1
|
Wiggins KB, Winston SM, Reeves IL, Gaevert J, Spence Y, Brimble MA, Livingston B, Morton CL, Thomas PG, Sant AJ, Ross TM, Davidoff AM, Schultz-Cherry S. rAAV expressing a COBRA-designed influenza hemagglutinin generates a protective and durable adaptive immune response with a single dose. J Virol 2024; 98:e0078124. [PMID: 39078191 PMCID: PMC11338075 DOI: 10.1128/jvi.00781-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 07/31/2024] Open
Abstract
Influenza remains a worldwide public health threat. Although seasonal influenza vaccines are currently the best means of preventing severe disease, the standard-of-care vaccines require frequent updating due to antigenic drift and can have low efficacy, particularly in vulnerable populations. Here, we demonstrate that a single administration of a recombinant adenovirus-associated virus (rAAV) vector expressing a computationally optimized broadly reactive antigen (COBRA)-derived influenza H1 hemagglutinin (HA) induces strongly neutralizing and broadly protective antibodies in naïve mice and ferrets with pre-existing influenza immunity. Following a lethal viral challenge, the rAAV-COBRA vaccine allowed for significantly reduced viral loads in the upper and lower respiratory tracts and complete protection from morbidity and mortality that lasted for at least 5 months post-vaccination. We observed no signs of antibody waning during this study. CpG motif enrichment of the antigen can act as an internal adjuvant to further enhance the immune responses to allow for lower vaccine dosages with the induction of unique interferon-producing CD4+ and CD8+ T cells specific to HA head and stem peptide sequences. Our studies highlight the utility of rAAV as an effective platform to improve seasonal influenza vaccines. IMPORTANCE Developing an improved seasonal influenza vaccine remains an ambitious goal of researchers and clinicians alike. With influenza routinely causing severe epidemics with the potential to rise to pandemic levels, it is critical to create an effective, broadly protective, and durable vaccine to improve public health worldwide. As a potential solution, we created a rAAV viral vector expressing a COBRA-optimized influenza hemagglutinin antigen with modestly enriched CpG motifs to evoke a robust and long-lasting immune response after a single intramuscular dose without needing boosts or adjuvants. Importantly, the rAAV vaccine boosted antibody breadth to future strains in ferrets with pre-existing influenza immunity. Together, our data support further investigation into the utility of viral vectors as a potential avenue to improve our seasonal influenza vaccines.
Collapse
Affiliation(s)
- Kristin B. Wiggins
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Stephen M. Winston
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Isaiah L. Reeves
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Jessica Gaevert
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Yunyu Spence
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Mark A. Brimble
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Brandi Livingston
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Christopher L. Morton
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Paul G. Thomas
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Andrea J. Sant
- David H. Smith Center
for Vaccine Biology and Immunology, Department of Microbiology and
Immunology, University of Rochester Medical
Center, Rochester, New
York, USA
| | - Ted M. Ross
- Department of
Infectious Biology, Cleveland Clinic,
Cleveland, Ohio, USA
- Cleveland Clinic,
Florida Research and Innovation Center,
Port St. Lucie, Florida,
USA
| | - Andrew M. Davidoff
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Stacey Schultz-Cherry
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| |
Collapse
|
2
|
Greig JA, Martins KM, Breton C, Lamontagne RJ, Zhu Y, He Z, White J, Zhu JX, Chichester JA, Zheng Q, Zhang Z, Bell P, Wang L, Wilson JM. Integrated vector genomes may contribute to long-term expression in primate liver after AAV administration. Nat Biotechnol 2024; 42:1232-1242. [PMID: 37932420 PMCID: PMC11324525 DOI: 10.1038/s41587-023-01974-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/29/2023] [Indexed: 11/08/2023]
Abstract
The development of liver-based adeno-associated virus (AAV) gene therapies is facing concerns about limited efficiency and durability of transgene expression. We evaluated nonhuman primates following intravenous dosing of AAV8 and AAVrh10 vectors for over 2 years to better define the mechanism(s) of transduction that affect performance. High transduction of non-immunogenic transgenes was achieved, although expression declined over the first 90 days to reach a lower but stable steady state. More than 10% of hepatocytes contained single nuclear domains of vector DNA that persisted despite the loss of transgene expression. Greater reductions in vector DNA and RNA were observed with immunogenic transgenes. Genomic integration of vector sequences, including complex concatemeric structures, were detected in 1 out of 100 cells at broadly distributed loci that were not in proximity to genes associated with hepatocellular carcinoma. Our studies suggest that AAV-mediated transgene expression in primate hepatocytes occurs in two phases: high but short-lived expression from episomal genomes, followed by much lower but stable expression, likely from integrated vectors.
Collapse
Affiliation(s)
- Jenny A Greig
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly M Martins
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Camilo Breton
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - R Jason Lamontagne
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yanqing Zhu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhenning He
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John White
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jing-Xu Zhu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica A Chichester
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qi Zheng
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhe Zhang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lili Wang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Liu S, Chowdhury EA, Xu V, Jerez A, Mahmood L, Ly BQ, Le HK, Nguyen A, Rajwade A, Meno-Tetang G, Shah DK. Whole-Body Disposition and Physiologically Based Pharmacokinetic Modeling of Adeno-Associated Viruses and the Transgene Product. J Pharm Sci 2024; 113:141-157. [PMID: 37805073 DOI: 10.1016/j.xphs.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
To facilitate model-informed drug development (MIDD) of adeno-associated virus (AAV) therapy, here we have developed a physiologically based pharmacokinetic (PBPK) model for AAVs following preclinical investigation in mice. After 2E11 Vg/mouse dose of AAV8 and AAV9 encoding a monoclonal antibody (mAb) gene, whole-body disposition of both the vector and the transgene mAb was evaluated over 3 weeks. At steady-state, the following tissue-to-blood (T/B) concentration ratios were found for AAV8/9: ∼50 for liver; ∼10 for heart and muscle; ∼2 for brain, lung, kidney, adipose, and spleen; ≤1 for bone, skin, and pancreas. T/B values for mAb were compared with the antibody biodistribution coefficients, and five different clusters of organs were identified based on their transgene expression profile. All the biodistribution data were used to develop a novel AAV PBPK model that incorporates: (i) whole-body distribution of the vector; (ii) binding, internalization, and intracellular processing of the vector; (iii) transgene expression and secretion; and (iv) whole-body disposition of the secreted transgene product. The model was able to capture systemic and tissue PK of the vector and the transgene-produced mAb reasonably well. Pathway analysis of the PBPK model suggested that liver, muscle, and heart are the main contributors for the secreted transgene mAb. Unprecedented PK data and the novel PBPK model developed here provide the foundation for quantitative systems pharmacology (QSP) investigations of AAV-mediated gene therapies. The PBPK model can also serve as a quantitative tool for preclinical study design and preclinical-to-clinical translation of AAV-based gene therapies.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Ekram Ahmed Chowdhury
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Vivian Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Anthony Jerez
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Leeha Mahmood
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Bao Quoc Ly
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Huyen Khanh Le
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Anne Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Aneesh Rajwade
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Guy Meno-Tetang
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
4
|
Zhao L, Yang Z, Zheng M, Shi L, Gu M, Liu G, Miao F, Chang Y, Huang F, Tang N. Recombinant adeno-associated virus 8 vector in gene therapy: Opportunities and challenges. Genes Dis 2024; 11:283-293. [PMID: 37588223 PMCID: PMC10425794 DOI: 10.1016/j.gendis.2023.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/26/2022] [Accepted: 02/08/2023] [Indexed: 04/09/2023] Open
Abstract
In recent years, significant breakthroughs have been made in the field of gene therapy. Adeno-associated virus (AAV) is one of the most promising gene therapy vectors and a powerful tool for delivering the gene of interest. Among the AAV vectors, AAV serotype 8 (AAV8) has attracted much attention for its efficient and stable gene transfection into specific tissues. Currently, recombinant AAV8 has been widely used in gene therapy research on a variety of diseases, including genetic diseases, cancers, autoimmune diseases, and viral diseases. This paper reviewed the applications and challenges of using AAV8 as a vector for gene therapy, with the aim of providing a valuable resource for those pursuing the application of viral vectors in gene therapy.
Collapse
Affiliation(s)
- Liyuan Zhao
- Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230000, China
- Yangtze Delta Drug Advanced Research Institute, Yangtze Delta Pharmaceutical College, Nantong, Jiangsu 226133, China
- InnoStar Bio-tech Nantong Co., Ltd., Nantong, Jiangsu 226133, China
| | - Zixuan Yang
- Shanghai Innostar Bio-Technology Co., Ltd, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Minhui Zheng
- Shanghai Innostar Bio-Technology Co., Ltd, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Lei Shi
- Shanghai Innostar Bio-Technology Co., Ltd, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Mengyun Gu
- Shanghai Innostar Bio-Technology Co., Ltd, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Gang Liu
- InnoStar Bio-tech Nantong Co., Ltd., Nantong, Jiangsu 226133, China
| | - Feng Miao
- InnoStar Bio-tech Nantong Co., Ltd., Nantong, Jiangsu 226133, China
| | - Yan Chang
- Shanghai Innostar Bio-Technology Co., Ltd, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Fanghua Huang
- Center for Drug Evaluation, National Medical Products Administration, Beijing 100022, China
| | - Naping Tang
- Yangtze Delta Drug Advanced Research Institute, Yangtze Delta Pharmaceutical College, Nantong, Jiangsu 226133, China
- Shanghai Innostar Bio-Technology Co., Ltd, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| |
Collapse
|
5
|
Furuno K, Elvitigala KCML, Suzuki K, Sakai S. Local delivery of adeno-associated viral vectors with electrospun gelatin nanofiber mats. J Biomed Mater Res B Appl Biomater 2024; 112:e35345. [PMID: 37902433 DOI: 10.1002/jbm.b.35345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/28/2023] [Accepted: 10/14/2023] [Indexed: 10/31/2023]
Abstract
Adeno-associated viral (AAV) vectors play a significant role in gene therapy, yet the typical delivery methods, like systemic and local AAV injections, often lead to unintended off-target distribution and tissue damage due to injection. In this study, we propose a localized delivery approach for AAV vectors utilizing electrospun gelatin nanofiber mats, which are cross-linked with glutaraldehyde. The AAV vectors, which encoded a green fluorescent protein (GFP), were loaded onto the mats by immersing them in a solution containing the vectors. The amount of AAV vector loaded onto the mats increased as the vector concentration in the solution increased. The loaded AAV vector was steadily released into the cell culture medium over 3 days. The mats incubated for 3 days also showed the ability to transduce into the cells cultured on them. We evaluated the effectiveness of this delivery system by attaching the mats to mouse livers. GFP expression was visible on the surface of the liver beneath the attached mats, but not in areas in direct contact with the mats. These findings suggest that the attachment of AAV vector-loaded electrospun gelatin nanofiber mats to a target site present a promising solution for localized gene delivery while reducing off-target distribution.
Collapse
Affiliation(s)
- Kotoko Furuno
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | | | - Keiichiro Suzuki
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Osaka, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan
- Graduate School of Frontier Bioscience, Osaka University, Osaka, Japan
| | - Shinji Sakai
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| |
Collapse
|
6
|
Mehta N, Gilbert R, Chahal PS, Moreno MJ, Nassoury N, Coulombe N, Lytvyn V, Mercier M, Fatehi D, Lin W, Harvey EM, Zhang LH, Nazemi-Moghaddam N, Elahi SM, Ross CJD, Stanimirovic DB, Hayden MR. Preclinical Development and Characterization of Novel Adeno-Associated Viral Vectors for the Treatment of Lipoprotein Lipase Deficiency. Hum Gene Ther 2023; 34:927-946. [PMID: 37597209 DOI: 10.1089/hum.2023.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2023] Open
Abstract
Lipoprotein lipase deficiency (LPLD) results from mutations within the lipoprotein lipase (LPL) gene that lead to a complete lack of catalytically active LPL protein. Glybera was one of the first adeno-associated virus (AAV) gene replacement therapy to receive European Medicines Agency regulatory approval for the treatment of LPLD. However, Glybera is no longer marketed potentially due to a combination of economical, manufacturing, and vector-related issues. The aim of this study was to develop a more efficacious AAV gene therapy vector for LPLD. Following preclinical biodistribution, efficacy and non-Good Laboratory Practice toxicity studies with novel AAV1 and AAV8-based vectors in mice, we identified AAV8 pVR59. AAV8 pVR59 delivered a codon-optimized, human gain-of-function hLPLS447X transgene driven by a CAG promoter in an AAV8 capsid. AAV8 pVR59 was significantly more efficacious, at 10- to 100-fold lower doses, compared with an AAV1 vector based on Glybera, when delivered intramuscularly or intravenously, respectively, in mice with LPLD. Efficient gene transfer was observed within the injected skeletal muscle and liver following delivery of AAV8 pVR59, with long-term correction of LPLD phenotypes, including normalization of plasma triglycerides and lipid tolerance, for up to 6 months post-treatment. While intramuscular delivery of AAV8 pVR59 was well tolerated, intravenous administration augmented liver pathology. These results highlight the feasibility of developing a superior AAV vector for the treatment of LPLD and provide critical insight for initiating studies in larger animal models. The identification of an AAV gene therapy vector that is more efficacious at lower doses, when paired with recent advances in production and manufacturing technologies, will ultimately translate to increased safety and accessibility for patients.
