1
|
Watson-Levings RS, Palmer GD, Levings PP, Dacanay EA, Evans CH, Ghivizzani SC. Gene Therapy in Orthopaedics: Progress and Challenges in Pre-Clinical Development and Translation. Front Bioeng Biotechnol 2022; 10:901317. [PMID: 35837555 PMCID: PMC9274665 DOI: 10.3389/fbioe.2022.901317] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
In orthopaedics, gene-based treatment approaches are being investigated for an array of common -yet medically challenging- pathologic conditions of the skeletal connective tissues and structures (bone, cartilage, ligament, tendon, joints, intervertebral discs etc.). As the skeletal system protects the vital organs and provides weight-bearing structural support, the various tissues are principally composed of dense extracellular matrix (ECM), often with minimal cellularity and vasculature. Due to their functional roles, composition, and distribution throughout the body the skeletal tissues are prone to traumatic injury, and/or structural failure from chronic inflammation and matrix degradation. Due to a mixture of environment and endogenous factors repair processes are often slow and fail to restore the native quality of the ECM and its function. In other cases, large-scale lesions from severe trauma or tumor surgery, exceed the body’s healing and regenerative capacity. Although a wide range of exogenous gene products (proteins and RNAs) have the potential to enhance tissue repair/regeneration and inhibit degenerative disease their clinical use is hindered by the absence of practical methods for safe, effective delivery. Cumulatively, a large body of evidence demonstrates the capacity to transfer coding sequences for biologic agents to cells in the skeletal tissues to achieve prolonged delivery at functional levels to augment local repair or inhibit pathologic processes. With an eye toward clinical translation, we discuss the research progress in the primary injury and disease targets in orthopaedic gene therapy. Technical considerations important to the exploration and pre-clinical development are presented, with an emphasis on vector technologies and delivery strategies whose capacity to generate and sustain functional transgene expression in vivo is well-established.
Collapse
Affiliation(s)
- Rachael S. Watson-Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Glyn D. Palmer
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Padraic P. Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - E. Anthony Dacanay
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Christopher H. Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MI, United States
| | - Steven C. Ghivizzani
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
- *Correspondence: Steven C. Ghivizzani,
| |
Collapse
|
2
|
|
3
|
Teo KYC, Lee SY, Barathi AV, Tun SBB, Tan L, Constable IJ. Surgical Removal of Internal Limiting Membrane and Layering of AAV Vector on the Retina Under Air Enhances Gene Transfection in a Nonhuman Primate. ACTA ACUST UNITED AC 2018; 59:3574-3583. [DOI: 10.1167/iovs.18-24333] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Kelvin Yi Chong Teo
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Shu Yen Lee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Amutha Veluchamy Barathi
- Translational Pre-clinical Model Platform, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sai Bo Bo Tun
- Translational Pre-clinical Model Platform, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Licia Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Ian Jeffery Constable
- Centre of Ophthalmology and Visual Science (incorporating Lions Eye Institute), The University of Western Australia, Perth, Western Australia
- Department of Ophthalmology, Sir Charles Gairdner Hospital, Perth, Western Australia
| |
Collapse
|
4
|
Hickey DG, Edwards TL, Barnard AR, Singh MS, de Silva SR, McClements ME, Flannery JG, Hankins MW, MacLaren RE. Tropism of engineered and evolved recombinant AAV serotypes in the rd1 mouse and ex vivo primate retina. Gene Ther 2017; 24:787-800. [PMID: 28872643 PMCID: PMC5746594 DOI: 10.1038/gt.2017.85] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 07/19/2017] [Accepted: 08/23/2017] [Indexed: 11/09/2022]
Abstract
There is much debate on the adeno-associated virus (AAV) serotype that best targets specific retinal cell types and the route of surgical delivery-intravitreal or subretinal. This study compared three of the most efficacious AAV vectors known to date in a mouse model of retinal degeneration (rd1 mouse) and macaque and human retinal explants. Green fluorescent protein (GFP) driven by a ubiquitous promoter was packaged into three AAV capsids: AAV2/8(Y733F), AAV2/2(quad Y-F) and AAV2/2(7m8). Overall, AAV2/2(7m8) transduced the largest area of retina and resulted in the highest level of GFP expression, followed by AAV2/2(quad Y-F) and AAV2/8(Y733F). AAV2/2(7m8) and AAV2/2(quad Y-F) both resulted in similar patterns of transduction whether they were injected intravitreally or subretinally. AAV2/8(Y733F) transduced a significantly smaller area of retina when injected intravitreally compared with subretinally. Retinal ganglion cells, horizontal cells and retinal pigment epithelium expressed relatively high levels of GFP in the mouse retina, whereas amacrine cells expressed low levels of GFP and bipolar cells were infrequently transduced. Cone cells were the most frequently transduced cell type in macaque retina explants, whereas Müller cells were the predominant transduced cell type in human retinal explants. Of the AAV serotypes tested, AAV2/2(7m8) was the most effective at transducing a range of cell types in degenerate mouse retina and macaque and human retinal explants.
Collapse
Affiliation(s)
- D G Hickey
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
| | - T L Edwards
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
| | - A R Barnard
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
| | - M S Singh
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK.,Moorfields Eye Hospital NHS Foundation Trust NIHR Biomedical Research Centre, London, UK
| | - S R de Silva
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
| | - M E McClements
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
| | - J G Flannery
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - M W Hankins
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK.,Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, UK
| | - R E MacLaren
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK.,Moorfields Eye Hospital NHS Foundation Trust NIHR Biomedical Research Centre, London, UK.,Oxford University Hospitals NHS Trust Biomedical Research Centre, Oxford, UK
| |
Collapse
|
5
|
Fujita K, Nishiguchi KM, Shiga Y, Nakazawa T. Spatially and Temporally Regulated NRF2 Gene Therapy Using Mcp-1 Promoter in Retinal Ganglion Cell Injury. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 5:130-141. [PMID: 28480312 PMCID: PMC5415330 DOI: 10.1016/j.omtm.2017.04.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/12/2017] [Indexed: 02/03/2023]
Abstract
Retinal ganglion cell degeneration triggered by axonal injury is believed to underlie many ocular diseases, including glaucoma and optic neuritis. In these diseases, retinal ganglion cells are affected unevenly, both spatially and temporally, such that healthy and unhealthy cells coexist in different patterns at different time points. Herein, we describe a temporally and spatially regulated adeno-associated virus gene therapy aiming to reduce undesired off-target effects on healthy retinal neurons. The Mcp-1 promoter previously shown to be activated in stressed retinal ganglion cells following murine optic nerve injury was combined with the neuroprotective intracellular transcription factor Nrf2. In this model, Mcp-1 promoter-driven NRF2 expression targeting only stressed retinal ganglion cells showed efficacy equivalent to non-selective cytomegalovirus promoter-driven therapy for preventing cell death. However, cytomegalovirus promoter-mediated NRF2 transcription induced cellular stress responses and death of Brn3A-positive uninjured retinal ganglion cells. Such undesired effects were reduced substantially by adopting the Mcp-1 promoter. Combining a stress-responsive promoter and intracellular therapeutic gene is a versatile approach for specifically targeting cells at risk of degeneration. This strategy may be applicable to numerous chronic ocular and non-ocular conditions.
