Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (62)

Search Parameters:
Keywords = AAV engineering

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 604 KiB  
Review
Implications of AAV Serotypes in Neurological Disorders: Current Clinical Applications and Challenges
by Sachin Sharma, Vibhuti Joshi and Vivek Kumar
Clin. Transl. Neurosci. 2025, 9(3), 32; https://doi.org/10.3390/ctn9030032 - 15 Jul 2025
Viewed by 446
Abstract
Adeno-associated virus (AAV) vectors have emerged as powerful tools for in vivo gene therapy, enabling long-term transgene expression in targeted tissues with minimal pathogenicity. This review examines the AAV serotypes used in clinical gene therapy trials for neurodegenerative (central nervous system, CNS) diseases, [...] Read more.
Adeno-associated virus (AAV) vectors have emerged as powerful tools for in vivo gene therapy, enabling long-term transgene expression in targeted tissues with minimal pathogenicity. This review examines the AAV serotypes used in clinical gene therapy trials for neurodegenerative (central nervous system, CNS) diseases, highlighting their tropisms, engineering advances, and translational progress. We discuss how capsid modifications, cell-specific promoters, and novel delivery routes are enhancing AAV tropism and reducing immunogenicity to overcome current limitations. Key clinical trials in neurodegenerative disorders (such as Parkinson’s, Alzheimer’s, and Huntington’s disease) are summarized, including delivery methods (intravenous, intracoronary, intrathecal, etc.) and outcomes. We further outline the regulatory landscape with recent approvals of AAV-based therapies and ongoing efforts to address safety challenges like immune responses and vector dose toxicity. A more translational, forward-looking perspective is adopted to consider combination therapies (e.g., AAV with immune modulation or genome editing) and strategic directions to improve the next generation of AAV vectors. Overall, continued innovation in AAV vector design and delivery, alongside careful clinical evaluation, is accelerating the translation of gene therapies for neurodegenerative diseases. Full article
Show Figures

Figure 1

28 pages, 2110 KiB  
Review
Adeno-Associated Virus Vectors in Retinal Gene Therapy: Challenges, Innovations, and Future Directions
by Jiayu Huang, Jiajun Li, Xiangzhong Xu and Keran Li
Biomolecules 2025, 15(7), 940; https://doi.org/10.3390/biom15070940 - 28 Jun 2025
Viewed by 835
Abstract
Adeno-associated virus (AAV) vectors have emerged as the leading platform for retinal gene therapy due to their favorable safety profile, low immunogenicity, and ability to mediate long-term transgene expression within the immune-privileged ocular environment. By integrating diverse strategies such as gene augmentation and [...] Read more.
Adeno-associated virus (AAV) vectors have emerged as the leading platform for retinal gene therapy due to their favorable safety profile, low immunogenicity, and ability to mediate long-term transgene expression within the immune-privileged ocular environment. By integrating diverse strategies such as gene augmentation and gene editing, AAV-based therapies have demonstrated considerable promise in treating both inherited and acquired retinal disorders. However, their clinical translation remains limited by several key challenges, including restricted packaging capacity, suboptimal transduction efficiency, the risk of gene therapy-associated uveitis, and broader societal concerns such as disease burden and ethical oversight. This review summarizes recent advances aimed at overcoming these barriers, with a particular focus on delivery route-specific disease applicability, multi-vector systems, and capsid engineering approaches to enhance payload capacity, targeting specificity, and biosafety. By synthesizing these developments, we propose a conceptual and technical framework for a more efficient, safer, and broadly applicable AAV platform to accelerate clinical adoption in retinal gene therapy. Full article
(This article belongs to the Special Issue Retinal Diseases: Molecular Mechanisms and Therapies)
Show Figures