Collapse
Affiliation(s)
- Neel Mehta
- Department of Medical Genetics, Center for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Rénald Gilbert
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, Canada
- Department of Bioengineering, McGill University, Montréal, Canada
| | - Parminder S Chahal
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, Canada
| | - Maria J Moreno
- Department of Translational Biosciences, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, Canada
| | - Nasha Nassoury
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, Canada
| | - Nathalie Coulombe
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, Canada
| | - Viktoria Lytvyn
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, Canada
| | - Mario Mercier
- Department of Translational Biosciences, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, Canada
| | - Dorothy Fatehi
- Department of Translational Biosciences, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, Canada
| | - Wendy Lin
- Department of Medical Genetics, Center for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Emily M Harvey
- Department of Medical Genetics, Center for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Lin-Hua Zhang
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Nazila Nazemi-Moghaddam
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, Canada
| | - Seyyed Mehdy Elahi
- Department of Production Platforms and Analytics, Human Health Therapeutics Research Center, National Research Council Canada, Montréal, Canada
| | - Colin J D Ross
- Department of Medical Genetics, Center for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Danica B Stanimirovic
- Department of Translational Biosciences, Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, Canada
| | - Michael R Hayden
- Department of Medical Genetics, Center for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| |
Collapse
|
7
|
Martino RA, Wang Q, Xu H, Hu G, Bell P, Arroyo EJ, Sims JJ, Wilson JM. Vector Affinity and Receptor Distribution Define Tissue-Specific Targeting in an Engineered AAV Capsid. J Virol 2023; 97:e0017423. [PMID: 37199615 PMCID: PMC10308920 DOI: 10.1128/jvi.00174-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/20/2023] [Indexed: 05/19/2023] Open
Abstract
Unbiased in vivo selections of diverse capsid libraries can yield engineered capsids that overcome gene therapy delivery challenges like traversing the blood-brain barrier (BBB), but little is known about the parameters of capsid-receptor interactions that govern their improved activity. This hampers broader efforts in precision capsid engineering and is a practical impediment to ensuring the translatability of capsid properties between preclinical animal models and human clinical trials. In this work, we utilize the adeno-associated virus (AAV)-PHP.B-Ly6a model system to better understand the targeted delivery and BBB penetration properties of AAV vectors. This model offers a defined capsid-receptor pair that can be used to systematically define relationships between target receptor affinity and in vivo activity of engineered AAV vectors. Here, we report a high-throughput method for quantifying capsid-receptor affinity and demonstrate that direct binding assays can be used to organize a vector library into families with varied affinity for their target receptor. Our data indicate that efficient central nervous system transduction requires high levels of target receptor expression at the BBB, but it is not a requirement for receptor expression to be limited to the target tissue. We observed that enhanced receptor affinity leads to reduced transduction of off-target tissues but can negatively impact on-target cellular transduction and penetration of endothelial barriers. Together, this work provides a set of tools for defining vector-receptor affinities and demonstrates how receptor expression and affinity interact to impact the performance of engineered AAV vectors in targeting the central nervous system. IMPORTANCE Novel methods for measuring adeno-associated virus (AAV)-receptor affinities, especially in relation to vector performance in vivo, would be useful to capsid engineers as they develop AAV vectors for gene therapy applications and characterize their interactions with native or engineered receptors. Here, we use the AAV-PHP.B-Ly6a model system to assess the impact of receptor affinity on the systemic delivery and endothelial penetration properties of AAV-PHP.B vectors. We discuss how receptor affinity analysis can be used to isolate vectors with optimized properties, improve the interpretation of library selections, and ultimately translate vector activities between preclinical animal models and humans.
Collapse
Affiliation(s)
- R. Alexander Martino
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qiang Wang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hao Xu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gui Hu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edgardo J. Arroyo
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joshua J. Sims
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James M. Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Wilson DR, Tzeng SY, Rui Y, Neshat SY, Conge MJ, Luly KM, Wang E, Firestone JL, McAuliffe J, Maruggi G, Jalah R, Johnson R, Doloff JC, Green JJ. Biodegradable Polyester Nanoparticle Vaccines Deliver Self-Amplifying mRNA in Mice at Low Doses. ADVANCED THERAPEUTICS 2023; 6:2200219. [PMID: 37743930 PMCID: PMC10516528 DOI: 10.1002/adtp.202200219] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Indexed: 02/19/2023]
Abstract
Delivery of self-amplifying mRNA (SAM) has high potential for infectious disease vaccination due its self-adjuvating and dose-sparing properties. Yet a challenge is the susceptibility of SAM to degradation and the need for SAM to reach the cytosol fully intact to enable self-amplification. Lipid nanoparticles have been successfully deployed at incredible speed for mRNA vaccination, but aspects such as cold storage, manufacturing, efficiency of delivery, and the therapeutic window would benefit from further improvement. To investigate alternatives to lipid nanoparticles, we developed a class of >200 biodegradable end-capped lipophilic poly(beta-amino ester)s (PBAEs) that enable efficient delivery of SAM in vitro and in vivo as assessed by measuring expression of SAM encoding reporter proteins. We evaluated the ability of these polymers to deliver SAM intramuscularly in mice, and identified a polymer-based formulation that yielded up to 37-fold higher intramuscular (IM) expression of SAM compared to injected naked SAM. Using the same nanoparticle formulation to deliver a SAM encoding rabies virus glycoprotein, the vaccine elicited superior immunogenicity compared to naked SAM delivery, leading to seroconversion in mice at low RNA injection doses. These biodegradable nanomaterials may be useful in the development of next-generation RNA vaccines for infectious diseases.
Collapse
Affiliation(s)
- David R Wilson
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuan Rui
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sarah Y Neshat
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Marranne J Conge
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kathryn M Luly
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ellen Wang
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | - Joshua C Doloff
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Departments of Chemical & Biomolecular Engineering, Materials Science & Engineering, Neurosurgery, Oncology, and Ophthalmology, Sidney Kimmel Comprehensive Cancer Center and Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD 21231, USA
| |
Collapse
|
9
|
Davis-Gardner ME, Weber JA, Xie J, Pekrun K, Alexander EA, Weisgrau KL, Furlott JR, Rakasz EG, Kay MA, Gao G, Farzan M, Gardner MR. A strategy for high antibody expression with low anti-drug antibodies using AAV9 vectors. Front Immunol 2023; 14:1105617. [PMID: 37153616 PMCID: PMC10161250 DOI: 10.3389/fimmu.2023.1105617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Use of adeno-associated virus (AAV) vectors is complicated by host immune responses that can limit transgene expression. Recent clinical trials using AAV vectors to deliver HIV broadly neutralizing antibodies (bNAbs) by intramuscular administration resulted in poor expression with anti-drug antibodies (ADA) responses against the bNAb. Methods Here we compared the expression of, and ADA responses against, an anti-SIV antibody ITS01 when delivered by five different AAV capsids. We first evaluated ITS01 expression from AAV vectors three different 2A peptides. Rhesus macaques were selected for the study based on preexisiting neutralizing antibodies by evaluating serum samples in a neutralization assay against the five capsids used in the study. Macaques were intramuscularly administered AAV vectors at a 2.5x10^12 vg/kg over eight administration sites. ITS01 concentrations and anti-drug antibodies (ADA) were measured by ELISA and a neutralization assay was conducted to confirm ex vivo antibody potency. Results We observed that ITS01 expressed three-fold more efficiently in mice from AAV vectors in which heavy and light-chain genes were separated by a P2A ribosomal skipping peptide, compared with those bearing F2A or T2A peptides. We then measured the preexisting neutralizing antibody responses against three traditional AAV capsids in 360 rhesus macaques and observed that 8%, 16%, and 42% were seronegative for AAV1, AAV8, and AAV9, respectively. Finally, we compared ITS01 expression in seronegative macaques intramuscularly transduced with AAV1, AAV8, or AAV9, or with the synthetic capsids AAV-NP22 or AAV-KP1. We observed at 30 weeks after administration that AAV9- and AAV1-delivered vectors expressed the highest concentrations of ITS01 (224 µg/mL, n=5, and 216 µg/mL, n=3, respectively). The remaining groups expressed an average of 35-73 µg/mL. Notably, ADA responses against ITS01 were observed in six of the 19 animals. Lastly, we demonstrated that the expressed ITS01 retained its neutralizing activity with nearly the same potency of purified recombinant protein. Discussion Overall, these data suggest that the AAV9 capsid is a suitable choice for intramuscular expression of antibodies in nonhuman primates.
Collapse
Affiliation(s)
- Meredith E. Davis-Gardner
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Jesse A. Weber
- Department of Immunology and Microbiology, University of Florida (UF) Scripps Biomedical Research, University of Florida, Jupiter, FL, United States
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Katja Pekrun
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA, United States
| | - Eric A. Alexander
- Wisconsin National Primate Research Center, University of Madison-Wisconsin, Madison, WI, United States
| | - Kim L. Weisgrau
- Wisconsin National Primate Research Center, University of Madison-Wisconsin, Madison, WI, United States
| | - Jessica R. Furlott
- Wisconsin National Primate Research Center, University of Madison-Wisconsin, Madison, WI, United States
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Madison-Wisconsin, Madison, WI, United States
| | - Mark A. Kay
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Michael Farzan
- Department of Immunology and Microbiology, University of Florida (UF) Scripps Biomedical Research, University of Florida, Jupiter, FL, United States
| | - Matthew R. Gardner
- Department of Medicine, Division of Infectious Diseases, Emory University, Atlanta, GA, United States
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| |
Collapse
|
10
|
Esmagambetov IB, Ryabova EI, Derkaev AA, Shcheblyakov DV, Dolzhikova IV, Favorskaya IA, Grousova DM, Dovgiy MA, Prokofiev VV, Gosudarev AI, Byrikhina DV, Zorkov ID, Iliukhina AA, Kovyrshina AV, Shelkov AY, Naroditsky BS, Logunov DY, Gintsburg AL. rAAV expressing recombinant antibody for emergency prevention and long-term prophylaxis of COVID-19. Front Immunol 2023; 14:1129245. [PMID: 37063833 PMCID: PMC10098153 DOI: 10.3389/fimmu.2023.1129245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
IntroductionNumerous agents for prophylaxis of SARS-CoV-2-induced diseases are currently registered for the clinical use. Formation of the immunity happens within several weeks following vaccine administration which is their key disadvantage. In contrast, drugs based on monoclonal antibodies, enable rapid passive immunization and therefore can be used for emergency pre- and post-exposure prophylaxis of COVID-19. However rapid elimination of antibody-based drugs from the circulation limits their usage for prolonged pre-exposure prophylaxis.MethodsIn current work we developed a recombinant adeno-associated viral vector (rAAV), expressing a SARS-CoV-2 spike receptor-binding domain (RBD)-specific antibody P2C5 fused with a human IgG1 Fc fragment (P2C5-Fc) using methods of molecular biotechnology and bioprocessing.Results and discussionsA P2C5-Fc antibody expressed by a proposed rAAV (rAAV-P2C5-Fc) was shown to circulate within more than 300 days in blood of transduced mice and protect animals from lethal SARS-CoV-2 virus (B.1.1.1 and Omicron BA.5 variants) lethal dose of 105 TCID50. In addition, rAAV-P2C5-Fc demonstrated 100% protective activity as emergency prevention and long-term prophylaxis, respectively. It was also demonstrated that high titers of neutralizing antibodies to the SARS-CoV-2 virus were detected in the blood serum of animals that received rAAV-P2C5-Fc for more than 10 months from the moment of administration.Our data therefore indicate applicability of an rAAV for passive immunization and induction of a rapid long-term protection against various SARS-CoV-2 variants.
