readministration studies have been done in order to determine if a systemic humoral response would interfere with transgene expression in the eye. In a study by Bennett et al., they found that after initial injection neutralizing antibodies were formed (Bennett et al., 1999). When the same AAV-GFP vector was injected into the contralateral eye 7 months after the initial injection, no inflammatory response was detected and transgene expression was evident, despite the presence of neutralizing antibodies and a 128-fold increase in serum anti-AAV antibody after readministration (Bennett et al., 1999). In a similar study, mouse eyes were given subretinal injections of the vector, at repeat administration transgene expression was still observed even though there was a significant anti-AAV humoral response present in the mice (Anand et al., 2000). 40 1.2.8.c. Safety of Lentivirus Vectors The two main concerns when using Ientiviruses for gene therapy are: 1) the potential for insertional oncogenesis and 2) spontaneous recombination occurring creating a wild-type virus (Lever, 1999). The phenomenon of insertional oncogenesis is pertinent, though rare, because the random insertion of the viral genome next to active genes of the host cell genome can interfere with the normal cellular functioning (Recchia et al., 2006). If near a promoter of a tumor-suppressor gene or oncogene, oncogenesis and consequential disease could result for the patient. New vectors are being designed that will help alleviate this issue by directing viral genome insertion to a consistent and safe location within the genome. Non-integrating HIV vectors are also being designed that will remain in the nucleus to allow transcription to proceed but will not undergo genome insertion (Yanez-Munoz, 2006). The issue of recombination occurring and leading to self-sufficient wild-type virus regeneration can be easily overcome by designing a plasmid system that plans for this potential hazard. Several methods have been created that are specifically designed to inhibit insertional complications (Lever, 1999; Lu et al., 2004). 41 1.2.8.d. Adenovirus Vectors in the Eye- Safety and Host Immune Response Although adenovirus vectors have been shown to be efficient for gene therapy, the recombinant adenovirus vectors have induced inflammatory and other immune responses. It is theorized that inflammation is induced for several reasons: size of the virus, capsid structure, and transgenes such as LacZ and GFP. Three distinct phases in recombinant adenovirus induced inflammation have been described (Figure 1.15). The first phase is a Short-lived response to the capsid, which induces cytokine release. The second phase occurs after internalization of the virus and is thought to be due to the adenovirus genes. The third phase is immune recognition of the adenovims gene products and also possibly the transgene product. Antibodies are then formed which might result in elimination of the transgene and viral vector (Schaak, 2005). While the first and second generation adenoviruses Show a substantial immune response complete with inflammation and a secondary immune response, the Av-3, also known as encapsidated adenovirus mini-Chromosomes (EAMS), has Shown a significantly lower secondary immune response since they lack all viral genes (Muruve, 2004). 42 @ Monocytes Granuloc es \ , . Chemokines/ CytokInes @ NK Cells Leukocyte recruitment Tissue Macropahge/ DC __.————-> Chemokines/ Cy10kines M3531: x}\ Target Tissue 3‘43} Adhesion molecules I l I I I J Endothelium Figure 1.15. The typical host immune response to recombinant adenovirus vectors. NK= natural killer cells, 00: dendritic cells, In the eye after subretinal injection of an adenovirus (Av-2) vector, there seems to be a deviant immune response (Hoffman et al., 1997). Subretinal injection of the vector has shown to induce a systemic humoral response, yet after readministration of an adenovirus vector, transgene expression did not appear to be inhibited by the host immune response (Isenmann et al., 2001). Interestingly, in another study, vaccination of the animals in order to create an immune response and then followed by adenovirus vector intraocular administration every two weeks thereafter showed to have little if any effect on transgene expression within the eye (Hamilton et al., 2006). 43 1.2.8.e. Host Immune Response to Expressed Transgene in the Eye Few studies have focused on the immune response to the transgene after intraocular injection. In a study done by Isenmann et al., there was a difference in immune response based on injection site. Following intravitreal injections, an immune response to the transgene was detectable where as following subretinal injection an immune response was not detected (Isenmann et al., 2001). Two studies have looked Specifically at the humoral immune response to GFP after intraocular injection of a viral vector carrying the GFP transgene. Both of the studies qualitatively analyzed the presence of anti-GF P antibodies via Western Blot. Dudus et al. used pre- and 7 week post-injection serum samples and found that 1 out of 3 mice had an antibody response to GFP (Dudus et al., 1999). Another study showed similar results with a high Circulating anti-GFP antibody level detected in 2 out of 4 mice after subretinal injection of an AAV- GFP vector (Bennett et al., 1999). 1.2.8.f. Biodistribution of the Viral Vector within the Host after Intraocular Treatment AAV delivered by subretinal or intravitreal injection has been shown to spread along the optic nerve to the optic tract in several species: dog, primate, and mouse (LeMeur et al., 2005; Jacobson et al 2006; Dudus et al.1999; Shen et al., 2003; Provost et al., 2005). Both gross investigation using reporter genes as well 44 as quantitative PCR (qPCR) have been used in order to demonstrate the distribution of the vector in the host after intraocular injection. In the mouse, GFP fluorescence has been observed in the layers of the neuroretina and also along the optic nerve after subretinal injection. GFP was observed in the retinal ganglion cells, the optic nerve fibers, and the optic disc. Along the visual pathway, GFP was observed in the optic nerve, lateral geniculate body, optic radiation all the way to the occipital lobe (Shen et al., 2003). Interestingly, the type of injection performed had an effect on the distribution of the vector. Comparing intravitreal and subretinal injections in both dogs and mice, GFP fluorescence was detected along the visual pathway to a greater extent after intravitreal injection than after subretinal injection as might be expected (Dudus et al., 1999). PCR has also been used to determine the relative amount and location of the vector after spread from an intraocular injection in the rat, dog, and nonhuman primate. Two studies have worked to address this issue, but with different results. Provost et al. injected, via intravitreal or subretinal routes, an AAV-GFP vector into the eyes of rats, dogs, and nonhuman primates; PCR for the GFP transgene sequence was performed to detect the sequence within many body tissues including retina, Optic nerve, Optic chiasm, optic tract, lateral geniculate nuclei, optic radiation, visual cortex, superior colliculus, inferior colliculus, temporal lobe, thalamus, cerebellum, spinal cord, cerebrospinal fluid, pituitary 45 gland, lachrymal gland, amygdala, salivary gland, submandibular lymph node, preauricular lymph node, thymus, axillary lymph nodes, gonads, liver, and peripheral blood mononuclear cells (Provost et al., 2005). Sequence was detected occasionally in peripheral blood mononuclear cells. As expected, vector sequence was found in the optic nerve, and along the visUal pathway after intravitreal injection, but interestingly the vector sequence was also found in the optic nerve after subretinal injection (Provost et al., 2005). In another study done by Jacobson, subretinal injection of a vector carrying the RPE65 transgene led to distributionand thus viral sequence detection by qPCR in various organs. Tissues that were analyzed included: optic nerve, optic chiasm, optic tract, Iatertal geniculate nucleus, optic radiation, visual cortex, superior colliculus, mandibular lymph node, parotid lymph node, heart, lung, diaphragm, liver, pancreas, spleen, kidney, jejunum, gonad, and Skeletal muscle. The spread of the vector was found not to be dose-dependent (Jacobson etal., 2006). Vector sequence assayed by amplifying a portion of the CMV enhancer/ CBA promoter was found at a level of 100 copies/pg or higher of DNA in the mandibular lymph nodes, heart, diaphragm, optic nerve, and optic chiasm. 1.2.8.9. Genotoxicity Issues of genotoxocity remain a concern when introducing a vector that has a ubiquitous promoter or when a foreign protein such as GFP, LacZ, or in gene addition therapy if the transgenic protein is introduced into an animal that is 46 deficient for the particular protein. For example, very high doses of the AAV vector carrying the RPE65 transgene has Shown to produce toxic effects in the retina when the transgene is under the control of a ubiquitous promoter (Jacobson et al., 2006). Thus, dosage toxicity studies are necessary before Clinical trials begin to determine the safe dosage range. In most studies, there is little systemic toxicity and retinal damage after intraocular injection of a vector (Jacobson et al., 2006; LeMeur et al., 2005). For example, subretinal injection of AAV-2, -4, and -5 with the GFP transgene in 14 beagles and 9 macaques showed normal retinal angiography images and ERG amplitudes (Le Meur et al., 2005). In another study, AAV-GFP vectors were injected into the eye and high levels of GFP in retinal cells up to 270uM Showed no toxic effect and did not seem to affect normal photoreceptor function based on ERG results (Rex et al. 2005; Shen et al., 2003). One way to reduce the risk of genotoxicity is to use a tissue-specific promoter. For the retina, a mouse opsin promoter has been developed that only expresses the transgene in rods and cones (Glushakova et. al., 2006). Another way to solve this issue is by using inducible promoters such as tetracycline- or erythropoietin-inducible promoters that are activated Upon systemic administration of the appropriate drug. These methods have even been Shown to work in the eye despite the blood-ocular barriers (Auricchio et al., 2002; Blau et aL,1999) 47 1.3. Objectives, Hypotheses, and Experimental Design 1.3.1. Objectives The purpose of this work was to investigate in detail the humoral immune response to GFP after treatment with viral vectors carrying the GFP transgene administered intraocularly in dogs. We focused primarily on AAV constructs since these are the most widely used vector types in retinal gene therapy. The systemic immune response was investigated for both GFP and AAV capsid antibodies while the local immune was investigated for GFP antibodies. The systemic immune response to GFP after intraocular treatment with adenovirus and lentivirus vectors was also performed. 1.3.2. Hypotheses In previous studies, intraocular administration of AAV-GFP in mice and dogs was reported to induce a minor, individual systemic and intraocular humoral immune response to both the viral capsid and GFP. We hypothesize that the same response will be observed and we wish to perform a more detailed analysis of the timing of the humoral immune response in dogs. The systemic AAV antibody response Should be detectable at 2 weeks post-treatment while the GFP antibody response should be detected systemically 4 weeks post-treatment due to slow expression of the transgene that is typical for AAV vectors. The local GFP antibody response Should be detectable at approximately 4 weeks post- treatment for similar reasons. Variables such as the promoter used in the 48 construct and the age of the animal at treatment are expected to influence the immune response. We anticipate that GFP expression resulting after intraocular treatment with lentivirus and Av-1 vectors will also result in a detectable systemic humoral immune response. 1.3.3. Experimental Design Intraocular injections with vectors carrying the GFP transgene were performed: AAV vectors were used in 22 dogs, lentivims was used in 3 dogs, and Av-1 was used in 1 dog. For the AAV-treated pups, one of two AAV constructs were used: AAV-CBA-GF P or AAV-mOPS-GFP and injections performed at either ~3 days of age or ~3 weeks of age (figure 1.16). For lentivirus- and Av-1- treated dogs, subretinal injections were performed. Aqueous samples were collected from AAV-treated dogs pre-injection and then at the time Of euthanasia. Sera samples were collected from each dog pre- and post-treatment. Sera samples were collected from AAV- and lentivirus-treated dogs every two weeks up to 8 weeks for most dogs. The Av-1-treated dog sera samples were collected at 2 and 3 weeks post- treatment. These serum and aqueous samples were then analyzed using canine-specific lgG ELISAS (GFP and AAV) in order to understand the systemic and local humoral responses. 49 Age of Pup at Treatment / \ 3 day treatment 3 week treatment Injection of AAV Injection Of AAV Aqueous/serum Aqueous/serum sample collection pre- sample collection pre- treatment treatment Serum Collection 2, 4, 6, 8, Monitor GFP and >8 weeks post-injection, other further time points Euthanasia Serum collection Aqueous cOllection AAV EL'SA GFP ELISA Figure 1.16. Flow chart of experimental design for the AAV—treated dogs. 50 Sera from AAV-treated dogs were analyzed using the AAV and GFP ELISAS while the aqueous samples were analyzed using the GFP ELISA. Sera from the lentivirus- and Av-1-treated dogs were analyzed using the GFP ELISA. The optical density (OD) values from these ELISAS were then analyzed using SAS. 51 Chapter 2. DEVELOPMENT OF ELISA ASSAYS 2.1. Introduction Two canine-Specific enzyme-linked immunosorbent assays (ELISAS) were developed in order to analyze the humoral immune response after intraocular injection of viral vectors carrying the GF P transgene : rAAV2/5, Av-1, and lentivirus. The first ELISA designed was the GFP ELISA to detect anti-GFP antibodies and the second was the AAV ELISA to detect the anti-AAV capsid antibodies. Previous studies have analyzed the humoral immune response in mice and dogs based on human serum controls which could create bias and distorted results. By creating canine- Specific ELISAS, the humoral immune response can be more accurately assessed. 