Figure 1

19 pages, 3864 KiB  
Article
Development and Characterization of Adeno-Associated Virus-Loaded Coaxial Electrospun Scaffolds for Potential Viral Vector Delivery
by Haiguang Zhang, Bing Zhou, Wei Dong, Yongteng Song, Qingxi Hu, Heng Zhang, Min Yu, Guanglang Zhu, Yudong Sun and Jiaxuan Feng
Polymers 2025, 17(10), 1381; https://doi.org/10.3390/polym17101381 - 17 May 2025
Viewed by 538
Abstract
Gene therapy, which treats genetic diseases by fixing defective genes, has gained significant attention. Viral vectors show great potential for gene delivery but face limitations like poor targeting, uncontrolled release, and risks from high-dose delivery which can lower efficiency and trigger immune responses. [...] Read more.
Gene therapy, which treats genetic diseases by fixing defective genes, has gained significant attention. Viral vectors show great potential for gene delivery but face limitations like poor targeting, uncontrolled release, and risks from high-dose delivery which can lower efficiency and trigger immune responses. Loading viral vectors onto tissue engineered scaffolds presents a promising strategy to address these challenges, but their widespread application remains limited due to concerns regarding viral vector bioactivity, scaffold biocompatibility, and the stability of sustained release. An adeno-associated virus (AAV), recognized for its safety, high efficiency, and low immunogenicity, was employed as a model virus. In this study, we developed an electrospun scaffold (AAV/PCL-PEO@Co-ES) by encapsulating the AAV within core–shell fibers composed of polycaprolactone (PCL) and polyethylene oxide (PEO) via coaxial electrospinning. This configuration ensures viral vector protection while enabling controlled and sustained release. The physicochemical characterization results indicated that the scaffold exhibited excellent mechanical properties (tensile strength: 3.22 ± 0.48 MPa) and wettability (WCA: 67.90 ± 8.45°). In vitro release and cell transduction assays demonstrated that the AAV-loaded scaffold effectively controls viral vector release and transduction. Furthermore, both in vitro and in vivo evaluations demonstrated good biocompatibility and efficient viral vector delivery. These findings highlight the potential of the AAV/PCL-PEO@Co-ES scaffold as a safe and effective platform for sustained gene delivery, offering valuable insights for the future design of clinically relevant viral vector delivery systems. Full article
(This article belongs to the Special Issue Polymer-Based Materials for Drug Delivery and Biomedical Applications)
Show Figures

Figure 1

20 pages, 986 KiB  
Review
Past, Present, and Future of Viral Vector Vaccine Platforms: A Comprehensive Review
by Justin Tang, Md Al Amin and Jian L. Campian
Vaccines 2025, 13(5), 524; https://doi.org/10.3390/vaccines13050524 - 15 May 2025
Viewed by 2525
Abstract
Over the past several decades, viral vector-based vaccines have emerged as some of the most versatile and potent platforms in modern vaccinology. Their capacity to deliver genetic material encoding target antigens directly into host cells enables strong cellular and humoral immune responses, often [...] Read more.
Over the past several decades, viral vector-based vaccines have emerged as some of the most versatile and potent platforms in modern vaccinology. Their capacity to deliver genetic material encoding target antigens directly into host cells enables strong cellular and humoral immune responses, often superior to what traditional inactivated or subunit vaccines can achieve. This has accelerated their application to a wide array of pathogens and disease targets, from well-established threats like HIV and malaria to emerging infections such as Ebola, Zika, and SARS-CoV-2. The COVID-19 pandemic further highlighted the agility of viral vector platforms, with several adenovirus-based vaccines quickly authorized and deployed on a global scale. Despite these advances, significant challenges remain. One major hurdle is pre-existing immunity against commonly used vector backbones, which can blunt vaccine immunogenicity. Rare but serious adverse events, including vector-associated inflammatory responses and conditions like vaccine-induced immune thrombotic thrombocytopenia (VITT), have raised important safety considerations. Additionally, scaling up manufacturing, ensuring consistency in large-scale production, meeting rigorous regulatory standards, and maintaining equitable global access to these vaccines present profound logistical and ethical dilemmas. In response to these challenges, the field is evolving rapidly. Sophisticated engineering strategies, such as integrase-defective lentiviral vectors, insect-specific flaviviruses, chimeric capsids to evade neutralizing antibodies, and plug-and-play self-amplifying RNA approaches, seek to bolster safety, enhance immunogenicity, circumvent pre-existing immunity, and streamline production. Lessons learned from the COVID-19 pandemic and prior outbreaks are guiding the development of platform-based approaches designed for rapid deployment during future public health emergencies. This review provides an exhaustive, in-depth examination of the historical evolution, immunobiological principles, current platforms, manufacturing complexities, regulatory frameworks, known safety issues, and future directions for viral vector-based vaccines. Full article
(This article belongs to the Special Issue Strategies of Viral Vectors for Vaccine Development)
Show Figures