Collapse
Affiliation(s)
- Kosuke Fujita
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Yukihiro Shiga
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Toru Nakazawa
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.,Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.,Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| |
Collapse
|
6
|
Taking Stock of Retinal Gene Therapy: Looking Back and Moving Forward. Mol Ther 2017; 25:1076-1094. [PMID: 28391961 DOI: 10.1016/j.ymthe.2017.03.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/04/2017] [Accepted: 03/04/2017] [Indexed: 11/23/2022] Open
Abstract
Over the past 20 years, there has been tremendous progress in retinal gene therapy. The safety and efficacy results in one early-onset severe blinding disease may lead to the first gene therapy drug approval in the United States. Here, we review how far the field has come over the past two decades and speculate on the directions that the field will take in the future.
Collapse
|
7
|
Chen JL, Walton KL, Qian H, Colgan TD, Hagg A, Watt MJ, Harrison CA, Gregorevic P. Differential Effects of IL6 and Activin A in the Development of Cancer-Associated Cachexia. Cancer Res 2016; 76:5372-82. [PMID: 27328730 DOI: 10.1158/0008-5472.can-15-3152] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/13/2016] [Indexed: 11/16/2022]
Abstract
Cachexia is a life-threatening wasting syndrome lacking effective treatment, which arises in many cancer patients. Although ostensibly induced by multiple tumor-produced cytokines (tumorkines), their functional contribution to initiation and progression of this syndrome has proven difficult to determine. In this study, we used adeno-associated viral vectors to elevate circulating levels of the tumorkines IL6 and/or activin A in animals in the absence of tumors as a tactic to evaluate hypothesized roles in cachexia development. Mice with elevated levels of IL6 exhibited 8.1% weight loss after 9 weeks, whereas mice with elevated levels of activin A lost 11% of their body weight. Co-elevation of both tumorkines to levels approximating those observed in cancer cachexia models induced a more rapid and profound body weight loss of 15.4%. Analysis of body composition revealed that activin A primarily triggered loss of lean mass, whereas IL6 was a major mediator of fat loss. Histologic and transcriptional analysis of affected organs/tissues (skeletal muscle, fat, and liver) identified interactions between the activin A and IL6 signaling pathways. For example, IL6 exacerbated the detrimental effects of activin A in skeletal muscle, whereas activin A curbed the IL6-induced acute-phase response in liver. This study presents a useful model to deconstruct cachexia, opening a pathway to determining which tumorkines are best targeted to slow/reverse this devastating condition in cancer patients. Cancer Res; 76(18); 5372-82. ©2016 AACR.
Collapse
Affiliation(s)
- Justin L Chen
- Hudson Institute of Medical Research, Clayton, Australia. Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Kelly L Walton
- Hudson Institute of Medical Research, Clayton, Australia
| | - Hongwei Qian
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Timothy D Colgan
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia. Department of Physiology, The University of Melbourne, Melbourne, Australia
| | - Adam Hagg
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Matthew J Watt
- The Obesity and Metabolism Program of the Biomedicine Discovery Institute, Monash University, Clayton, Australia. Department of Physiology, Monash University, Clayton, Australia
| | - Craig A Harrison
- Hudson Institute of Medical Research, Clayton, Australia. Department of Physiology, Monash University, Clayton, Australia. Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Paul Gregorevic
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia. Department of Physiology, The University of Melbourne, Melbourne, Australia. Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia. Department of Neurology, The University of Washington School of Medicine, Seattle, Washington.
| |
Collapse
|
8
|
Abstract
The prognosis of patients with coronary artery disease and stroke has improved substantially over the last decade as a result of advances in primary and secondary preventive care as well as novel interventional approaches, including the development of drug-eluting stents and balloons. Despite this progress, however, cardiovascular disease remains the leading cause of death in industrialized nations. Sustained efforts to elucidate the underlying mechanisms of atherogenesis, reperfusion-induced cardiac injury, and ischemic heart failure have led to the identification of several target genes as key players in the development and progression of atherosclerotic vascular disease. This knowledge has now enabled genetic therapeutic modulation not only for inherited diseases with a single gene defect, such as familial hypercholesterolemia, but also for multifactorial disorders. This review will focus on approaches in adeno-associated viral (AAV)-mediated gene therapy for atherosclerosis and its long-term sequelae.
Collapse
|
9
|
Le Guiner C, Stieger K, Toromanoff A, Guilbaud M, Mendes-Madeira A, Devaux M, Guigand L, Cherel Y, Moullier P, Rolling F, Adjali O. Transgene regulation using the tetracycline-inducible TetR-KRAB system after AAV-mediated gene transfer in rodents and nonhuman primates. PLoS One 2014; 9:e102538. [PMID: 25248159 PMCID: PMC4172479 DOI: 10.1371/journal.pone.0102538] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 06/19/2014] [Indexed: 11/19/2022] Open
Abstract
Numerous studies have demonstrated the efficacy of the Adeno-Associated Virus (AAV)-based gene delivery platform in vivo. The control of transgene expression in many protocols is highly desirable for therapeutic applications and/or safety reasons. To date, the tetracycline and the rapamycin dependent regulatory systems have been the most widely evaluated. While the long-term regulation of the transgene has been obtained in rodent models, the translation of these studies to larger animals, especially to nonhuman primates (NHP), has often resulted in an immune response against the recombinant regulator protein involved in transgene expression regulation. These immune responses were dependent on the target tissue and vector delivery route. Here, using AAV vectors, we evaluated a doxycyclin-inducible system in rodents and macaques in which the TetR protein is fused to the human Krüppel associated box (KRAB) protein. We demonstrated long term gene regulation efficiency in rodents after subretinal and intramuscular administration of AAV5 and AAV1 vectors, respectively. However, as previously described for other chimeric transactivators, the TetR-KRAB-based system failed to achieve long term regulation in the macaque after intramuscular vector delivery because of the development of an immune response. Thus, immunity against the chimeric transactivator TetR-KRAB emerged as the primary limitation for the clinical translation of the system when targeting the skeletal muscle, as previously described for other regulatory proteins. New developments in the field of chimeric drug-sensitive transactivators with the potential to not trigger the host immune system are still needed.
Collapse
Affiliation(s)
- Caroline Le Guiner
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes University Hospital, Nantes, France
| | - Knut Stieger
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes University Hospital, Nantes, France
- Department of Ophthalmology, Faculty of Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Alice Toromanoff
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes University Hospital, Nantes, France
| | - Mickaël Guilbaud
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes University Hospital, Nantes, France
| | | | - Marie Devaux
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes University Hospital, Nantes, France
| | - Lydie Guigand
- INRA UMR 703 and Atlantic Gene Therapies, ONIRIS, Nantes, France
| | - Yan Cherel
- INRA UMR 703 and Atlantic Gene Therapies, ONIRIS, Nantes, France
| | - Philippe Moullier
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes University Hospital, Nantes, France
- Department of Molecular Genetics and Microbiology department, University of Florida, Gainesville, Florida, United States of America
| | - Fabienne Rolling
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes University Hospital, Nantes, France
| | - Oumeya Adjali
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes University Hospital, Nantes, France
| |
Collapse
|
10
|
Abstract
Adeno-associated virus (AAV) is a member of the family Parvoviridae that has been widely used as a vector for gene therapy because of its safety profile, its ability to transduce both dividing and non-dividing cells, and its low immunogenicity. AAV has been detected in many different tissues of several animal species but has not been associated with any disease. As a result of natural infections, antibodies to AAV can be found in many animals including humans. It has been shown that pre-existing AAV antibodies can modulate the safety and efficacy of AAV vector-mediated gene therapy by blocking vector transduction or by redirecting distribution of AAV vectors to tissues other than the target organ. This review will summarize antibody responses against natural AAV infections, as well as AAV gene therapy vectors and their impact in the clinical development of AAV vectors for gene therapy. We will also review and discuss the various methods used for AAV antibody detection and strategies to overcome neutralizing antibodies in AAV-mediated gene therapy.