2.2. Materials and Methods 2.2.1. Immunization of Dogs Colony dogs were immunized with the intent Of using serum samples as controls for the GFP ELISA and AAV ELISA. Each dog was injected using a multi- channel LectraJet injector (D’Antonio Consultants International, East Syracuse, NY) with a vector preparation, described below, that was intended to create an immune response to both the vector capsid as well as the GFP transgene (Carter and Kerr, 2003). 52 The pHR’-CMV-GFP vector was created in the laboratory of Vincente Planelles (University of Utah) and was used for immunizations because it induced a high level of circulating anti-GFP antibodies. The protocol for creation of these vectors has been published (Zhu et al., 2001: Planelles, 2003). The lentivirus vector (pHR-CMV-GFP) was prepared for injection by diluting the virus to ~10"6 infectious units of the vector in sterile saline. The AAV-GFP vector was created in the laboratory of William Hauswirth at the University of Florida. Another member of our research group, Danielle Eifler, prepared the UF 12 type 5 AAV vector to a concentration of ~10"13 vp/mL in sterile saline. A total of 4 dogs were injected with the appropriate virus and 2 negative control dogs were injected with sterile saline as outlined in Table 2.1 below. Each dog was sedated with 0.03-0.05 mg/kg of acepromazine (Boehringer lngelheim Verrndica, Inc., St. Joseph, MO) and 0.2mg/kg of butorphanol (Fort Dodge Animal Health, Fort Dodge, Iowa). The area on the medial thigh was then shaved and scrubbed. Topical lidocaine was applied to area to be injected. Two separate injection Sites on the medial thigh of each leg were used with a total of four Sites per injection day. A vector preparation volume of 0.5 mL was injected per site. After, the injection protocol was finished, each dog received 100mL saline subcutaneously following the injection procedure. Immunizations were first performed for all dogs on 7/21/04 and then repeated on 8/8/04 and 8/18/04. Dogs were re-immunized on 4/20/05. 53 Mica Negative control dog-0.5ml of saline Snap Negative control dog-0.5ml of saline Pluto Lentivirus-GFP-0.5ml of virus solution Spruce Lentivirus-GFP-0.5ml of virus solution Red AAV-GFP- 0.5ml of virus solution Roscoe AAV-GFP- 0.5ml of virus solution Table 2.1. lmmunized dogs. The vector and amount of virus injected per dog is described. Sixty milliliters of blood was collected from each dog prior to immunization and then serum was collected every week for the following Six weeks. After the Six weeks, blood was collected every two weeks. The blood was placed on ice following collection, allowed to Sit at 4°C for twenty-four hours, and then serum was collected and stored at -20°C. After dogs were re-innoculated on 4/20/05, serum was collected 1 and 2 weeks after innoculation. 2.2.2. Development of GFP ELISA During the initial phases of development, GFP and antibodies were used at varying concentrations in order to create optimal assay conditions. GFP was used to coat the plate (BD Falcon, ref 353279, Bedford, MA) using dilutions of 1:100, 1:200, 1:500, 121,000, 1210,000, and 1:30.000 in a bicarbonate solution (see methods) with the initial concentration GFP at 1mg/mL (Clontech, Mountain View, CA), resulting in working concentrations of 0.01 mg/mL, 0.005mg/mL, 0.002mg/mL, 0.001 mg/mL, 0.0001 mg/mL, and 0.000033mg/ml, respectively 54 (Figure 2.2). The optimal GFP concentration range was found by using GFP hyperimmune rabbit serum (HIRS) as a positive control (Abcam, Cambridge, MA), normal rabbit serum as a negative control, and anti-rabbit-Specific HRP- conjugated lgG as the secondary antibody (Caltag. L42007, Carlsbad, CA). Varying dilutions of the normal and HIRS were tested: 1:500, 1:1,000, and 1:2,500, with the secondary antibody at a dilution of 120,000. The immunized dog serum was then used to create a canine-specific assay (Figure 2.3.). The positive control dog sera collected on 8/25 was Chosen because it produced the highest OD values and was then used to finally optimize the assay for dog sera samples. After analyzing serum dilutions of1:5, 1:10, 1:25, and 1:50, the optimal positive control sera with the highest OD being ~1.2 was a 1:1 mixture of a both positive control dog sera at a 1:25 dilution using PBS-T. The same 1:1 mixture of negative control dog sera was used at a 1:25 dilution using PBS-T which gave an OD of ~0.1 (Figure 2.4.). Positive and negative control sera were used for each plate. Upon completion of the assay design, the test dog serum samples were then analyzed using the finalized protocol described next. 2.2.2.a. Reagents (see Appendix A for detailed instructions) 0 Phosphate-buffered solution (PBS) 0 0.05% PBS-Tween wash buffer 0 Bicarbonate solution (Pierce, Rockford, IL) 55 o GFP antigen solution for coating wells at 0.002ug/mL (Clontech, Mountain View, CA) . HRP-anti-Dog lgG (H+L) (Bethyl, Montgomery, TX) was made fresh for each day of use from stock solution 0 1-Step ABTS used as substrate (Pierce, Rockford, IL) 2.2.2.b. Detailed GFP ELISA Protocol The GFP ELISA (Figure 2.1.) was designed in order to determine if a systemic antibody (IgG) response was induced following intraocular injection of viral vectors [AAV2/5, Av-1, lentivirus] with a GFP transgene. (see Appendix A for laboratory protocol). 0 O O 0 Wells washed, dog 0 O O O —'> 8 8 8 8 serum dIlutIons O O O O incubated for 1 hour 96-well plate coated with 100uLof 0.002 mg/ml GFP overnight 0 O O O @4deg.Celsius OOQOvOOOO / Substrate ABTS galls wfiéhe-Ctifi applied, incubated for n 89’ w' . . . . 1hour @ 37C HRP’ Incubated O O O O for1 hr Color change \ O O O 0 Color change occurs for pOSItIve Q O O 0 measured with result spectrophotometer to get OD values Figure 2.1. GFP ELISA Protocol Flow Chart 56 96-well flat- bottom ELISA plates (BD Falcon, Bedford, MA) were coated with 100pL per well of GFP antigen solution, covered with clear adhesive Slips (Nunc, Denmark) and left for 15 hours at 4°C. The wells were then washed and emptied three times with 200pL PBS-T solution Serum dilutions were prepared and applied to the wells at 100pL per well 0 Serum dilutions were prepared using a serial doubling dilution that ranged from 1:5 to 1:320 in PBS-T down the plate column and each dilution was run in duplicate o Aqueous dilutions in PBS-T were done at 1:20 and were run only once due to sample size limitations 0 Positive and negative control sera samples at 1:25 in PBS-T were used for every plate Plate re-covered with plastic Slip and placed on shaker, setting #5 (Wellmix, Therrno LabSystemS), and left at room temperature for one hour The plates was removed from the shaker and the wells were then washed and emptied three times with 200pL PBS-T solution The secondary antibody was added at a dilution of 1:20,000, the plate re- covered with the Slip, and left at room temperature on a Shaker (setting #5) for one hour The plate was removed from the Shaker and the wells were then washed and emptied three times with 200pL PBS-T solution 57 o 125uL of 1-Step ABTS was added to each well, the plate was re-covered with the Slip and was left at 37°C for one hour without shaking o The plate was read at dual wavelengths of 405nm and 450nm using a plate reader (Bio-rad microplate reader, Hercules, CA) to determine optical density (OD) values 2.2.2.c. Results of GFP ELISA Development The GFP ELISA was optimized through a series of steps. First, the optimal GFP concentration was determined using anti-GFP rabbit serum (Figure 2.2). The three best GFP dilutions for rabbit samples were found to be 1:10, 1:100, and 1:1000. Next, GFP dilutions in the range determined above were tested at 1:100, 1:200, 1:500, and 1:1000 and ELISA conditions optimized for dog samples using immunized dog serum at dilutions of1:10, 1:100, 1:500, and 121000 (Figure 2.3). The GFP dilution of 1:500 was Chosen from this trial for the canine GFP ELISA. The next step involved determining the optimal canine control sera dilutions (Figure 2.4). Sera from positive and negative control dogs as well a pool of the control dog sera were tested at dilutions of 1:5,1 :10,1 :25,1 :50, 1:250, 1:500, 1:750, and 1:1000. The 1:25 dilution provided the best dilution tested Since it produced the highest OD for the positive control samples with little the least amount of background from the negative control samples. 58 2.2.3. Development of AAV ELISA During the initial phases Of development, the AAV2/5 vector (Hauswirth Laboratory, University of Florida, UF11 T5 #E894) was tested at several concentrations in order to create an optimal assay. Using flat-bottom plates (BD Falcon, ref 353279, Bedford, MA) and sterile saline (Butler, Columbus, OH) as the diluent, the AAV2/5 virus (5.6E14Vp/ml stock) was tested at concentrations of 1,000vp/mL, 5,000vp/mL, 10,000vp/mL, 20,000vp/mL, 40,000vp/mL, 100,000vp/Ml, and at several dilutions of the virus of 1:50, 1:100, 1:200, 1:400, 1:800, and 1:1600. Sera from dogs that had been vaccinated with an AAV2/5 virus were used as positive controls for the development of the assay only. Varying dilutions of the control dog sera were used at: 1:5, 1:10, 1:20, 1:40, 1:80, 1:160, and 1:320. The greatest OD values were Observed at the highest virus concentrations, but were affected by the prozone effect (concentration of antibody exceeds the concentration of antigen resulting in a false negative result) at the highest concentrations of sera. A virus concentration of 100,000vp/mL was Chosen for the assay Since there was a limited amount of virus available and the OD values were comparatively high with no prozone influence (Figure 2.5). A serum dilution of 1:5 was run in duplicate for test samples since this provided the highest OD value per virus used. The negative control serum was not run due to background interference observed at the 1:5 dilution, but a negative control without sera was run per plate. 59 2.2.3.a. Reagents Phosphate-buffered solution (PBS) 0.05% PBS-Tween wash buffer A sterile saline solution (Butler, Columbus, OH) was used for the virus solution preparation The recombinant virus (made by the Hauswirth Lab at the University of Florida, UF 11 AAV Type 5, 5.60E+13 vp/mL). The negative control consisted of wells with all reagents except serum HRP-anti-Dog lgG (H+L) (Bethyl, Montgomery, TX) 1-Step ABTS used as substrate (Pierce, Rockford, IL) 2.2.3.b. Detailed AAV ELISA Protocol The AAV ELISA was designed in order to determine the systemic antibody (lgG) response created after intraocular injection of the AAV5 vectors into the eye. (see Appendix B for laboratory protocol) 96-well flat- bottom ELISA plates (BD Falcon, Bedford, MA) were coated with 100uL per well of virus solution, covered with Clear adhesive slips (Nunc, Denmark) and left for 15 hours at 4°C. The wells were then washed with 200uL PBS-T solution and emptied three times Serum dilutions were prepared and applied to the wells at 100uL per well 0 Serum dilutions of 1:5, 1:10, and 1:20 in PBS-T and were run O Positive control serum was run on each plate 60 O Negative control consisted of all reagents except sera to account for plate variability Plate re-covered with plastic Slip and placed on shaker, setting #5 (Wellmix, Thenno LabSystems), and left at room temperature for one hour The plate was removed and washed with PBS-T and then emptied three times as before The secondary antibody was added at a dilution of 1:20.000, plate re- covered with the Slip, and left at room temperature on a Shaker (setting #5) for one hour without Shaking The plate was removed and washed with PBS-T and then emptied three times as before 125uL of 1-Step ABTS was added to each well, re-covered with the slip, and left at 37°C for one hour The plate was read at dual wavelengths of 405nm and 450nm using a plate reader (Bio-rad microplate reader, Hercules, CA) to determine optical density (OD) values 2.2.3.a. Results of AAV ELISA Development The AAV ELISA was optimized using a range of control dog sera and virus concentrations (Figure 2.5). The optimal virus concentration was determined using the AAV-GFP immunized dogs. The optimal virus concentration ranged between 2.8x1011vp/mL and 100,000vp/mL. A range of positive control serum dilutions were used in order to determine the optimal serum dilution for the test 61 sera. The final assay design utilized 100,000vp/mL and a 1:5 serum dilution Since this used the least amount of virus per well while resulting in high enough OD values to analyze the anti-AAV antibody level with little wony of a prozone effect occurring. 62 .anm :39 oczEETBaEumEI .EEom gang. _mEo: nwmz .OD =_moa "Boa . Bow _ozcoo o>=mmoc "9Q .mEEEo. 968.98 9.0 £5, UON_::EE_ moon E0: Bow :0 ocean cozgcoocoo nEO _mEzao 05 950% €90 wE... .n.u 959". 5:25 E200 OOOTF 00m; Gov; O_L_. , _U I _HTI . -H_U -I . o No v.0 58 -89; “to D . 68 .08; new I o o Boa .08; 9.0 J wd m 58 .8: 9.0 I A 98-80: among F m ea .83 3.0 D m; ea .08; E0 I 9a -83 9.0 I =_woa 9.: 9505 Lama m2... .ed 0.59". 8:25 E200 COO—.2. omhnw 00m; 0mm; om; mm; or; m2. Juli: .. E. n 1 w i i- s . s , Nd v.0 ®.O md oc_ouo>-aw0n_ -Um_ooa I fl r , oc_oom>-um0n_ -ooaaw I , . N. _. cc_oom>.fion_ 65?. D, , oc_oom>-oi -Oo_OOn_ D e . V. _. o:_oom>-o._n_ 603.35 I L oc_owm>-9n_ .92."... i 0.? =wmoz ocm o>=_moh_ 05 052.580 2 520 E m; 0.0 83 Ba: 25:25 anm moo 6:80 5 co_a_a> €38.28 << o5 ouEzao 2 now: 96:26 3.3 new Bow 05 950% E50 mEH .m.~ 0.59". 5:25 Eamw 0mm; 00: om; 0.3 0m; 0: m; I - I. o . I._E,\a>m282 ><<-_E< _mEEO 9: BESS S ><< 0cm Bow 000 E mco_c.m:m> Eoan_o>o0 << . Po 9009A GD 66 Chapter 3 HUMORAL IMMUNE RESPONSES DETECTED IN DOGS GIVEN INTRAOCULAR TREATMENT OF VIRAL VECTOR 3.1. Introduction The humoral immune response induced from three different viral vectors after intraocular administration was investigated. AAV-GFP vectors were studied extensively since the AAV vectors have been most commonly used for intraocular gene therapy. Systemic and intraocular immune responses to both GFP and the AAV capsid were investigated. Lentivirus and Av-1 vectors were being assessed as potential vectors for ocular gene therapy by using the GFP reporter transgene. The anti-GFP response was monitored in dogs tested with these vectors. 3.1.1. Intraocular Injections In puppies after eyelid opening a subretinal injection was made under direct visualization using an operating microscope via a transvitreal approach. Subretinal injections were performed on puppies approximately 3 weeks of age or older while transscleral injections were performed on neonatal pups 3 to 8 days of age. In neonates the eyelids are fused shut and the cornea is not yet clear so injection under direct visualization is not possible. The neonatal pups have immature retinas and the aim of the injection was to reach the developing neurosensory retina to introduce the transgene at an early stage of retinal development. This timing may be required for very early-onset retinal 67 dystrophies. The precise site of injection, subretinal, intraretinal, or intravitreal, cannot be visualized when transscleral injections are performed prior to eyelid opening. 3.1.2. Anesthesia Anesthesia was induced in three day old puppies with isoflurane (Abbott Laboratories, North Chicago, IL) delivered by oxygen mask and anesthesia was maintained in the same manner. For three week old puppies, induction was by masking with isoflurane; they were then intubated and maintained at a surgical level of anesthesia with isoflurane (Abbott Laboratories, North Chicago, IL) delivered in oxygen. For dogs older than 9 weeks of age, intravenous thiopental sodium (PentothalQ, 6-12 mg/kg) was used to induce anesthesia; dogs were then intubated and maintained using isoflurane (Abbott Laboratories, North Chicago, IL) delivered in oxygen for the rest of the procedure. A pulse-oximeter (Vet/Ox 4400, Heska Corporation, Fort Collins, CO) was used to monitor the animal while under anesthesia, recording both blood oxygenation and pulse rate. A heating pad was used to maintain the dog’s body temperature. Pulse rate, oxygen saturation, and body temperature were recorded every 5 minutes during the injection procedure. 3.1.3. rAAV2/5, Lentivirus, and Av-1 rAAV2/5 vectors with one of two different promoters driving the GFP transgene expression were used; one promoter was the chicken-beta actin promoter (AAV- 68 CBA-GFP) which results in ubiquitous expression of the transgene and the other was the mouse opsin promoter (AAV-mOPS-GFP) which results in photoreceptor-specific expression of the transgene. Both vectors were used in both neonate and 3 week old puppies. These rAAV vectors were created in the laboratory of William Hauswirth (University of Florida). The protocol for creation of these vectors has been published (Zolotukhin et al., 2002). The Av-1 vector was a serotype 5 vector that was used at a concentration of 3x10"12 vp/mL (Amalfrtano Laboratory, Michigan State University). The lentivirus vector (pHR’-CMV—GFP, Planelles, University of Utah) was used at a concentration of 10"7 to 5x10"7 infectious units‘(Zhu et al., 2001: Planelles, 2003) 3.1.4. Injection Technique Used in Puppies Where the Eyelids Have Opened Subretinal injections were performed by Simon Petersen-Jones under aseptic surgical conditions. A subretinal injector as seen in Figure 3.1. (RetinaJect Injector, Surrnodics, Irvine, CA ), with Hamilton syringe (Hamilton Company, Reno, Nevada) and loaded with the vector solution, was used to enter the eye through the sclera (Figure 3.2.). The injector was positioned just anterior to the retina in the tapetal region dorsal to the optic disc. 69 Subretinal in'e tion ./’¥\k ., Figure 3.1. Path taken by the injector for the subretinal injection procedure. The arrow indicates the area of injection, the subretinal space between the RPE and photoreceptor layers of the retina. Silicone tubing 399 cannula \ 7 , / l, 25g needle \ ' I l I Delrin handpiece Figure 3.2. Subretinal injector (RetinaJect Injector, Surrnodics, Irvine, CA) used to perform the subretinal injections. (htt ://www.surmodics.com/ a eDetail.as x? a eld=93&menuld=95, 3/6/07) The small internal cannula inside the injector was then advanced so that it was pressed against the retinal surface. The vector was then injected and the force of the injected fluid created a retinotomy allowing the fluid, ~50pL, to pass into the subretinal space resulting in a retinal detachment (see Figure 3.3. for fundus 70 photographs showing before and after injection). The cannula was withdrawn into the needle and the injector withdrawn from the eye. Figure 3.3. Before and after subretinal injection in a dog’s eye. The top image is prior to injection. In the bottom image, a retinal detachment is evident in the tapetal fundus dorsal to the optic disc. This is the site of subretinal injection. 3.1.5. Injection Technique Used in Neonates These injections were performed by Simon Petersen-Jones on pups prior to eyelid opening. A small area on the upper eyelid was cleaned with diluted betadine, and a small skin incision was then made. Blunt dissection of the area 71 was performed using tenotomy scissors until the sclera of the eye was identified. A small incision was made through the sclera posterior to the ciliary body region to expose the choroid. A 309 blunt cannula was introduced through the choroid and an injection was made, On average, a 25uLvolume of vector was injected into the eye (Figure 3.4.). The subcuticular tissues and skin incision were closed using 6-0 vicryl (Ethicon, Piscataway, NJ) sutures. Figure 3.4. Injection site in neonatal puppies. After incision into the eye globe, the vector was injected into the region of the developing retina as is shown by the arrow. 3.1.6. Dogs Used in Study and Sample Collection The dogs used in this study were from a purpose-bred colony of mixed and purebred (Cardigan Welsh Corgis and Briards) dogs at Michigan State University. All animals were treated with care under IACUC specifications. See Tables 3.1 and 3.2 for a list of dogs in the AAV anti-GFP study and in the anti- AAV study, respectively. Adult dogs that underwent intraocular lentivirus (Planelles, University of Utah) treatments included Haley, Cornet, and Jackie. All dogs were treated with 50pL of the vector through subretinal injection. 72 One dog undenrvent intraocular treatment with 50uL of an Av-1 through subretinal injection. 3 Weeks of Age Injections (0-8 week serum samples) ID Eye Dog Number Injected Vector Injection Type Kumquat 8 Left AAV-CBA-GFP Subretinal Lazarus 1 2 Left AAV-CBA-GFP Subretinal Martha 1 3 Left AAV-CBA-GFP Subretinal* Magdalene 1 4 Left AAV-CBA-GF P Subretinal* Mary 1 5 Left AAV-CBA-GFP Subretinal Andrew 1 6 Left AAV-CBA-GFP Subretinal Moses 28 Right AAV-mOPS-GFP Subretinal Barbie 29 Left AAV-mOPS-GFP Subretinal 3 Da 5 of Age Injections (0-8 week serum samples) ID Eye Dog Number Injected Vector Jude 1 0 Right AAV-CBA-GFP Transscleral Bartholomew 1 1 Right AAV-CBA-GFP Transscleral Steve 26 Right AAV-mOPS-GFP Transscleral Kelly 27 Right AAV-mOPS-GFP Transscleral 3 Weeks of Age Injections (0-4 weeks serum samples) ID Eye Dog Number Injected Vector Chinook 1 Left AAV-CBA-GFP Subretinal Hyde 2 Right AAV-CBA-GFP Subretinal Aramis 3 Left AAV-CBA-GFP Subretinal Frenchy 4 Right AAV-CBA-GFP Subretinal Penny 5 Left AAV-CBA-GFP Subretinal Shaila 6 Left AAV-mOPS—GFP Subretinal Merry 7 Left AAV-CBA-GFP Subretinal Lucy 1 7 Right AAV-mOPS-GFP Subretinal Marty 1 9 Left AAV-CBA-GFP Subretinal Tywyn 30 Right AAV-CBA-GF P Subretinal Table 3.1. Colony dogs injected with AAV2/5-CBA-GFP or AAV2I5-mOPS- GFP vectors for the GFP ELISA protocol. Subretina|* = some vector went intravitreal. 73 Anti-AAV Dog Serum Samples for Anti-AAV ELISA Eye Injection Dog Number Injected Vector Type Chinook 1 Left AAV-CBA-GFP Subretinal Aramis 3 Left AAV-CBA-GFP Subretinal Frenchy 4 Left AAV-CBA-GFP Subretinal Penny 5 Left AAV-CBA-GFP Subretinal Shaila 6 Left AAV-mOPS-GFP Subretinal Merry 7 Left AAV-CBA-GFP Subretinal Bartholomew 1 1 Right AAV-CBA-GFP Transscleral Lazarus 1 2 Left AAV-CBA-GFP Subretinal Martha 1 3 Left AAV-CBA-GF P Subretinal* Magdalene 14 Left AAV-CBA-GFP Subretina|* Mary 1 5 Left AAV-CBA-GFP Subretinal Andrew 1 6 Left AAV-CBA-GF P Subretinal Marty 19 Left AAV-CBA-GFP Subretinal Steve 26 Right AAV-mOPS-GFP Transscleral Kelly 27 Right AAV-mOPS-GFP Transscleral Moses 28 Right AAV-mOPS-GFP Subretinal Barbie 29 Left AAV-mOPS-GFP Subretinal Tywyn 30 Right AAV-CBA-GF P Subretinal Braedon 31 Both Both Subretinal Table 3.2. Colony dogs injected with AAV2/5 vectors that were used for the AAV ELISA protocol. Subretinal” = some vector went intravitreal. Both aqueous and serum samples were taken from all dogs injected with the GFP vectors. Aqueous samples were collected pre-injection on the day of surgery and then post-injection at the time of euthanasia. Serum samples were collected in two different groups of sampling. The first group of dogs had serum collected on the day of injection and then every two weeks until four weeks post- injection and at the time of euthanasia. The second group of dogs had serum samples taken the day of injection and then every two weeks until eight weeks post-injection and at the time of euthanasia. Several dogs had samples taken 74 past the eight week limit that were not ‘day of euthanasia’ serum samples. Serum samples were centrifuged for 30 minutes in at 3500 rpm at room temperature and then stored in 1 mL aliquots at -80°C. Serum collection \ Day 29 Y1 $ \L Ni 3 week treatment 3 day treatment I Birth \ Y J = 2 weeks Serum collection Figure 3.5. Serum and Aqueous Sampling Time Line. 3.1.7. Monitoring GFP Expression GFP expression in the eye was monitored using a RetCam (Clarity Medical Systems, Inc., Pleasanton, CA). Every two weeks post-injection until sacrifice, all of the injected dog eye(s) were examined under regular white light and then using the fluorescein angiography setting. This setting provides light of a wavelength to cause fluorescein to fluoresce and a barrier filter in the viewing pathway to enhance the visualization of the fluorescence. The images were used to observe the GFP fluorescence after intraocular injection and were stored for further assessment at a later date (Figure 3.6.). 75 Figure 3.6. GFP expression was monitored using a RetCam. In the top image, a white light image of the fundus in which the retina appears normal with a small trace of a subretinal injection scar. In the bottom image, the fluorescein angiography capabilities of the RetCam are used to view GFP expression. 3.1.8. Data Analysis using SAS Statistical Analysis Software (SAS) was used to analyze data sets. All data sets (AAV-GFP, Av-1, and lentivirus) were analyzed separately. The anti-GFP AAV-GFP serum and aqueous data sets were not normally distributed but were still analyzed using a ‘mixed’ model analysis. The statistical model to analyze this data set included fixed effects of injection age, injection method, and promoter. The dog (based on ID number) was the random effect for the model. The positive and negative control sera were used as covariates in the analysis in order to account for individual plate variations. The least squares mean (LSM) antibody level was reported for pre- and post-treatment, time points collected, injection method, injection age, and promoter. All 3 week serum samples were included in the 4 week serum sample category for this analysis. 76 The anti-AAV data set was not normally distributed but was still analyzed using a ‘mixed’ model analysis. The statistical model to analyze this data set included fixed effects of injection age and injection method. The dog (based on ID number) was the random effect for the model. The assay date was used to account for plate variation. The LSM antibody level was reported for pre- and post-treatment, time points collected, injection method, and injection age. The anti-GFP lentivirus data set was normally distributed and was analyzed using a ‘mixed’ model analysis. The dog (based on ID number) was the random effect for the model. The positive and negative control sera were used as covariates in the analysis in order to account for individual plate variations. The LSM antibody level was reported for pre- and post-treatment and time points collected. The anti-GFP Av-1 data set included only one dog for analysis and thus was not analyzed using SAS. All original values were reported. A ‘PROC CORR’ analysis was used to compare the OD values of two data sets: anti-AAV and anti-GFP AAV-GFP. The correlation value was reported. A ‘PROC CORR’ analysis was used to compare the OD values of two data sets: Anti-GFP AAV-GFP aqueous OD values and anti-GFP AAV-GFP serum 00s. The correlation value was reported. 77 A p-value of <0.05 was considered significant for all statistical analyses performed. 78 3.2. Results Two ELISAs were developed in this work and used to determine if gene therapy administered in the canine eye induced local and/or systemic antibody responses either to the therapy gene and/or the virus vector. In this study, GFP was used as a reporter gene, and AAV, Av-1, and lentivirus were the vectors for subretinal and intravitreal injection methods. Thus, ELISAS developed utilize either GFP or AAV viral capsid as antigens. The GFP ELISA was used for the samples collected from each dog in order to detect a systemic humoral response to GFP after treatment with AAV-GFP, Av-1, and lentivirus. The AAV ELISA was used for the serum samples in order to detect a systemic humoral response to the AAV viral capsid. 