Figure 1

16 pages, 4385 KiB  
Article
Adeno-Associated Virus-Engineered Umbilical Cord-Derived Mesenchymal Stromal Cells Overexpressing Human sFlt-1 for Anti-Angiogenesis
by Ewa Yee-Wa Choy, Chee-Onn Leong, Soon-Keng Cheong, Khong-Lek Then and Kong-Yong Then
Life 2025, 15(5), 728; https://doi.org/10.3390/life15050728 - 30 Apr 2025
Viewed by 930
Abstract
Purpose. Genetic engineering of mesenchymal stromal cells (MSCs) using viral vectors has emerged as a promising approach to enhance the efficacy of anti-angiogenic gene therapies. Umbilical cord-derived MSCs are an attractive cell source due to their easy accessibility and potential for genetic modification. [...] Read more.
Purpose. Genetic engineering of mesenchymal stromal cells (MSCs) using viral vectors has emerged as a promising approach to enhance the efficacy of anti-angiogenic gene therapies. Umbilical cord-derived MSCs are an attractive cell source due to their easy accessibility and potential for genetic modification. Adeno-associated viruses (AAVs) have been utilized in clinical settings to deliver therapeutic genes due to its characteristic of transient integration into the genome. In this study, we investigated the efficacy of using recombinant AAV-engineered umbilical cord-derived MSCs overexpressing anti-angiogenic factor, hsFlt-1 (MSCs.hsFlt1). Methods. The plasmid containing the hsFlt-1 gene was cloned into the AAV2 target backbone and validated using Sanger sequencing. The transduction process was studied to determine the optimal conditions, including the effect of MOI, media serum percentage, and attachment of MSCs, to achieve higher transduction efficiency. The functionality of MSCs.hsFtl1 was analyzed using qPCR, ELISA, and tube formation assays. Results. MSCs.hsFtl1 transduced at an MOI of 1 × 106 demonstrated high transduction efficiency and exhibited robust gene and protein expression of hsFlt-1. The results revealed significant inhibition of growth in human umbilical vein endothelial cells (HUVECs) using a remarkably low dose of MSCs.hsFlt1 at 12.3 ng/mL. This observed anti-angiogenic effect was comparable to the clinically used Bevacizumab. Conclusions. The anti-angiogenic potential of MSCs.hsFlt1 effectively demonstrated in this study suggests their promising utility for targeted anti-angiogenic gene therapy approaches. Full article
(This article belongs to the Special Issue Molecular and Cellular Biology of Angiogenesis)
Show Figures

Figure 1

17 pages, 4759 KiB  
Article
Exosome-Modified AAV Gene Therapy Attenuates Autoimmune Hepatitis via Enhanced Regulatory T Cell Targeting and Immune Modulation
by Wenwei Shao, Weilin Huang, Yixuan Wang, Helin Sima, Kai Ma, Rongtao Chen, Heqiao Han, Yixuan Yang, Yuchen Bao, Xiaolei Pei and Lei Zhang
Microorganisms 2025, 13(4), 823; https://doi.org/10.3390/microorganisms13040823 - 4 Apr 2025
Cited by 1 | Viewed by 1185
Abstract
Autoimmune hepatitis (AIH) is a chronic liver disorder driven by immune dysregulation, marked by reduced regulatory T cells (Tregs) and unchecked inflammation. Current therapies lack specificity and efficacy, necessitating novel approaches. This study explores gene therapy using exosome-associated adeno-associated virus (exo-AAV) to deliver [...] Read more.
Autoimmune hepatitis (AIH) is a chronic liver disorder driven by immune dysregulation, marked by reduced regulatory T cells (Tregs) and unchecked inflammation. Current therapies lack specificity and efficacy, necessitating novel approaches. This study explores gene therapy using exosome-associated adeno-associated virus (exo-AAV) to deliver the Foxp3 gene, aiming to restore Treg-mediated immune tolerance in AIH. We engineered exosomes expressing the CD4-targeting antibody on their surface, encapsulating AAV6/Foxp3, to enhance lymphoid cell specificity. In a ConA-induced murine AIH model, engineered exo-AAV administration significantly increased hepatic Treg proportions while reducing Th17 cells and inflammatory cytokines (IFN-γ, TNF-α, IL-6), compared to control groups (unmodified exo-AAV or empty exosomes). Liver histopathology and serum ALT levels also improved in engineered exo-AAV treated mice. Mechanistically, engineered exo-AAV demonstrated superior targeting via CD4 binding, validated by immunofluorescence and nanoparticle tracking. Despite transient reductions in splenic Tregs, localized hepatic immune modulation underscored exo-AAV’s efficacy. These findings highlight engineered exo-AAV as a promising strategy for precision gene therapy in AIH, overcoming limitations of traditional AAV delivery by enhancing lymphocyte-specific transduction and immune balance restoration. This approach presents a novel therapeutic avenue for systemic autoimmune diseases reliant on Treg reinforcement. Full article
(This article belongs to the Section Medical Microbiology)
Show Figures