Collapse
Affiliation(s)
- Roberto Calcedo
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | | |
Collapse
|
11
|
Lipinski DM, Thake M, MacLaren RE. Clinical applications of retinal gene therapy. Prog Retin Eye Res 2013; 32:22-47. [DOI: 10.1016/j.preteyeres.2012.09.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 09/04/2012] [Accepted: 09/04/2012] [Indexed: 02/08/2023]
|
12
|
Bennett J, Maguire AM. Gene Therapy for Retinal Disease. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00034-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
13
|
Photoreceptor degeneration in mice: adeno-associated viral vector-mediated delivery of erythropoietin. Methods Mol Biol 2013; 982:237-63. [PMID: 23456874 DOI: 10.1007/978-1-62703-308-4_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The exogenous delivery of erythropoietin (EPO) and EPO derivatives (EPO-Ds) represents a valuable strategy to protect the retina from degeneration. In this chapter we describe a method to deliver EPO and the EPO derivative S100E in the light-damage model of induced retinal degeneration using adeno--associated viral (AAV) vectors and to evaluate the functional and morphological protection of the retina from light damage.
Collapse
|
14
|
Ferreira JR, Hirsch ML, Zhang L, Park Y, Samulski RJ, Hu WS, Ko CC. Three-dimensional multipotent progenitor cell aggregates for expansion, osteogenic differentiation and 'in vivo' tracing with AAV vector serotype 6. Gene Ther 2012; 20:158-68. [PMID: 22402320 PMCID: PMC3374053 DOI: 10.1038/gt.2012.16] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Multipotent adult progenitor cells (MAPC) are bone marrow-derived stem cells with a high growth rate suitable for therapeutical applications as three-dimensional (3D) aggregates. Combined applications of osteogenically differentiated MAPC (OD-MAPC) aggregates and adeno-associated viral vectors (AAV) in bone bioengineering are still deferred until information regarding expansion technologies, osteogenic potential, and AAV cytotoxicity and transduction efficiency is better understood. In this study, we tested whether self-complementary AAV (scAAV) can potentially be used as a gene delivery system in a OD-MAPC-based “in vivo” bone formation model in the craniofacial region. Both expansion of rat MAPC (rMAPC) and osteogenic differentiation with dexamethasone were also tested in 3D aggregate culture systems “in vitro” and “vivo”. Rat MAPCs (rMAPCs) grew as undifferentiated aggregates for 4 days with a population doubling time of 37h. After expansion, constant levels of Oct4 transcripts, and Oct4 and CD31 surface markers were observed, which constitute a hallmark of rMAPCs undifferentiated stage. Dexamethasone effectively mediated rMAPC osteogenic differentiation by inducing the formation of a mineralized collagen type I network, and facilitated the activation of the wnt/β-catenin, a crucial pathway in skeletal development. To investigate the genetic modification of rMAPCs grown as 3D aggregates prior to implantation, scAAV serotypes 2, 3, and 6 were evaluated. scAAV6 packaged with the enhanced green fluorescent protein expression cassette efficiently mediated long-term transduction (10 days) “in vitro” and “vivo”. The reporter transduction event allowed the tracing of OD-rMAPC (induced by dexamethasone) aggregates following OD-rMAPC transfer into a macro-porous hydroxyapatite scaffold implanted in a rat calvaria model. Furthermore, the scAAV6-transduced OD-rMAPC generated a bone-like matrix with a collagenous matrix rich in bone specific proteins (osteocalcin and osteopontin) in the scaffold macro-pores 10 days post-implantation. Newly formed bone was also observed in the interface between native bone and scaffold. The collective work supports future bone tissue engineering applications of 3D MAPC cultures for expansion, bone formation, and the ability to genetically alter these cells using scAAV vectors.
Collapse
Affiliation(s)
- J R Ferreira
- School of Dentistry, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
With the recent progress in identifying disease-causing genes in humans and in animal models, there are more and more opportunities for using retinal gene transfer to learn more about retinal physiology and also to develop therapies for blinding disorders. Success in preclinical studies for one form of inherited blindness have led to testing in human clinical trials. This paves the way to consider a number of other retinal diseases as ultimate gene therapy targets in human studies. The information presented here is designed to assist scientists and clinicians to use gene transfer to probe the biology of the retina and/or to move appropriate gene-based treatment studies from the bench to the clinic.
Collapse
Affiliation(s)
- Jean Bennett
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
16
|
Mussolino C, della Corte M, Rossi S, Viola F, Di Vicino U, Marrocco E, Neglia S, Doria M, Testa F, Giovannoni R, Crasta M, Giunti M, Villani E, Lavitrano M, Bacci ML, Ratiglia R, Simonelli F, Auricchio A, Surace EM. AAV-mediated photoreceptor transduction of the pig cone-enriched retina. Gene Ther 2011; 18:637-45. [PMID: 21412286 PMCID: PMC3131697 DOI: 10.1038/gt.2011.3] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 08/16/2010] [Accepted: 09/19/2010] [Indexed: 01/04/2023]
Abstract
Recent success in clinical trials supports the use of adeno-associated viral (AAV) vectors for gene therapy of retinal diseases caused by defects in the retinal pigment epithelium (RPE). In contrast, evidence of the efficacy of AAV-mediated gene transfer to retinal photoreceptors, the major site of inherited retinal diseases, is less robust. In addition, although AAV-mediated RPE transduction appears efficient, independently of the serotype used and species treated, AAV-mediated photoreceptor gene transfer has not been systematically investigated thus so far in large animal models, which also may allow identifying relevant species-specific differences in AAV-mediated retinal transduction. In the present study, we used the porcine retina, which has a high cone/rod ratio. This feature allows to properly evaluate both cone and rod photoreceptors transduction and compare the transduction characteristics of AAV2/5 and 2/8, the two most efficient AAV vector serotypes for photoreceptor targeting. Here we show that AAV2/5 and 2/8 transduces both RPE and photoreceptors. AAV2/8 infects and transduces photoreceptor more efficiently than AAV2/5, similarly to what we have observed in the murine retina. The use of the photoreceptor-specific rhodopsin promoter restricts transgene expression to porcine rods and cones, and results in photoreceptor transduction levels similar to those obtained with the ubiquitous promoters tested. Finally, immunological, toxicological and biodistribution studies support the safety of AAV subretinal administration to the large porcine retina. The data presented here on AAV-mediated transduction of the cone-enriched porcine retina may affect the development of gene-based therapies for rare and common severe photoreceptor diseases.