3.2.1. Presence of Circulating Anti-GFP Antibodies after Intraocular Treatment with AAV-GFP Three serum dilutions were tested using the GFP ELISA previously described (Figure 3.7.). All dilutions followed a parallel pattern and therefore serum dilution 1:5 is reported. The least squares mean (LSM) was reported for each dilution; therefore, a negative value may be reported for the pre-treatment sera since the mean has been calculated using the positive and negative covariates from each plate. The anti-GFP antibody response was found to vary between individual dogs. A high anti-GFP level, comparatively, was observed in 4 out of the 22 dogs tested 79 (Figure 3.8.). Overall, there was a statistically significant systemic humoral response detected to GFP after intraocular injection of the AAV-GFP viral vectors with a p-value of 0.0029 for the comparison of pre- versus all post-treatment time points (Figure 3.9.). Time points analyzed in this assay were: 0, 2, 4, 6, 8, and >8 weeks post— injection (Figure 3.10.). Using paired t-tests, antibody levels for time points 4 weeks post—injection showed a significant difference from zero (p= 0.0409). The results from samples collected at 6, 8, and >8 weeks post-injection were not significant (p= 0.1038, p= .190, and p= 0.115, respectively) based on the LSM compared to zero. Overall, the trend observed in dogs was a detectable immune response at 4 weeks post-treatment and was found to decrease starting at 6 weeks post-treatment. One dog that was monitored long-term had an antibody response that was maintained up to 26 weeks post-injection. Additional independent variables included in the statistical model were: promoter (CBA vs. mOPS) and injection age (3 days vs. 3 weeks). The animals treated with the AAV-CBA-GFP vector had a significant antibody response while those treated with the AAV-MOPS-GFP construct did not. Dogs treated at 3 weeks of age had a significant anti-GFP antibody response while the group at 3 days of age did not (see Table 3.3). 80 3.2.2. Presence of Anti-GFP Antibodies in Aqueous after Intraocular Treatment with AAV-GFP An aqueous dilution of 1:20 was used for assays for two reasons: 1) limited sample volume and 2) higher aqueous concentrations did not result in an increased antibody observation capability based on sample tn'als (not shown). The anti-GFP antibody response was found to vary between individual dogs with some dogs having an aqueous anti-GFP response while others did not. Overall, there was not a significant local humoral response detected to GFP after intraocular injection of the AAV-GFP viral vectors with a p-value of 0.0667 for the comparison of pre- versus post-treatment samples (Figure 3.11.). Based on euthanasia schedules, a range of 13 to 96 weeks post-injection aqueous samples were analyzed with an equally wide range of individual antibody responses observed (Figure 3.12.). Two independent variables were analyzed including: promoter (CBA vs. mOPS) and injection age (3 days or 3 weeks). Unlike the case for serum anti-GFP antibody responses, none of these variables were shown to have a significant effect on the local immune response in the eye (Table 3.4.). 81 35:5 8; 55% m; 9.: >_co .m_m>_mcm 85...: Lo". 65:26 65 comics 95:26.2 6:93 a m_ 29.: .9: 293868 2 moon nofimz-><< Eo: << 9 m>:m_mm wet. _ mod- 7 m m e m \m _ ll . . llllr .I- . . l - Ll - ll-‘i :“T o A no.0 W 9 B . Po W ON; 0 or; mfio w: No R can 5 6 .v .N .c 9.025 :0 0202.00 5.667.. 33002:: auO-><< Lora “52383 cm; can .2.; .m# 23:25 .2 EEom E 205.. >uo£E< 9.0.2.? .2 moo cams. mafia—um «map.— 82 .589 ><< £3. E953: 538:5 Lots mop 33229.. some .3 << 0. 9.6.2”. 3.82. :ll , l: ,- l--- l , coma- wwwmvm «mommrosimsos m o v m e ,. ...|- I . _ . . til- : . :lll ; -all'Lll |L|- LII}; . z , ..- r. ll- 000.0 :5, F .o 3:50 m>=mmmz $1994.. .03an waOE 5253.: x33 m H II. .53an K . o 9.... .o.o~ 2 .>.m._ 2.. 2o comrades". co momma 830:.ch 65 $522.. new 526%. m. .3 some .2 33>-.. 2.... dd 95m."— . / \ ..mmood E32...“ E33 m. 5 E053... “mo... new 0.... p200=oo 5.62:. 3.302.... mu0-><< .23 E33 5 265.. 23223 9.0-354 .2 30 22mm .3 caps. moan—um ammo... m...o- _..o- mo .o- 0 mod enjeA ClO ueew _..o Nd mNd 84 0:005:90 2.02.0220 n. . E505 0.00 .2 00.00.: 0 5.2. 0300 00.000. 0. 0:... .0 0.00:? .:0U:000_0c. 0:. :0 00000 020?: __0.0>o 0:.r .o.0N 2 $5.. 0:. .o :00...qu00 :0 00mm: 00:00...:m.m 0... 02005:. 0:0 00.000. 0. .00 :000 .2 020?: 0...... .36 0.39". E0200; n.n.0-><< o. 0>..m_0m. v.025 . - m 0 . v N N o 0A 0 to- H .||..|F_.|. l. l l O to N0 . f . 0.0 m.. v.0 omwd word 9.0.0.. ..mmd nmwod .V.O . 0.0 / \ 3 0.0 vmood... @.0 II) 0 9mm 00 ueew ..... N... m... E320 83.00 m. 5 0A 0:0 .0 .o .e .N .0 9.00.5 :0 00.00.60 5200?. .0.:000..:_ 9.0 -><< .020 E:.0m :. 0_0>0.. >00:..:< 9.0-..:< .2 «DO .e.—mm 2+. :00... 0.0.03.6 .000. 85 .8 5... ~33 0E0... .880 .23 :6: .530... :00»... 0:.0> A. 808.0 803 5:... 008°... 3:0 0.8520 28... «$8.? 085... 82. $8.... 0.. 0.3; n 2...... n 0.0.... <00 stud"... mu... 8.8.... .203"... .2050... 0.... ..mau. 0030...; 30050005 .0. Am. 5 00.0E0m E:.0w won. 20.... 00:_0> DO on0 00.000. 0. 0:0 00:_0> DO EOE.00.E0o0 0:0 -90 0o :O0_.00E00 :o 0000.. 0:_0>-0 __0.0>o 05H .o._0N o. 5.0-. 05 00 50:00:50 :o 0000.. 00:000.:90 05 00.00.05 0:0 00.000. 0. .00 :000 .2 0:.0>-0 0.? .36 0.50.". :20-00.50000 5:00.505 F .0- 00.0- 00.0- 3.0- . No.0- .1.-0...: A . ,. ,u an... 0...... -. 1: . O . . . ,. .H «80.0 No.0 00.0 / \ . 00.0 -- $8.0 . 00.0 to 0:00:00 no 502% on” 5 :o_.00_:_..0o0 0:0 .20 00000.30 03003.2 5 0.96.. €8.05 000-05. .2 000 0.00.3 :85. 020300 .08.. enleA go ueew 87 ..0.00> ><< 5.2. 20:000.. 03000.2: .000 000 03.20:. :000 .0. . 00 2.0. 0:. 030:0 500.0 0.....- .N...n 2:0... 2.05.00: 0.0-3.2 0. 00.00.. x82. 00 00 00 8 00 00 0.. 00,00 0. 0.0. 0. 0. 0. 09.0 ”.0080 02.0002 000.03. 00.. u.o.Eoo 02:00.". 0m0.0>< . 30.300 .0 02...; 00 <0_._m 0.0 0:833 no: 0:220... 0.. F.0- 0 v.0 - N0 0.0 - 0.0 i- 0.0 0.0 0.0 0.0 0.0 _- ..é anIRA (10 88 340... E a... 00.0... 83.0 .22 E2. 2.22.... . :00:.v 0:_0> .0 00000... 503.0 .003... 023.0 .20 202.30 808.? 808.... 53...... 0 as... :8: 0.. 0.0.. 0 9.00; 0 0.0.2 <00 .8000»... ou< 508.... .3000»... .2022. 00.00..0> 0:00:00005 .0. SN" 5 00.0800 0:00:0< 000 50.. 00:_0> 00 <0...m 000 .0 0:00.... 00.0:00 «000.. .0...” 030... 89 3.2.3. Correlation between Anti-GFP Antibody Responses in Serum and Aqueous Samples A correlation analysis (PROC CORR) was performed in SAS for the GFP ELISA results between the serum and aqueous samples for 17 dogs (Figure 3.13.). A correlation (R"2) of 0.30062 was observed based on this analysis with a p-value of 0.2255; therefore, a relationship between the systemic and local antibody responses against the GFP reporter gene was not determined from this sample of dogs. One dog had a high level of both aqueous and circulating anti-GFP antibodies at euthanasia (seen in Figure 3.13.). The data were analyzed with and without this dog’s data, which was removed for the final analysis since they were found to significantly bias the analysis. 3.2.4. Presence of Circulating Anti-AAV Antibodies after Intraocular Treatment with AAV-6F P A single dog out of the 19 dogs tested had a high anti-AAV OD value. The data were analyzed both with and without this dog included, and the dog was found to bias the data set and was therefore removed. Overall, there was a statistically significant systemic anti-AAV antibody level after intraocular injection of the AAV- GFP viral vectors with a p-value 0f 0.0157 for the comparison of pre- versus all post-treatment time points (Figure 3.14). Time points analyzed were: 0, 2, and 4 weeks post-treatment (Figure 3.15). Overall, the trend observed started with a significant immune response detectable at 2 weeks post-treatment which then tapered off slightly at 4 weeks post-treatment. 90 Additional independent variables were included in this statistical analysis which included: promoter and injection age (3 days vs. 3 weeks). Neither of the variables was observed to significantly influence the anti-AAV antibody response (Table 3.5.). 3.2.5. Correlation between Anti-AAV and Anti-GFP Antibodies in the Serum A correlation analysis (PROC CORR) was performed in SAS for the GFP ELISA and AAV ELISA results for serum samples at time points 0, 2, and 4 weeks post- treatment (Figure 3.16.). A correlation (R"2) of 0.07859 was observed based on this analysis with a p-value of 0.6390; therefore, no relationship between the systemic anti-AAV and anti-GFP was detected in this sample of dogs. 3.2.6. Presence of Circulating Anti-GFP Antibodies after Intraocular Treatment with Lentivirus Overall, there was a systemic humoral response detected to GFP after intraocular injection of the lentivirus viral vectors with a p-value of 0.0200 for the comparison of pre- versus all post-treatment time points (Figure 3.17.). Individual time points analyzed in this assay were: 0, 2, 4, 6, and 8 weeks post- injection (Figure 3.18.). Using paired t-tests, antibody levels for time points 2, 4, 6, and 8 weeks post-injection showed a significant difference. Overall, the trend observed starts with the largest peak at 2 weeks which then tapers off slowly until 8 weeks post-treatment. 91 3.2.7. Presence of Circulating Anti-GFP Antibodies after Intraocular Treatment with Av-1 One dog was treated with Av-1 in order to study the potential systemic antibody response to GFP. Time points analyzed in this assay were 0, 2, and 3 weeks post-injection using a 1:20 serum dilution since the 1:5 and 1:10 semm dilution 003 were influenced by a prozone. At 0 weeks, the OD value was 0.116 while at 2 and 3 weeks post-treatment the OD values were 1.055 and 1.078, respectively. Overall, the antibody response observed was high at 2 weeks post-treatment and increased further at 3 weeks post-treatment (at which time the trial was terminated). 92 w:_m> 00 anw amo-=c< F0. 00““... 000._. 000.0 000.0 00v.0 00N.0 00 .0 L _ _ _ . _ O v W m... I _..0 d V b . m 1 N0 0 n S . O r mo 0 A mo: 0 r «.0 m 0.0 moi—tom .5980 one mzoozod. E mo:_o> 00 o 0;... .93 o. .20.. 0;. 00 8280600 :0 0003 00:85:90 0..: $8205 0:0 00.000. w_ :3 some .2 029-0 9:. .36 0.59". 00.0- 600 2n. . 3.0- No.0- 0 886 N0.0 m W a B U O B v0.0 m 9 00.0 00.0 .858 f \ to .050 , - Nfio Egon... 22$ .6 522:. m” 5 E0-><< 53. Ed “co—:32... 20.300035 ..otm cozooEimom 0:0 .95 0900:00 £3.00 5 226.. 33:5 ><<._E< .2 80 250.3 58.2 8.2.3 6.8.. 94 0:005:90 2.00:0...00 u. . 56:0 000 :0. .3005 0 :Es 0>0n0 00.000. 0. 0E: .0 0.00.:0> E35002. 0.... :0 00000 0=_0>-0 .003 0:... .90N o. .20.. 0:. .0 50:00:50 :0 00000 8:005:90 0:. 00.00.05 0:0 00.000. 0. .00 :000 .0. 0=_0>-0 0.: .m...m 050.“. 205.00: 9.0.32. 2 02.000 0.025 V N 0 ..mmmod \ finned E320 E300 00.0- V0.0- No.0- 0 No.0 V0.0 00.0 00.0 v.0 N _. .0 v _. .0 emeA (10 ueew no cows—:0 9 5 000$: 5:5 acwEu00:._.-umom 9.003 v 0:0 .N .0 0302.00 E300 :_ 0_0>0.. 2000.050. 32.0.3 :0.— 000 Emmi. c005. 00.0300 “000.. 95 96 0000.0 0000.0 0000.0 090.0 020..-: 00000.0 .0000 00000.0 00000.0 .200 0000.0 00000.0 00000.0 00000.0 000.2 0.. 30:30.00. 3:00.530 9.003 n 0.30 n .0000»... 0...... 00.08.... .0020"... 0...: 00.08.... 00.0500 £200 000 ..o. 00:_0> no <0.._m ><< ..o :00... 00.0300 000.. .00 030... $6 No 0:_m> DO <<-_E< 0.0 md Yo md 00.05% E:.0m E0: Nd 0 oomd .iIllT 000.0 0 . oomd m0:_m> DO <<.._u:< ho 0.030.230 9.0 050E enIRA CIO VSI'IE d:|'9'!1UV 97 0:005:90 0.0000090 n. . E0600 200 .2 000.0000 0 5.3 0>o00 00.000. 0. 5.0.. 000800000000 0:0 -05 0o 000000008 05 co 00000 00.0?0 .0205 00... 6.0.0 2 .20.. 05 00 5000088 :0 00000 000005020 05 0200.00. 000 00.000. 0. .00 0000 .6., 00.0?0 00.0 Eur. 050E “won— mhm : . x O . no.0 ,_ O *0NN00 m _. .O (:10 UBGW 9! o mmd «0000.0 .00N0.0 £2600 , 0.0 E3000 3 00.5..0 m" 5 000-030.2000.. 5.3 000E002... 0000. 0:0 0.0. 00000.60 E3000 c. 0.05.. >0o0=c< 0.00-0.2 ..2 000 €30.10 :00...— 0000300 00000 98 00005090 0.0000030 n. . 02600 0.00 .o. 0.9.00.0 0 5.3 0>o00 00.000. 0. 00... .o 0.000? 0.0003000. 00. co 00000 03.01. ..0.0>o 00... .o.0N o. .20.. 00. .0 000000050 00 00000 000005000 00. 00.00.00. 000 00.000. 0. .00 0000 .2 03.0?0 00... .00....“ 0.00.". .00E.00.... 02.20.00 0. 02.0.00. x00>> m 0 0 0 0 . . O . . mod . to , W . . mwd W ._. .00000 u Nd O . . 0 0000.0 .— 00.0 ..0000 0.0 iNOOO-O . mmuo .02500 05.00 .0 02.2.0 0” 0 0.002.053 5.; 205.00; .00.. 0 0:0 .0 .0 .0 .0 9.00.5 00 00.00.60 0.3.00 0. 0.050 >00000< 0.00-050 3.00.02.50.10 0005. 00.0000 .000._ 99 3.3. Discussion and Future Work 3.3.1. Discussion and Summary of Results Circulatory anti-GFP and anti-AAV antibodies were observed in some but not all of the dogs treated with the AAV-GFP vector. The single dog treated with the Av—1 vector had a high immune response to GFP and the lentivirus treated dogs all maintained a circulatory antibody response. Variations in antibody response differed between vectors based on time of onset and level of antibodies detected. The AAV-GFP vectors induced an anti-GFP antibody response approximately 4 weeks post-treatment whereas the lentivirus and Av—1 vectors induced an anti- GFP antibody response as soon as two weeks post-treatment. 3.3.1.a. Anti-GFP Antibody Response after Treatment with AAV-GFP As we hypothesized, an anti-GFP humoral immune response was detectable in serum as early as 4 weeks post-treatment. This delay in development of a humoral immune response correlates with the relatively slow onset of transgene expression typical of AAV vectors. The anti-GFP antibody response was also detectable up to 26 weeks post-treatment (the last time point that was collected). An anti-GFP antibody response could be detected in post-treatment aqueous samples as late as 96 weeks post AAV-GFP treatment. 3.3.1.b. Anti-AAV Antibody Response after Treatment with AAV-GFP The AAV ELISA detected an anti-AAV humoral immune response in serum samples from some of the treated dogs, and this response was first detected 2 100 weeks post-injection. The antibody response was detected two weeks sooner than for GFP, as would be expected since the antigen is present at injection and does not require transgene expression to be occur. The antibody response was low compared to the anti-GFP response but was still significant over the time points analyzed. 