Figure 1

21 pages, 1382 KiB  
Review
Small Genomes, Big Disruptions: Parvoviruses and the DNA Damage Response
by Rhiannon R. Abrahams and Kinjal Majumder
Viruses 2025, 17(4), 494; https://doi.org/10.3390/v17040494 - 29 Mar 2025
Viewed by 1028
Abstract
Parvoviruses are small, single-stranded DNA viruses that have evolved sophisticated mechanisms to hijack host cell machinery for replication and persistence. One critical aspect of this interaction involves the manipulation of the host’s DNA Damage Response (DDR) pathways. While the viral genome is comparatively [...] Read more.
Parvoviruses are small, single-stranded DNA viruses that have evolved sophisticated mechanisms to hijack host cell machinery for replication and persistence. One critical aspect of this interaction involves the manipulation of the host’s DNA Damage Response (DDR) pathways. While the viral genome is comparatively simple, parvoviruses have developed strategies that cause significant DNA damage, activate DDR pathways, and disrupt the host cell cycle. This review will explore the impact of parvovirus infections on host genome stability, focusing on key viral species such as Adeno-Associated Virus (AAV), Minute Virus of Mice (MVM), and Human Bocavirus (HBoV), and their interactions with DDR proteins. Since parvoviruses are used as oncolytic agents and gene therapy vectors, a better understanding of cellular DDR pathways will aid in engineering potent anti-cancer agents and gene therapies for chronic diseases. Full article
(This article belongs to the Special Issue Advances in Parvovirus Research 2024)
Show Figures

Figure 1

26 pages, 2222 KiB  
Review
Progress in AAV-Mediated In Vivo Gene Therapy and Its Applications in Central Nervous System Diseases
by Shuming Wang and Lin Xiao
Int. J. Mol. Sci. 2025, 26(5), 2213; https://doi.org/10.3390/ijms26052213 - 28 Feb 2025
Cited by 3 | Viewed by 2630
Abstract
As the blood–brain barrier (BBB) prevents molecules from accessing the central nervous system (CNS), the traditional systemic delivery of chemical drugs limits the development of neurological drugs. However, in recent years, innovative therapeutic strategies have tried to bypass the restriction of traditional drug [...] Read more.
As the blood–brain barrier (BBB) prevents molecules from accessing the central nervous system (CNS), the traditional systemic delivery of chemical drugs limits the development of neurological drugs. However, in recent years, innovative therapeutic strategies have tried to bypass the restriction of traditional drug delivery methods. In vivo gene therapy refers to emerging biopharma vectors that carry the specific genes and target and infect specific tissues; these infected cells and tissues then undergo fundamental changes at the genetic level and produce therapeutic proteins or substances, thus providing therapeutic benefits. Clinical and preclinical trials mainly utilize adeno-associated viruses (AAVs), lentiviruses (LVs), and other viruses as gene vectors for disease investigation. Although LVs have a higher gene-carrying capacity, the vector of choice for many neurological diseases is the AAV vector due to its safety and long-term transgene expression in neurons. Here, we review the basic biology of AAVs and summarize some key issues in recombinant AAV (rAAV) engineering in gene therapy research; then, we summarize recent clinical trials using rAAV treatment for neurological diseases and provide translational perspectives and future challenges on target selection. Full article
(This article belongs to the Special Issue Application of Genetic Engineering in Treatments for Human Diseases)
Show Figures

