Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Cell ; 186(2): 446-460.e19, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36638795

ABSTRACT

Precise targeting of large transgenes to T cells using homology-directed repair has been transformative for adoptive cell therapies and T cell biology. Delivery of DNA templates via adeno-associated virus (AAV) has greatly improved knockin efficiencies, but the tropism of current AAV serotypes restricts their use to human T cells employed in immunodeficient mouse models. To enable targeted knockins in murine T cells, we evolved Ark313, a synthetic AAV that exhibits high transduction efficiency in murine T cells. We performed a genome-wide knockout screen and identified QA2 as an essential factor for Ark313 infection. We demonstrate that Ark313 can be used for nucleofection-free DNA delivery, CRISPR-Cas9-mediated knockouts, and targeted integration of large transgenes. Ark313 enables preclinical modeling of Trac-targeted CAR-T and transgenic TCR-T cells in immunocompetent models. Efficient gene targeting in murine T cells holds great potential for improved cell therapies and opens avenues in experimental T cell immunology.


Subject(s)
Dependovirus , Genetic Engineering , T-Lymphocytes , Animals , Mice , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Gene Targeting , Genetic Engineering/methods
2.
J Virol ; 95(19): e0058721, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34232726

ABSTRACT

Adeno-associated viruses utilize different glycans and the AAV receptor (AAVR) for cellular attachment and entry. Directed evolution has yielded new AAV variants; however, structure-function correlates underlying their improved transduction are generally overlooked. Here, we report that infectious cycling of structurally diverse AAV surface loop libraries yields functionally distinct variants. Newly evolved variants show enhanced cellular binding, uptake, and transduction, but through distinct mechanisms. Using glycan-based and genome-wide CRISPR knockout screens, we discover that one AAV variant acquires the ability to recognize sulfated glycosaminoglycans, while another displays receptor switching from AAVR to integrin ß1 (ITGB1). A previously evolved variant, AAVhum.8, preferentially utilizes the ITGB1 receptor over AAVR. Visualization of the AAVhum.8 capsid by cryoelectron microscopy at 2.49-Å resolution localizes the newly acquired integrin recognition motif adjacent to the AAVR footprint. These observations underscore the new finding that distinct AAV surface epitopes can be evolved to exploit different cellular receptors for enhanced transduction. IMPORTANCE Understanding how viruses interact with host cells through cell surface receptors is central to discovery and development of antiviral therapeutics, vaccines, and gene transfer vectors. Here, we demonstrate that distinct epitopes on the surface of adeno-associated viruses can be evolved by infectious cycling to recognize different cell surface carbohydrates and glycoprotein receptors and solve the three-dimensional structure of one such newly evolved AAV capsid, which provides a roadmap for designing viruses with improved attributes for gene therapy applications.


Subject(s)
Dependovirus/genetics , Dependovirus/metabolism , Directed Molecular Evolution , Receptors, Virus/metabolism , Amino Acid Motifs , CRISPR-Cas Systems , Capsid/chemistry , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cryoelectron Microscopy , Dependovirus/chemistry , Dependovirus/ultrastructure , Genetic Variation , Glycosaminoglycans/metabolism , Humans , Integrin beta1/chemistry , Integrin beta1/metabolism , Polysaccharides/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/chemistry , Virus Internalization
3.
J Virol ; 94(19)2020 09 15.
Article in English | MEDLINE | ID: mdl-32669336