3.3.1.c. Discussion of Results from AAV Treated Dogs Unlike other studies, we were able to study the antibody response to GFP in enough detail to understand the approximate onset of the anti-GFP antibody . response and also duration of up to 26 weeks for serum and 96 weeks for aqueous samples. One area of interest in this study was the variation in anti- GFP antibody responses between individual dogs. Previous studies have observed an antibody response to GFP in both serum and aqueous post- treatment samples after intraocular treatment of AAV-GFP (Bennett et al., 1999; Dudus et al., 1999). In those studies, 2 out of 4 and 1 out 3 mice, respectively, had a similar circulating high level of anti-GFP antibodies after intraocular treatment with AAV-GFP (Bennett et al., 1999; Dudus et al., 1999). Specific for the AAV-GFP treated dogs in this study, two independent variables in the study were found to significantly influence the anti-GFP response: the CBA promoter led to a greater immune response compared to the mOPS promoter and injections given at 3 weeks of age resulted in a greater response than those given at 3 days of age. 101 Since the eye is an immune-privileged environment, antibodies would be unable to cross the blood-retinal barrier due to the size of the molecule; therefore, an intraocular immune response has been explored. Recently, the iris was found to produce a homing effect which led monocytic bone marrow cells to the iris which then participated in a local immune response (Li et al., 2006). Furthermore, the iris monocytic cells may participate in the systemic induction of regulatory T cells; thus, systemic viral transduction may not need to occur to induce a systemic humoral response. Viral escape from the eye could potentially allow for viral systemic transduction of host organs and tissues. Virus escape could occur through many avenues which include escape through the drainage apparatus as well as escape through a degraded blood-retinal barrier. Upon aqueous sampling from the eye, breakdown of the blood-retinal barrier occur this could allow virus to escape into the bloodstream (Strubbe and Gelatt, 1998). lntravitreal or subretinal injection can also allow viral vectors to reach the central visual pathway. The highly significant influence of the CBA promoter on the anti-GFP response could be because the mOPS promoter limits expression of GFP to the photoreceptor; thus, if the viral vector construct reached other tissues, GFP expression would not have occurred in those tissues, whereas with the CBA promoter it would. The CBA promoter is a ubiquitously expressed promoter whereas the mOPS promoter is photoreceptor-specific. 102 The influence of the age of 3 weeks at the time of treatment is also fascinating since the pup’s immune system at 3 days and 3 weeks of age are different. At 3 days of age, the pup’s immune system is less mature and is more dependent on the passive immunity from mother’s colostrum and is in the process of recognizing “self” (T izard, 2004). By three weeks of age, the pup is starting to lose maternal antibodies and therefore its immune system is more able to recognize and produce antibodies to foreign antigens. Also to consider, since only an lgG antibody response was analyzed, genetic variability between dogs may account as well for the individual response observed between dogs. 3.3.1.d Correlation Analyses No correlation was observed between the anti-AAV antibody response and the anti-GF P response after intraocular treatment of AAV-GFP. The other correlation, between the anti-GFP antibody responses in serum and aqueous samples, also showed no significant relationship. A small number of euthanasia serum and aqueous samples were collected from a large range of time points post-treatment and therefore could account for the lack of correlation observed. 3.3.1.a. Anti-GFP Antibody Response after Treatment with Lentivirus The serum samples from dogs treated with lentivirus undenrvent analysis using the GFP ELISA. As we hypothesized, an anti-GFP systemic humoral immune response was detectable in serum from all dogs as early as 2 weeks post- treatment up to 8 weeks post-treatment (the last time point at which samples 103 were collected). These results showed the highest antibody level at 2 weeks post-treatment with a slow decrease over the 8 week time period. These results are limited since the serum samples were collected every two weeks, and thus an antibody response may be detectable sooner than 2 weeks post-treatment due to fast expression of the GFP transgene. Further work should be done in order to understand in more detail the antibody response produced against the virus as well as the transgene. 3.3.1 .f. Anti-GFP Antibody Response after Treatment with Av-1 The serum sample from the dog treated with Av-1 underwent analysis using the GFP ELISA. This vector results in rapid, strong GFP expression within 24 hours post-treatment and also induces some degree intraocular inflammation. As we hypothesized, an anti-GFP systemic humoral immune response was detectable in serum at 2 weeks post-treatment and remained high at 3 weeks post-treatment (the last time point at which samples were collected). More work should be done using this vector in order to understand the antibody response in dogs and it should also be studied over a longer period of time. 3.3.1.9. AAV-GFP, Lentivirus, and Av-1 Antibody Responses Compared Each of the vectors induced a different anti-GFP response after intraocular treatment (Figure 3.19.). The AAV vectors, overall, seemed to produce a low antibody level, but this response did vary between the dogs. The lentivirus vector produced a slightly higher antibody level in the three dogs studied, and the 104 antibody levels did seem to decrease over time. Finally, one dog was studied after intraocular treatment of Av-1 which induced the highest antibody level. These results are important for continued research using viral vectors for gene addition therapy within the eye. A humoral immune response could limit the therapeutic effectiveness over time and could also inhibit success of repeated dosing. 3.3.2. Future Research The AAV vector ELISA results allow for the continuation of research in several connected areas. The potential for genotoxicity and other adverse long-term effects such as waning of the transgene or limited therapy potential remain important issues for the feasibility and success of ocular gene addition therapy. . The spread of the vector within the host may be correlated with the systemic antibody response. Real-Time PCR analysis should be performed in order to detect the spread of the vector from the treatment site. Many filtering organs, such as kidney, liver, and lung, as well as tissues from along the central visual pathway, should be included in this analysis. . Since, from previous studies, no inflammation or decreased transgene expression was observed upon repeat administration of an AAV vector into the eye, tolerance may be occurring (Bennett et al., 1999; Anand et 105 al., 2000; Isenmann et al., 2001). Using a constant dose of vector for intraocular injections, the level of GFP expression after one or several injections could be quantified for individual photoreceptors (using a photon-counting confocal laser scanning microscope as described in Rex et al., 2005) and averaged together at each injection time point for individual animals. This would allow for a more detailed analysis of the neutralizing antibody effects on the transgene expression over time and after repeat administration. . As mentioned previously, an antibody response may not significantly affect the transgene expression in the eye. Since ubiquitous promoters are used, the concern about genotoxicity within organs remains a concern. Further studies to observe the possible cytotoxic effect of either single or repeated administration over time would create a more thorough analysis of the effects of using ubiquitous promoters as opposed to tissue-specific promoters for gene therapy. Further work into the lentivirus and Av-1 vectors could also be done in order to further understand the antibody response in the host after intraocular treatment. . An ELISA could be developed to study the antibody response to the lentivirus packaging. 106 . An ELISA could be developed to study the antibody response to the Av-1 capsid. 3.3.3. Conclusion A variable degree of humoral immune response to the transgene and AAV capsid was observed after intraocular injection of AAV vectors. For lentivims and adenovirus vectors, a humoral immune response was observed after intraocular injection. This information is imperative for the safety of intraocular gene addition therapy trials since every patient will have a unique response to the foreign transgene. 107 0.0; 00 000000. 000 T>< 000 .0. 00:.0> 00 30m. 00.3000. 000 ><< 000 .0. 0.00; .00 000000. 5.00 00.3 0:.0> .20.. .2.» 0.30.“. 0002.00: 0.0.32. 2 02.000 0.002, . 0.0- 0.. w 0 V 4 q . - m, o A .0 “w Nd 0.0 o G 0.0 M 0.0-32 u - 0 023000.. u I r 0.0 _->< u I. F ----0......._......._m.m...._.._.....m0.0% ...................................................... . N. . 3-2 .0.— 02600 ow; .00603030000 000 ><< .2 03000 E200 m" 5 0008000.... 5.33000. .000 05.00 0. 0.20.. €8.05 000.05 .0. 000 000-03.300.00 000 ..->< .0.0-><< 108 APPENDICES Appendix A. ELISA GFP Laboratory Protocol MATERIALS NEEDED: For solutions: -N32HPO4 -KH2PO4 -KCL -NaCl -Tween (Polyoxyethylene- Sorbitan Monolauvate) -gelatin- -bicarbonate packets- from Pierce -access to milli-Q water -pH meter -Probind ELISA plates For ELISA protocol: -GFP from Clonetech -secondary antibody- -HRP- anti-dog lgG -HRP- anti-rabbit lgG -ABTS (acts as the substrate) PREPARATION OF EQUIPMENT AND SOLUTIONS: Cleaning of Conboy: Rinse conboy thoroughly with tap water five times, distilled water five times, and with milli-Q water five times. Preparation of PBS: - In a 1000ml beaker: NazHPO4 — 23g KH2PO4 — 4g KCI - 4g NaCl — 1609 Added 800ml milli-Q water to mix Added ~ 200ml to make 1L - Add solution to large conboy - Add milIi-Q water until 20L - Calibrate pH meter 0 Want pH to be 7.2 - add drops of HCI or NaOH solutions until desired pH attained ‘ 000000 109 Preparation of Bicarbonate solution for dilution of GFP: - Bicarb will last a long time in cabinet- but need to watch out for contamination! - Add 500mL nanopure water to one packet of powdered buffer Preparation of GFP for coating ELISA plates: GFP concentration (from Clonetech) = 1mg/mL = 1000ug/mL 0 Aliquoted into 20uL/ per tube -) 1 tube/ 96-well plate To dilute GFP to coat ELISA plates: Need 100uLper well at a concentration of 1pg/mL 20uLof GFP and add Bicarb to a total volume of 10,000uL(10mL) -Only thaw and dilute GFP to be used that day -Do NOT pre-dilute GFP Control serum samples Dog serum was tested for the best results on varying concentrations of GFP. The serum from 8/25 was combined from both dogs (either + or - control dogs) in a 1:1 mixture and then diluted to 1:25 using wash buffer. The mixture and 1:25 control dilutions were then stored in the freezer. + control = Spruce and Pluto - control = Mica and Snap WASH/ DILUENT solution = 0.05% PBS-T 10L BULK-add 5mL of Tween-20 to 10L of PBS 1 500mL bottle- add .25mL Tween to 500 mL bottle of PBS ELISA Procedure: 1- Coating Plate with GFP: DO THIS 15 HOURS BEFORE OTHER STEPS! a. 100pl/well of GFP- bicarbonate solution b. Place clear, adhesive coverslip on plate c. Incubate overnight at 4°C (in fridge) on a flat surface, do not tip plate 2- Wash Plate: a. Following ovemight incubation at 4°C, dump solution in wells and wash plate with PBS-T i. Pipette 200ul/well of PBS-T — repeat this for a total of 3 rinses- empty wells of solution 3- “Whack” Plate: a. Slam plate upside down onto a stack of paper-towels until paper- towels do not have any moisture on them (plate is essentially dry) 4- Serum Dilutions: a. During 1 hour block, prepare serum dilutions: i. Using a 96 round bottom well plate, pipette 115p|lwell for doubling dilutions into second thru last rows. ii. Pipette 230ul/well of the starting dilution of serum in the first row of wells. 110 iii. Pipette 115ul out of the first row and pipette this into the second row, remembering to mix well before pipetting 115p| into the next row. iv. Continue in this manner to the end of the plate. v. From the last row, pipette 115ul out and discard so that all rows end up with 115p| volume. "Start with 1 :5 dilution“ 230pLin first well: 46uLserum and 184 prash buffer . If using pooled normal dog serum for a negative control, dilution used is 1:25. Use wash buffer to dilute. o If using pooled hyperimmune serum, dilution used is 1:25. Use wash buffer to dilute. . If using rabbit Hyperimmune serum (rabbit anti-GFP from abcam), dilution used is: 1:2500. Use PBS-T to dilute. o If using normal rabbit serum for a negative control, dilution used is 1: 2,500. Use PBS-T to dilute. 5- Add Serum Dilutions: a. Pipette 100pl of serum dilution from round well plate into corresponding well of ELISA plate 6- Incubate: a. Leave for 1 hour at room temperature with vigorous shaking (setting #5 on shaker) 7- Preparation of Secondary Antibodies : Prepare secondary detection antibodies while plate is incubating with serum samples HRP-Anti-Dog: o 100uI/well 0 A dilution of 1/20,000 is used- use PBS-T to dilute 0 1 uU20ml PBS-T HRP-Anti-Rabbit: . 100ul/well 0 A dilution of 1/20,000 is used- use was buffer to dilute. 0 1 uL/20ml wash buffer. 8- Wash and ‘Whack’ plate: a. Following incubation, wash plate with 200ul/well of PBS-T (3X) and ‘wack’ dry on a stack of paper towels 9- Addition of secondary detection antibody: a. Add 100ullwell of the appropriate (dog or rabbit) HRP-conjugated secondary detection antibody 10- Incubation: a. Incubate plate with secondary antibodies for 1 hour at room temperature with vigorous shaking (setting #5 on shaker) 111 11- Wash and ‘Whack’ plate: a. Following incubation, wash plate with 200ul/well of PBS-T (3X) and ‘wack’ dry on a stack of paper towels. 