Collapse
Affiliation(s)
- C Mussolino
- Telethon Institute of Genetics and Medicine, Naples, Italy
- SEMM-European School of Molecular Medicine, Naples site, Italy
| | - M della Corte
- Department of Ophthalmology, Second University of Naples, Naples, Italy
| | - S Rossi
- Department of Ophthalmology, Second University of Naples, Naples, Italy
| | - F Viola
- UO Oculistica, Fondazione IRCCS, Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, University of Milan, Milan, Italy
| | - U Di Vicino
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - E Marrocco
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - S Neglia
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - M Doria
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - F Testa
- Department of Ophthalmology, Second University of Naples, Naples, Italy
| | - R Giovannoni
- Department of Surgical Sciences. University of Milano-Bicocca, Monza, Italy
| | - M Crasta
- Visionvet, Eye Clinic for Animal, Bologna, Italy
| | - M Giunti
- Department of Veterinary Morphophysiology and Animal Production, University of Bologna, Bologna, Italy
| | - E Villani
- UO Oculistica, Fondazione IRCCS, Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, University of Milan, Milan, Italy
| | - M Lavitrano
- Department of Surgical Sciences. University of Milano-Bicocca, Monza, Italy
| | - M L Bacci
- Department of Veterinary Morphophysiology and Animal Production, University of Bologna, Bologna, Italy
| | - R Ratiglia
- UO Oculistica, Fondazione IRCCS, Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, University of Milan, Milan, Italy
| | - F Simonelli
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Ophthalmology, Second University of Naples, Naples, Italy
| | - A Auricchio
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Medical Genetics, Department of Pediatrics, ‘Federico II' University, Naples, Italy
| | - E M Surace
- Telethon Institute of Genetics and Medicine, Naples, Italy
| |
Collapse
|
17
|
Colella P, Iodice C, Di Vicino U, Annunziata I, Surace EM, Auricchio A. Non-erythropoietic erythropoietin derivatives protect from light-induced and genetic photoreceptor degeneration. Hum Mol Genet 2011; 20:2251-62. [PMID: 21421996 PMCID: PMC3090200 DOI: 10.1093/hmg/ddr115] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/01/2011] [Accepted: 03/15/2011] [Indexed: 11/21/2022] Open
Abstract
Given the high genetic heterogeneity of inherited retinal degenerations (IRDs), a wide applicable treatment would be desirable to halt/slow progressive photoreceptor (PR) cell loss in a mutation-independent manner. In addition to its erythropoietic activity, erythropoietin (EPO) presents neurotrophic characteristics. We have previously shown that adeno-associated viral (AAV) vector-mediated systemic EPO delivery protects from PR degeneration. However, this is associated with an undesired hematocrit increase that could contribute to PR protection. Non-erythropoietic EPO derivatives (EPO-D) are available which allow us to dissect erythropoiesis's role in PR preservation and may be more versatile and safe than EPO as anti-apoptotic agents. We delivered in animal models of light-induced or genetic retinal degeneration either intramuscularly or subretinally AAV vectors encoding EPO or one of the three selected EPO-D: the mutant S100E, the helix A- and B-derived EPO-mimetic peptides. We observed that (i) systemic expression of S100E induces a significantly lower hematocrit increase than EPO and provides similar protection from PR degeneration, and (ii) intraocular expression of EPO-D protects PR from degeneration in the absence of significant hematocrit increase. On the basis of this, we conclude that erythropoiesis is not required for EPO-mediated PR protection. However, the lower efficacy observed when EPO or S100E is expressed intraocularly rather than systemically suggests that hormone systemic effects contribute to PR protection. Unlike S100E, EPO-mimetic peptides preserve PR only when given locally, suggesting that different EPO-D have a different potency or mode of action. In conclusion, our data show that subretinal delivery of AAV vectors encoding EPO-D protects from light-induced and genetic PR degeneration.
Collapse
Affiliation(s)
- Pasqualina Colella
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
- The Open University, Milton Keynes, UK and
| | - Carolina Iodice
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | - Umberto Di Vicino
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | - Ida Annunziata
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | - Enrico M. Surace
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
- Medical Genetics, Department of Pediatrics, Federico II University, Naples, Italy
| |
Collapse
|
18
|
Yin L, Greenberg K, Hunter JJ, Dalkara D, Kolstad KD, Masella BD, Wolfe R, Visel M, Stone D, Libby RT, DiLoreto D, Schaffer D, Flannery J, Williams DR, Merigan WH. Intravitreal injection of AAV2 transduces macaque inner retina. Invest Ophthalmol Vis Sci 2011; 52:2775-83. [PMID: 21310920 PMCID: PMC3088562 DOI: 10.1167/iovs.10-6250] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 10/18/2010] [Accepted: 10/21/2010] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Adeno-associated virus serotype 2 (AAV2) has been shown to be effective in transducing inner retinal neurons after intravitreal injection in several species. However, results in nonprimates may not be predictive of transduction in the human inner retina, because of differences in eye size and the specialized morphology of the high-acuity human fovea. This was a study of inner retina transduction in the macaque, a primate with ocular characteristics most similar to that of humans. METHODS In vivo imaging and histology were used to examine GFP expression in the macaque inner retina after intravitreal injection of AAV vectors containing five distinct promoters. RESULTS AAV2 produced pronounced GFP expression in inner retinal cells of the fovea, no expression in the central retina beyond the fovea, and variable expression in the peripheral retina. AAV2 vector incorporating the neuronal promoter human connexin 36 (hCx36) transduced ganglion cells within a dense annulus around the fovea center, whereas AAV2 containing the ubiquitous promoter hybrid cytomegalovirus (CMV) enhancer/chicken-β-actin (CBA) transduced both Müller and ganglion cells in a dense circular disc centered on the fovea. With three shorter promoters--human synapsin (hSYN) and the shortened CBA and hCx36 promoters (smCBA and hCx36sh)--AAV2 produced visible transduction, as seen in fundus images, only when the retina was altered by ganglion cell loss or enzymatic vitreolysis. CONCLUSIONS The results in the macaque suggest that intravitreal injection of AAV2 would produce high levels of gene expression at the human fovea, important in retinal gene therapy, but not in the central retina beyond the fovea.
Collapse
Affiliation(s)
- Lu Yin
- From the Flaum Eye Institute
- the Center for Visual Science
| | - Kenneth Greenberg
- the Helen Wills Neuroscience Institute and
- the Departments of Molecular and Cell Biology
- Vision Science, and
| | | | | | - Kathleen D. Kolstad
- the Helen Wills Neuroscience Institute and
- the Departments of Molecular and Cell Biology
- Vision Science, and
| | | | | | - Meike Visel
- the Helen Wills Neuroscience Institute and
- the Departments of Molecular and Cell Biology
- Vision Science, and
| | - Daniel Stone
- the Helen Wills Neuroscience Institute and
- Chemical Engineering, University of California, Berkeley, Berkeley, California
| | | | | | - David Schaffer
- the Helen Wills Neuroscience Institute and
- Chemical Engineering, University of California, Berkeley, Berkeley, California
| | - John Flannery
- the Helen Wills Neuroscience Institute and
- the Departments of Molecular and Cell Biology
- Vision Science, and
| | - David R. Williams
- the Center for Visual Science
- Institute of Optics, University of Rochester, Rochester, New York; and
| | | |
Collapse
|
19
|
Weller ML, Amornphimoltham P, Schmidt M, Wilson PA, Gutkind JS, Chiorini JA. Epidermal growth factor receptor is a co-receptor for adeno-associated virus serotype 6. Nat Med 2010; 16:662-4. [PMID: 20473307 PMCID: PMC2885716 DOI: 10.1038/nm.2145] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 04/07/2010] [Indexed: 01/08/2023]
Abstract
A key step in gene therapy is the efficient transfer of genes in a cell type- and tissue-specific manner. To better understand the mechanism of adeno-associated virus serotype 6 (AAV6) transduction, we used comparative gene analysis (CGA) combined with pathway visualization software to identify a positive correlation between AAV6 transduction and epidermal growth factor receptor (EGFR) expression. Subsequent experiments suggested that EGFR is necessary for vector internalization and probably functions as a co-receptor for AAV6.