Collapse
|
11
|
Pires Ferreira D, Gruntman AM, Flotte TR. Gene therapy for alpha-1 antitrypsin deficiency: an update. Expert Opin Biol Ther 2023; 23:283-291. [PMID: 36825473 DOI: 10.1080/14712598.2023.2183771] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
INTRODUCTION Altering the human genetic code has been explored since the early 1990s as a definitive answer for the treatment of monogenic and acquired diseases which do not respond to conventional therapies. In Alpha-1 antitrypsin deficiency (AATD) the proper synthesis and secretion of alpha-1 antitrypsin (AAT) protein is impaired, leading to its toxic hepatic accumulation along with its pulmonary insufficiency, which is associated with parenchymal proteolytic destruction. Because AATD is caused by mutations in a single gene whose correction alone would normalize the mutant phenotype, it has become a popular target for both augmentation gene therapy and gene editing. Although gene therapy products are already a reality for the treatment of some pathologies, such as inherited retinal dystrophy and spinal muscular atrophy, AATD-related pulmonary and, especially, liver diseases still lack effective therapeutic options. AREAS COVERED Here, we review the course, challenges, and achievements of AATD gene therapy as well as update on new strategies being developed. EXPERT OPINION Reaching safe and clinically effective expression of the AAT is currently the greatest challenge for AATD gene therapy. The improvement and emergence of technologies that use gene introduction, silencing and correction hold promise for the treatment of AATD.
Collapse
Affiliation(s)
- Debora Pires Ferreira
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Alisha M Gruntman
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Terence R Flotte
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
12
|
Hahn PA, Martins MA. Adeno-associated virus-vectored delivery of HIV biologics: the promise of a "single-shot" functional cure for HIV infection. J Virus Erad 2023; 9:100316. [PMID: 36915910 PMCID: PMC10005911 DOI: 10.1016/j.jve.2023.100316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
The ability of immunoglobulin-based HIV biologics (Ig-HIV), including broadly neutralizing antibodies, to suppress viral replication in pre-clinical and clinical studies illustrates how these molecules can serve as alternatives or adjuncts to antiretroviral therapy for treating HIV infection. However, the current paradigm for delivering Ig-HIVs requires repeated passive infusions, which faces both logistical and economic challenges to broad-scale implementation. One promising way to overcome these obstacles and achieve sustained expression of Ig-HIVs in vivo involves the transfer of Ig-HIV genes to host cells utilizing adeno-associated virus (AAV) vectors. Because AAV vectors are non-pathogenic and their genomes persist in the cell nucleus as episomes, transgene expression can last for as long as the AAV-transduced cell lives. Given the long lifespan of myocytes, skeletal muscle is a preferred tissue for AAV-based immunotherapies aimed at achieving persistent delivery of Ig-HIVs. Consistent with this idea, recent studies suggest that lifelong immunity against HIV can be achieved from a one-time intramuscular dose of AAV/Ig-HIV vectors. However, realizing the promise of this approach faces significant hurdles, including the potential of AAV-delivered Ig-HIVs to induce anti-drug antibodies and the high AAV seroprevalence in the human population. Here we describe how these host immune responses can hinder AAV/Ig-HIV therapies and review current strategies for overcoming these barriers. Given the potential of AAV/Ig-HIV therapy to maintain ART-free virologic suppression and prevent HIV reinfection in people living with HIV, optimizing this strategy should become a greater priority in HIV/AIDS research.
Collapse
Affiliation(s)
- Patricia A. Hahn
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458, USA
- The Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Mauricio A. Martins
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458, USA
| |
Collapse
|
13
|
Shitik EM, Shalik IK, Yudkin DV. AAV- based vector improvements unrelated to capsid protein modification. Front Med (Lausanne) 2023; 10:1106085. [PMID: 36817775 PMCID: PMC9935841 DOI: 10.3389/fmed.2023.1106085] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) is the leading platform for delivering genetic constructs in vivo. To date, three AAV-based gene therapeutic agents have been approved by the FDA and are used in clinical practice. Despite the distinct advantages of gene therapy development, it is clear that AAV vectors need to be improved. Enhancements in viral vectors are mainly associated with capsid protein modifications. However, there are other structures that significantly affect the AAV life cycle and transduction. The Rep proteins, in combination with inverted terminal repeats (ITRs), determine viral genome replication, encapsidation, etc. Moreover, transgene cassette expression in recombinant variants is directly related to AAV production and transduction efficiency. This review discusses the ways to improve AAV vectors by modifying ITRs, a transgene cassette, and the Rep proteins.
Collapse
|
14
|
Greig JA, Breton C, Ashley SN, Martins KM, Gorsuch C, Chorazeczewski JK, Furmanak T, Smith MK, Zhu Y, Bell P, Shoop W, Li H, Smith J, Tomberlin G, Clark P, Mitchell TW, Buza EL, Yan H, Jantz D, Wilson JM. Treating Transthyretin Amyloidosis via Adeno-Associated Virus Vector Delivery of Meganucleases. Hum Gene Ther 2022; 33:1174-1186. [PMID: 36375122 PMCID: PMC9700363 DOI: 10.1089/hum.2022.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transthyretin amyloidosis (ATTR) is a progressive and fatal disease caused by transthyretin (TTR) amyloid fibril accumulation in tissues, which disrupts organ function. As the TTR protein is primarily synthesized by the liver, liver transplantation can cure familial ATTR but is not an option for the predominant age-related wild-type ATTR. Approved treatment approaches include TTR stabilizers and an RNA-interference therapeutic, but these require regular re-administration. Gene editing could represent an effective one-time treatment. We evaluated adeno-associated virus (AAV) vector-delivered, gene-editing meganucleases to reduce TTR levels. We used engineered meganucleases targeting two different sites within the TTR gene. AAV vectors expressing TTR meganuclease transgenes were first tested in immunodeficient mice expressing the human TTR sequence delivered using an AAV vector and then against the endogenous TTR gene in rhesus macaques. Following a dose of 3 × 1013 genome copies per kilogram, we detected on-target editing efficiency of up to 45% insertions and deletions (indels) in the TTR genomic DNA locus and >80% indels in TTR RNA, with a concomitant decrease in serum TTR levels of >95% in macaques. The significant reduction in serum TTR levels following TTR gene editing indicates that this approach could be an effective treatment for ATTR.
Collapse
Affiliation(s)
- Jenny A. Greig
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Camilo Breton
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Scott N. Ashley
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kelly M. Martins
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Joanna K. Chorazeczewski
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas Furmanak
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Melanie K. Smith
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yanqing Zhu
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Bell
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wendy Shoop
- Precision BioSciences, Inc., Durham, North Carolina, USA
| | - Hui Li
- Precision BioSciences, Inc., Durham, North Carolina, USA
| | - Jeff Smith
- Precision BioSciences, Inc., Durham, North Carolina, USA
| | | | - Peter Clark
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas W. Mitchell
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth L. Buza
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hanying Yan
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Derek Jantz
- Precision BioSciences, Inc., Durham, North Carolina, USA
| | - James M. Wilson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Correspondence: Dr. James M. Wilson, Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, 125 South 31st Street, Suite 1200, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Derkaev AA, Ryabova EI, Esmagambetov IB, Shcheblyakov DV, Godakova SA, Vinogradova ID, Noskov AN, Logunov DY, Naroditsky BS, Gintsburg AL. rAAV expressing recombinant neutralizing antibody for the botulinum neurotoxin type A prophylaxis. Front Microbiol 2022; 13:960937. [PMID: 36238585 PMCID: PMC9551282 DOI: 10.3389/fmicb.2022.960937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Botulinum neurotoxin (BoNT) is one of the most dangerous bacterial toxins and a potential biological weapon component. BoNT mechanism of pathological action is based on inhibiting the release of neurotransmitters from nerve endings. To date, anti-BoNT therapy is reduced to the use of horse hyperimmune serum, which causes many side effects, as well as FDA-approved drug BabyBig which consists of human-derived anti-BoNT antibodies (IgG) for infant botulinum treatment. Therapeutics for botulism treatment based on safer monoclonal antibodies are undergoing clinical trials. In addition, agents have been developed for the specific prevention of botulism, but their effectiveness has not been proved. In this work, we have obtained a recombinant adeno-associated virus (rAAV-B11-Fc) expressing a single-domain antibody fused to the human IgG Fc-fragment (B11-Fc) and specific to botulinum toxin type A (BoNT/A). We have demonstrated that B11-Fc antibody, expressed via rAAV-B11-Fc treatment, can protect animals from lethal doses of botulinum toxin type A, starting from day 3 and at least 120 days after administration. Thus, our results showed that rAAV-B11-Fc can provide long-term expression of B11-Fc-neutralizing antibody in vivo and provide long-term protection against BoNT/A intoxication. Consequently, our study demonstrates the applicability of rAAV expressing protective antibodies for the prevention of intoxication caused by botulinum toxins.
Collapse
|
16
|
Tong D, Zhang M, Yang Y, Xia H, Tong H, Zhang H, Zeng W, Liu M, Wu Y, Ma H, Hu X, Liu W, Cai Y, Yao Y, Yao Y, Liu K, Shan S, Li Y, Gao G, Guo W, Peng Y, Chen S, Rao J, Zhao J, Min J, Zhu Q, Zheng Y, Liu L, Shan C, Zhong K, Qiu Z, Jin T, Chiu S, Yuan Z, Xue T. Single-dose AAV-based vaccine induces a high level of neutralizing antibodies against SARS-CoV-2 in rhesus macaques. Protein Cell 2022; 14:69-73. [PMID: 36726759 PMCID: PMC9871966 DOI: 10.1093/procel/pwac020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/13/2022] [Accepted: 05/08/2022] [Indexed: 02/04/2023] Open
Affiliation(s)
| | | | | | | | - Haiyang Tong
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Huajun Zhang
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Weihong Zeng
- Hefei National Research Center for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, CAS Key Laboratory of Innate Immunity and Chronic Disease, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230026, China
| | - Muziying Liu
- Anhui Institute of Pediatric Research, Anhui Provincial Children’s Hospital, Hefei 230051, China
| | - Yan Wu
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Huan Ma
- Hefei National Research Center for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, CAS Key Laboratory of Innate Immunity and Chronic Disease, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230026, China
| | - Xue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Weiyong Liu
- First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yuan Cai
- First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yanfeng Yao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yichuan Yao
- First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China,Hefei National Research Center for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, CAS Key Laboratory of Innate Immunity and Chronic Disease, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230026, China
| | - Kunpeng Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Shifang Shan
- Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yajuan Li
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Ge Gao
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Weiwei Guo
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yun Peng
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Shaohong Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Juhong Rao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jiaxuan Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Juan Min
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Qingjun Zhu
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yanmin Zheng
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Lianxin Liu
- First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Chao Shan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Kai Zhong
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | | | | | | | | | | |
Collapse
|
17
|
Greig JA, Jennis M, Dandekar A, Chorazeczewski JK, Smith MK, Ashley SN, Yan H, Wilson JM. Muscle-directed AAV gene therapy rescues the maple syrup urine disease phenotype in a mouse model. Mol Genet Metab 2021; 134:139-146. [PMID: 34454844 DOI: 10.1016/j.ymgme.2021.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/04/2021] [Accepted: 08/07/2021] [Indexed: 11/19/2022]
Abstract
Maple syrup urine disease (MSUD) is a rare, inherited metabolic disorder characterized by a dysfunctional mitochondrial enzyme complex, branched-chain alpha-keto acid dehydrogenase (BCKDH), which catabolizes branched-chain amino acids (BCAAs). Without functional BCKDH, BCAAs and their neurotoxic alpha-keto intermediates can accumulate in the blood and tissues. MSUD is currently incurable and treatment is limited to dietary restriction or liver transplantation, meaning there is a great need to develop new treatments for MSUD. We evaluated potential gene therapy applications for MSUD in the intermediate MSUD (iMSUD) mouse model, which harbors a mutation in the dihydrolipoamide branched-chain transacylase E2 (DBT) subunit of BCKDH. Systemic delivery of an adeno-associated virus (AAV) vector expressing DBT under control of the liver-specific TBG promoter to the liver did not sufficiently ameliorate all aspects of the disease phenotype. These findings necessitated an alternative therapeutic strategy. Muscle makes a larger contribution to BCAA metabolism than liver in humans, but a muscle-specific approach involving a muscle-specific promoter for DBT expression delivered via intramuscular (IM) administration only partially rescued the MSUD phenotype in mice. Combining the muscle-tropic AAV9 capsid with the ubiquitous CB7 promoter via IM or IV injection, however, substantially increased survival across all assessed doses. Additionally, near-normal serum BCAA levels were achieved and maintained in the mid- and high-dose cohorts throughout the study; this approach also protected these mice from a lethal high-protein diet challenge. Therefore, administration of a gene therapy vector that expresses in both muscle and liver may represent a viable approach to treating patients with MSUD.
Collapse
Affiliation(s)
- Jenny A Greig
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Jennis
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aditya Dandekar
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joanna K Chorazeczewski
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Melanie K Smith
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott N Ashley
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hanying Yan
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Hordeaux J, Buza EL, Jeffrey B, Song C, Jahan T, Yuan Y, Zhu Y, Bell P, Li M, Chichester JA, Calcedo R, Wilson JM. MicroRNA-mediated inhibition of transgene expression reduces dorsal root ganglion toxicity by AAV vectors in primates. Sci Transl Med 2021; 12:12/569/eaba9188. [PMID: 33177182 DOI: 10.1126/scitranslmed.aba9188] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/07/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022]
Abstract
Delivering adeno-associated virus (AAV) vectors into the central nervous system of nonhuman primates (NHPs) via the blood or cerebral spinal fluid is associated with dorsal root ganglion (DRG) toxicity. Conventional immune-suppression regimens do not prevent this toxicity, possibly because it may be caused by high transduction rates, which can, in turn, cause cellular stress due to an overabundance of the transgene product in target cells. To test this hypothesis and develop an approach to eliminate DRG toxicity, we exploited endogenous expression of microRNA (miR) 183 complex, which is largely restricted to DRG neurons, to specifically down-regulate transgene expression in these cells. We introduced sequence targets for miR183 into the vector genome within the 3' untranslated region of the corresponding transgene messenger RNA and injected vectors into the cisterna magna of NHPs. Administration of unmodified AAV vectors resulted in robust transduction of target tissues and toxicity in DRG neurons. Consistent with the proposal that immune system activity does not mediate this neuronal toxicity, we found that steroid administration was ineffective in alleviating this pathology. However, including miR183 targets in the vectors reduced transgene expression in, and toxicity of, DRG neurons without affecting transduction elsewhere in the primate's brain. This approach might be useful in reducing DRG toxicity and the associated morbidity and should facilitate the development of AAV-based gene therapies for many central nervous system diseases.
Collapse
Affiliation(s)
- Juliette Hordeaux
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth L Buza
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brianne Jeffrey
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chunjuan Song
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tahsin Jahan
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuan Yuan
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yanqing Zhu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mingyao Li
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jessica A Chichester
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roberto Calcedo
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
19
|
Muhuri M, Zhan W, Maeda Y, Li J, Lotun A, Chen J, Sylvia K, Dasgupta I, Arjomandnejad M, Nixon T, Keeler AM, Manokaran S, He R, Su Q, Tai PWL, Gao G. Novel Combinatorial MicroRNA-Binding Sites in AAV Vectors Synergistically Diminish Antigen Presentation and Transgene Immunity for Efficient and Stable Transduction. Front Immunol 2021; 12:674242. [PMID: 33995418 PMCID: PMC8113644 DOI: 10.3389/fimmu.2021.674242] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/07/2021] [Indexed: 12/26/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) platforms hold promise for in vivo gene therapy but are undermined by the undesirable transduction of antigen presenting cells (APCs), which in turn can trigger host immunity towards rAAV-expressed transgene products. In light of recent adverse events in patients receiving high systemic AAV vector doses that were speculated to be related to host immune responses, development of strategies to mute innate and adaptive immunity is imperative. The use of miRNA binding sites (miR-BSs) to confer endogenous miRNA-mediated regulation to detarget transgene expression from APCs has shown promise for reducing transgene immunity. Studies have shown that designing miR-142BSs into rAAV1 vectors were able to repress costimulatory signals in dendritic cells (DCs), blunt the cytotoxic T cell response, and attenuate clearance of transduced muscle cells in mice to allow sustained transgene expression in myofibers with negligible anti-transgene IgG production. In this study, we screened individual and combinatorial miR-BS designs against 26 miRNAs that are abundantly expressed in APCs, but not in skeletal muscle. The highly immunogenic ovalbumin (OVA) transgene was used as a proxy for foreign antigens. In vitro screening in myoblasts, mouse DCs, and macrophages revealed that the combination of miR-142BS and miR-652-5pBS strongly mutes transgene expression in APCs but maintains high myoblast and myocyte expression. Importantly, rAAV1 vectors carrying this novel miR-142/652-5pBS cassette achieve higher transgene levels following intramuscular injections in mice than previous detargeting designs. The cassette strongly inhibits cytotoxic CTL activation and suppresses the Th17 response in vivo. Our approach, thus, advances the efficiency of miRNA-mediated detargeting to achieve synergistic reduction of transgene-specific immune responses and the development of safe and efficient delivery vehicles for gene therapy.
Collapse
Affiliation(s)
- Manish Muhuri
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Yukiko Maeda
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Jia Li
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Anoushka Lotun
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Jennifer Chen
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Katelyn Sylvia
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Ishani Dasgupta
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Motahareh Arjomandnejad
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Thomas Nixon
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Allison M. Keeler
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Sangeetha Manokaran
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Ran He
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Qin Su
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Phillip W. L. Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
20
|
Rodríguez-Márquez E, Meumann N, Büning H. Adeno-associated virus (AAV) capsid engineering in liver-directed gene therapy. Expert Opin Biol Ther 2020; 21:749-766. [PMID: 33331201 DOI: 10.1080/14712598.2021.1865303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Gene therapy clinical trials with adeno-associated virus (AAV) vectors report impressive clinical efficacy data. Nevertheless, challenges have become apparent, such as the need for high vector doses and the induction of anti-AAV immune responses that cause the loss of vector-transduced hepatocytes. This fostered research focusing on development of next-generation AAV vectors capable of dealing with these hurdles.Areas Covered: While both the viral vector genome and the capsid are subjects to engineering, this review focuses on the latter. Specifically, we summarize the principles of capsid engineering strategies, and describe developments and applications of engineered capsid variants for liver-directed gene therapy.Expert Opinion: Capsid engineering is a promising strategy to significantly improve efficacy of the AAV vector system in clinical application. Reduction in vector dose will further improve vector safety, lower the risk of host immune responses and the cost of manufacturing. Capsid engineering is also expected to result in AAV vectors applicable to patients with preexisting immunity toward natural AAV serotypes.
Collapse
Affiliation(s)
- Esther Rodríguez-Márquez
- Universidad Autónoma De Madrid, Madrid, Spain.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Nadja Meumann
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,German Center for Infection Research (DZIF, Partner Site Hannover-Braunschweig, Germany
| |
Collapse
|
21
|
Salabarria SM, Nair J, Clement N, Smith BK, Raben N, Fuller DD, Byrne BJ, Corti M. Advancements in AAV-mediated Gene Therapy for Pompe Disease. J Neuromuscul Dis 2020; 7:15-31. [PMID: 31796685 PMCID: PMC7029369 DOI: 10.3233/jnd-190426] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pompe disease (glycogen storage disease type II) is caused by mutations in acid α-glucosidase (GAA) resulting in lysosomal pathology and impairment of the muscular and cardio-pulmonary systems. Enzyme replacement therapy (ERT), the only approved therapy for Pompe disease, improves muscle function by reducing glycogen accumulation but this approach entails several limitations including a short drug half-life and an antibody response that results in reduced efficacy. To address these limitations, new treatments such as gene therapy are under development to increase the intrinsic ability of the affected cells to produce GAA. Key components to gene therapy strategies include the choice of vector, promoter, and the route of administration. The efficacy of gene therapy depends on the ability of the vector to drive gene expression in the target tissue and also on the recipient's immune tolerance to the transgene protein. In this review, we discuss the preclinical and clinical studies that are paving the way for the development of a gene therapy strategy for patients with early and late onset Pompe disease as well as some of the challenges for advancing gene therapy.
Collapse
Affiliation(s)
- S M Salabarria
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| | - J Nair
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| | - N Clement
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| | - B K Smith
- Department of Physical Therapy and Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida, USA
| | - N Raben
- Laboratory of Protein Trafficking and Organelle Biology, Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - D D Fuller
- Department of Physical Therapy and Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida, USA
| | - B J Byrne
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| | - M Corti
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| |
Collapse
|
22
|
Engineered AAV8 capsid acquires heparin and AVB sepharose binding capacity but has altered in vivo transduction efficiency. Gene Ther 2020; 30:236-244. [PMID: 33028973 PMCID: PMC8024426 DOI: 10.1038/s41434-020-00198-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022]
Abstract
Naturally occurring adeno-associated virus (AAV) serotypes that bind to ligands such as AVB sepharose or heparin can be purified by affinity chromatography, which is a more efficient and scalable method than gradient ultracentrifugation. Wild type AAV8 does not bind effectively to either of these molecules, which constitutes a barrier to using this vector when a high throughput design is required. Previously, AAV8 was engineered to contain a SPAKFA amino acid sequence to facilitate purification using AVB sepharose resin; however, in vivo studies were not conducted to examine whether these capsid mutations altered the transduction profile. To address this gap in knowledge, a mutant AAV8 capsid was engineered to bind to AVB sepharose and heparan sulfate (AAV8-AVB-HS), which efficiently bound to both affinity columns, resulting in elution yields of >80% of the total vector loaded compared to <5% for wild type AAV8. However, in vivo comparison by intramuscular, intravenous, and intraperitoneal vector administration demonstrated a significant decrease in AAV8-AVB-HS transduction efficiency without alteration of the transduction profile. Therefore, although it is possible to engineer AAV capsids to bind various affinity ligands, the consequences associated with mutating surface exposed residues have the potential to negatively impact other vector characteristics including in vivo potency and production yield. This study demonstrates the importance of evaluating all aspects of vector performance when engineering AAV capsids.
Collapse
|
23
|
Zhao J, Yue Y, Patel A, Wasala L, Karp JF, Zhang K, Duan D, Lai Y. High-Resolution Histological Landscape of AAV DNA Distribution in Cellular Compartments and Tissues following Local and Systemic Injection. Mol Ther Methods Clin Dev 2020; 18:856-868. [PMID: 32953935 PMCID: PMC7479330 DOI: 10.1016/j.omtm.2020.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022]
Abstract
Adeno-associated virus (AAV) is one of the most important gene delivery vehicles for in vivo gene therapy. Intramuscular (i.m.) and intravascular (i.v.) injection are commonly used for AAV gene transfer. Unfortunately, the fate of AAV vectors following administration remains unclear at the histological level. Taking advantage of RNAscope, a recently developed in situ hybridization technique that can reveal high-resolution viral DNA localization information, in this study, we evaluated body-wide distribution of an AAV9 vector in the context of the cell and tissue microenvironments. We observed distinctive kinetics of cell and nuclear entry of the AAV DNA in striated muscle and liver following i.m. and i.v. injection. We also found characteristic distribution patterns of the AAV DNA in various histological structures in internal organs, including gonads and lymph nodes, following i.v. injection. Finally, we showed significantly body-wide spreading of the AAV DNA following i.m. injection. These results add a new dimension to our understanding of AAV transduction biology and provide a basis for assessing the full impact of AAV gene therapy.
Collapse
Affiliation(s)
- Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Aman Patel
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | - Lakmini Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Jacob F. Karp
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Department of Bioengineering, University of Missouri, Columbia, MO 65212, USA
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
24
|
Lind LA, Andel EM, McCall AL, Dhindsa JS, Johnson KA, Stricklin OE, Mueller C, ElMallah MK, Lever TE, Nichols NL. Intralingual Administration of AAVrh10-miR SOD1 Improves Respiratory But Not Swallowing Function in a Superoxide Dismutase-1 Mouse Model of Amyotrophic Lateral Sclerosis. Hum Gene Ther 2020; 31:828-838. [PMID: 32498636 DOI: 10.1089/hum.2020.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease characterized by degeneration of motor neurons and muscles, and death is usually a result of impaired respiratory function due to loss of motor neurons that control upper airway muscles and/or the diaphragm. Currently, no cure for ALS exists and treatments to date do not significantly improve respiratory or swallowing function. One cause of ALS is a mutation in the superoxide dismutase-1 (SOD1) gene; thus, reducing expression of the mutated gene may slow the progression of the disease. Our group has been studying the SOD1G93A transgenic mouse model of ALS that develops progressive respiratory deficits and dysphagia. We hypothesize that solely treating the tongue in SOD1 mice will preserve respiratory and swallowing function, and it will prolong survival. At 6 weeks of age, 11 SOD1G93A mice (both sexes) received a single intralingual injection of gene therapy (AAVrh10-miRSOD1). Another 29 mice (both sexes) were divided into two control groups: (1) 12 SOD1G93A mice that received a single intralingual vehicle injection (saline); and (2) 17 non-transgenic littermates. Starting at 13 weeks of age, plethysmography (respiratory parameters) at baseline and in response to hypoxia (11% O2) + hypercapnia (7% CO2) were recorded and videofluoroscopic swallow study testing were performed twice monthly until end-stage disease. Minute ventilation during hypoxia + hypercapnia and mean inspiratory flow at baseline were significantly reduced (p < 0.05) in vehicle-injected, but not AAVrh10-miRSOD1-injected SOD1G93A mice as compared with wild-type mice. In contrast, swallowing function was unchanged by AAVrh10-miRSOD1 treatment (p > 0.05). AAVrh10-miRSOD1 injections also significantly extended survival in females by ∼1 week. In conclusion, this study indicates that intralingual AAVrh10-miRSOD1 treatment preserved respiratory (but not swallowing) function potentially via increasing upper airway patency, and it is worthy of further exploration as a possible therapy to preserve respiratory capacity in ALS patients.
Collapse
Affiliation(s)
- Lori A Lind
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Ellyn M Andel
- Department of Otolaryngology, University of Missouri, Columbia, Missouri, USA
| | - Angela L McCall
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Justin S Dhindsa
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Katherine A Johnson
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Olivia E Stricklin
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Christian Mueller
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Pediatrics, University of Massachusetts Medical School, Worcester Massachusetts, USA
| | - Mai K ElMallah
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Teresa E Lever
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA.,Department of Otolaryngology, University of Missouri, Columbia, Missouri, USA
| | - Nicole L Nichols
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
25
|
Fuchs SP, Martinez-Navio JM, Rakasz EG, Gao G, Desrosiers RC. Liver-Directed but Not Muscle-Directed AAV-Antibody Gene Transfer Limits Humoral Immune Responses in Rhesus Monkeys. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 16:94-102. [PMID: 31890736 PMCID: PMC6923507 DOI: 10.1016/j.omtm.2019.11.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/09/2019] [Indexed: 12/19/2022]
Abstract
A number of publications have described the use of adeno-associated virus (AAV) for the delivery of anti-HIV and anti-simian immunodeficiency virus (SIV) monoclonal antibodies (mAbs) to rhesus monkeys. Anti-drug antibodies (ADAs) have been frequently observed, and long-term AAV-mediated delivery has been inconsistent. Here, we investigated different AAV vector strategies and delivery schemes to rhesus monkeys using the rhesus monkey mAb 4L6. We compared 4L6 immunoglobulin G1 (IgG1) delivery using the AAV1 versus the AAV8 serotype with a cytomegalovirus (CMV) promoter and the use of a muscle-specific versus a liver-specific promoter. Long-term expression levels of 4L6 IgG1 following AAV8-mediated gene transfer were comparable to those following AAV1-mediated gene transfer. AAV1-mediated gene transfer, using a muscle-specific promoter, showed robust ADAs and transiently low 4L6 IgG1 levels that ultimately declined to below detectable levels. Intravenous AAV8-mediated gene transfer, using a liver-specific promoter, also resulted in low levels of delivered 4L6 IgG1, but those low levels were maintained in the absence of any detectable ADAs. Booster injections using AAV1-CMV allowed for increased 4L6 IgG1 serum levels in animals that were primed with AAV8 but not with AAV1. Our results suggest that liver-directed expression may help to limit ADAs and that re-administration of AAV of a different serotype can result in successful long-term delivery of an immunogenic antibody.
Collapse
Affiliation(s)
- Sebastian P Fuchs
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - José M Martinez-Navio
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ronald C Desrosiers
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
26
|
Gupta V, Cadieux CL, McMenamin D, Medina-Jaszek CA, Arif M, Ahonkhai O, Wielechowski E, Taheri M, Che Y, Goode T, Limberis MP, Li M, Cerasoli DM, Tretiakova AP, Wilson JM. Adeno-associated virus-mediated expression of human butyrylcholinesterase to treat organophosphate poisoning. PLoS One 2019; 14:e0225188. [PMID: 31765413 PMCID: PMC6876934 DOI: 10.1371/journal.pone.0225188] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/30/2019] [Indexed: 02/05/2023] Open
Abstract
Rare diseases defined by genetic mutations are classic targets for gene therapy. More recently, researchers expanded the use of gene therapy in non-clinical studies to infectious diseases through the delivery of vectorized antibodies to well-defined antigens. Here, we further extend the utility of gene therapy beyond the “accepted” indications to include organophosphate poisoning. There are no approved preventives for the multi-organ damage resulting from acute or chronic exposure to organophosphates. We show that a single intramuscular injection of adeno-associated virus vector produces peak expression (~0.5 mg/ml) of active human butyrylcholinesterase (hBChE) in mice serum within 3–4 weeks post-treatment. This expression is sustained for up to 140 days post-injection with no silencing. Sustained expression of hBChE provided dose-dependent protection against VX in male and female mice despite detectable antibodies to hBChE in some mice, thereby demonstrating that expression of hBChE in vivo in mouse muscle is an effective prophylactic against organophosphate poisoning.
Collapse
Affiliation(s)
- Vibhor Gupta
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - C. Linn Cadieux
- United States Army Medical Research Institute of Chemical Defense, Maryland, United States of America
| | - Deirdre McMenamin
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - C. Angelica Medina-Jaszek
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Muhammad Arif
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Omua Ahonkhai
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Erik Wielechowski
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Maryam Taheri
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yan Che
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tamara Goode
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Maria P. Limberis
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mingyao Li
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Douglas M. Cerasoli
- United States Army Medical Research Institute of Chemical Defense, Maryland, United States of America
| | - Anna P. Tretiakova
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - James M. Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
27
|
van Lieshout LP, Soule G, Sorensen D, Frost KL, He S, Tierney K, Safronetz D, Booth SA, Kobinger GP, Qiu X, Wootton SK. Intramuscular Adeno-Associated Virus-Mediated Expression of Monoclonal Antibodies Provides 100% Protection Against Ebola Virus Infection in Mice. J Infect Dis 2019; 217:916-925. [PMID: 29365142 DOI: 10.1093/infdis/jix644] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/30/2017] [Indexed: 01/14/2023] Open
Abstract
The 2013-2016 West Africa outbreak demonstrated the epidemic potential of Ebola virus and highlighted the need for counter strategies. Monoclonal antibody (mAb)-based therapies hold promise as treatment options for Ebola virus infections. However, production of clinical-grade mAbs is labor intensive, and immunity is short lived. Conversely, adeno-associated virus (AAV)-mediated mAb gene transfer provides the host with a genetic blueprint to manufacture mAbs in vivo, leading to steady release of antibody over many months. Here we demonstrate that AAV-mediated expression of nonneutralizing mAb 5D2 or 7C9 confers 100% protection against mouse-adapted Ebola virus infection, while neutralizing mAb 2G4 was 83% protective. A 2-component cocktail, AAV-2G4/AAV-5D2, provided complete protection when administered 7 days prior to challenge and was partially protective with a 3-day lead time. Finally, AAV-mAb therapies provided sustained protection from challenge 5 months following AAV administration. AAV-mAb may be a viable alternative strategy for vaccination against emerging infectious diseases.
Collapse
Affiliation(s)
| | - Geoff Soule
- Zoonotic Diseases and Special Pathogens Program, Canada
| | - Debra Sorensen
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Kathy L Frost
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Shihua He
- Zoonotic Diseases and Special Pathogens Program, Canada
| | - Kevin Tierney
- Zoonotic Diseases and Special Pathogens Program, Canada
| | - David Safronetz
- Zoonotic Diseases and Special Pathogens Program, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Stephanie A Booth
- Molecular Pathobiology, National Microbiology Laboratory, Public Health Agency of Canada, Canada
| | - Gary P Kobinger
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, Laval University, Québec City, Canada
| | - Xiangguo Qiu
- Zoonotic Diseases and Special Pathogens Program, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
28
|
Hordeaux J, Hinderer C, Buza EL, Louboutin JP, Jahan T, Bell P, Chichester JA, Tarantal AF, Wilson JM. Safe and Sustained Expression of Human Iduronidase After Intrathecal Administration of Adeno-Associated Virus Serotype 9 in Infant Rhesus Monkeys. Hum Gene Ther 2019; 30:957-966. [PMID: 31017018 DOI: 10.1089/hum.2019.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many neuropathic diseases cause early, irreversible neurologic deterioration, which warrants therapeutic intervention during the first months of life. In the case of mucopolysaccharidosis type I, a recessive lysosomal storage disorder that results from a deficiency of the lysosomal enzyme α-l-iduronidase (IDUA), one of the most promising treatment approaches is to restore enzyme expression through gene therapy. Specifically, administering pantropic adeno-associated virus (AAV) encoding IDUA into the cerebrospinal fluid (CSF) via suboccipital administration has demonstrated remarkable efficacy in large animals. Preclinical safety studies conducted in adult nonhuman primates supported a positive risk-benefit profile of the procedure while highlighting potential subclinical toxicity to primary sensory neurons located in the dorsal root ganglia (DRG). This study investigated the long-term performance of intrathecal cervical AAV serotype 9 gene transfer of human IDUA administered to 1-month-old rhesus monkeys (N = 4) with half of the animals tolerized to the human transgene at birth via systemic administration of an AAV serotype 8 vector expressing human IDUA from the liver. Sustained expression of the transgene for almost 4 years is reported in all animals. Transduced cells were primarily pyramidal neurons in the cortex and hippocampus, Purkinje cells in the cerebellum, lower motor neurons, and DRG neurons. Both tolerized and non-tolerized animals were robust and maintained transgene expression as measured by immunohistochemical analysis of brain tissue. However, the presence of antibodies in the non-tolerized animals led to a loss of measurable levels of secreted enzyme in the CSF. These results support the safety and efficiency of treating neonatal rhesus monkeys with AAV serotype 9 gene therapy delivered into the CSF.