12-Addition of ABTS: a. Add 125uI/well 1-step ABTS and incubate for 1 hour at 37°C without shaking 13-Use Plate Reader to determine absorbance (set filter at 405nm and 450nm) Storage of antibodies/substrates etc: Goat Anti-Rabbit lgG (H+L) a. Caltag - L42007 b. Store at 2-8 °C, dilute only what is needed at a time HRP-Anti-Dog lgG (H+L) c. Bethyl — A40-123P cl. Store at 2-8 °C Rabbit Anti-GFP (Rabbit Polyclonal to GFP) e. Store at 4 °C short term f. Long term: -20 or -80°C - add 1:1 glycerol for stability i. Discussed with Dr. Burton —at 4 °C GFP - 1 mglmL g. Clontech #632373 h. Aliquots of 20u| at -80 °C 1-STEP ABTS for ELISA from Pierce Biotechnology i. Store at 4 °C Serum samples from Subretinal Injection Dogs and Positive and Negative Control from lmmunized Dogs: j. Stored at -80 °C - Plates- BD Falcon 353279 sold through VWR 112 Appendix B. Anti-AAV ELISA Protocol and Summary of References MATERIALS NEEDED: For solutions: -Na2HPO4 -KH2PO4 -KCL -NaCl -Tween (Polyoxyethylene- Sorbitan Monolauvate) -access to milIi-Q water -pH meter -Sterile saline -Probind ELISA plates from VWR For ELISA protocol: -recombinant UF11 virus from Hauswirth Lab, FL -secondary antibody- -HRP— anti-dog lgG -ABTS (acts as the substrate) PREPARATION OF EQUIPMENT AND SOLUTIONS: Cleaning of Conboy: Rinse conboy thoroughly with tap water five times, distilled water five times, and with milIi-Q water five times. Preparation of PBS: - In a 1000m| beaker: NazHPO4 — 23g KHzPO4 - 4g KCI — 4g NaCl - 160g Added 800ml milli-Q water to mix Added ~ 200ml to make 1L - Add solution to large conboy - Add milli-Q water until 20L - Calibrate pH meter 0 Want pH to be 7.2 — add drops of HCI or NaOH solutions until desired pH attained OOOOOO Virus concentration math-) 100.000 vplml Er 100uLIweII Use saline for dilutions 5.6E+13 -) 1:1000 = 5.6E+10 -) 1:1000 = 5.6E+7 [Virus] dilute dilute “vortex between dilutions“ (5.6E+7) X = (4.5ml)x(100,000vp/ml) X= 8.03 vairus in 4.5 ml saline solution 113 Add 100prer each well used for Anti-AAV ELISA WASH/ DILUENT solution = 0.05% PBS-T BULK-add 5mL of Tween-20 to 10L of PBS 1 500mL bottle at a time- add .25mL Tween to 500 mL bottle of PBS ELISA Procedure: 1 Coating Plate with virus: DO THIS 15 HOURS BEFORE OTHER STEPS! a. 100uI/well of virus-saline solution b. Place clear, adhesive coverslip on plate c. Incubate overnight at 4°C (in fridge) on a flat surface, do not tip plate Wash Plate: a. Following ovemight incubation at 4°C, pour off solution from wells and wash plate with PBS-T i. Pipette 200pI/well of PBS-T - repeat this for a total of 3 rinses- empty wells of solution “Whack” Plate: a. Slam plate upside down onto a stack of paper-towels until paper- towels do not have any moisture on them (plate is essentially dry) 4- Serum Dilutions: a. During 1 hour block, prepare serum dilutions- use 1:5 dilutions for all samples: i. 115/5= 23 uL ii. 23pLserum and 92uLwash buffer per well in round bottom plate iii. Aliquot 100uLinto each well 2 w I Control Dog Serum - If using pooled normal dog serum for a negative control, dilution used is 1:25. Use wash buffer to dilute. . If using pooled hyperimmune serum, dilution used is 1:25. Use wash buffer to dilute. 5- Add Serum Dilutions: a. Pipette 100u| of serum from round well plate into corresponding well of ELISA plate 6- Incubate: a. Leave for 1 hour at room temperature with vigorous shaking (setting #5 on shaker) 7- Preparation of Secondary Antibodies : Prepare secondary detection antibodies while plate is incubating with serum samples 114 HRP-Anti-Dog: . 100pllwe|| o A dilution of 1/20,000 is used- use PBS-T to dilute o 1 W 20ml PBS-T 8- Wash and ‘Whack’ plate: a. Following incubation, wash plate with 200pI/well of PBS-T (3X) and ‘wack’ dry on a stack of paper towels 9- Addition of secondary detection antibody: a. Add 100uI/well of the appropriate HRP—conjugated secondary detection antibody 10- Incubation: a. Incubate plate with secondary antibodies for 1 hour at room temperature with vigorous shaking (setting #5 on shaker) 11-Wash and ‘Whack’ plate: a. Following incubation, wash plate with 200uI/well of PBS-T (3X) and ‘wack’ dry on a stack of paper towels. 12-Addition of ABTS: a. Add 125ullwell 1-step ABTS and incubate for 1 hour at 37°C without shaking 13-Use Plate Reader to determine absorbance (set filter at 405nm and 450nm) 115 Appendix C. AAV2I5-GFP Vector Construction AAV2/5 Plasmid Construction The AAV2 vector pseudotyped with the capsid from AAV5 was produced in the laboratory of Dr. Hauswirth at the University of Florida. An adenovirus helper plasmid was used to pseudotype the vector with the AAV5 capsid. Adenovirus Helper Plasmid Production The creation of the adenovirus helper plasmid with AAV5 capsid was performed in a series of steps. The open reading frame (ORF) of AAV5 was amplified with PCR and was then subcloned into the plasmid pACGZ resulting in the hybrid plasmid pACG2R5C which contains the ORF for the AAV2 Rep proteins and the ORF for the AAV5 capsid proteins. A fragment from the pACGZRSC plasmid was subcloned into the pXYZ adenovirus helper plasmid to create the pXYZS plasmid complete with the AAV5 capsid gene (Zolotukhin et al., 2002). AAV Vector Plasmid Production The AAV vector plasmid was constructed using the pTR-UF backbone and consists of AAV2 flanking inverted terminal repeat (ITR) regions (Zolotukhin et al., 2002). Two different AAV vector plasmids were used in this study to create two different AAV2/5 vectors. The first AAV vector consisted of the chicken beta actin promoter, CMV enhancer regions, and GFP transgene. The second AAV vector consisted of the mouse opsin promoter and GFP transgene (Glushakova etaL,2006) 116 Promoters Chicken Beta Actin (CBA) The CBA promoter is a ubiquitous promoter that allows a constant and powerful expression of the transgene. This promoter is active in any host cell and thus transcribes the transgene in any cell of the body. 1 51 101 151 201 251 catggtcgag cctccccacc gcgatggggg 9990939999 agagcggcgc cggccctata gtgagcccca cccaattttg ngggggggg cggggcgggg gctccgaaag aaaagcgaag cgttctgctt tatttattta gggggggcgc cgaggcggag tttcctttta cgcgcggcgg cactctcccc atctcccccc ttttttaatt attttgtgca gcgccaggcg gggcggggcg aggtgcggcg gcagccaatc tggcgaggcg gcggcggcgg 9C9 Figure A. The CBA promoter sequence (provided by the Hauswirth Lab). This sequence was Blasted (RID: 1171314962-8045-14060912295.BLASTQ3, 2/12/07) with hits found only in the chicken genome. Mouse Opsin (mOPSSOO) The mOP8500 promoter is a tissue-specific promoter present in some of the AAV vectors that was characterized by the Hauswirth laboratory (Glushakova et al., 2006). The promoter was shown to be specific for photoreceptors and thus the likelihood that the transgene would be expressed outside the eye is low. 1 51 101 151 201 251 301 351 401 451 cgtcaagtga tcctgggaag gagccattgt tgaggccacc gacaagatga gtcaccttgg attagcgtta 9939393399 gcgctgtggg gtctacgaag gccattgtca agatgggata ggagaagtga agactgacat gacacccttt cccctctgca gtatgatatc tcactttata agccgtcagt agcccgtggg gggcttgggg ggtgagttca atttagggcc ggggaggaaa cctttcttta agccaattag tcgcggatgc agggtctggg ggctgagctc gcagcct actggataag ggaggagaca caagggttcc tcccagagga cctaagggcc gccccggtgg tgaatcagcc gggggtcagt gccaagcagc tcagggggtc ttgtcaactg agtcgcagcc ctctggggca tccacccgat cagcagtggg tctggcttag gcctggagtt cttggtctct Figure B. The mOPS500 promoter sequence (provided by the Hauswirth Lab). This sequence was blasted (RID: 1171314867-26111-185748596015.BLASTQ3, 2/12/07) with hits only found in the mouse genome on chromosome 6. 117 The Green Fluorescent Protein (GFP) Transgene The GFP transgene is a reporter gene commonly used to determine the efficacy of the gene transfer event. GFP originates from the jellyfish, Aequorea Victoria, and fluoresces a bright green color when under a UV or blue light and has been shown to be effective in many species and systems. 1 51 101 151 201 251 301 351 401 451 501 551 601 651 701 caccatgagc tggaactgga ggtgaaggtg cactggaaag atggcgtgca tttttcaaga tttcaaagat gtgacaccct gatggaaaca tgtgtacatc agatcagaca caacagaaca ttacctgtcc accacatggt atggacgagc aagggcgagg tggcgatgtg atgccacata ctccctgtgc gtgcttttcc gcgccatgcc gacgggaact ggtgaataga ttctcggcca atggccgaca caacattgag ctccaatcgg acccagtctg cctgctggag tgtacaagtg aactgttcac aatgggcaca cggaaagctc catggccaac agatacccag cgagggctat acaagacccg atcgagctga caagctggaa agcaaaagaa gatggatccg cgacggccct ccctgtctaa tttgtgaccg agc tggcgtggtc aattttctgt accctgaaat actggtcact accatatgaa gtgcaggaga cgctgaagtc agggcattga tacaactata tggcatcaag tgcagctggc gtgctcctcc agatcccaac ctgctgggat ccaattctcg cagcggagag tcatctgcac accctgacct gcagcatgac gaaccatctt aagttcgaag ctttaaggag actcccacaa gtcaacttca cgaccattat cagacaacca gaaaagagag cacacatggc Figure C. The GFP cDNA sequence used in the AAV viral vector constructs (provided by the Hauswirth Lab). Protocol for AAV2/5 vector production Cell transfection 293 cells (ATCC, Manassas, VA) were used for the transfection with the above plasmid constructs. The cells were cultured with Dulbecco’s modified Eagle’s medium (DMEM) which was supplemented with 5% fetal bovine serum (FBS). Cells were split 1:3 one day prior to transfection and were allowed to reach ~75- 80% cell confluency. The plasmid is CsCI-purified and was then transfected using a CaPO4 precipitate. 1.8mg of pXY25 and 0.6mg of AAV vector plasmid in a total volume of 50mL of 0.25M CaClz followed by the addition of 50mL of 2x 118 HBS. pH7.05, to the DNA/CaClz (Zolotukhin et al., 2002). After 1-2 minutes of room temperature incubation, 1100mL of warm DMEM with 5% PBS was added lPstI(1) ' SrmI(87) ,TR ./ f1(+) origin , //Srm1(98) \wmi Id <~\.;\\” AEnRI(171) [5"‘5‘A'_I:’W;;»..::L ’- ' \\\.L\\¥\ 54"” > Iqm [(181) ._ k . / >4 CMV Ie enhancer \\ Chiken b-actin promoter ApR/-, \ Exon1 \\ ,5 ‘r, Intmn r2 pTR-UF11 \ Q L I — \\\\\\ XbaI(1798) W 72oobp ._ NKEDRI(1894) PS? I (4370) - ,'- , i' ‘ "NotI (1922) 81111101280) \r: GFPh TR‘ . ~’ ‘Bamm(2455) SnuI(4269) // ~. . \ \.\ NotI(2654) bGH po|y(A) \ z i, \ BamI-II(2663) SaII(3960) ‘1‘ \ swo poly(A) BamHI(3954) 1, \ .\ S011 (2861) 1 ‘ ‘1, \ ‘._ i]: PYF441 enhancer 1‘ “\ EDRI(3044) i ' ' HSV-tk I “PstI(3141) ‘PstI(3338) neoR Figure D. The pTR-UF11 plasmid with CMV enhancer, CBA promoter, and GFP transgene (Vince Chiodo, Hauswirth Lab, University of Florida). 119 The medium was removed from the cells and then the plasmid solution was added to the cells and allowed to incubate for 60h at 37°C at 5% C02. Cells were then washed with PBS and then harvested. The cells then undenNent centrifugation for 10 minutes at 10009, were resuspended in 60mL of lysis solution (150mM NaCl, 50mM Tris, pH 8.4), and then stored at -20°C (Zolotukhin etaL,2002) AAV2/5 Purification Purification of the AAV5 vector occurred through a series of steps: cell lysis, iodixanol gradients, and column chromatography. Cells were lysed using a freeze/thaw method that involved dry ice in ethanol and a 37°C water bath. Benzonase (Sigma, St. Louis, MO) was then added to the cell Iysate and incubated for 30 minutes (Zolotukhin et al., 2002). The lysate was then centrifuged for 30 minutes at 40009 in order to clarify the crude Iysate. The supernatant was then divided 4 ways into iodixanol gradients. Iodixanol gradients are created by using quick-seal tubes (25 x 89 mm, Beckman) and iodixanol (5,5’-[(2-hydroxy-1,3-propanedin)bisacetyI-amino)] bis[N,N’-bis (2,3dihydroxypropyI-2,4,6-triiodo-1,3-benzenecarboxamide] using a 60% sterile solution of OptiPrep (Nycomed) and PBS-MK buffer (1 x PBS containing 1mM MgCIz and 2.5mM KCI). The cell lysate solution was added then centrifuged for 1 hour at 18C at 69,000rpm (Zolotukhin et al., 2002). 5mL of the 120 60-40% step interface was aspirated with an 189 needle, and the bands of the gradient were stored either in the freezer or refrigerator (Zolotukhin et al., 2002). Column chromatography of the iodixanol gradient further purifies the vector. A 4- mL HiTrap Q column (Pharmacia) was used. First, the column was equilibrated at 5mL/min with 5 column volumes of Buffer A (20mM Tris, 15mM NaCl, pH 8.5) followed by 25mL of Buffer A using the Pharrnacia ATKA FPLC system (Zolotukhin et al., 2002). The iodixanol fraction was diluted 1:1 with Buffer A and then added to the column at a rate of 3-5 ml/minute. The column was then washed with 10 column volumes (50mL) of Buffer A. The vector was eluted with Buffer B (Zolotukhin et al., 2002). AAV2/5 Concentration The vector was concentrated through a three cycle-centrifugation method involving a Biomax 100 K concentrator (Millipore, Bedford, MA). The virus is concentrated to 1 mL and is then stored at -8OC in 1 x PBS (Zolotukhin et al., 2002) AAV2/5 Quality Control Assays Quality control measures are taken using an assay to determine the protein purity of the AAV2/5, another assay for the infectiousness, and then a dot-blot assay for the physical particle titer (Zolotukhin et al., 2002). The protein purity of the AAV2/5 vector was determined by running a 10% SDS-polyacrylamide gel with silver staining. A western blot was then done using an anti-AAV2 capsid 121 monoclonal antibody B1 (American Research Products) that recognizes AAV5 capsids at a dilution of 1:2000. A horseradish peroxidase-conjugated sheep anti- mouse (Amersham) was used for detection at a dilution of 1:5000 and Super Signal (Pierce, Rockford, IL) was used as the substrate (Zolotukhin et al., 2002). The infectious center assay (ICA) was used to determine the infectiousness of the vector. 96-well plates were coated with 2 x 104 C12 cells and infected 16 hours after coating using 10-fold dilutions of the AAV and also infected with adenovirus 5 at a multiplicity of infection of 10 (Zolotukhin et al., 2002). The C12 cells were then harvested and suspended in 5 mL of 1 x PBS. The cells are then vacuum filtered onto nylon membranes (0.45pm) which were then transferred to filter paper moistened with 0.5 N NaOH/1.5 M NaCl and 1M Tris-Cl. The filter paper was then probed for the transgene. Titer was calculated by using the dilution factor from the infectious centers (spots) observed on the filter paper, accurate in the range of 10-200 spots (Zolotukhin et al., 2002). Another assay was used to determine the infectiousness of the AAV2/5 vectors that have the GFP transgene. The single-cell fluorescence assay (SCFA) determined the vector titer based on the expression of GFP. 2 x 104 293 or 012 cells are seeded into a 96 well plate. Serial dilutions of the AAV2/5 and Ad5 (MOI of 10) were added to wells. After thirty hours, cells infected with the GFP vector were visually scored using a fluorescent microscope. Titer is calculated based on the dilution factor (Zolotukhin et al., 2002). 122 The last quality assurance assay used is the dot-blot assay. This assay determines the number of vectors with a viral genome. DNase I (Roche) was used to digest all plasmid and unpackaged vector DNA for 1 hour at 37°C in a volume of 200pL containing 5U of the enzyme, 10mM Tris-Cl, pH 7.5, and mM MgCIz. Then, in order to free the viral genomes from the capsids, an equal volume of proteinase K buffer (20mM Tris-Cl, pH 8.0, 20mM EDTA, pH 8.0, 1% SDS) was added as well as proteinase K (30ug). This solution was incubated for 1 hour at 37°C. The DNA was then extracted using phenol and precipitated using ethanol and a glycogen carrier (Zolotukhin et al., 2002). DNA was then dissolved in 40pL of distilled water. 10pL of this was diluted in 400pL 0.4 N NaOH/10mM EDTA. A twofold dilution series was prepared from the plasmid DNA as above. The viral DNA was immobilized onto a nylon membrane. The plasmid DNA was also immobilized onto the membrane using a dot-blot apparatus (Bio-Rad, Hercules, CA). The membrane was then probed for the transgene and exposed to film. Comparing the plasmid DNA standard curve to the vector DNA allows for extrapolation of the viral titer. 