Figure 1

32 pages, 8096 KiB  
Article
Reversing Pathology in an Aggravated Fabry Mouse Model Using Low-Dose Engineered Human Alpha-Galactosidase A AAV Gene Therapy
by Wanida Ruangsiriluk, Mugdha Deshpande, Natalia Boukharov, Girija Rajarshi, Shreya Mukherji, Shipeng Yuan, Jennifer Wiseman, Nancy Chen, Eric Park, Hyelim Cho and Rizwana Islam
Biomedicines 2025, 13(3), 577; https://doi.org/10.3390/biomedicines13030577 - 25 Feb 2025
Viewed by 1732
Abstract
Background/Objectives: Fabry disease is an X-linked disorder caused by lysosomal accumulation of glycosphingolipids due to the deficiency of α-Galactosidase (α-GAL), which leads to pathology in multiple organ systems. The standard of care is enzyme replacement therapy (ERT) with recombinant native α-GAL protein. [...] Read more.
Background/Objectives: Fabry disease is an X-linked disorder caused by lysosomal accumulation of glycosphingolipids due to the deficiency of α-Galactosidase (α-GAL), which leads to pathology in multiple organ systems. The standard of care is enzyme replacement therapy (ERT) with recombinant native α-GAL protein. Shortcomings of the native α-GAL include low stability, a short circulating half-life, and inadequate uptake by affected tissues that limits the efficacy of ERT and could potentially reduce AAV gene therapy (GT) benefits. Cross-correction by secreted α-GAL is essential for liver-targeted as well as ubiquitous AAV GT due to poor transduction and/or short half-life of some of the significantly affected cell types. Methods: To overcome potential limitations of AAV GT delivering native α-GAL, we used an engineered GLA transgene product to improve enzyme stability and reduce predicted immunogenicity. Results: The stabilized α-GAL variant, Eng-C, had an extended circulatory half-life, allowing for enhanced distribution and efficient uptake by target organs. AAV gene therapy with Eng-C demonstrated significantly greater substrate reduction in the severe Fabry G3Stg/GlaKO mouse model across all affected tissues. Efficacy of the Eng-C AVV GT was equal to or greater than the efficacy of the higher doses of the AAV GT with native α-GAL. Furthermore, this study is the first to demonstrate that the pre-existing pathology in some tissues in G3Stg/GlaKO mice can be reversed with efficient treatment. Conclusions: Our findings demonstrate that an AAV-based gene therapy expressing an engineered α-GAL with improved stability and lower immunogenicity could be effective at lower doses than other AAV GTs, potentially offering lower safety risks typically associated with high AAV vector doses. Full article
Show Figures

Figure 1

11 pages, 9934 KiB  
Article
Tropism of the AAV6.2 Vector in the Murine Retina
by Ryo Suzuki, Yusaku Katada, Momo Fujii, Naho Serizawa, Kazuno Negishi and Toshihide Kurihara
Int. J. Mol. Sci. 2025, 26(4), 1580; https://doi.org/10.3390/ijms26041580 - 13 Feb 2025
Viewed by 1429
Abstract
Retinitis pigmentosa (RP) is a progressive inherited retinal dystrophy (IRD) that primarily affects rod photoreceptor cells, leading to the degeneration of photoreceptors and the gradual loss of vision. While RP is one of the most studied IRDs, other neurodegenerative diseases affecting the retina [...] Read more.
Retinitis pigmentosa (RP) is a progressive inherited retinal dystrophy (IRD) that primarily affects rod photoreceptor cells, leading to the degeneration of photoreceptors and the gradual loss of vision. While RP is one of the most studied IRDs, other neurodegenerative diseases affecting the retina and optic nerve, such as glaucoma, also involve common mechanisms of cellular stress and degeneration. Current therapeutic approaches under investigation include gene therapy, retina prosthesis, and neuroprotection. Among these approaches, gene therapy has shown promise, though challenges related to viral vector tropism and transduction efficiency persist. The adeno-associated virus (AAV) vector is commonly employed for gene delivery, but novel serotypes and engineered variants are being explored to improve specificity and efficacy. This study evaluates the gene transfer efficiency of the AAV6.2 vector following intravitreal injection into the murine retina. Male C57BL/6 mice (9 weeks old) were intravitreally injected with 1 µL of AAV2-CMV-EGFP, AAV6-CMV-EGFP, or AAV6.2-CMV-EGFP at a titer of 3.2 × 1012 vg/mL per eye. Retinal transduction was assessed using in vivo fluorescence imaging, flat-mount imaging, and immunohistochemistry. EGFP expression in retinal ganglion cells, Müller cells, amacrine cells, and bipolar cells was quantitatively analyzed. All three AAV serotypes effectively transduced retinal ganglion cells, but AAV6.2 exhibited enhanced transduction in Müller cells and other neuronal retinal cells, including bipolar and amacrine cells. AAV6.2 demonstrated more localized expression around retinal blood vessels compared to the diffuse expression observed with AAV2. Immunohistochemical analysis revealed that AAV6.2 had significantly higher transduction efficiency in Müller cells (p < 0.001) compared to AAV2 and AAV6. AAV6.2 shows superior transduction efficiency in Müller cells, positioning it as a promising vector for gene therapies targeting retinal degenerative diseases such as RP. Its ability to effectively transduce Müller cells suggests potential applications in neuroprotection and gene replacement therapies. Full article
Show Figures

Figure 1

33 pages, 2768 KiB  
Review
Adeno-Associated Virus Vectors: Principles, Practices, and Prospects in Gene Therapy
by Limor Zwi-Dantsis, Saira Mohamed, Giulia Massaro and Emad Moeendarbary
Viruses 2025, 17(2), 239; https://doi.org/10.3390/v17020239 - 9 Feb 2025
Cited by 3 | Viewed by 5420
Abstract
Gene therapy offers promising potential as an efficacious and long-lasting therapeutic option for genetic conditions, by correcting defective mutations using engineered vectors to deliver genetic material to host cells. Among these vectors, adeno-associated viruses (AAVs) stand out for their efficiency, versatility, and safety, [...] Read more.
Gene therapy offers promising potential as an efficacious and long-lasting therapeutic option for genetic conditions, by correcting defective mutations using engineered vectors to deliver genetic material to host cells. Among these vectors, adeno-associated viruses (AAVs) stand out for their efficiency, versatility, and safety, making them one of the leading platforms in gene therapy. The enormous potential of AAVs has been demonstrated through their use in over 225 clinical trials and the FDA’s approval of six AAV-based gene therapy products, positioning these vectors at the forefront of the field. This review highlights the evolution and current applications of AAVs in gene therapy, focusing on their clinical successes, ongoing developments, and the manufacturing processes required for the rapid commercial growth anticipated in the AAV therapy market. It also discusses the broader implications of these advancements for future therapeutic strategies targeting more complex and multi-systemic conditions and biological processes such as aging. Finally, we explore some of the major challenges currently confronting the field. Full article
(This article belongs to the Section General Virology)
Show Figures

Figure 1

21 pages, 3110 KiB  
Article
Improved Recombinant Adeno-Associated Viral Vector Production via Molecular Evolution of the Viral Rep Protein
by Thomas Steininger, Veronika Öttl, Linda E. Franken, Cornelius Frank, Philip Ohland, Miriam Lopez Ferreiro, Stefan Klostermann, Johannes Fritsch, Evelyn Hirschauer, Anna Sandmeir, Luisa D. Hilgenfeld, Florian Semmelmann, Marie-Sofie Dürr, Fabian Konkel, Gregor Pechmann, Sabine Linder, Markus Haindl, Mustafa N. Yazicioglu, Philippe Ringler, Matthias E. Lauer, Denis Phichith, Stefan Seeber and Julia Fakhiriadd Show full author list remove Hide full author list
Int. J. Mol. Sci. 2025, 26(3), 1319; https://doi.org/10.3390/ijms26031319 - 4 Feb 2025
Cited by 1 | Viewed by 5194
Abstract
In the dynamic field of gene therapy, recombinant adeno-associated viruses (rAAVs) have become leading viral vectors due to their safety, long-term expression, and wide-ranging cell and tissue tropism. With numerous FDA approvals and commercial products underscoring their potential, there is a critical need [...] Read more.
In the dynamic field of gene therapy, recombinant adeno-associated viruses (rAAVs) have become leading viral vectors due to their safety, long-term expression, and wide-ranging cell and tissue tropism. With numerous FDA approvals and commercial products underscoring their potential, there is a critical need for efficient production processes to achieve high vector titers and quality. A major challenge in rAAV production is the efficient packaging of the genome into the viral capsid, with empty or partially filled capsids often representing over 90% of the produced material. To tackle this issue, we engineered the replication and packaging proteins of an AAV (Rep) to boost their functionality and improve vector titers. We subjected a complex Rep library derived from the AAV serotypes 1–13 to directed evolution in an AAV producer cell line. After each round of selection, single clones were analyzed, showing enrichment of specific hybrid Rep domains. Comparative analysis of these selected clones revealed considerable differences in their ability to package AAV2-based viral genomes, with hybrid Rep proteins achieving up to a 2.5-fold increase in packaging efficiency compared to their parental counterparts. These results suggest that optimizing rep gene variants through directed evolution is an effective strategy to enhance rAAV production efficiency. Full article
(This article belongs to the Special Issue Virus Engineering and Applications)
Show Figures

Figure 1

18 pages, 7517 KiB  
Article
Conventional and Tropism-Modified High-Capacity Adenoviral Vectors Exhibit Similar Transduction Profiles in Human iPSC-Derived Retinal Organoids
by Andrew McDonald, Carmen Gallego, Charlotte Andriessen, Michaela Orlová, Manuel A. F. V. Gonçalves and Jan Wijnholds
Int. J. Mol. Sci. 2025, 26(1), 55; https://doi.org/10.3390/ijms26010055 - 24 Dec 2024
Cited by 1 | Viewed by 1194
Abstract
Viral vector delivery of gene therapy represents a promising approach for the treatment of numerous retinal diseases. Adeno-associated viral vectors (AAV) constitute the primary gene delivery platform; however, their limited cargo capacity restricts the delivery of several clinically relevant retinal genes. In this [...] Read more.
Viral vector delivery of gene therapy represents a promising approach for the treatment of numerous retinal diseases. Adeno-associated viral vectors (AAV) constitute the primary gene delivery platform; however, their limited cargo capacity restricts the delivery of several clinically relevant retinal genes. In this study, we explore the feasibility of employing high-capacity adenoviral vectors (HC-AdVs) as alternative delivery vehicles, which, with a capacity of up to 36 kb, can potentially accommodate all known retinal gene coding sequences. We utilized HC-AdVs based on the classical adenoviral type 5 (AdV5) and on a fiber-modified AdV5.F50 version, both engineered to deliver a 29.6 kb vector genome encoding a fluorescent reporter construct. The tropism of these HC-AdVs was evaluated in an induced pluripotent stem cell (iPSC)-derived human retinal organoid model. Both vector types demonstrated robust transduction efficiency, with sustained transgene expression observed for up to 110 days post-transduction. Moreover, we found efficient transduction of photoreceptors and Müller glial cells, without evidence of reactive gliosis or loss of photoreceptor cell nuclei. However, an increase in the thickness of the photoreceptor outer nuclear layer was observed at 110 days post-transduction, suggesting potential unfavorable effects on Müller glial or photoreceptor cells associated with HC-AdV transduction and/or long-term reporter overexpression. These findings suggest that while HC-AdVs show promise for large retinal gene delivery, further investigations are required to assess their long-term safety and efficacy. Full article
Show Figures

Figure 1

22 pages, 3451 KiB  
Review
Molecular Engineering of Virus Tropism
by Bo He, Belinda Wilson, Shih-Heng Chen, Kedar Sharma, Erica Scappini, Molly Cook, Robert Petrovich and Negin P. Martin
Int. J. Mol. Sci. 2024, 25(20), 11094; https://doi.org/10.3390/ijms252011094 - 15 Oct 2024
Cited by 5 | Viewed by 4303
Abstract
Engineered viral vectors designed to deliver genetic material to specific targets offer significant potential for disease treatment, safer vaccine development, and the creation of novel biochemical research tools. Viral tropism, the specificity of a virus for infecting a particular host, is often modified [...] Read more.
Engineered viral vectors designed to deliver genetic material to specific targets offer significant potential for disease treatment, safer vaccine development, and the creation of novel biochemical research tools. Viral tropism, the specificity of a virus for infecting a particular host, is often modified in recombinant viruses to achieve precise delivery, minimize off-target effects, enhance transduction efficiency, and improve safety. Key factors influencing tropism include surface protein interactions between the virus and host-cell, the availability of host-cell machinery for viral replication, and the host immune response. This review explores current strategies for modifying the tropism of recombinant viruses by altering their surface proteins. We provide an overview of recent advancements in targeting non-enveloped viruses (adenovirus and adeno-associated virus) and enveloped viruses (retro/lentivirus, Rabies, Vesicular Stomatitis Virus, and Herpesvirus) to specific cell types. Additionally, we discuss approaches, such as rational design, directed evolution, and in silico and machine learning-based methods, for generating novel AAV variants with the desired tropism and the use of chimeric envelope proteins for pseudotyping enveloped viruses. Finally, we highlight the applications of these advancements and discuss the challenges and future directions in engineering viral tropism. Full article
(This article belongs to the Special Issue Virus Engineering and Applications: 2nd Edition)
Show Figures

Figure 1

20 pages, 4613 KiB  
Article
Redundancy in Innate Immune Pathways That Promote CD8+ T-Cell Responses in AAV1 Muscle Gene Transfer
by Ning Li, Sandeep R. P. Kumar, Di Cao, Maite Munoz-Melero, Sreevani Arisa, Bridget A. Brian, Calista M. Greenwood, Kentaro Yamada, Dongsheng Duan and Roland W. Herzog
Viruses 2024, 16(10), 1507; https://doi.org/10.3390/v16101507 - 24 Sep 2024
Cited by 3 | Viewed by 2277
Abstract
While adeno-associated viral (AAV) vectors are successfully used in a variety of in vivo gene therapy applications, they continue to be hampered by the immune system. Here, we sought to identify innate and cytokine signaling pathways that promote CD8+ T-cell responses against [...] Read more.
While adeno-associated viral (AAV) vectors are successfully used in a variety of in vivo gene therapy applications, they continue to be hampered by the immune system. Here, we sought to identify innate and cytokine signaling pathways that promote CD8+ T-cell responses against the transgene product upon AAV1 vector administration to murine skeletal muscle. Eliminating just one of several pathways (including DNA sensing via TLR9, IL-1 receptor signaling, and possibly endosomal sensing of double-stranded RNA) substantially reduced the CD8+ T-cell response at lower vector doses but was surprisingly ineffective at higher doses. Using genetic, antibody-mediated, and vector engineering approaches, we show that blockade of at least two innate pathways is required to achieve an effect at higher vector doses. Concurrent blockade of IL-1R1 > MyD88 and TLR9 > MyD88 > type I IFN > IFNaR pathways was often but not always synergistic and had limited utility in preventing antibody formation against the transgene product. Further, even low-frequency CD8+ T-cell responses could eliminate transgene expression, even in MyD88- or IL-1R1-deficient animals that received a low vector dose. However, we provide evidence that CpG depletion of vector genomes and including TLR9 inhibitory sequences can synergize. When this construct was combined with the use of a muscle-specific promoter, transgene expression in muscle was sustained with minimal local or systemic CD8+ T-cell response. Thus, innate immune avoidance/blockade strategies by themselves, albeit helpful, may not be sufficient to prevent destructive cellular responses in muscle gene transfer because of the redundancy of immune-activating pathways. Full article
(This article belongs to the Special Issue Virology and Immunology of Gene Therapy)
Show Figures

Graphical abstract

Back to TopTop