Collapse
Affiliation(s)
- Juliette Hordeaux
- 1Gene Therapy Program, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Christian Hinderer
- 1Gene Therapy Program, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elizabeth L Buza
- 1Gene Therapy Program, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jean-Pierre Louboutin
- 2Section of Anatomy, Department of Basic Medical Sciences, University of West Indies, Kingston, Jamaica
| | - Tahsin Jahan
- 1Gene Therapy Program, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Peter Bell
- 1Gene Therapy Program, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jessica A Chichester
- 1Gene Therapy Program, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Alice F Tarantal
- 3Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, California
| | - James M Wilson
- 1Gene Therapy Program, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Jin Q, Qiao C, Li J, Xiao B, Li J, Xiao X. A GDF11/myostatin inhibitor, GDF11 propeptide-Fc, increases skeletal muscle mass and improves muscle strength in dystrophic mdx mice. Skelet Muscle 2019; 9:16. [PMID: 31133057 PMCID: PMC6537384 DOI: 10.1186/s13395-019-0197-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/10/2019] [Indexed: 01/27/2023] Open
Abstract
Background Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor β superfamily. The GDF11 propeptide, which is derived from the GDF11 precursor protein, blocks the activity of GDF11 and its homolog, myostatin, which are both potent inhibitors of muscle growth. Thus, treatment with GDF11 propeptide may be a potential therapeutic strategy for diseases associated with muscle atrophy like sarcopenia and the muscular dystrophies. Here, we evaluate the impact of GDF11 propeptide-Fc (GDF11PRO-Fc) gene delivery on skeletal muscle in normal and dystrophic adult mice. Methods A pull-down assay was used to obtain physical confirmation of a protein-protein interaction between GDF11PRO-Fc and GDF11 or myostatin. Next, differentiated C2C12 myotubes were treated with AAV6-GDF11PRO-Fc and challenged with GDF11 or myostatin to determine if GDF11PRO-Fc could block GDF11/myostatin-induced myotube atrophy. Localized expression of GDF11PRO-Fc was evaluated via a unilateral intramuscular injection of AAV9-GDF11PRO-Fc into the hindlimb of C57BL/6J mice. In mdx mice, intravenous injection of AAV9-GDF11PRO-Fc was used to achieve systemic expression. The impact of GDF11PRO-Fc on muscle mass, function, and pathological features were assessed. Results GDF11PRO-Fc was observed to bind both GDF11 and myostatin. In C2C12 myotubes, expression of GDF11PRO-Fc was able to mitigate GDF11/myostatin-induced atrophy. Following intramuscular injection in C57BL/6J mice, increased grip strength and localized muscle hypertrophy were observed in the injected hindlimb after 10 weeks. In mdx mice, systemic expression of GDF11PRO-Fc resulted in skeletal muscle hypertrophy without a significant change in cardiac mass after 12 weeks. In addition, grip strength and rotarod latency time were improved. Intramuscular fibrosis was also reduced in treated mdx mice; however, there was no change seen in central nucleation, membrane permeability to serum IgG or serum creatine kinase levels. Conclusions GDF11PRO-Fc induces skeletal muscle hypertrophy and improvements in muscle strength via inhibition of GDF11/myostatin signaling. However, GDF11PRO-Fc does not significantly improve the dystrophic pathology in mdx mice. Electronic supplementary material The online version of this article (10.1186/s13395-019-0197-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Quan Jin
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - Chunping Qiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Jianbin Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Bin Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Juan Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Xiao Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
30
|
Greig JA, Nordin JML, Smith MK, Ashley SN, Draper C, Zhu Y, Bell P, Buza EL, Wilson JM. A Gene Therapy Approach to Improve Copper Metabolism and Prevent Liver Damage in a Mouse Model of Wilson Disease. HUM GENE THER CL DEV 2019; 30:29-39. [DOI: 10.1089/humc.2018.219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Jenny A. Greig
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jayme M. L. Nordin
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Melanie K. Smith
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott N. Ashley
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christine Draper
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yanqing Zhu
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth L. Buza
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James M. Wilson
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Welles HC, Jennewein MF, Mason RD, Narpala S, Wang L, Cheng C, Zhang Y, Todd JP, Lifson JD, Balazs AB, Alter G, McDermott AB, Mascola JR, Roederer M. Vectored delivery of anti-SIV envelope targeting mAb via AAV8 protects rhesus macaques from repeated limiting dose intrarectal swarm SIVsmE660 challenge. PLoS Pathog 2018; 14:e1007395. [PMID: 30517201 PMCID: PMC6296672 DOI: 10.1371/journal.ppat.1007395] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 12/17/2018] [Accepted: 10/10/2018] [Indexed: 01/17/2023] Open
Abstract
Gene based delivery of immunoglobulins promises to safely and durably provide protective immunity to individuals at risk of acquiring infectious diseases such as HIV. We used a rhesus macaque animal model to optimize delivery of naturally-arising, autologous anti-SIV neutralizing antibodies expressed by Adeno-Associated Virus 8 (AAV8) vectors. Vectored transgene expression was confirmed by quantitation of target antibody abundance in serum and mucosal surfaces. We tested the expression achieved at varying doses and numbers of injections. Expression of the transgene reached a saturation at about 2 x 1012 AAV8 genome copies (gc) per needle-injection, a physical limitation that may not scale clinically into human trials. In contrast, expression increased proportionately with the number of injections. In terms of anti-drug immunity, anti-vector antibody responses were universally strong, while those directed against the natural transgene mAb were detected in only 20% of animals. An anti-transgene antibody response was invariably associated with loss of detectable plasma expression of the antibody. Despite having atypical glycosylation profiles, transgenes derived from AAV-directed muscle cell expression retained full functional activity, including mucosal accumulation, in vitro neutralization, and protection against repeated limiting dose SIVsmE660 swarm challenge. Our findings demonstrate feasibility of a gene therapy-based passive immunization strategy against infectious disease, and illustrate the potential for the nonhuman primate model to inform clinical AAV-based approaches to passive immunization. Antibodies are the humoral component of an immune response against an invading pathogen or vaccine immunogen. For challenging vaccine targets, as an alternative to active vaccination to induce the immune system to generate antibodies, current research is exploring the delivery of these proteins to populations at high risk of infection as prophylactics against infectious diseases, like HIV, RSV, and Ebola, amongst others. Passive vaccination via purified protein will require periodic reinjection to retain protective levels in subjects, adding a barrier to large scale coverage. Alternatively, delivery of antibodies using gene therapy may provide a one-time passive vaccination alternative. This strategy comes with its own hurdles, including anti-vector immunity, anti-drug immunity, physical limitations of vector uptake and the need to confirm antibody functionality. To date, many passive vaccinations strategies remain untested in humans. Non-human primate models of infection are frequently useful for predicting the success of vaccine candidates or concepts. Here, we characterize and optimize a rhesus macaque model for the delivery of anti-viral antibodies via the gene therapy vector adeno-associated virus. Lastly, we demonstrate the ability of the mAbs to protect against viral challenge. Our work demonstrates the feasibility and utility of vectored delivery of antibody transgenes in rhesus macaques. We hope this model of antibody delivery may be applied to various disease models in non-human primates and will inform clinical trial design of passive vaccination against infectious diseases.
Collapse
Affiliation(s)
- Hugh C. Welles
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Institute for Biomedical Sciences, The George Washington University, Washington, DC, United States of America
- * E-mail: (HCW); (MR)
| | - Madeleine F. Jennewein
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Rosemarie D. Mason
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sandeep Narpala
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lingshu Wang
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cheng Cheng
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yi Zhang
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John-Paul Todd
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory of Cancer Research, Frederick, Maryland, United States of America
| | - Alejandro B. Balazs
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Adrian B. McDermott
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John R. Mascola
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mario Roederer
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (HCW); (MR)
| |
Collapse
|
32
|
Greig JA, Calcedo R, Kuri-Cervantes L, Nordin JML, Albrecht J, Bote E, Goode T, Chroscinski EA, Bell P, Richman LK, Betts MR, Wilson JM. AAV8 Gene Therapy for Crigler-Najjar Syndrome in Macaques Elicited Transgene T Cell Responses That Are Resident to the Liver. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:191-201. [PMID: 30547050 PMCID: PMC6282099 DOI: 10.1016/j.omtm.2018.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 10/25/2018] [Indexed: 11/29/2022]
Abstract
Systemic delivery of adeno-associated viral (AAV) vectors has been evaluated for the treatment of several liver diseases, including homozygous familial hypercholesterolemia, ornithine transcarbamylase deficiency, and hemophilia. Here, we evaluated this approach for the treatment of Crigler-Najjar syndrome. We administered wild-type rhesus macaques with 1.0 × 1013 or 2.5 × 1013 genome copies/kg of an AAV serotype 8 vector expressing a codon-optimized version of human uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) from a liver-specific promoter. We extensively studied vector biodistribution, transgene expression, and immune responses following vector administration. All rhesus macaques survived until their scheduled necropsy at day 56 and showed no clinical abnormalities during the course of the study. Macaques administered with either vector dose developed a T cell response to the AAV capsid and/or transgene. We mapped the immunodominant epitope in the human UGT1A1 sequence, and we found no correlation between peripheral and tissue-resident lymphocyte responses. Upon further investigation, we characterized CD107a+, granzyme B+, CD4+, and CD8+ transgene-specific cellular responses that were restricted to tissue-resident T cells. This study highlights the importance of studying immune responses at the vector transduction site and the limited usefulness of blood as a surrogate to evaluate tissue-restricted T cell responses.
Collapse
Affiliation(s)
- Jenny A Greig
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roberto Calcedo
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayme M L Nordin
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica Albrecht
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erin Bote
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tamara Goode
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Chroscinski
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura K Richman
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael R Betts
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Giles AR, Govindasamy L, Somanathan S, Wilson JM. Mapping an Adeno-associated Virus 9-Specific Neutralizing Epitope To Develop Next-Generation Gene Delivery Vectors. J Virol 2018; 92:e01011-18. [PMID: 30089698 PMCID: PMC6158442 DOI: 10.1128/jvi.01011-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 07/31/2018] [Indexed: 01/11/2023] Open
Abstract
Recent clinical trials have demonstrated the potential of adeno-associated virus (AAV)-based vectors for treating rare diseases. However, significant barriers remain for the translation of these vectors into widely available therapies. In particular, exposure to the AAV capsid can generate an immune response of neutralizing antibodies. One approach to overcome this response is to map the AAV-specific neutralizing epitopes and rationally design an AAV capsid able to evade neutralization. To accomplish this, we isolated a monoclonal antibody against AAV9 following immunization of BALB/c mice and hybridoma screening. This antibody, PAV9.1, is specific for intact AAV9 capsids and has a high neutralizing titer of >1:160,000. We used cryo-electron microscopy to reconstruct PAV9.1 in complex with AAV9. We then mapped its epitope to the 3-fold axis of symmetry on the capsid, specifically to residues 496-NNN-498 and 588-QAQAQT-592. Capsid mutagenesis demonstrated that even a single amino acid substitution within this epitope markedly reduced binding and neutralization by PAV9.1. In addition, in vivo studies showed that mutations in the PAV9.1 epitope conferred a "liver-detargeting" phenotype to the mutant vectors, unlike AAV9, indicating that the residues involved in PAV9.1 interactions are also responsible for AAV9 tropism. However, we observed minimal changes in binding and neutralizing titer when we tested these mutant vectors for evasion of polyclonal sera from mice, macaques, or humans previously exposed to AAV. Taken together, these studies demonstrate the complexity of incorporating mapped neutralizing epitopes and previously identified functional motifs into the design of novel capsids able to evade immune response.IMPORTANCE Gene therapy utilizing viral vectors has experienced recent success, culminating in U.S. Food and Drug Administration approval of the first adeno-associated virus vector gene therapy product in the United States: Luxturna for inherited retinal dystrophy. However, application of this approach to other tissues faces significant barriers. One challenge is the immune response to viral infection or vector administration, precluding patients from receiving an initial or readministered dose of vector, respectively. Here, we mapped the epitope of a novel neutralizing antibody generated in response to this viral vector to design a next-generation capsid to evade immune responses. Epitope-based mutations in the capsid interfered with the binding and neutralizing ability of the antibody but not when tested against polyclonal samples from various sources. Our results suggest that targeted mutation of a greater breadth of neutralizing epitopes will be required to evade the repertoire of neutralizing antibodies responsible for blocking viral vector transduction.
Collapse
Affiliation(s)
- April R Giles
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lakshmanan Govindasamy
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Suryanarayan Somanathan
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
van Haasteren J, Hyde SC, Gill DR. Lessons learned from lung and liver in-vivo gene therapy: implications for the future. Expert Opin Biol Ther 2018; 18:959-972. [PMID: 30067117 PMCID: PMC6134476 DOI: 10.1080/14712598.2018.1506761] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Ex-vivo gene therapy has had significant clinical impact over the last couple of years and in-vivo gene therapy products are being approved for clinical use. Gene therapy and gene editing approaches have huge potential to treat genetic disease and chronic illness. AREAS COVERED This article provides a review of in-vivo approaches for gene therapy in the lung and liver, exploiting non-viral and viral vectors with varying serotypes and pseudotypes to target-specific cells. Antibody responses inhibiting viral vectors continue to constrain effective repeat administration. Lessons learned from ex-vivo gene therapy and genome editing are also discussed. EXPERT OPINION The fields of lung and liver in-vivo gene therapy are thriving and a comparison highlights obstacles and opportunities for both. Overcoming immunological issues associated with repeated administration of viral vectors remains a key challenge. The addition of targeted small molecules in combination with viral vectors may offer one solution. A substantial bottleneck to the widespread adoption of in-vivo gene therapy is how to ensure sufficient capacity for clinical-grade vector production. In the future, the exploitation of gene editing approaches for in-vivo disease treatment may facilitate the resurgence of non-viral gene transfer approaches, which tend to be eclipsed by more efficient viral vectors.
Collapse
Affiliation(s)
- Joost van Haasteren
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen C. Hyde
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Deborah R. Gill
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Greig JA, Nordin JML, White JW, Wang Q, Bote E, Goode T, Calcedo R, Wadsworth S, Wang L, Wilson JM. Optimized Adeno-Associated Viral-Mediated Human Factor VIII Gene Therapy in Cynomolgus Macaques. Hum Gene Ther 2018; 29:1364-1375. [PMID: 29890905 DOI: 10.1089/hum.2018.080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hemophilia A is a common hereditary bleeding disorder that is characterized by a deficiency of human blood coagulation factor VIII (hFVIII). Previous studies with adeno-associated viral (AAV) vectors identified two liver-specific promoter and enhancer combinations (E03.TTR and E12.A1AT) that drove high level expression of a codon-optimized, B-domain-deleted hFVIII transgene in a mouse model of the disease. This study further evaluated these enhancer/promoter combinations in cynomolgus macaques using two different AAV capsids (AAVrh10 and AAVhu37). Each of the four vector combinations was administered intravenously at a dose of 1.2 × 1013 genome copy/kg into five macaques per group. Delivery of the hFVIII transgene via the AAVhu37 capsid resulted in a substantial increase in hFVIII expression compared to animals administered with AAVrh10 vectors. Two weeks after administration of E03.TTR packaged within the AAVhu37 capsid, average hFVIII expression was 20.2 ± 5.0% of normal, with one animal exhibiting peak expression of 37.1% of normal hFVIII levels. The majority of animals generated an anti-hFVIII antibody response by week 8-10 post vector delivery. However, two of the five macaques administered with AAVhu37.E03.TTR were free of a detectable antibody response for 30 weeks post vector administration. Overall, the study supports the continued development of AAV-based gene therapeutics for hemophilia A using the AAVhu37 capsid.
Collapse
Affiliation(s)
- Jenny A Greig
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Jayme M L Nordin
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - John W White
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Qiang Wang
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Erin Bote
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Tamara Goode
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Roberto Calcedo
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | | | - Lili Wang
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - James M Wilson
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
36
|
Greig JA, Nordin JML, Draper C, McMenamin D, Chroscinski EA, Bell P, Gray JT, Richman LK, Wilson JM. Determining the Minimally Effective Dose of a Clinical Candidate AAV Vector in a Mouse Model of Crigler-Najjar Syndrome. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 10:237-244. [PMID: 30112420 PMCID: PMC6090885 DOI: 10.1016/j.omtm.2018.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/17/2018] [Indexed: 11/12/2022]
Abstract
Liver metabolism disorders are attractive targets for gene therapy, because low vector doses can reverse the buildup of toxic metabolites in the blood. Crigler-Najjar syndrome is an inherited disorder of bilirubin metabolism that is caused by the absence of uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) activity. This syndrome is characterized by hyperbilirubinemia and jaundice. Unfortunately, current phototherapy treatment is not effective long term. We intravenously injected phototherapy-rescued adult UGT1 knockout mice with 2.5 × 1010–2.5 × 1013 genome copies (GC)/kg of a clinical candidate vector, AAV8.TBG.hUGT1A1co, to study the treatment of disease compared to vehicle-only control mice. There were no apparent vector-related laboratory or clinical sequelae; the only abnormalities in clinical pathology were elevations in liver transaminases, primarily in male mice at the highest vector dose. Minimal to mild histopathological findings were present in control and vector-administered male mice. At vector doses greater than 2.5 × 1011 GC/kg, we observed a reversal of total bilirubin levels to wild-type levels. Based on a significant reduction in serum total bilirubin levels, we determined the minimally effective dose in this mouse model of Crigler-Najjar syndrome to be 2.5 × 1011 GC/kg.
Collapse
Affiliation(s)
- Jenny A Greig
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayme M L Nordin
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christine Draper
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Deirdre McMenamin
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Chroscinski
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John T Gray
- Audentes Therapeutics, San Francisco, CA, USA
| | - Laura K Richman
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
37
|
Lin H, Hu H, Duan W, Liu Y, Tan G, Li Z, Liu Y, Deng B, Song X, Wang W, Wen D, Wang Y, Li C. Intramuscular Delivery of scAAV9-hIGF1 Prolongs Survival in the hSOD1 G93A ALS Mouse Model via Upregulation of D-Amino Acid Oxidase. Mol Neurobiol 2018; 55:682-695. [PMID: 27995572 DOI: 10.1007/s12035-016-0335-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022]
Abstract
Self-complementary adeno-associated viral vector 9 (scAAV9) has been confirmed to be an efficient AAV serotype for gene transfer to the central nervous system (CNS). Neurotrophic factors have been considered to be therapeutic targets for amyotrophic lateral sclerosis (ALS). In the present study, we intramuscularly injected scAAV9 encoding human insulin-like growth factor 1 (hIGF1) into an hSOD1G93A ALS mouse model. We observed that scAAV9-hIGF1 significantly reduced the loss of motor neurons of the anterior horn in the lumbar spinal cord and delayed muscle atrophy in ALS mice. Importantly, IGF1 significantly delayed disease onset and prolonged the life span of ALS mice. In addition, scAAV9-hIGF1 protected motor neurons from apoptosis through upregulation of D-amino acid oxidase (DAO), which controls the level of D-serine. Moreover, to further verify these results, we used CRISPR-Cas9 system to target the central nervous system knockdown of IGF1. This experiment supported the continued investigation of neurotrophic factor gene therapies targeting the central nervous system as a potential treatment for ALS.
Collapse
Affiliation(s)
- HuiQian Lin
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Neurology, The First Hospital of Shijiazhuang City, Shijiazhuang, China
| | - HaoJie Hu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - WeiSong Duan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Hebei Neurology, Shijiazhuang, China
| | - YaLing Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Hebei Neurology, Shijiazhuang, China
| | - GuoJun Tan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Hebei Neurology, Shijiazhuang, China
| | - ZhongYao Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Hebei Neurology, Shijiazhuang, China
| | - YaKun Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Hebei Neurology, Shijiazhuang, China
| | - BinBin Deng
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - XueQin Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Hebei Neurology, Shijiazhuang, China
| | - Wan Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Di Wen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - ChunYan Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
- Key Laboratory of Hebei Neurology, Shijiazhuang, China.
| |
Collapse
|
38
|
Greig JA, Limberis MP, Bell P, Chen SJ, Calcedo R, Rader DJ, Wilson JM. Nonclinical Pharmacology/Toxicology Study of AAV8.TBG.mLDLR and AAV8.TBG.hLDLR in a Mouse Model of Homozygous Familial Hypercholesterolemia. HUM GENE THER CL DEV 2017; 28:28-38. [PMID: 28319445 DOI: 10.1089/humc.2017.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The homozygous form of familial hypercholesterolemia (HoFH) is an excellent model for developing in vivo gene therapy in humans. The success of orthotropic liver transplantation in correcting the metabolic abnormalities in HoFH suggests that the correction of low-density lipoprotein receptor (LDLR) expression in hepatocytes via gene therapy should be sufficient for therapeutic efficacy. Vectors based on adeno-associated virus serotype 8 (AAV8) have been previously developed for liver-targeted gene therapy of a number of genetic diseases, including HoFH. In preparation for initiating a Phase 1 clinical trial of AAV8-mediated LDLR gene therapy for HoFH, a combined pharmacology/toxicology study was conducted in a mouse model of HoFH. No dose-limiting toxicities were found at or below 6.0 × 1013 GC/kg. Therefore, the maximally tolerated dose is greater than the highest dose that was tested. Mild and transient liver pathology was noted at the highest dose. Therefore, the no effect dose was greater than or equal to the middle dose of 7.5 × 1012 GC/kg. The minimally effective dose was determined to be ≤7.5 × 1011 GC/kg, based on stable reductions in cholesterol that were considered to be clinically significant. This translates to a therapeutic window of ≥80-fold for the treatment of HoFH.
Collapse
Affiliation(s)
- Jenny A Greig
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Maria P Limberis
- 2 Department of Pathology and Laboratory Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Peter Bell
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Shu-Jen Chen
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Roberto Calcedo
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Daniel J Rader
- 3 Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,4 Department of Genetics, University of Pennsylvania , Philadelphia, Pennsylvania
| | - James M Wilson
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
39
|
Greig JA, Wang Q, Reicherter AL, Chen SJ, Hanlon AL, Tipper CH, Clark KR, Wadsworth S, Wang L, Wilson JM. Characterization of Adeno-Associated Viral Vector-Mediated Human Factor VIII Gene Therapy in Hemophilia A Mice. Hum Gene Ther 2017; 28:392-402. [PMID: 28056565 DOI: 10.1089/hum.2016.128] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Adeno-associated viral (AAV) vectors are promising vehicles for hemophilia gene therapy, with favorable clinical trial data seen in the treatment of hemophilia B. In an effort to optimize the expression of human coagulation factor VIII (hFVIII) for the treatment of hemophilia A, an extensive study was performed with numerous combinations of liver-specific promoter and enhancer elements with a codon-optimized hFVIII transgene. After generating 42 variants of three reduced-size promoters and three small enhancers, transgene cassettes were packaged within a single AAV capsid, AAVrh10, to eliminate performance differences due to the capsid type. Each hFVIII vector was administered to FVIII knockout (KO) mice at a dose of 1010 genome copies (GC) per mouse. Criteria for distinguishing the performance of the different enhancer/promoter combinations were established prior to the initiation of the studies. These criteria included prominently the level of hFVIII activity (0.12-2.12 IU/mL) and the pattern of development of anti-hFVIII antibodies. In order to evaluate the impact of capsid on hFVIII expression and antibody formation, one of the enhancer and promoter combinations that exhibited high hFVIII immunogenicity was evaluated using AAV8, AAV9, AAVrh10, AAVhu37, and AAVrh64R1 capsids. The capsids subdivided into two groups: those that generated anti-hFVIII antibodies in ≤20% of mice (AAV8 and AAV9), and those that generated anti-hFVIII antibodies in >20% of mice (AAVrh10, AAVhu37, and AAVrh64R1). The results of this study, which entailed extensive vector optimization and in vivo testing, demonstrate the significant impact that transcriptional control elements and capsid can have on vector performance.
Collapse
Affiliation(s)
- Jenny A Greig
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Qiang Wang
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Amanda L Reicherter
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Shu-Jen Chen
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Alexandra L Hanlon
- 2 School of Nursing, University of Pennsylvania , Philadelphia, Pennsylvania
| | | | - K Reed Clark
- 3 Dimension Therapeutics , Cambridge, Massachusetts
| | | | - Lili Wang
- 4 Department of Pathology and Laboratory Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - James M Wilson
- 1 Gene Therapy Program, Department of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
40
|
Fuchs SP, Desrosiers RC. Promise and problems associated with the use of recombinant AAV for the delivery of anti-HIV antibodies. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16068. [PMID: 28197421 PMCID: PMC5289440 DOI: 10.1038/mtm.2016.68] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/11/2016] [Indexed: 02/07/2023]
Abstract
Attempts to elicit antibodies with potent neutralizing activity against a broad range of human immunodeficiency virus (HIV) isolates have so far proven unsuccessful. Long-term delivery of monoclonal antibodies (mAbs) with such activity is a creative alternative that circumvents the need for an immune response and has the potential for creating a long-lasting sterilizing barrier against HIV. This approach is made possible by an incredible array of potent broadly neutralizing antibodies (bnAbs) that have been identified over the last several years. Recombinant adeno-associated virus (rAAV) vectors are ideally suited for long-term delivery for a variety of reasons. The only products made from rAAV are derived from the transgenes that are put into it; as long as those products are not viewed as foreign, expression from muscle tissue may continue for decades. Thus, use of rAAV to achieve long-term delivery of anti-HIV mAbs with potent neutralizing activity against a broad range of HIV-1 isolates is emerging as a promising concept for the prevention or treatment of HIV-1 infection in humans. Experiments in mice and monkeys that have demonstrated protective efficacy against AIDS virus infection have raised hopes for the promise of this approach. However, all published experiments in monkeys have encountered unwanted immune responses to the AAV-delivered antibody, and these immune responses appear to limit the levels of delivered antibody that can be achieved. In this review, we highlight the promise of rAAV-mediated antibody delivery for the prevention or treatment of HIV infection in humans, but we also discuss the obstacles that will need to be understood and solved in order for the promise of this approach to be realized.
Collapse
Affiliation(s)
- Sebastian P Fuchs
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, USA; Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ronald C Desrosiers
- Department of Pathology, Miller School of Medicine, University of Miami , Miami, Florida, USA
| |
Collapse
|
41
|
Greig JA, Calcedo R, Grant RL, Peng H, Medina-Jaszek CA, Ahonkhai O, Qin Q, Roy S, Tretiakova AP, Wilson JM. Intramuscular administration of AAV overcomes pre-existing neutralizing antibodies in rhesus macaques. Vaccine 2016; 34:6323-6329. [PMID: 27817961 DOI: 10.1016/j.vaccine.2016.10.053] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 10/20/2022]
Abstract
The seroprevalence of neutralizing antibodies (NAbs) to adeno-associated viral (AAV) vector capsids may preclude a percentage of the population from receiving gene therapy, particularly following systemic vector administration. We hypothesized that the use of intramuscular (IM) administration of AAV vectors might circumvent this issue. IM injections were used to administer AAV8 vectors expressing either secreted or non-secreted transgenes into mice and the influence of NAbs supplied by pre-administration of pooled human IgG on transgene expression was evaluated. We then studied the impact of naturally occurring pre-existing AAV8 NAbs on expression of a secreted transgene following IM vector delivery in rhesus macaques. Finally, we evaluated the ability to readminister AAV vectors via IM injections in rhesus macaques. In mice, the presence of AAV8 NAbs had no effect on gene expression in the injected skeletal muscle. However, liver transgene expression following hepatic distribution of the vector was ablated. In rhesus macaques, naturally occurring pre-existing AAV8 NAb titers of ⩽1:160 had no effect on expression levels of a secreted transgene after IM delivery of the vector. Additionally, readministration of AAV vectors was possible by IM injection into the previously injected muscle groups, with no effect on transgene expression by the original vector. Therefore, the presence of pre-existing NAbs in the human population should not preclude subjects from receiving gene therapy by IM administration of the vector so long as sufficient levels of secreted transgene expression can be produced without the involvement of liver.
Collapse
Affiliation(s)
- Jenny A Greig
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - Roberto Calcedo
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - Rebecca L Grant
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - Hui Peng
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - C Angelica Medina-Jaszek
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - Omua Ahonkhai
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - Qiuyue Qin
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - Soumitra Roy
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - Anna P Tretiakova
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, University of Pennsylvania Perelman School of Medicine, TRL Suite 1200, 125 South 31st Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
42
|
Limberis MP, Tretiakova A, Nambiar K, Wong G, Racine T, Crosariol M, Xiangguo Q, Kobinger G, Wilson JM. Adeno-Associated Virus Serotype 9-Expressed ZMapp in Mice Confers Protection Against Systemic and Airway-Acquired Ebola Virus Infection. J Infect Dis 2016; 214:1975-1979. [PMID: 27683818 DOI: 10.1093/infdis/jiw460] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/21/2016] [Indexed: 11/12/2022] Open
Abstract
Adeno-associated viral vectors can be used as a platform for delivering biological countermeasures against pandemic and biological threats. We show that vector delivery of two antibody components of the ZMapp product is effective in mice against systemic and airway challenge with a mouse-adapted strain of Ebola virus. This platform provides a generic manufacturing solution and overcomes some of the delivery challenges associated with repeated administration of the protective protein.
Collapse
Affiliation(s)
- Maria P Limberis
- Gene Therapy Program.,Department of Pathology and Laboratory Medicine
| | - Anna Tretiakova
- Gene Therapy Program.,Department of Medicine, University of Pennsylvania, Philadelphia
| | - Kalyani Nambiar
- Gene Therapy Program.,Department of Medicine, University of Pennsylvania, Philadelphia
| | - Gary Wong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada.,Department of Medical Microbiology, University of Manitoba, Canada
| | - Trina Racine
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada.,Department of Medical Microbiology, University of Manitoba, Canada
| | - Marco Crosariol
- Gene Therapy Program.,Department of Medicine, University of Pennsylvania, Philadelphia
| | - Qiu Xiangguo
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada.,Department of Medical Microbiology, University of Manitoba, Canada
| | - Gary Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada.,Department of Medical Microbiology, University of Manitoba, Canada
| | - James M Wilson
- Gene Therapy Program.,Department of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
43
|
Geisler A, Fechner H. MicroRNA-regulated viral vectors for gene therapy. World J Exp Med 2016; 6:37-54. [PMID: 27226955 PMCID: PMC4873559 DOI: 10.5493/wjem.v6.i2.37] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 03/02/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
Safe and effective gene therapy approaches require targeted tissue-specific transfer of a therapeutic transgene. Besides traditional approaches, such as transcriptional and transductional targeting, microRNA-dependent post-transcriptional suppression of transgene expression has been emerging as powerful new technology to increase the specificity of vector-mediated transgene expression. MicroRNAs are small non-coding RNAs and often expressed in a tissue-, lineage-, activation- or differentiation-specific pattern. They typically regulate gene expression by binding to imperfectly complementary sequences in the 3' untranslated region (UTR) of the mRNA. To control exogenous transgene expression, tandem repeats of artificial microRNA target sites are usually incorporated into the 3' UTR of the transgene expression cassette, leading to subsequent degradation of transgene mRNA in cells expressing the corresponding microRNA. This targeting strategy, first shown for lentiviral vectors in antigen presenting cells, has now been used for tissue-specific expression of vector-encoded therapeutic transgenes, to reduce immune response against the transgene, to control virus tropism for oncolytic virotherapy, to increase safety of live attenuated virus vaccines and to identify and select cell subsets for pluripotent stem cell therapies, respectively. This review provides an introduction into the technical mechanism underlying microRNA-regulation, highlights new developments in this field and gives an overview of applications of microRNA-regulated viral vectors for cardiac, suicide gene cancer and hematopoietic stem cell therapy, as well as for treatment of neurological and eye diseases.
Collapse
|
44
|
Aravalli RN, Steer CJ. Gene editing technology as an approach to the treatment of liver diseases. Expert Opin Biol Ther 2016; 16:595-608. [DOI: 10.1517/14712598.2016.1158808] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
45
|
Aravalli RN, Belcher JD, Steer CJ. Liver-targeted gene therapy: Approaches and challenges. Liver Transpl 2015; 21:718-37. [PMID: 25824605 PMCID: PMC9353592 DOI: 10.1002/lt.24122] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/06/2015] [Accepted: 03/14/2015] [Indexed: 12/15/2022]
Abstract
The liver plays a major role in many inherited and acquired genetic disorders. It is also the site for the treatment of certain inborn errors of metabolism that do not directly cause injury to the liver. The advancement of nucleic acid-based therapies for liver maladies has been severely limited because of the myriad untoward side effects and methodological limitations. To address these issues, research efforts in recent years have been intensified toward the development of targeted gene approaches using novel genetic tools, such as zinc-finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats as well as various nonviral vectors such as Sleeping Beauty transposons, PiggyBac transposons, and PhiC31 integrase. Although each of these methods uses a distinct mechanism of gene modification, all of them are dependent on the efficient delivery of DNA and RNA molecules into the cell. This review provides an overview of current and emerging therapeutic strategies for liver-targeted gene therapy and gene repair.
Collapse
Affiliation(s)
- Rajagopal N. Aravalli
- Department of Radiology, University of Minnesota Medical School, Minneapolis, MN 54455
| | - John D. Belcher
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 54455
| | - Clifford J. Steer
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 54455,Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, MN 54455
| |
Collapse
|
46
|
Gardner MR, Kattenhorn LM, Kondur HR, von Schaewen M, Dorfman T, Chiang JJ, Haworth KG, Decker JM, Alpert MD, Bailey CC, Neale ES, Fellinger CH, Joshi VR, Fuchs SP, Martinez-Navio JM, Quinlan BD, Yao AY, Mouquet H, Gorman J, Zhang B, Poignard P, Nussenzweig MC, Burton DR, Kwong PD, Piatak M, Lifson JD, Gao G, Desrosiers RC, Evans DT, Hahn BH, Ploss A, Cannon PM, Seaman MS, Farzan M. AAV-expressed eCD4-Ig provides durable protection from multiple SHIV challenges. Nature 2015; 519:87-91. [PMID: 25707797 PMCID: PMC4352131 DOI: 10.1038/nature14264] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 01/27/2015] [Indexed: 12/25/2022]
Abstract
Long-term in vivo expression of a broad and potent entry inhibitor could circumvent the need for a conventional vaccine for HIV-1. Adeno-associated virus (AAV) vectors can stably express HIV-1 broadly neutralizing antibodies (bNAbs). However, even the best bNAbs neutralize 10-50% of HIV-1 isolates inefficiently (80% inhibitory concentration (IC80) > 5 μg ml(-1)), suggesting that high concentrations of these antibodies would be necessary to achieve general protection. Here we show that eCD4-Ig, a fusion of CD4-Ig with a small CCR5-mimetic sulfopeptide, binds avidly and cooperatively to the HIV-1 envelope glycoprotein (Env) and is more potent than the best bNAbs (geometric mean half-maximum inhibitory concentration (IC50) < 0.05 μg ml(-1)). Because eCD4-Ig binds only conserved regions of Env, it is also much broader than any bNAb. For example, eCD4-Ig efficiently neutralized 100% of a diverse panel of neutralization-resistant HIV-1, HIV-2 and simian immunodeficiency virus isolates, including a comprehensive set of isolates resistant to the CD4-binding site bNAbs VRC01, NIH45-46 and 3BNC117. Rhesus macaques inoculated with an AAV vector stably expressed 17-77 μg ml(-1) of fully functional rhesus eCD4-Ig for more than 40 weeks, and these macaques were protected from several infectious challenges with SHIV-AD8. Rhesus eCD4-Ig was also markedly less immunogenic than rhesus forms of four well-characterized bNAbs. Our data suggest that AAV-delivered eCD4-Ig can function like an effective HIV-1 vaccine.
Collapse
Affiliation(s)
- Matthew R. Gardner
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Lisa M. Kattenhorn
- Department of Microbiology and Immunobiology, Harvard Medical School, New England Primate Research Center, Southborough, MA 01772, USA
| | - Hema R. Kondur
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Markus von Schaewen
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Tatyana Dorfman
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Jessica J. Chiang
- Department of Microbiology and Immunobiology, Harvard Medical School, New England Primate Research Center, Southborough, MA 01772, USA
| | - Kevin G. Haworth
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Julie M. Decker
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael D. Alpert
- Department of Microbiology and Immunobiology, Harvard Medical School, New England Primate Research Center, Southborough, MA 01772, USA
- Immunathon Inc., Cambridge, MA 02141, USA
| | - Charles C. Bailey
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Ernest S. Neale
- Department of Microbiology and Immunobiology, Harvard Medical School, New England Primate Research Center, Southborough, MA 01772, USA
| | - Christoph H. Fellinger
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Vinita R. Joshi
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Sebastian P. Fuchs
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jose M. Martinez-Navio
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Brian D. Quinlan
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Annie Y. Yao
- Department of Microbiology and Immunobiology, Harvard Medical School, New England Primate Research Center, Southborough, MA 01772, USA
| | - Hugo Mouquet
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
- Department of Immunology, Institut Pasteur, Paris, 75015, France
| | - Jason Gorman
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Baoshan Zhang
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pascal Poignard
- Department of Immunology and Microbial Science, IAVI Neutralizing Antibody Center, and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michel C. Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Dennis R. Burton
- Department of Immunology and Microbial Science, IAVI Neutralizing Antibody Center, and Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Peter D. Kwong
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Incorporated, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Incorporated, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Ronald C. Desrosiers
- Department of Microbiology and Immunobiology, Harvard Medical School, New England Primate Research Center, Southborough, MA 01772, USA
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David T. Evans
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53711, USA
| | - Beatrice H. Hahn
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Paula M. Cannon
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | | | - Michael Farzan
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, FL 33458, USA
| |
Collapse
|