Collapse
Affiliation(s)
- Melodie L Weller
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, US National Institutes of Health (NIH), Bethesda, Maryland, USA
| | | | | | | | | | | |
Collapse
|
20
|
Epo delivery by genetically engineered C2C12 myoblasts immobilized in microcapsules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 670:54-67. [PMID: 20384218 DOI: 10.1007/978-1-4419-5786-3_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ver the last half century, the use of erythropoietin (Epo) in the management of malignancies has been extensively studied. Originally viewed as the renal hormone responsible for red blood cell production, many recent in vivo and clinical approaches demonstrate that various tissues locally produce Epo in response to physical or metabolic stress. Thus, not only its circulating erythrocyte mass regulator activity but also the recently discovered nonhematological actions are being thoroughly investigated in order to fulfill the specific Epo delivery requirements for each therapeutic approach.
Collapse
|
21
|
PiggyBac transposon-based inducible gene expression in vivo after somatic cell gene transfer. Mol Ther 2009; 17:2115-20. [PMID: 19809403 DOI: 10.1038/mt.2009.234] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Somatic cell gene transfer has permitted inducible gene expression in vivo through coinfection of multiple viruses. We hypothesized that the highly efficient plasmid-based piggyBac transposon system would enable long-term inducible gene expression in mice in vivo. We used a multiple-transposon delivery strategy to create a tetracycline-inducible expression system in vitro in human cells by delivering the two genes on separate transposons for inducible reporter gene expression along with a separate selectable transposon marker. Evaluation of stable cell lines revealed 100% of selected clones exhibited inducible expression via stable expression from three separate transposons simultaneously. We next tested and found that piggyBac-mediated gene transfer to liver or lung could achieve stable reporter gene expression in mice in vivo in either immunocompetent or immune deficient animals. A single injection of piggyBac transposons could achieve long-term inducible gene expression in the livers of mice in vivo, confirming our multiple-transposon strategy used in cultured cells. The plasmid-based piggyBac transposon system enables constitutive or inducible gene expression in vivo for potential therapeutic and biological applications without using viral vectors.
Collapse
|
22
|
Stieger K, Belbellaa B, Le Guiner C, Moullier P, Rolling F. In vivo gene regulation using tetracycline-regulatable systems. Adv Drug Deliv Rev 2009; 61:527-41. [PMID: 19394373 PMCID: PMC7103297 DOI: 10.1016/j.addr.2008.12.016] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Accepted: 12/15/2008] [Indexed: 10/26/2022]
Abstract
Numerous preclinical studies have demonstrated the efficacy of viral gene delivery vectors, and recent clinical trials have shown promising results. However, the tight control of transgene expression is likely to be required for therapeutic applications and in some instances, for safety reasons. For this purpose, several ligand-dependent transcription regulatory systems have been developed. Among these, the tetracycline-regulatable system is by far the most frequently used and the most advanced towards gene therapy trials. This review will focus on this system and will describe the most recent progress in the regulation of transgene expression in various organs, including the muscle, the retina and the brain. Since the development of an immune response to the transactivator was observed following gene transfer in the muscle of nonhuman primate, focus will be therefore, given on the immune response to transgene products of the tetracycline inducible promoter.
Collapse
Affiliation(s)
- Knut Stieger
- INSERM UMR U649, CHU-Hotel Dieu, Nantes, France
- Department of Ophthalmology, Justus-Liebig-University Giessen, Giessen, Germany
| | | | | | | | | |
Collapse
|
23
|
Cao L, Lin EJD, Cahill MC, Wang C, Liu X, During MJ. Molecular therapy of obesity and diabetes by a physiological autoregulatory approach. Nat Med 2009; 15:447-54. [PMID: 19270710 DOI: 10.1038/nm.1933] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Accepted: 01/23/2009] [Indexed: 12/31/2022]
Abstract
Hypothalamic brain-derived neurotrophic factor (BDNF) is a key element in the regulation of energy balance. Here we investigated the therapeutic efficacy of BDNF by gene transfer in mouse models of obesity and diabetes. Gene transfer of BDNF led to marked weight loss and alleviation of obesity-associated insulin resistance. To facilitate clinical translation and ensure that BDNF protein expression was appropriately decreased as weight loss progressed, thus preventing cachexia, we developed a molecular autoregulatory system involving a single recombinant adeno-associated virus vector harboring two expression cassettes, one constitutively driving BDNF and the other driving a specific microRNA targeting BDNF. The microRNA element was controlled by a promoter (that controlling the Agrp gene encoding agouti-related peptide) responsive to BDNF-induced physiological changes. Hence, as body weight decreased and agouti-related protein is induced, microRNA expression was activated, inhibiting transgene expression. In contrast to the progressive weight loss associated with a nonregulated approach, this microRNA-approach led to a sustainable plateau of body weight after notable weight loss was achieved. This strategy mimics the body's endogenous physiological feedback mechanisms, thereby resetting the hypothalamic set point to reverse obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Lei Cao
- Cancer Genetics and Neuroscience Program, Department of Molecular Virology, Immunology and Medical Genetics, and the Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | |
Collapse
|
24
|
Colella P, Cotugno G, Auricchio A. Ocular gene therapy: current progress and future prospects. Trends Mol Med 2009; 15:23-31. [PMID: 19097940 DOI: 10.1016/j.molmed.2008.11.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 11/04/2008] [Accepted: 11/04/2008] [Indexed: 12/16/2022]
Abstract
As gene therapy begins to produce its first clinical successes, interest in ocular gene transfer has grown owing to the favorable safety and efficacy characteristics of the eye as a target organ for drug delivery. Important advances also include the availability of viral and non-viral vectors that are able to efficiently transduce various ocular cell types, the use of intraocular delivery routes and the development of transcriptional regulatory elements that allow sustained levels of gene transfer in small and large animal models after a single administration. Here, we review recent progress in the field of ocular gene therapy. The first experiments in humans with severe inherited forms of blindness seem to confirm the good safety and efficacy profiles observed in animal models and suggest that gene transfer has the potential to become a valuable therapeutic strategy for otherwise untreatable blinding diseases.
Collapse
Affiliation(s)
- Pasqualina Colella
- Telethon Institute of Genetics and Medicine (TIGEM), Via Pietro Castellino 111, 80131 Naples, Italy
| | | | | |
Collapse
|
25
|
Stieger K, Schroeder J, Provost N, Mendes-Madeira A, Belbellaa B, Le Meur G, Weber M, Deschamps JY, Lorenz B, Moullier P, Rolling F. Detection of intact rAAV particles up to 6 years after successful gene transfer in the retina of dogs and primates. Mol Ther 2008; 17:516-23. [PMID: 19107120 DOI: 10.1038/mt.2008.283] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gene transfer to the retina using recombinant adeno-associated viral (rAAV) vectors has proven to be an effective option for the treatment of retinal degenerative diseases in several animal models and has recently advanced into clinical trials in humans. To date, intracellular trafficking of AAV vectors and subsequent capsid degradation has been studied only in vitro, but the fate of AAV particles in transduced cells following subretinal injection has yet to be elucidated. Using electron microscopy and western blot, we analyzed retinas of one primate and four dogs that had been subretinally injected with AAV2/4, -2/5, or -2/2 serotypes and that displayed efficient gene transfer over several years. We show that intact AAV particles are still present in retinal cells, for up to 6 years after successful gene transfer in these large animals. The persistence of intact vector particles in the target organ, several years postadministration, is totally unexpected and, therefore, represents a new and unanticipated safety issue to consider at a time when gene therapy clinical trials raise new immunological concerns.
Collapse
|
26
|
Buch PK, Bainbridge JW, Ali RR. AAV-mediated gene therapy for retinal disorders: from mouse to man. Gene Ther 2008; 15:849-57. [PMID: 18418417 DOI: 10.1038/gt.2008.66] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A wide range of retinal disorders can potentially be treated using viral vector-mediated gene therapy. The most widely used vectors for ocular gene delivery are based on adeno-associated virus (AAV), because they elicit minimal immune responses and mediate long-term transgene expression in a variety of retinal cell types. Proof-of-concept experiments have demonstrated the efficacy of AAV-mediated transgene delivery in a number of animal models of inherited and acquired retinal disorders. Following extensive preclinical evaluation in large animal models, gene therapy for one form of inherited retinal degeneration due to RPE65 deficiency is now being tested in three concurrent clinical trials. Here, we review different approaches for treating inherited retinal degenerations and more common acquired retinal disorders using AAV-based vectors.
Collapse
Affiliation(s)
- P K Buch
- Division of Molecular Therapy, UCL Institute of Ophthalmology and UCL/Moorfields Eye Hospital Biomedical Research Centre for Ophthalmology, London, UK
| | | | | |
Collapse
|
27
|
Abstract
Some of the most successful gene therapy results have been obtained using recombinant viral vectors to treat animal models of inherited and acquired ocular diseases. Clinical trials using adenovirus vector systems have been initiated for two ocular diseases. Adeno-associated viruses (AAVs) represent an attractive alternative to adenoviral vector systems as they enable stable and long-term expression and can target a variety of different ocular cell types depending on the capsid serotype; recently clinical trails for congenital blindness was initiated with a vector-based AAV serotype 2. High levels of retinal gene transfer have been achieved using vectors based on AAV serotypes 1, 2, 4 and 5. This report compares the gene transfer efficacy and stability of expression of vector systems based on three novel AAV serotypes: AAV7, 8, 9, with the established vectors AAV1, 2, 5. We show here that AAV7 and 8 enable superior long-term transduction of retinal and also anterior chamber structures.
Collapse
Affiliation(s)
- Corinna Lebherz
- Gene Therapy Program, Division of Medical Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Albert Maguire
- F.M. Kirby Center for Molecular Ophthalmology, Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Waixing Tang
- F.M. Kirby Center for Molecular Ophthalmology, Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean Bennett
- F.M. Kirby Center for Molecular Ophthalmology, Department of Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - James M. Wilson
- Gene Therapy Program, Division of Medical Genetics, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
28
|
Surace EM, Auricchio A. Versatility of AAV vectors for retinal gene transfer. Vision Res 2008; 48:353-9. [PMID: 17923143 DOI: 10.1016/j.visres.2007.07.027] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Revised: 07/31/2007] [Accepted: 07/31/2007] [Indexed: 12/21/2022]
Abstract
Gene therapy represents a promising therapeutic option for many inherited and acquired retinal diseases. Recombinant adeno-associated viral vectors (AAV) are the most efficient tools to transfer genes in vivo to the retina. The recent identification of dozens of novel AAV serotypes enormously expands on the versatility of AAV as vector system for in vivo somatic gene transfer. The results from the forthcoming trials with AAV in the retina of patients with Leber Congenital Amaurosis will be critical for the rapid development of AAV-based therapeutics for retinal diseases.
Collapse
|
29
|
Kiuru M, Crystal RG. Progress and prospects: gene therapy for performance and appearance enhancement. Gene Ther 2008; 15:329-37. [DOI: 10.1038/sj.gt.3303100] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
30
|
Abstract
This review will cover the state of the field in retinal degeneration and gene therapy with a focus on the great strides that have been made in retina gene therapy. Topics ranging from the development of animal models to clinical trials (for the treatment of Leber congenital amaurosis, age-related macular degeneration, and retinoblastoma) will be discussed. In addition, the results of gene therapy studies targeting the photoreceptors will be presented. Finally, strategies and progress in overcoming the challenges of photoreceptor-directed gene therapy will be presented.
Collapse
Affiliation(s)
- Tonia S Rex
- F.M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
31
|
Lamartina S, Cimino M, Roscilli G, Dammassa E, Lazzaro D, Rota R, Ciliberto G, Toniatti C. Helper-dependent adenovirus for the gene therapy of proliferative retinopathies: stable gene transfer, regulated gene expression and therapeutic efficacy. J Gene Med 2007; 9:862-74. [PMID: 17685494 DOI: 10.1002/jgm.1083] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Ocular neovascular disorders, such as diabetic retinopathy and age-related macular degeneration, are the principal causes of blindness in developed countries. Current treatments are of limited efficacy, whereas a therapy based on intraocular gene transfer of angiostatic factors represents a promising alternative. For the first time we have explored the potential of helper-dependent adenovirus (HD-Ad), the last generation of Ad vectors, in the therapy of retinal neovascularization. METHODS We first analyzed efficiency and stability of intraretinal gene transfer following intravitreous injection in mice. A HD-Ad vector expressing green fluorescent protein (GFP) under the control of the cytomegalovirus (CMV) promoter (HD-Ad/GFP) was compared with a first-generation (E1/E3-deleted) Ad vector carrying an identical GFP expression cassette (FG-Ad/GFP). We also constructed HD-Ad vectors expressing a soluble form of the VEGF receptor (sFlt-1) in a constitutive (HD-Ad/sFlt-1) or doxycycline (dox)-inducible (HD-Ad/S-M2/sFlt-1) manner and tested their therapeutic efficacy upon intravitreous delivery in a rat model of oxygen-induced retinopathy (OIR). RESULTS HD-Ad/GFP promoted long-lasting (up to 1 year) transgene expression in retinal Müller cells, in marked contrast with the short-term expression observed with FG-Ad/GFP. Intravitreous injection of HD-Ad vectors expressing sFlt-1 resulted in detectable levels of sFlt-1 and inhibited retinal neovascularization by more than 60% in a rat model of OIR. Notably, the therapeutic efficacy of the inducible vector HD-Ad/S-M2/sFlt-1 was strictly dox-dependent. CONCLUSIONS HD-Ad vectors enable stable gene transfer and regulated expression of angiostatic factors following intravitreous injection and thus are attractive vehicles for the gene therapy of neovascular diseases of the retina.
Collapse
|
32
|
Abstract
Gene transfer is being rigorously evaluated in the laboratory in the preparation for the development of clinical therapies. Many CNS diseases, which have proved more challenging to treat than peripheral disorders, are prime candidates for gene therapy. However, there are numerous considerations in the development of gene therapy, including delivery, maintenance of expression, transgene level regulation, toxicity of the viral vector system and safety of the gene product. The authors review these issues and discuss various approaches used in preclinical studies. Alzheimer's and Parkinson's disease are employed as models, in which much research has already been performed, to address disease-specific questions about gene therapy approaches.
Collapse
Affiliation(s)
- Deborah A Ryan
- University of Rochester School of Medicine & Dentistry, Interdepartmental Graduate Program in Neuroscience, Rochester, New York, USA
| | | |
Collapse
|
33
|
Ramaswamy S, McBride JL, Herzog CD, Brandon E, Gasmi M, Bartus RT, Kordower JH. Neurturin gene therapy improves motor function and prevents death of striatal neurons in a 3-nitropropionic acid rat model of Huntington's disease. Neurobiol Dis 2007; 26:375-84. [PMID: 17336076 DOI: 10.1016/j.nbd.2007.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Revised: 01/10/2007] [Accepted: 01/14/2007] [Indexed: 11/21/2022] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disease characterized by the selective loss of neurons in the striatum and cerebral cortex. This study tested the hypothesis that an adenoassociated viral (AAV2) vector encoding for the trophic factor neurturin (NTN) could provide neuroprotection in the rat 3-nitropropionic acid (3NP) model of HD. Rats received AAV2-NTN (CERE-120), AAV2-eGFP or Vehicle, followed 4 weeks later by the mitochondrial toxin 3NP. 3NP induced motor impairments were observed on the rotarod test, the platform test, and a clinical rating scale in all groups. However, each of these deficits was attenuated by AAV2-NTN (CERE-120). Stereological counts revealed a significant protection of NeuN-ir striatal neurons from 3NP toxicity by AAV2-NTN. These data support the concept that AAV2-NTN might be a valuable treatment for patients with Huntington's disease.
Collapse
Affiliation(s)
- Shilpa Ramaswamy
- Department of Neuroscience, Rush University Medical Center, 1735 West Harrison Street, Suite 300, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Nguyen M, Huan-Tu G, Gonzalez-Edick M, Rivera VM, Clackson T, Jooss KU, Harding TC. Rapamycin-regulated control of antiangiogenic tumor therapy following rAAV-mediated gene transfer. Mol Ther 2007; 15:912-20. [PMID: 17245354 DOI: 10.1038/mt.sj.6300079] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Regulated gene expression may be required for the clinical development of certain gene therapies. Several approaches have been developed that allow pharmacologic control of transgene expression, including the dimerizer-regulated transcriptional system in which rapamycin or its analogs function as transcriptional inducers. These compounds can also act as direct antitumor agents via inhibition of mammalian target of rapamycin (mTOR). We describe the development of an optimized recombinant adeno-associated virus (AAV) expression cassette that allows dimerizer-regulated gene expression from a single vector in vitro and in vivo. After demonstrating multiple cycles of rapamycin-dependent transgene induction following a single administration of an AAV vector in vivo, application of this regulated AAV gene expression system to the pharmacologic control of antiangiogenic therapy was evaluated in preclinical tumor models. Dimerizer-regulated vectors were constructed encoding a soluble inhibitor of the vascular endothelial growth factor (VEGF) pathway. In two subcutaneous models of glioblastoma, regulated expression of the VEGF inhibitor via recombinant AAV-mediated gene transfer, in combination with rapamycin, was shown to decrease tumor growth rate significantly. The dual properties of rapamycin--as a transcriptional inducer and mTOR inhibitor--are exploited in combination with an AAV-encoded antiangiogenic agent to provide a novel approach for the treatment of malignant diseases.
Collapse
Affiliation(s)
- Minh Nguyen
- Cell Genesys Inc., South San Francisco, California, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Nguyen M, Huan-Tu G, Gonzalez-Edick M, Rivera VM, Clackson T, Jooss KU, Harding TC. Rapamycin-regulated Control of Antiangiogenic Tumor Therapy Following rAAV-mediated Gene Transfer. Mol Ther 2007. [DOI: 10.1038/sj.mt.6300079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
36
|
Abstract
Controlling gene activity in space and time represents a cornerstone technology in gene and cell therapeutic applications, bioengineering, drug discovery as well as fundamental and applied research. This chapter provides a comprehensive overview of the different approaches for regulating gene activity and product protein formation at different biosynthetic levels, from genomic rearrangements over transcription and translation control to strategies for engineering inducible secretion and protein activity with a focus on the development during the past 2 years. Recent advances in designing second-generation gene switches, based on novel inducer administration routes (gas phase) as well as on the combination of heterologous switches with endogenous signals, will be complemented by an overview of the emerging field of mammalian synthetic biology, which enables the design of complex synthetic and semisynthetic gene networks. This article will conclude with an overview of how the different gene switches have been applied in gene therapy studies, bioengineering and drug discovery.
Collapse
Affiliation(s)
- W Weber
- Institute for Chemical and Bioengineering, ETH Zurich, ETH Hoenggerberg HCI F 115, Wolfgang-Pauli-Strasse 10, 8093 Zurich, Switzerland
| | | |
Collapse
|
37
|
Allocca M, Tessitore A, Cotugno G, Auricchio A. AAV-mediated gene transfer for retinal diseases. Expert Opin Biol Ther 2006; 6:1279-94. [PMID: 17223737 DOI: 10.1517/14712598.6.12.1279] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Vectors based on the adeno-associated virus (rAAV) are able to transduce the retina of animal models, including non-human primates, for a long-term period, safely and at sustained levels. The ability of the various rAAV serotypes to transduce retinal target cells has been exploited to successfully transfer genes to photoreceptors, retinal pigment epithelium and the inner retina, which are affected in many inherited and non-inherited blinding diseases. rAAV-mediated, constitutive and regulated gene expression at therapeutic levels has been achieved in the retina of animal models, thus providing proof-of-principle of gene therapy efficacy and safety in models of dominant and recessive retinal disorders. In addition, gene transfer of molecules with either neurotrophic or antiangiogenic properties provides useful alternatives to the classic gene replacement for treatment of both mendelian and complex traits affecting the retina. Years of successful rAAV-mediated gene transfer to the retina have resulted in restoration of vision in dogs affected with congenital blindness. This has paved the way to the first attempts at treating inherited retinal diseases in humans with rAAV. Although the results of rAAV clinical trials for non-retinal diseases give a warning that the outcome of viral-mediated gene transfer in humans may be different from that predicted based on results in other species, the immune privilege of the retina combined with the versatility of rAAV serotypes may ultimately provide the first successful treatment of human inherited diseases using rAAV.
Collapse
Affiliation(s)
- Mariacarmela Allocca
- Telethon Institute of Genetics and Medicine (TIGEM), Via P. Castellino, 111. 80131 Napoli, Italy.
| | | | | | | |
Collapse
|
38
|
Abstract
The eye has unique advantages as a target organ for gene therapy of both inherited and acquired ocular disorders and offers a valuable model system for gene therapy. The eye is readily accessible to phenotypic examination and investigation of therapeutic effects in vivo by fundus imaging and electrophysiological techniques. Considerable progress has been made in the development of gene replacement therapies for retinal degenerations resulting from gene defects in photoreceptor cells (rds, RPGRIP, RS-1) and in retinal pigment epithelial cells (MerTK, RPE65, OA1) using recombinant adeno-associated virus and lentivirus-based vectors. Gene therapy also offers a potentially powerful approach to the treatment of complex acquired disorders such as those involving angiogenesis, inflammation and degeneration, by the targeted sustained intraocular delivery of therapeutic proteins. Proposals for clinical trials of gene therapy for early-onset retinal degeneration owing to defects in the gene encoding the visual cycle protein RPE65 have recently received ethical approval.
Collapse
Affiliation(s)
- J W B Bainbridge
- Division of Molecular Therapy, Institute of Ophthalmology, University College London, London, UK
| | | | | |
Collapse
|
39
|
Shen WY, Lai CM, Graham CE, Binz N, Lai YKY, Eade J, Guidolin D, Ribatti D, Dunlop SA, Rakoczy PE. Long-term global retinal microvascular changes in a transgenic vascular endothelial growth factor mouse model. Diabetologia 2006; 49:1690-701. [PMID: 16752188 DOI: 10.1007/s00125-006-0274-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2005] [Accepted: 03/20/2006] [Indexed: 01/29/2023]
Abstract
AIMS/HYPOTHESIS Vascular endothelial growth factor (VEGF) plays a pivotal role in the pathogenesis of diabetic retinopathy. We investigated whether transgenic mice with moderate VEGF expression in photoreceptors (trVEGF029) developed changes similar to diabetic retinopathy and whether retinopathy progressed with time. MATERIALS AND METHODS Human VEGF(165) (hVEGF(165)) expression was analysed using ELISA and quantitative RT-PCR; serum glucose levels were also measured. Fundus fluorescein angiography (FA) was used to screen the degree of retinopathy from 6 weeks. Dynamic changes in the density of retinal microvasculature, as well as other changes similar to diabetic retinopathy, including retinal leucostasis, capillary endothelial cell and pericyte loss, and numbers of acellular capillaries, were quantified. RESULTS trVEGF029 mice were normoglycaemic and showed a moderate, short-term hVEGF(165) upregulation for up to 3 weeks. Changes in the retinal microvasculature not only mimicked those seen in diabetic retinopathy, but also showed similar pathological progression with time. FA at 6 weeks identified two phenotypes, mild and moderate, which were distinguished by the extent of vascular leakage. Quantitative analysis of diabetic retinopathy-like changes revealed that these parameters were tightly correlated with the initial degree of vascular leakage; low levels reflected slow and limited retinal microvascular changes in mild cases and high levels reflected more rapid and extensive changes in moderate cases. CONCLUSIONS/INTERPRETATION The data suggest that even an early short-term elevation in hVEGF(165) expression might set a train of events that lead to progressive retinopathy. Induction of many features characteristic of diabetic retinopathy in trVEGF029 enables mechanisms leading to the disease state to be examined, and provides a relevant animal model for testing novel therapeutics.
Collapse
Affiliation(s)
- W-Y Shen
- Centre of Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Perth, WA, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Stieger K, Le Meur G, Lasne F, Weber M, Deschamps JY, Nivard D, Mendes-Madeira A, Provost N, Martin L, Moullier P, Rolling F. Long-term doxycycline-regulated transgene expression in the retina of nonhuman primates following subretinal injection of recombinant AAV vectors. Mol Ther 2006; 13:967-75. [PMID: 16442848 DOI: 10.1016/j.ymthe.2005.12.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2005] [Revised: 12/13/2005] [Accepted: 12/13/2005] [Indexed: 10/25/2022] Open
Abstract
Adeno-associated viral gene therapy has shown promise for the treatment of inherited and acquired retinal disorders. In most applications, regulation of expression is a critical concern for both safety and efficacy. The purpose of our study was to evaluate the ability of the tetracycline-regulatable system to establish long-term transgene regulation in the retina of nonhuman primates. Three rAAV vectors expressing the tetracycline-dependent transactivator (rtTA) under the control of either the ubiquitous CAG promoter or the specific RPE65 promoter (AAV2/5.CAG.TetOn.epo, AAV2/4.CAG.TetOn.epo, and AAV2/4.RPE65.TetOn.epo) were generated and administered subretinally to seven macaques. We demonstrated that repeated inductions of transgene expression in the nonhuman primate retina can be achieved using a Tet-inducible system via rAAV vector administration over a long period (2.5 years). Maximum erythropoietin (EPO) secretion in the anterior chamber depends upon the rAAV serotype and the nature of the promoter driving rtTA expression. We observed that the EPO isoforms produced in the retina differ from one another based on the transduced cell type of origin within the retina and also differ from both the physiological EPO isoforms and the isoforms produced by AAV-transduced skeletal muscle.
Collapse
Affiliation(s)
- Knut Stieger
- INSERM UMR U649, CHU-Hotel Dieu, Bât. J. Monnet, 30 Avenue J. Monnet, 44035 Nantes Cedex 01, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Affiliation(s)
- Jean Bennett
- Ophthalmology, Cell and Developmental Biology at the University of Pennsylvania School of Medicine in Philadelphia, PA, USA
| |
Collapse
|
42
|
Le Bec C, Douar AM. Gene Therapy Progress and Prospects – Vectorology: design and production of expression cassettes in AAV vectors. Gene Ther 2006; 13:805-13. [PMID: 16453010 DOI: 10.1038/sj.gt.3302724] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adeno-associated virus (AAV) derived vectors are considered highly eligible vehicles for human gene therapy. Not only do they possess many great potential for clinical applications due to their wide range of tissue targets but also their excellent preclinical safety profile makes them particularly suitable candidates for treating serious diseases. Initial clinical trials have yielded encouraging results and prompted further improvements in their design and methods of production. Many studies have been performed to modify the tropism of recombinant (r)AAV by capsid modification. However, the precise control of spatial and temporal gene expression, which may be important in determining the safety and efficacy of gene transfer, lies in a rational choice and a subtle combination of various regulatory genetic elements to be inserted into the expression cassette. Moreover, new strategies based on such genetic sequences open new perspectives for enhancing vector genome persistence, disrupting or reducing pathogenic gene expression and even targeting genes.
Collapse
Affiliation(s)
- C Le Bec
- CNRS UMR 8115, Généthon, Evry, France
| | | |
Collapse
|
43
|
Mandel RJ, Manfredsson FP, Foust KD, Rising A, Reimsnider S, Nash K, Burger C. Recombinant adeno-associated viral vectors as therapeutic agents to treat neurological disorders. Mol Ther 2006; 13:463-83. [PMID: 16412695 DOI: 10.1016/j.ymthe.2005.11.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2005] [Revised: 11/12/2005] [Accepted: 11/13/2005] [Indexed: 12/11/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) is derived from a small human parvovirus with an excellent safety profile. In addition, this viral vector efficiently transduces and supports long-term transgene expression in the nervous system. These properties make rAAV a reasonable candidate vector for treating neurological disorders. Indeed, rAAV is currently being used in five early stage clinical trials for various neurodegenerative disorders. Therefore, we will review the currently available preclinical data using rAAV in animal models of central nervous system (CNS) disorders. Moreover, potential caveats for rAAV-based gene therapy in the CNS are also presented.
Collapse
Affiliation(s)
- Ronald J Mandel
- Department of Neuroscience, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Pharmacologic transgene-expression dosing is considered essential for future gene therapy scenarios. Genetic interventions require precise transcription or translation fine-tuning of therapeutic transgenes to enable their titration into the therapeutic window, to adapt them to daily changing dosing regimes of the patient, to integrate them seamlessly into the patient's transcriptome orchestra, and to terminate their expression after successful therapy. In recent years, decisive progress has been achieved in designing high-precision trigger-inducible mammalian transgene control modalities responsive to clinically licensed and inert heterologous molecules or to endogenous physiologic signals. Availability of a portfolio of compatible transcription control systems has enabled assembly of higher-order control circuitries providing simultaneous or independent control of several transgenes and the design of (semi-)synthetic gene networks, which emulate digital expression switches, regulatory transcription cascades, epigenetic expression imprinting, and cellular transcription memories. This review provides an overview of cutting-edge developments in transgene control systems, of the design of synthetic gene networks, and of the delivery of such systems for the prototype treatment of prominent human disease phenotypes.
Collapse
Affiliation(s)
- Wilfried Weber
- Institute for Chemical and Bio-Engineering, Swiss Federal Institute of Technology Zurich-ETH Zurich, ETH Hoenggerberg HCI F 115, Wolfgang-Pauli-Strasse 10, CH-8093 Zurich, Switzerland
| | | |
Collapse
|