123 BIBLIOGRAPHY Acland, G. M., Aguirre, G. D., Ray, J., Zhang, 0., Aleman, T. S., Cideciyan, A. V., Pearce-Kelling, S. E., Anand, V., Zeng, Y., Maguire, A. M., Jacobson, S. G., Hauswirth, W. W. & Bennett, J. (2001) Gene therapy restores vision in a canine model of childhood blindness. Nature Genetics. 28, 92-95 Acland, G. M., Aguirre, G. D., Bennett, J., Aleman, T. S., Cideciyan, A. V., Bennicelli, J., Dejneka, N. S., Pearce-Kelling, S. E., Maguire, A. M., Palczewski, K., Hauswirth, W. W., Jacobson, S. G. Long-Term Restoration of Rod and Cone Vision by Single Dose rAAV-Mediated Gene Transfer to the Retina In a Canine Model of Childhood Blindness. (2005) Molecular Therapy 10,1-11. Aguirre, G. D., Rubin, L. F. & Bistner, S. l. (1998) Congenital stationary night blindness in the dog: common mutation in the RPE65 gene indicates founder effect. Molecular Vision. 4, 23. Ali, R. R., Sarra, G. M., Stephens, C., Alwis, M. D., Bainbridge, J. W. B., Munro, P. M., Fauser, S., Reichel, M. B., Kinnon, 0., Hunt, D. M., Bhattacharya, S. S. & Thrasher, A. J. (2000) Restoration of photoreceptor ultrastmcture and function in retinal degeneration slow mice by gene therapy. Nature Genetics. 25, 306-310 Ali, R. R., Reichel, M., De Alwis, M., Kanuga, N., Kinnon, C., Levinsky, R., Hunt, D. M., Bhattacharya, S. S., Thrasher, A. J. Adeno-Associated Virus Gene Transfer to Mouse Retina. (1998) Human Gene Therapy 9, 81-86. American Society of Gene Therapy. Serious Adverse Event in a Clinical Trial of Gene Therapy for the X-Linked Form of Severe Combined Immune Deficiency Disease(XSCID). (2002) American Society of Gene Therapy Anand, V., Walsh, C. E., Fersh, M., Maguire, A. M., Bennett, J. Additional Transduction Events after Subretinal Readministration of Recombinant Adeno-Associated Virus. (2000) Human Gene Therapy, 449-457. Anand, V., Duffy, B., Yang, 2., Dejneka, N. S., Maguire, A. M. & Bennett, J. (2002) A Deviant Immune Response to Viral Proteins and Transgene Product is Generated on Subretinal Administration of Adenovirus and Adeno-Associated Virus. Molecular Therapy. 5, 125-132 124 Arruda, V., Fields, P. A., Milner, R., Wainwright, L., De Miguel, M. P., Donovan, P. J., Herzog, R. W., Nichols, T. C., Biegel, J. A., Razavi, M., Dake, M., Huff, D., Flake, A. W., Couto, L., Kay, M. A. & High, K. A. (2001) Lack of germline transmission of vector sequences following systemic administration of recombinant AAV-2 vector in males. Molecular Therapy. 4, 586-592 Auricchio, A., Rivera, V. M., Clackson, T., O' Connor, E., Maguire, A. M., Tolentino, M. J., Bennett, J. & Wilson, J. M. (2002) Pharmacological Regulation of Protein Expression from Adeno-Associated Viral Vectors in the Eye. Molecular Therapy. 6, 238-242 Auricchio, A., Kobinger, G., Anand, V., Hildinger, M., O'Connor, E., Maguire, A. M., Wilson, J. M., Bennett, J. Exchange of surface proteins impacts on viral vector cellular specificity and transduction characteristics: the retina as a model. (2001) Human Molecular Genetics 10, 3075-3081. Auricchio, A., Rolling, F. Adeno-Associated Viral Vectors for Retinal Gene Transfer and Treatment of Retinal Diseases. (2005) Current Gene Therapy 5, 339-348. Bainbridge, J., Mistry, A., Schlichtenbrede, FC., Smith, A., Broderick, C., De Alwis, M., Georgiadis, A., Taylor, PM., Squires, M., Sethi, C., Charteris, D., Thrasher, AJ., Sargan, D. 8 Ali, RR. (2003) Stable rAAV-mediated transduction of rod and cone photoreceptors in the canine retina. Gene Therapy. 10, 1336-1344 Baldridge, W. H. & Kurennvi, D. E. (1998) Calcium-sensitive calcium influx in photoreceptor inner segments. Journal of Neurophysiology. 79, 3012-3018 Bemelmans, A.-P., Bonnel, 8., Houhou, L., Dufour, N., Nandrot, E., Helminger, D., Sarkis, C., Abitbol, M., Mallet, J. Retinal cell type expression specificity of HIV-1-derived gene transfer vectors upon subretinal injection in the adult rat: influence of pseudotyping and promoter. (2005) The Journal of Gene Medicine 7, 1367-1374. Bemelmans, A.-P., Kostic, C., Crippa, S. V., Hauswirth, W. W., Lem, J., Munier, F. L., Seeliger, M. W., Wenzel, A. & Arsenijevic, Y. (2006) Lentiviral gene transfer of RPE65 rescues survival and function of cones in a mouse model of Leber Congenital Amaurosis. Public Library of Science, Medicine. 3, 1-26 Bennett, J., Wilson, J. M., Sun, D., Forbes, B. & Maguire, A. M. (1994) Adenovirus Vector-Mediated In Vivo Gene Transfer Into Adult Murine Retina. Investigative Ophthalmology and Visual Science. 35, 2535-2542 125 Bennett, J., Tanabe, T., Sun, D., Zeng, Y., Kjeldbye, H., Gouras, P. & Maguire, A. M. (1996) Photoreceptor cell rescue in retinal degeneration (rd) mice by in vivo gene therapy. Nature Medicine. 2, 649-654 Bennett, J., Maguire, A. M., Cideciyan, A. V., Schnell, M., Glover, E., Anand, V., Aleman, T. 8., Chirrnule, N., Gupta, A. R., Huang, Y., Gao, G.-P., Nyberg, W. C., Tazelaar, J., Hughes, J., Wilson, J. M., Jacobson, S. G. Stable transgene expression in rod photoreceptors after recombinant adeno- associated virus-mediated gene transfer to monkey retina. (1999) Proc. Natl.Acad. Sci 96, 9920-9925. Bessant, D. A., Ali, R. R. & Bhattacharya, S. S. (2001) Molecular genetics and prospects for therapy of the inherited retinal dystrophies. Current Opinion in Genetics & Development. 1 1, 307-316 Bundey, S. & Crews, S. J. (1984) A study of retinitis pigmentosa in the city of Birmingham- I. Prevalance. Journal of Medical Genetics. 21, 417-420 Carter, E. W. & Kerr, D. E. (2003) Optimization of DNA-based Vaccination in Cows Using Green Fluorescent Protein and Protein A as a Prelude to Immunization against Staphylococcal Mastitis. Journal of Dairy Science. 86, 1177-1186 Cavazzana-Calvo, M., Hacein-Bey, S., de Saint Basile, G., Gross, F., Yvon, E., Nusbaum, P., Selz, F., Hue, 0., Certain, S., Casanova, J.-L., Bousso, P., Le Desit, F., Fischer, A. Gene Therapy of Human Severe Combined Immunodeficiency (SCID)—X1 Disease. (2000) Science 288, 669-672. Cavazzana-Calvo, M., Lagresle, C., Hacein-Bey-Abina, S. & Fischer, A. (2005) Gene Therapy for Severe Combined Immunodeficiency. Annual Review of Medicine. 56, 585-602 Chader, G. J. (2002) Animal models in research on retinal degenerations: past progress and future hope. Vision Research. 42, 393-399 Chao, H. & Walsh, C. J. (2004) AAV Vectors for Hemophilia B Gene Therapy. The Mount Sinai Journal of Medicine. 71, 305-313 Chen, Y., Moiseyev, G., Takahashi, Y. & Ma, J.-X. (2006) RPE65 Gene Delivery Restores lsomerohydrolase Activity and Prevents Early Cone Loss in RPE65-I- Mice. Investigative Ophthalmology and Visual Science. 47, 1 177-1 184 Connelly S. Adenoviral Vectors. Anthony Meager. Gene Therapy Technologies, Applications, and Regulations. [5], 87-107. 1999. West Sussex, John Wiley and Sons Ltd. 126 Cremers, F. P. M., van den Hurk, J. A. J. M. & den Hollander, A. I. (2002) Molecular genetics of Leber congenital amaurosis. Human Molecular Genetics. 11 , 1 169-1 176 Cronin, J., Zhang, X.-Y. & Reiser, J. (2005) Altering the Tropism of Lentiviral Vectors through Pseudotyping. Current Gene Therapy. 5, 387-398 D'Cruz, P. M., Yasumura, D., Weir, J., Matthes, M. T., Abderrahim, H., La Vail, M. M. & Vollrath, D. (2000) Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Human Molecular Genetics. 9, 645-651 Dejneka, N. 8., Rex, T. 8. & Bennett, J. (2003) Gene Therapy and Animal Models for Retinal Disease. Developmental Ophthalmology. 37, 188-198 Dudus, L., Anand, V., Acland, G. M., Chen, 8.-J., Wilson, J. M., Fisher, K. J., Maguire, A. M., Bennett, J. Persistent transgene product in retina, optic nerve and brain after intraocular injection of rAAV. (1999) Vision Research 39, 2545-2553. Everett, R. 8., Evans, H. K., Hodges, B. L., Ding, E. Y., Serra, D. M. & Amalfitano, A. (2004) Strain-specific rate of shutdown of CMV enhancer activity in murine liver confirmed by use of persistent [E1-, E2b-] adenoviral vectors. Virology. 325, 96-105 Favre, D., Provost, N., Blouin, V., Blancho, G., Cherel, Y., Salvetti, A. & Moullier, P. (2001) Immediate and Long-Term Safety of Recombinant Adeno- associated Virus Injection into the Nonhuman Primate Muscle. Molecular Therapy. 4, 559-566 Flannery, J. G., Zolotukhin, 8., Vaquero, M. l., LaVail, M., Muzyczka, N., Hauswirth, W. W. Efficient photoreceptor-targeted gene expression in vivo by recombinant adeno-associated virus. (2005) Proceedings of the National.Academy of Science. 94, 6919-6921 . Flint SJ, Enquist LW, Racaniello VR, Skalka AM. Principles of Virology. 2nd. 2004. Washington, DC, American Society for Microbiology. Ford, M., Bragadottir, R., Rakoczy, P. E. & Narfstrom, K. (2003) Gene transfer in the RPE65 null mutation dog: relationship between construct volume, visual behavior and electroretinographic (ERG) results. Ford, M. Documenta Ophthalmoligica. 107, 79-86 Forrester J, Dick A, McMenamin P, Lee W. The Eye: Basic Sciences in Practice. 1996. London, Saunders. 127 Gal, A., Li, Y., Thompson, D. A., Weir, J., Orth, U., Jacobson, S. G., Apfelstedt, E., Vollrath, 8. & Vollrath, D. (2000) Mutations in MERTK, the human orthologue of the RC8 rat retinal dystrophy gene, cause retinitis pigmentosa. Nature Genetics. 26, 270-271 Glushakova, L., Timmers, A. M., lssa, T. M., Cortez, N. G., Pang, J., Teusner, J. T. & Hauswirth, W. W. (2006) Does recombinant adeno-associated virus- vectored proximal region of mouse rhodopsin promoter support only rod- type specific expression in vivo? Molecular Vision. 12, 298-302 Grant, C. A., Ponnazhagan, 8., Wang, X.-8., Srivastava, A. & Li, T. (1997) Evaluation of recombinant adeno-associated virus as a gene transfer vector for the retina. Current Eye Research.949-956 Gruter, 0., Kostic, C., Crippa, 8. V., Perez, M.-T. R., Zografos, L., Schorderet, D. F., Munier, F. L. & Arsenijevic, Y. (2005) Lentiviral vector-mediated gene transfer in adult mouse photoreceptors is impaired by the presence of a physical barrier. Gene Therapy. 12, 942-947 Haase, W., Friese, W., Gordon, R. D., Muller, H. & Cook, N. J. (1990) Immunological characterization and localization of the Na+/Ca2(+)- exchanger in bovine retina. Journal of Neuroscience. 10, 1486-1494 Hacein-Bey-Abina, 8., von Kalle, C., Schmidt, M., McConnack, MP., Wulffraat, N., Leboulch, P., Lim, A., Osborne, C8., Pawliuk, R., Morillon, E., Sorensen, R., Forster, A., Fraser, P., Cohen, Jl., de Saint Basile, G., Alexander, |., Wintergerst, U., Frebourg, T., Aurias, A., Stoppa-Lyonnet, D., Romana, 8., Redford-Weiss, l., Gross, F., Valensi, F., Delabesse, E., Macintyre, E., Sigaux, F., Soulier, J., Leiva, LE., Wissler, M., Prinz, C., Rabbitts, TH., Le Deist, F., Fischer, A. & Cavazzana-Calvo, M. (2003) LMOZ-associated clonal T cell proliferation in two patients after gene therapy for SClD-X1 . Science. 302, 415-419 Hack, |., Peichl, L. & Brandstatter, J. H. (1999) An alternative pathway for rod signals in the rodent retina: rod photoreceptors, cone bipolar cells, and the localization of glutamate receptors. Proceedings of the National.Academy of Science. 96, 14130-14135 Hamilton, M., Brough, D. E., McVey, D., Bruder, J. T., King, C. R. & Wei, L. I. (2006) Repeated Administration of Adenovector in the Eye Results in Efficient Gene Delivery. Investigative Ophthalmology and Visual Science. 47, 299-305 Hartogs P. Gene therapy researchers defend trial after death of patient. CNN . 1 999. Heckenlively, J. R., Foxman, 8. G. & Parellhoff, E. S. (1988) Retinal dystrophy and macular coloboma. Documenta Ophthalmologica. 68, 257-271 128 Hoffman, L. M., Maguire, A. M. & Bennett, J. (1997) Cell-Mediated Immune Response and Stability of Intraocular Transgene Expression After Adenovirus-Mediated Delivery. Investigative Ophthalmology and Visual Science. 38, 2224-2233 Huang, 8. H., Pittler, S. J., Huang, X., Oliveira, L., Berson, E. L. & Dryja, T. P. (1995) Autosomal recessive retinitis pigmentosa caused by mutations in the alpha subunit of rod cGMP phosphodiesterase. Nature Genetics. 11, 468-470 Isenmann, 8., Engel, 8., Kugler, 8., Gravel, C., Weller, M. & Bahr, M. (20061) lntravitreal adenoviral gene transfer evokes an immune response in the retina that is directed against the heterologous IacZ transgene product but does not limit transgene expression. Brain Research. 892, 229-240 Jacobson, S. G., Bove, S. L., Aleman, T. 8., Conlon, T. J., Zeiss, C. J., Roman, A. J., Cideciyan, A. V., Schwartz, 8. B., Komaromy, A. M., Doobraih, M., Cheung, A. Y., Sumaroka, A., Pearce-Kelling, 8. E., Aguirre, G. D., Kaushal, 8., Maguire, A. M., Flotte, T. R. & Hauswirth, W. W. (2006) Safety in nonhuman primates of ocular AAV2-RPE65, a candidate treatment for blindness in Leber congenital amaurosis. Human Gene Therapy. 17, 845-858 Janeway CA, Travers P, Walport M, Schlomchik MJ. Immunobiology. 6th. 2005. New York, Garland Science. Jomary, C., Vincent, K., Gn’st, J., Neal, M. & Jones, 8. (1997) Rescue of photoreceptor function by AAV-mediated gene transfer in a mouse model of inherited retinal degeneration. Gene Therapy. 4, 683-690 Kay, M. A., Glorioso, J. C. 8 Naldini, L. (2001) Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics. Nature Medicine. 7, 33-40 Koch, S. A. & Rubin, L. F. (1972) Distribution of cones in retina of the normal dog. American Journal of Veterinary Research. 33, 361 -363 Kotin, R. M., Siniscalco, M., Samulski, R. J., Zhu, X., Hunter, L., Laughlin, C. A., McLaughlin, 8., Muzyczka, N., Rocchi, M., Bems, K. I. Site-specific integration be adeno-associated virus. (1990) Proceedings of the National.Academy of Science. 87, 221 1-2215. Kumar-Singh, R., Farber, D. B. Encapsidated adenovirus mini-chromosome- mediated delivery of genes to the retina: application to the rescue of photoreceptor degeneration. (1998) Human Molecular Genetics 7, 1893- 1 900. 129 Lai, C.-M., Yu, M. J., Brankov, M., Barnett, N. L., Zhou, X., Redmond, T. M., Narfstrom, K., Rakoczy, P. E. Recombinant adeno-associated virus type 2-mediated gene delivery into the Rpe65-I- knockout mouse eye results in limited rescue. (2004) Genetic Vaccines and Therapy 2, 1-15. Le Meur, G., Weber, M., Pereon, Y., Mendes—Madeira, A., Nivard, D., Deschamps, J.-Y., Moullier, P. & Rolling, F. (2005) Postsurgical Assessment and Long-tenn Safety of Recombinant Adeno-Associated Virus-Mediated Gene Transfer Into the Retinas of Dogs and Primates. Archives of Ophthalmology. 123, 500-506 Lever AML. Lentiviral Vectors. A.Meager. Gene Therapy Technologies, Applications, and Regulations. [4], 61-86. 1999. West Sussex, Wiley. Lewin, A. 8., Drenser, K. A., Hauswirth, W. W., Nishikawa, 8., Yasumura, D., Flannery, J. G. 8 La Vail, M. M. (1998) Ribozyme rescue of photoreceptor cells in a transgenic rat model of autosomal dominant retinitis pigmentosa. Nature Medicine. 4, 967-971 Li, X., Shen, 8., Urso, D., Kalique, 8., Park, S.H., Sharafieh, R., O’Rourke, J., Cone, RE. (2006) Phenotypic and immunoregulatory characteristics of monocytic iris cells. Immunology. 117, 566-575. Loewen, N., Leske, D. A., Cameron, J. D., Chen, Y., Whitwam, T., Simari, R. D., Teo, W.-L., Fautsch, M. P., Poeschla, E. M., Holmes, J. M. Long-term retinal transgene expression with FIV versus adenoviral vectors. (2004) Molecular Vision 1 0, 272-280. Lotery, A. J., Yang, G. 8., Mullins, R. F., Russell, 8. R., Schmidt, M., Stone, E. M., Lindbloom, J. D., Chiorini, J. A., Kotin, R. M. & Davidson, B. L. (2003) Adeno-Associated Virus Type 5: Transduction Efficiency and Cell-Type Specificity in the Primate Retina. Human Gene Therapy. 14, 1663-1671 Lu, X., Humeua, L., Slepushkin, V., Binder, 6., Yu, 0., Slepushkina, T., Chen, Z., Merling, R., Davis, B., Chang, Y., Dropulic, B. Safe two-plasmid production for the first clinical lentivirus vector that achieves >99% transduction in primary cells using a one-step protocol (2004) The Journal of Gene Medicine. 6, 963-973. Mallam, J. N., Hurwitz, M. Y., Mahoney, T., Chevez—Barrios, P., Hunrvitz, R. I. Efficient Gene Transfer into Retinal Cells Using Adenoviral Vectors: Dependence on Receptor Expression. (2004) Investigative Ophthalmology and Visual Science. 45, 1680-1687. ' 130 Manno, C. 8., Chew, A. J., Hutchinson, 8., Larson, P. J., Herzog, R. W., Arruda, V. R., Tai, S. J., Ragni, M. V., Thompson, A., Ozelo, M., Couto, L. B., Leonard, D. G., Johnson, F. A., McClelland, A., Scallan, C., Skarsgard, E., Flake, A. W., Kay, M. A., High, K. A. & Glader, B. (2002) AAV-mediated factor IX gene transfer to skeletal muscle in patients with severe hemophilia B. Blood. 101, 2963-2972 Mata, N. L., Moghrahi, W. N., Lee, J. 8., Bui, T. V., Radu, R. A., Honrvitz, J. & Travis, G. H. (2004) RPE65 Is a Retinyl Binding Protein That Presents Insoluble Substrate to the lsomerase in Retinal Pigment Epithelial Cells. The Journal of Biological Chemistry. 279, 635-643 Miyoshi, H., Takahashi, M., Gage, F. H., Verrna, I. M. Stable abd efficient gene transfer into retina using an HIV-based Ientiviral vector. (1997) Proceedings of the National.Academy of Science. 94, 10319-10323. Morimura, H., Fishman, G. A., Grover, 8. A., Fulton, A. B., Berson, E. L. & Dryja, T. P. (1998) Mutations in the RPE65 gene in patients with autosomal recessive retinitis pigmentosa or Leber congenital amaurosis. Proceedings of the National.Academy of Science. 95, 3088-3093 Muruve, D. A. (2004) Helper-dependent Adenovirus Vectors Elicit Intact Innate but Attenuated Adaptive Host Immune Responses In Vivo. Virology. 78, 5966-5972 Naldini, L., Blomer, U., Gallay, P., Ory, D., Mulligan, R., Gage, F. H., Verrna, I. M. & Trono, D. ( 1996) In Vivo Gene Delivery and Stable Transduction of Nondividing Cells by a Lentiviral Vector. Science. 272, 263-267 Naldini, L., Blomer, U., Gage, F. H., Trono, D. & VemIa, I. M. (2006) Efficient transfer, integration, and sustained Iong-terrn expression of the transgene in adult rat brains injected with a Ientiviral vector. Proceedings of the National.Academy of Science. 93, 1 1382-1 1388 Narfstrom, K., Katz, M. L., Ford, M., Redmond, T. M., Rakoczy, E. & Bragadottir, R. (2003) In Vivo Gene Therapy in Young and Adult RPE65-I- Dogs Produces Long-Term Visual Improvement. Journal of Heredity. 94, 31-37 Narfstrom K. Retinal Degeneration in a Strain of Abyssinian Cats. 1985. Linkoping. Narfstrom, K., Katz, M. L., Bragadottir, R., Seeliger, M., Boulanger, A., Redmond, T. M., Caro, L., Lai, C.-M. 8r Rakoczy, P. E. (2003) Functional and Structural Recovery of the Retina after Gene Therapy in the RPE65 Null Mutation Dog. Investigative Ophthalmology and Visual Science. 44, 1663- 1672 131 Pang, J., Chang.Bo, Kumar, A., Nusinowitz, 8., Noorwez, S. M., Li, J., Rani, A., Foster, T. C., Chiodo, V. A., Doyle, T., Li, H., Malhotra, R., Teusner, J. T., McDowell, J. H., Min, S.-H., Li, Q., Kaushal, S. & Hauswirth, W. W. (2006a) Gene Therapy Restores Vision-Dependent Behavior as Well as Retinal Structure and Function in a Mouse Model of RPE65 Leber Congenital Amaurosis. Molecular Therapy. 13, 565-571 Pang, J., Chang, M., Haire, S. E., Barker, E., Planelles, V. 8 Blanks, J. C. (2006b) Efficiency of Ientiviral transduction during development in normal and rd mice. Molecular Vision. 12, 756-767 Petersen-Jenes, S. M., Entz, D., Sargan, D. R. PDE6A Mutation Causes PRA in Cardigan Welsh Corgis. ( 1999) Investigative Ophthalmology and Visual Science 40, 1637-1644. Petersen-Jones, 8. M., Entz, D., Sargan, D. R. cGMP Phosphodiesterase-alpha Mutation Causes Progressive Retinal Atrophy in the Cardigan Welsh Corgi Dog. (1999) Investigative Ophthalmology and Visual Science. 40, 1637- 1644. Pittler, S. J., Baehr, W. Identification of a nonsense mutation in the rod photoreceptor cGMP phosphodiesterase B-subunit gene of the rd mouse. (1991) Proc. NatI.Acad. Sci 88, 8322-8326. Planelles, V. (2003) Hybrid Lentivirus Vectors. Methods in Molecular Biology. 229, 273-284 Provost, M, Le Meur, G., Weber, M., Mendes—Madeira, A., Podevin, G., Cherel, Y., Colle, M.-A., Deschamps, J.-Y., Moullier, P. & Rolling, F. (2005) Biodistribution of rAAV Vectors Following Intraocular Administration: Evidence for the Presence and Persistence of Vector DNA in the Optic Nerve and in the Brain. Molecular Therapy. 1 1, 275-283 Pugh Jr EN, Lamb TD. Phototransduction in Vertebrate Rods and Cones: Molecular Mechanisms of Amplification, Recovery and Light Adaptation. D.G.8tavenga, W.J.de Grip, and E.N.Pugh Jr. Molecular Mechanisms of Visual Transduction. [5], 183-255. 2005. Elsevier Science. Recchia, A., Bonini, C., Magnani, Z., Urbinati, F., Sartori, D., Muraro, 8., Tagliafico, E., Bondanza, A., Stanghellini, M. T. L., Bemardi, M., Pescarollo, A., Ciceri, F., Bordignon, C. 8 Mavilio, F. (2006) Retroviral vector integration deregulates gene expression but has no consequence on the biology and function of transplanted T cells. Proceedings of the National.Academy of Science. 103, 1457-1462 Recombinant DNA Advisory Committee (2002) Assessment of Adenoviral Vector Safety and Toxicity: Report of the National Institutes of Health Recombinant DNA Advisory Committee. Human Gene Therapy. 13, 3-13 132 Redmond, T. M., Shirley Yu, Eric Lee, Dean Bok, Duco Hamasaki, Ning Chen, Patrice Goletz, Jian-Xing Ma, Rosalie K.Crouch & Karl Pfeifer (1998) RPE65 is necessary for production of 11-cis-vitamin A in the retinal visual cycle. Nature Genetics. 20, 344-351 Reichel, M. B., Bainbridge, J., Baker, D., Thrasher, A. J., Bhattacharya, S. 8., Ali, R. R. An immune response after intraocular administration of an adenoviral vector containing a Beta-galactosidase reporter gene slows retinal degeneration in the rd mouse. (2001) British Journal of Ophthalmology 85, 341 -344. RetNet. http://www.sph.uth.tmc.edu_/Retnet/; April, 2007. Rex, T. 8., Feet, J. A., Surace, E. M., Calvert, P. D., Nikonov, S. 8., Lyubarsky, A. L., Bendo, E., Hughes, T., Pugh Jr, E. N. & Bennett, J. (2005) The distribution, concentration, and toxicity of enhanced green fluorescent protein in retinal cells after genomic or somatic (virus-mediated) gene transfer. Molecular Vision. 1 1 , 1236-1245 Sadelain, M. (2004) Insertional oncogenesis in gene therapy: how much of a risk? Nature. 11, 569-573 Sarra, G. M., Stephens, C., Schlichtenbrede, F. C., Bainbridge, J. W. B., Thrasher, A. J., Luthert, P. J. & Ali, R. R. (2002) Kinetics of transgene expression in mouse retina following sub-retinal injection of recombinant adeno-associated virus. Vision Research. 42, 541 -549 Schaack, J. (2005) Induction and Inhibition of Innate Inflammatory Responses by Adenovirus Early Gene Promoters. Viral Immunology. 18, 79-88 Schuettrumph, J., Liu, J.-H., Couto, L. B., Addya, K., Leonard, D. G. B., Zhen, 2., Summer, J. & Arruda, V. R. (2006) Inadvertent Gennline Transmission of AAV2 Vector: Findings in a Rabbit Model Correlate with Those in a Human Clinical Trial. Molecular Therapy. 13, 1064-1073 Shen, W.-Y., Lai, C.-M., Lai, Y. K. Y., Zhang, D., Zaknich, T., Sutano, E. N., Constable, I. J., Rakoczy, P. E. Practical Considerations of recombinant adeno-associated virus-mediated gene transfer for treatment of retinal degenerations. (2003) The Journal of Gene Medicine. 5, 576-587. Smith, A. J., Schlichtenbrede, F. C., Tschemutter, M., Bainbridge, J. W., Thrasher, A. J. & Ali, R. R. (2003) AAV-Mediated Gene Transfer Slows Photoreceptor Loss in the RCS Rat Model of Retinitis Pigmentosa. Molecular Therapy. 8, 188-195 Song, 8., Lu, Y., Choi, Y., Han, Y., Tang, 0., Zhao, G., Bems, K. & Flotte, T. (2004) DNA-dependent PK inhibits adeno-associated virus DNA 133 integration. Proceedings of the National.Academy of Science. 101, 21 12- 21 16 Soucy, B, Wang, Y., Nirenberg, 8., Nathans, J. & Meister, M. (1998) A novel signaling pathway from rod photoreceptors to ganglion cells in mammalian retina. Neuron.481-493 Stolberg 8G. The Biotech Death of Jesse Gelsinger. (2006, July 19) New York Times Sunday Magazine. 1999. New York. Strubbe D, Gelatt K. Ophthalmic Examination and Diagnostic Procedures. Gelatt, KN. Veterinary Ophthalmology. 3rd[10], 427-466. 1998. Baltimore, Williams and Wilkins. Stryer, L. (1991) Visual excitation and recovery. Journal of Biological Chemistry. 266, 10711-10714 Suber, M., Pittler, S. J., Qin, N., Wright, G. C., Holcombe, V., Lee, R. H., Craft, C. M., Lolley, R. N., Baehr, W., Hunrvitz, R. L. Irish setter dogs affected with rod/ cone dysplasia contain a nonsense mutation in the rod cGMP phosphodiesterase beta-subunit gene. (1993) Proceedings of the National.Academy of Science. 90, 3968-3972. Tizard, |,R. Immunity in the Fetus and Newborn. Veterinary Immunology. 7th[19], 223-233. 2004. Philadelphia, Saunders. Takahashi, M., Miyoshi, H., Verrna, I. M. & Gage, F. H. (1999) Rescue from Photoreceptor Degeneration in the rd Mouse by Human Immunodeficiency Virus Immunodeficiency Virus Vector-Mediated Gene Transfer. Journal of Virology. 73, 7812-7816 Tripathy, S. K., Black, H. B., Goldwasser, E. & Leiden, J. M. (1996) Immune response to transgene-encoded proteins limit the stability of gene expression after injection of replication-defective adenovirus vectors. Nature Medicine. 2, 545-550 Tschemutter, M., Schlichtenbrede, F. C., Hower, S., Balaggan, K. 8., Munro, P. M., Bainbridge, J. W. B., Thrasher, A. J., Smith, A. J. & Ali, R. R. (2005) Long-term preservation of retinal function in the RCS rat model of retinitis pigmentosa following lentivirus-mediated gene therapy. Gene Therapy. 12, 694-701 Tuntivanich N. Phenotypic Characterization of Progressive Retinal Atrophy in the Cardigan Welsh Corgi with a Mutation in the PDE6A Gene. 2006. Vanduffel, W., Tootell, R. B., Schoups, A. A. & Orban, G. A. (2002) The organization of orientation selectivity throughout macaque visual cortex. Cerebral Cortex. 12, 647-662 134 Veske, A., Nilsson, S. E. G., Narfstrom, K. & Gal, A. (1999) Retinal Dystrophy of Swedish Briard/Briard-Beagle Dogs is Due to a 4-bp Deletion in RPE65. Genomics. 57, 57-61 Vihinen-Ranta, M., Suikkanen, 8. & Parrish, C. R. (2004) Pathways of Cell Infection by Parvoviruses and Adeno-Associated Viruses. Journal of Virology. 78, 6709-6714 Vollrath, D., Feng, W., Duncan, J. L., Yasumura, D., D'Cruz, P. M., Chappelow, A., Matthes, M. T., Kay, M. A., La Vail, M. M. Correction of the retinal dystrophy phenotype of the RCS rat by viral gene transfer of Mertk. (2001) Proceedings of the National.Academy of Science. 98, 12584—12589. Von Seggem, D. J., Aguilar, E., Kinder, K., Fleck, S. K., Gonzalez Arrnas, J. C., Stevenson, S. C., Ghazal, P., Nemerow, G. R. & Friedlander, M. (2003) In Vivo Transduction of Photoreceptors or Ciliary Body by Intravitral Injection of Pseudotyped Adenoviral Vectors. Molecular Therapy. 7, 27-34 Walters, R. W., Agbandje-McKenna, M., Bowman, V., Moninger, T. 0., Olson, N. H., Seiler.Michael, Chiorini, J. A., Baker, T. S. & Zabner, J. (2004) Structure of Adeno-Associated Virus Serotype 5. Journal of Virology. 78, 3361-3371 Wenkel, H., Streilein, J. W. Analysis of Immune Deviation Elicited by Antigens Injected into the Subretinal Space. (1998) Investigative Ophthalmology and Visual Science. 39, 1823-1834. Wrigstad A. Hereditary Dystrophy of the Retina and the Retinal Pigment Epithelium in a Strain of Briard Dogs: A clinical, morphological, and electrophysiological study. 1994. Linkoping. Yanez—Munoz, R. J., Balaggan, K. 8., MacNeil, A., Howe, S. J., Schmidt, M., Smith, A. J., Buch, P., MacLaren, R. B, Anderson, P. N., Barker, 8. E., Duran, Y., Bartholomae, C., von Kalle, C., Heckenlively, J. R., Kinnon, C., All, R. R. & Thrasher, A. J. (2006) Effective gene therapy with nonintegrating Ientiviral vectors. Nature Medicine. 12, 348-353 Yang, G. S., Schmidt, M., Yan, Z., Lindbloom, J. D., Harding, T. C., Donahue, B. A., Engelhardt, J. F., Kotin, R. & Davidson, B. L. (2002) Virus-Mediated Transduction of Murine Retina with Adeno-Associated Virus: Effects of Viral Capsid and Genome Size. Journal of Virology. 76, 7651-7660 Zeiss, C. J., Johnson, E. A. Proliferation of Microglia, but not Photoreceptors, in the Outer Nuclear Layer of the rd-1 mouse. (2004) 3. Investigative Ophthalmology and Visual Science. 45, 971 -976. 135 Zeiss, C. J., Neal, J., Johnson, E. A. Caspase—3 in Postnatal Retinal Development and Degeneration. (2004) Investigative Ophthalmology and Visual Science .45, 964-970. Zhu, Y., Feuer, G., Day, 8. L., Wrzesinski, 8. & Planelles, V. (2001) Multigene Lentiviral Vectors Based on Differential Splicing and Translational Control. Molecular Therapy. 4, 375-382 Zolotukhin, 8., Potter, M., Zolotukhin, I., Sakai, Y., Loiler, 8., Fraites Jr., T. J., Chiodo, V. A., Phillipsberg, T., Muzyczka, N., Hauswirth, W. W., Flotte, T. R., Byme, B. J. & Snyder, R. O. (2002) Production and purification of serotype 1, 2, and 5 recombnant adeno-associated viral vectors. Methods. 28, 158-167 136 ERS "'iiiiiiiiiii(Militiaiiiiii'“ 291