ABSTRACT

Adeno-associated viruses (AAV) are composed of nonenveloped, icosahedral protein shells that can be adapted to package and deliver recombinant therapeutic DNA. Approaches to engineer recombinant capsids for gene therapy applications have focused on rational design or library-based approaches that can address one or two desirable attributes; however, there is an unmet need to comprehensively improve AAV vector properties. Such cannot be achieved by utilizing sequence data alone but requires harnessing the three-dimensional (3D) structural properties of AAV capsids. Here, we solve the structures of a natural AAV isolate complexed with antibodies using cryo-electron microscopy and harness this structural information to engineer AAV capsid libraries through saturation mutagenesis of different antigenic footprints. Each surface loop was evolved by infectious cycling in the presence of a helper adenovirus to yield a new AAV variant that then serves as a template for evolving the next surface loop. This stepwise process yielded a humanized AAV8 capsid (AAVhum.8) displaying nonnatural surface loops that simultaneously display tropism for human hepatocytes, increased gene transfer efficiency, and neutralizing antibody evasion. Specifically, AAVhum.8 can better evade neutralizing antisera from multiple species than AAV8. Further, AAVhum.8 displays robust transduction in a human liver xenograft mouse model with expanded tropism for both murine and human hepatocytes. This work supports the hypothesis that critical properties, such as AAV capsid antibody evasion and tropism, can be coevolved by combining rational design and library-based evolution for clinical gene therapy.IMPORTANCE Clinical gene therapy with recombinant AAV vectors has largely relied on natural capsid isolates. There is an unmet need to comprehensively improve AAV tissue tropism, transduction efficiency, and antibody evasion. Such cannot be achieved by utilizing capsid sequence data alone but requires harnessing the 3D structural properties of AAV capsids. Here, we combine rational design and library-based evolution to coevolve multiple, desirable properties onto AAV by harnessing 3D structural information.


Subject(s)
Capsid Proteins/immunology , Capsid/immunology , Dependovirus/immunology , Tropism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line , Cryoelectron Microscopy , Dependovirus/genetics , Genetic Therapy , Hepatocytes/metabolism , Humans , Mice , Molecular Docking Simulation
4.
Proc Natl Acad Sci U S A ; 114(24): E4812-E4821, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28559317

ABSTRACT

Preexisting neutralizing antibodies (NAbs) against adeno-associated viruses (AAVs) pose a major, unresolved challenge that restricts patient enrollment in gene therapy clinical trials using recombinant AAV vectors. Structural studies suggest that despite a high degree of sequence variability, antibody recognition sites or antigenic hotspots on AAVs and other related parvoviruses might be evolutionarily conserved. To test this hypothesis, we developed a structure-guided evolution approach that does not require selective pressure exerted by NAbs. This strategy yielded highly divergent antigenic footprints that do not exist in natural AAV isolates. Specifically, synthetic variants obtained by evolving murine antigenic epitopes on an AAV serotype 1 capsid template can evade NAbs without compromising titer, transduction efficiency, or tissue tropism. One lead AAV variant generated by combining multiple evolved antigenic sites effectively evades polyclonal anti-AAV1 neutralizing sera from immunized mice and rhesus macaques. Furthermore, this variant displays robust immune evasion in nonhuman primate and human serum samples at dilution factors as high as 1:5, currently mandated by several clinical trials. Our results provide evidence that antibody recognition of AAV capsids is conserved across species. This approach can be applied to any AAV strain to evade NAbs in prospective patients for human gene therapy.


Subject(s)
Dependovirus/genetics , Dependovirus/immunology , Directed Molecular Evolution/methods , Immune Evasion/genetics , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/genetics , Antigen-Antibody Complex/immunology , Antigenic Variation/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Dependovirus/classification , Female , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors , HEK293 Cells , Humans , Macaca mulatta , Mice , Mice, Inbred BALB C , Models, Molecular , Serotyping
5.
Nat Commun ; 13(1): 5947, 2022 10 10.
Article in English | MEDLINE | ID: mdl-36210364

ABSTRACT

Recombinant adeno-associated viral (AAV) vectors are a promising gene delivery platform, but ongoing clinical trials continue to highlight a relatively narrow therapeutic window. Effective clinical translation is confounded, at least in part, by differences in AAV biology across animal species. Here, we tackle this challenge by sequentially evolving AAV capsid libraries in mice, pigs and macaques. We discover a highly potent, cross-species compatible variant (AAV.cc47) that shows improved attributes benchmarked against AAV serotype 9 as evidenced by robust reporter and therapeutic gene expression, Cre recombination and CRISPR genome editing in normal and diseased mouse models. Enhanced transduction efficiency of AAV.cc47 vectors is further corroborated in macaques and pigs, providing a strong rationale for potential clinical translation into human gene therapies. We envision that ccAAV vectors may not only improve predictive modeling in preclinical studies, but also clinical translatability by broadening the therapeutic window of AAV based gene therapies.


Subject(s)
Dependovirus , Gene Editing , Animals , Dependovirus/metabolism , Genetic Therapy , Genetic Vectors/genetics , Humans , Macaca/genetics , Mice , Swine , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL