1
|
Liu H, Liu N, Zhou C, Du A, Kapadia M, Tai PWL, Barton E, Gao G, Wang D. High-purity AAV vector production utilizing recombination-dependent minicircle formation and genetic coupling. EMBO Mol Med 2025; 17:1475-1494. [PMID: 40379974 PMCID: PMC12162853 DOI: 10.1038/s44321-025-00248-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/19/2025] Open
Abstract
Triple transfection of HEK293 cells is the most widely used method for producing recombinant adeno-associated virus (rAAV), a leading gene delivery vector for human gene therapy. Despite its tremendous success, this approach generates several vector-related impurities that could potentially compromise the safety and potency of rAAV. In this study, we introduce a method for high-purity AAV vector production utilizing recombination-dependent minicircle formation and genetic coupling (AAVPureMfg). Compared with traditional triple transfection, AAVPureMfg substantially improves vector purity by reducing prokaryotic DNA contaminants by 10- to 50-fold and increasing the full capsid ratio up to threefold. Mechanistically, Bxb1-mediated excision of the transgene cassette generates a minicircle cis construct devoid of bacterial sequences and ensures synchronized colocalization of trans and cis constructs in productive cells. Furthermore, we developed iterations that enhance vector genome homogeneity and streamline the production of rAAV with various transgenes, serotypes, and ITR configurations. Overall, our findings demonstrate that AAVPureMfg overcomes the inherent limitations associated with triple transfection, offering a broadly applicable and easy-to-implement method for producing high-purity rAAV with reduced plasmid costs.
Collapse
Affiliation(s)
- Hao Liu
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Nan Liu
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Chen Zhou
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Ailing Du
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Mayank Kapadia
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Phillip W L Tai
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Erik Barton
- Pfizer Inc., Worldwide Research, Development and Medical, Bioprocess Research and Development, Chesterfield, MO, 63017, USA
| | - Guangping Gao
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| | - Dan Wang
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
2
|
Yamaguchi Y, Shimojo S, Shibuya R, Bandoh K, Matsushita A, Fukuhara M, Tsunaka Y, Torisu T, Uchiyama S. Direct Identification and Quantification of Recombinant Adeno-Associated Virus in Crude Cell Lysate and Conditioned Medium by Mass Photometry. Anal Chem 2025; 97:10405-10416. [PMID: 40326171 PMCID: PMC12096344 DOI: 10.1021/acs.analchem.5c00793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Recombinant adeno-associated virus (rAAV) has attracted attention as a gene therapy vector. Monitoring the percentage of full particles (FPs) to the sum of empty particles (EPs) and FPs (F/E ratio) is required to optimize the rAAV production conditions; however, there is a lack of analytical methods to identify FPs and EPs and quantify the F/E ratio of rAAV without purification. Here, we established a direct analysis method for identifying FPs and EPs and quantifying the F/E ratio and genomic titer of unpurified rAAV in crude cell lysate and conditioned medium by mass photometry (MP). MP can detect the events of both molecules that bind to the glass surface and molecules that unbind from the glass surface. Few unbinding molecules were detected in the cell lysate and conditioned medium, but unbinding particles were as prevalent as binding particles in rAAV. By analyzing the unbinding side of the histogram, the F/E ratio of rAAV in the cell lysate was directly quantified with accuracy comparable to that of purified rAAV, which showed there was no interference from impurities. The genomic titer of rAAV in cell lysate was also estimated using particle counts of the unbinding side. This method can successfully determine the F/E ratio and estimate genomic titers of rAAV in crude cell lysate and conditioned medium during the manufacturing process. Direct quantification by MP is a convenient, rapid, and accurate method for quantifying unpurified rAAV and will be useful for improving rAAV production processes, for example, by screening manufacturing conditions.
Collapse
Affiliation(s)
- Yuki Yamaguchi
- Department
of Biotechnology, Graduate School of Engineering, The University of Osaka, 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
| | - Saki Shimojo
- Department
of Biotechnology, Graduate School of Engineering, The University of Osaka, 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
| | - Risa Shibuya
- Department
of Biotechnology, Graduate School of Engineering, The University of Osaka, 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
| | - Karin Bandoh
- Department
of Biotechnology, Graduate School of Engineering, The University of Osaka, 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
| | | | - Mitsuko Fukuhara
- Department
of Biotechnology, Graduate School of Engineering, The University of Osaka, 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
- U-Medico
Inc., 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
| | - Yasuo Tsunaka
- Department
of Biotechnology, Graduate School of Engineering, The University of Osaka, 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
| | - Tetsuo Torisu
- Department
of Biotechnology, Graduate School of Engineering, The University of Osaka, 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
| | - Susumu Uchiyama
- Department
of Biotechnology, Graduate School of Engineering, The University of Osaka, 2-1 Yamadaoka, Suita, Osaka565-0871, Japan
| |
Collapse
|
3
|
Suarez-Amaran L, Song L, Tretiakova AP, Mikhail SA, Samulski RJ. AAV vector development, back to the future. Mol Ther 2025; 33:1903-1936. [PMID: 40186350 DOI: 10.1016/j.ymthe.2025.03.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025] Open
Abstract
Adeno-associated virus (AAV) has become a pivotal tool in gene therapy, providing a safe and efficient platform for long-term transgene expression. This review presents a comprehensive analysis of AAV's historical development, from its initial identification as a "contaminant" to its current clinical applications. We examine the molecular evolution of AAV, detailing advancements in vector engineering, rational design, directed evolution platforms, and computational modeling, which have expanded its therapeutic potential across diverse disease areas. Additionally, we explore AAV genome regulation, with a particular focus on inverted terminal repeats (ITRs) and AAV capsid-genome interactions, which play a crucial role in vector transduction efficiency and host adaptation. An assessment of past and present clinical trials as well as future directions is provided to illustrate the field's trajectory. Finally, another unique milestone in AAV research is also reported; namely, a pool of AAV libraries has been successfully administered to human decedents and analyzed, representing a transformative step in AAV evolution and selection for human applications. These studies should pave the way for more refined AAV vector optimization, accelerating the development of next-generation gene therapies with enhanced clinical translatability, potentially accelerating the gene therapy revolution.
Collapse
Affiliation(s)
- Lester Suarez-Amaran
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA; Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Liujiang Song
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Anna P Tretiakova
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Sheila A Mikhail
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Richard Jude Samulski
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA; Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
4
|
Herzog CR, Zhang J, Feng X, Dang TT, Yu X, Huang J, Fang F, Gao H, Yu X, Wang Y, Han R, Liu Y, Cornetta K, Xiao W, Xu W. Bioinformatic Analysis of the Genetic Basis of Differential Adeno-Associated Virus Production Capability of 293 Variants. Hum Gene Ther 2025; 36:801-813. [PMID: 40293710 DOI: 10.1089/hum.2025.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Human embryonic kidney 293 (HEK 293) cells are the main producer cell line for recombinant adeno-associated virus (rAAV) production. However, AAV vector yields among 293 clones vary considerably. To elucidate the biological basis for these differences, whole genomes of an adherent and a suspension 293 cell clone with high-yield rAAV were sequenced using nanopore technology. All 293 cell derivative lines showed a twofold copy number gain at the adenoviral integration site across, suggesting a genome duplication event. To our surprise, the two high-producer clones, despite having been separately developed, are biologically closely grouped together as compared to other commonly used 293 clones. Their genomes contain a similar adenoviral gene integration region, which likely leads to high expression of proteins that facilitate AAV replication and packaging. Thus, genome duplication in the adenovirus integration locus may be a key factor affecting AAV production yield.
Collapse
Affiliation(s)
- Christopher R Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Junping Zhang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Xiaomin Feng
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| | - Thao Thi Dang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
| | - Xiangping Yu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jie Huang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Medical Genomics, Indiana University, Indianapolis, Indiana, USA
| | - Fang Fang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
- Center for Medical Genomics, Indiana University, Indianapolis, Indiana, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
- Center for Medical Genomics, Indiana University, Indianapolis, Indiana, USA
| | - Xuhong Yu
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
- Center for Medical Genomics, Indiana University, Indianapolis, Indiana, USA
| | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
- Center for Medical Genomics, Indiana University, Indianapolis, Indiana, USA
| | - Renzhi Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yulong Liu
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
- Center for Medical Genomics, Indiana University, Indianapolis, Indiana, USA
| | - Kenneth Cornetta
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, USA
| | - Weidong Xiao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Weihong Xu
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Stanford Genome Technology Center, Palo Alto, California, USA
| |
Collapse
|
5
|
Zhou K, Yuan M, Sun J, Zhang F, Li X, Xiao X, Wu X. Co-delivery of IL-1Ra and SOX9 via AAV inhibits inflammation and promotes cartilage repair in surgically induced osteoarthritis animal models. Gene Ther 2025; 32:211-222. [PMID: 39833570 DOI: 10.1038/s41434-025-00515-y] [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: 10/29/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Osteoarthritis (OA), a prevalent joint disorder, can lead to disability, with no effective treatment available. Interleukin-1 (IL-1) plays a crucial role in the progression of OA, and its receptor antagonist (IL-1Ra), a natural IL-1 inhibitor, represents a promising therapeutic target by obstructing the IL-1 signaling pathway. This study delivered IL-1Ra via adeno-associated virus (AAV), a gene therapy vector enabling long-term protein expression, to treat knee osteoarthritis (KOA) in animal models. scAAV-oIL-1Ra-I1/2 injected directly into the joint in both MMT/ACLT-induced KOA model rat improved abnormal gait (increasing footprint area and pressure), subchondral bone lesions, and significantly reduced cartilage wear and pathological scores. In the MMT-induced KOA rabbit model, weight-bearing asymmetry (indicating pain) improved after 8 weeks of scAAV-oIL-1Ra-I1/2 administration, and X-ray showed decreased K-L scores (severity grade), reduced cartilage loss, and lower pathology scores compared to untreated animals. Additionally, sex-determining region Y-type high mobility group box 9 (SOX9) was co-delivered with IL-1Ra via AAV in ACLT + MMT-induced KOA rats. The combined treatment significantly alleviated subchondral bone lesions, cartilage destruction, synovial inflammation, and pathological scores, demonstrating superior efficacy compared to either treatment administered alone. Co-delivering IL-1Ra and SOX9 inhibited IL-1 mediated inflammatory signaling, maintained cartilage homeostasis, and promoted its repair in KOA models, suggesting potential for clinical use.
Collapse
Affiliation(s)
- Kaiyi Zhou
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Meng Yuan
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiabao Sun
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Feixu Zhang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Xinting Li
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Xiao Xiao
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China.
| | - Xia Wu
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
6
|
Xia K, Liu S, Wu Z, Jiang JH. Research Status and Applications of Adeno-Associated Virus. Chembiochem 2025; 26:e202400856. [PMID: 39724465 DOI: 10.1002/cbic.202400856] [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: 10/15/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Adeno-associated virus (AAV) has emerged as a powerful and effective tool for the delivery of exogenous genes into various cells or tissues. To improve the gene delivery efficiency, as well as the safety and specificity of AAV's cell-targeting capabilities, extensive investigations have been conducted into its molecular biological characteristics, including capsid structure, cellular tropism, and the mechanisms underlying its entry, replication, DNA packaging, and capsid assembly. Significant differences exist between human and non-human primate AAVs regarding tissue targeting and transduction efficiency. These differences are primarily attributed to the amino acid sequences of AAV capsid proteins, the structural characteristics of these proteins, and the interactions of AAV with surface factors on host cells, such as cell surface receptors, signaling molecules, and associated proteins. This review primarily focuses on several key aspects of AAV, including its genome, coat proteins and their structures, genome replication, virus assembly, and the role of helper viruses. Additionally, it examines the utilization of recombinant adeno-associated viruses (rAAV), detailing their production methods, mechanisms of cell entry and trafficking, and various serotypes. The review further interprets the role of rAAV by analyzing its current applications in research and therapy.
Collapse
Affiliation(s)
- Ke Xia
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Shuangling Liu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Zhenkun Wu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
7
|
Ladiwala P, Ndahiro N, Hauk P, Wen J, Sargunas J, Chen YJ, Barton E, Betenbaugh MJ. Unraveling Cytotoxicity in HEK293 Cells During Recombinant AAV Production for Gene Therapy Applications. Biotechnol J 2025; 20:e202400501. [PMID: 40079705 DOI: 10.1002/biot.202400501] [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: 08/14/2024] [Revised: 01/05/2025] [Accepted: 01/23/2025] [Indexed: 03/15/2025]
Abstract
Transient transfection of HEK293 cells represents the dominant technique for the production of recombinant adeno-associated virus (AAV) vectors. However, recombinant AAV (rAAV) production is cytotoxic, potentially impacting process performance, product yields, and quality, complicating downstream processing. This study characterizes cell death response for rAAV producing HEK293 cells and explores the potential to control cytotoxicity. Initial analysis of triple transfected cells revealed caspase-mediated apoptosis as a likely mechanism of cellular death. Next, the causes of this cytotoxicity were investigated by dissecting transfection steps. Exposing cells to polyethyleneimine (PEI) alone or complexed with a blank plasmid at typical concentrations had a limited impact on cell growth. However, the inclusion of plasmid constructs containing genes to produce rAAVs triggered significant cell death, with the helper plasmid being the most toxic both independently and in combination with packaging and transgene plasmids. Additionally, apoptosis in transfected cultures could be inhibited using the pan-caspase inhibitor, N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (Z-VAD.fmk), leading to a 65% increase in peak viable cell density (VCD). Although the rAAV genome titer remained relatively unaltered, capsid levels declined upon cell death inhibition. Consequently, the ratio of full to empty capsids, an important product quality attribute (PQA) for rAAVs increased following caspase inhibition. This study provides insights into apoptosis activation in rAAVs and uncovers avenues for its modulation to alter PQAs.
Collapse
Affiliation(s)
- Pranay Ladiwala
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nelson Ndahiro
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pricila Hauk
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Junneng Wen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justin Sargunas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yu-Ju Chen
- Pfizer Inc., Chesterfield, Missouri, USA
| | | | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Wang S, Xiao L. Progress in AAV-Mediated In Vivo Gene Therapy and Its Applications in Central Nervous System Diseases. Int J Mol Sci 2025; 26:2213. [PMID: 40076831 PMCID: PMC11899905 DOI: 10.3390/ijms26052213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
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 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.
Collapse
Affiliation(s)
- Shuming Wang
- Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China;
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Lin Xiao
- Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China;
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
9
|
Deng Z, Lv YL, Wang XT, Yuan LH, Zhao K, Du ZM, Xiao X. Production of Recombinant Adeno-Associated Virus Through High-Cell-Density Transfection of HEK293 Cells Based on Fed-Perfusion Culture. Hum Gene Ther 2025; 36:116-127. [PMID: 39761134 DOI: 10.1089/hum.2024.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Adeno-associated virus (AAV)-associated gene therapy has been increasingly promising, in light of the drugs progressed to clinical trials or approved for medications internationally. Therefore, scalable and efficient production of recombinant AAV is pivotal for advancing gene therapy. Traditional methods, such as the triple-plasmid transfection of human embryonic kidney 293 cells in suspension culture, have been widely employed but often hampered by low unit yield. In this study, we optimized the cell culture process with high cell density up to 2 × 107 cells/mL by employing a perfusion culture system with centrifugation and medium exchange in shake flasks and perfusion device in bioreactor. Furthermore, we utilized a design of experiments strategy to systematically modulate a series of transfection-related variables including the quantity of plasmid DNA, the DNA-to-polyethylenimine ratio, incubation duration, and the impact of post-transfection feeding strategies on the yield of recombinant AAV (rAAV). Our comprehensive analysis and subsequent optimizations actualized a remarkable unit yield reaching nearly 2 × 1012 vector genomes (vg)/mL. Importantly, the resulting single-cell yield and biological activity were found to be comparable with those obtained from fed-batch cultures, underscoring the efficacy of our approach. Based on these findings, we investigated rAAV yield via high-density suspend culture in bioreactor, particularly focusing on cell aggregation and the use of perfusion technology. Intriguingly, we attempted to elevate the yield of an oversized recombinant coagulation factor VIII AAV843 vector by 3.5-fold, reaching a yield of 1 × 1012 vg/mL. Concurrently, the medium usage rate was only double that of batch feeding, thereby significantly shrinking the upstream cost of rAAV manufacture. In summary, this strategy significantly benefits large-scale AAV production for both commercial and clinical applications.
Collapse
Affiliation(s)
- Zhe Deng
- School of Bioengineering, East China University of Science and Technology, Shanghai, China
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Yan-Ling Lv
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Xin-Tao Wang
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Long-Hui Yuan
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Kai Zhao
- School of Bioengineering, East China University of Science and Technology, Shanghai, China
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Zeng-Min Du
- School of Bioengineering, East China University of Science and Technology, Shanghai, China
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Xiao Xiao
- School of Bioengineering, East China University of Science and Technology, Shanghai, China
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
10
|
Yang R, Tran NT, Chen T, Cui M, Wang Y, Sharma T, Liu Y, Zhang J, Yuan X, Zhang D, Chen C, Shi Z, Wang L, Dai Y, Zaidi H, Liang J, Chen M, Jaijyan D, Hu H, Wang B, Xu C, Hu W, Gao G, Yu D, Tai PWL, Wang Q. AAVone: A Cost-Effective, Single-Plasmid Solution for Efficient AAV Production with Reduced DNA Impurities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631712. [PMID: 39829756 PMCID: PMC11741346 DOI: 10.1101/2025.01.07.631712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Currently, the most common approach for manufacturing GMP-grade adeno-associated virus (AAV) vectors involves transiently transfecting mammalian cells with three plasmids that carry the essential components for production. The requirement for all three plasmids to be transfected into a single cell and the necessity for high quantities of input plasmid DNA, limits AAV production efficiency, introduces variability between production batches, and increases time and labor costs. Here, we developed an all-in-one, single-plasmid AAV production system, called AAVone. In this system, the adenovirus helper genes ( E2A , E4orf6 , and VA RNA ), packaging genes ( rep and cap ), and the vector transgene cassette are consolidated into a single compact plasmid with a 13-kb backbone. The AAVone system achieves a two- to four-fold increase in yields compared to the traditional triple-plasmid system. Furthermore, the AAVone system exhibits low batch-to-batch variation and eliminates the need for fine-tuning the ratios of the three plasmids, simplifying the production process. In terms of vector quality, AAVs generated by the AAVone system show similar in vitro and in vivo transduction efficiency, but a substantial reduction in sequences attributed to plasmid backbones and a marked reduction in non-functional snap-back genomes. In Summary, the AAVone platform is a straightforward, cost-effective, and highly consistent AAV production system - making it particularly suitable for GMP-grade AAV vectors.
Collapse
|
11
|
Wei L, Yu P, Wang H, Liu J. Adeno-associated viral vectors deliver gene vaccines. Eur J Med Chem 2025; 281:117010. [PMID: 39488197 DOI: 10.1016/j.ejmech.2024.117010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Adeno-associated viruses (AAVs) are leading platforms for in vivo delivery of gene therapies, with six licensed AAV-based therapeutics attributed to their non-pathogenic nature, low immunogenicity, and high efficiency. In the realm of gene-based vaccines, one of the most vital therapeutic areas, AAVs are also emerging as promising delivery tools. We scrutinized AAVs, focusing on their virological properties, as well as bioengineering and chemical modifications to demonstrate their significant potential in gene vaccine delivery, and detailing the preparation of AAV particles. Additionally, we summarized the use of AAV vectors in vaccines for both infectious and non-infectious diseases, such as influenza, COVID-19, Alzheimer's disease, and cancer. Furthermore, this review, along with the latest clinical trial updates, provides a comprehensive overview of studies on the potential of using AAV vectors for gene vaccine delivery. It aims to deepen our understanding of the challenges and limitations in nucleic acid delivery and pave the way for future clinical success.
Collapse
Affiliation(s)
- Lai Wei
- College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Peng Yu
- College of Biotechnology, Tianjin University of Science & Technology, 300457 Tianjin, China
| | - Haomeng Wang
- CanSino (Shanghai) Biological Research Co., Ltd, 201208, Shanghai, China.
| | - Jiang Liu
- Rosalind Franklin Institute, Harwell Campus, OX11 0QS, Oxford, United Kingdom; Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT, Oxford, United Kingdom.
| |
Collapse
|
12
|
Nie Y, Pan H, Li Q, Na H, Figueroa B, Vincent K. Characterization of the function of Adenovirus L4 gene products and their impact on AAV vector production. Mol Ther Methods Clin Dev 2024; 32:101370. [PMID: 39640223 PMCID: PMC11617683 DOI: 10.1016/j.omtm.2024.101370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024]
Abstract
Efficient manufacturing of recombinant adenovirus-associated virus (AAV) vectors is critical to the successful development of genomic medicines. We attempted to optimize AAV vector production in a producer cell line platform. In this system, helper functions required for AAV replication and production are provided via infection with a replication-competent wild-type Adenovirus. To evaluate strategies for the reduction of replication and packaging of adenovirus and to understand the interplay of recombinant AAV and the helper virus during AAV vector production, wild-type adenovirus was compared to a mutant (Ad5ts149) containing a temperature-sensitive mutation in the DNA polymerase gene. Infection of a producer cell line with Ad5ts149 at the restrictive temperature reduced recombinant AAV titer and altered the pattern of AAV protein expression. Further investigation revealed that the adenoviral late L4-22K/33K gene products regulated both AAV rep/cap gene transcription and splicing of the rep/cap transcripts. Furthermore, the L4-33K gene products were found to impact AAV production in both the producer cell line and transient transfection platforms. Optimization of Adenovirus L4-22K/33K expression to facilitate efficient expression and splicing of AAV rep/cap transcripts therefore represents a unique opportunity to optimize AAV vector production.
Collapse
Affiliation(s)
- Yingchao Nie
- Genomic Medicine Unit CMC, Global CMC Development, Sanofi R&D, 225 2nd Avenue, Waltham, MA 02451, USA
| | - Hao Pan
- Genomic Medicine Unit CMC, Global CMC Development, Sanofi R&D, 225 2nd Avenue, Waltham, MA 02451, USA
| | - Qingliang Li
- Genomic Medicine Unit CMC, Global CMC Development, Sanofi R&D, 225 2nd Avenue, Waltham, MA 02451, USA
| | - Huimin Na
- Genomic Medicine Unit CMC, Global CMC Development, Sanofi R&D, 225 2nd Avenue, Waltham, MA 02451, USA
| | - Bruno Figueroa
- Genomic Medicine Unit CMC, Global CMC Development, Sanofi R&D, 225 2nd Avenue, Waltham, MA 02451, USA
| | - Karen Vincent
- Genomic Medicine Unit CMC, Global CMC Development, Sanofi R&D, 225 2nd Avenue, Waltham, MA 02451, USA
| |
Collapse
|
13
|
Doshi J, Couto E, Staiti J, Vandenberghe LH, Zabaleta N. E2A, VA RNA I, and L4-22k adenoviral helper genes are sufficient for AAV production in HEK293 cells. Mol Ther Methods Clin Dev 2024; 32:101376. [PMID: 39670178 PMCID: PMC11635002 DOI: 10.1016/j.omtm.2024.101376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024]
Abstract
The replication-defective adeno-associated virus (AAV) is extensively utilized as a research tool or vector for gene therapy. The production process of AAV remains intricate, expensive, and mechanistically underexplored. With the aim of enhancing AAV manufacturing efficiencies in mammalian cells, we revisited the questions and optimization surrounding the requirement of the various adenoviral helper genes in enabling AAV production. First, we refined the minimal set of adenoviral genes in HEK293 AAV production to E2A, L4-22 K /33 K, and VA RNA I. These findings challenge the previously accepted necessity of adenoviral E4orf6 in AAV production. In addition, we identified L4-22 K genes as crucial helpers for AAV production. Next, a revised minimal adenoviral helper plasmid comprising E2A, L4-22 K, and VA RNA I genes was designed and demonstrated to yield high titer and potent AAV preps in HEK293 transient transfection. Lastly, stable packaging cells harboring inducible E2A and L4-22 K genes were shown to maintain AAV production yields comparable to transient transfection over a culture period of ∼10 weeks. Combined, these findings further our understanding of adenoviral helper function in mammalian AAV production and provide novel plasmid and cell-line reagents with an improved safety profile for potential broad applicability in the research and gene therapy community.
Collapse
Affiliation(s)
- Jiten Doshi
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Emma Couto
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary, Boston, MA, USA
| | - Jillian Staiti
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary, Boston, MA, USA
| | - Luk H. Vandenberghe
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Nerea Zabaleta
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| |
Collapse
|
14
|
Powers TW, Sloan CD, Stano D, Evans B, Liu K, Mariani S, Campbell JA, Lerch TF, Mo JJ. Implementing a robust platform analytical procedure for measuring adeno-associated virus vector genome titer. Mol Ther Methods Clin Dev 2024; 32:101381. [PMID: 39670179 PMCID: PMC11634990 DOI: 10.1016/j.omtm.2024.101381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024]
Abstract
The vector genome (vg) titer measurement, which is used to control patient dosing and ensure control over drug product manufacturing, is essential for the development of recombinant adeno-associated virus (AAV) gene therapy products. While qPCR and droplet digital PCR technologies are commonly implemented for measuring vg titer, chromatographic techniques with UV detectors represent promising future approaches, in line with traditional biotherapeutics. Here, we introduce a novel vg titer measurement approach using size-exclusion high-performance liquid chromatography with UV detection, which achieves excellent method precision (<2% relative SD), demonstrates linearity across a range of concentrations and varied particle content, is stability indicating, and can be bridged with existing vg titer methods. As there is no bias between this procedure and existing vg titer procedures, such as qPCR, this method can be implemented even at late stages during pharmaceutical development. The procedure was demonstrated to be applicable across serotypes and transgenes, enabling the approach to be used as a platform method for AAV. Given the method performance and criticality of vg titer measurements for AAV, this approach represents a beneficial technology for AAV therapeutics.
Collapse
Affiliation(s)
- Thomas W. Powers
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Courtney D.K. Sloan
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Don Stano
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Brad Evans
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Kang Liu
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Shawn Mariani
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Jessica A. Campbell
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Thomas F. Lerch
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Jim J. Mo
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| |
Collapse
|
15
|
Cruz FM, Moreno-Manuel AI, Pérez PS, Ruiz-Robles JM, Socuellamos PG, Gutiérrez LK, Vera-Pedrosa ML, Gutierrez AT, Mondéjar Parreño G, Macías Á, Martínez-Carrascoso I, Bermúdez-Jiménez FJ, Arias Santiago S, Martínez de Benito F, Braza-Boils A, Valenzuela C, Morillo CA, Zorio E, Jiménez-Jaimez J, Jalife J. Kir2.1 mutations differentially increase the risk of flecainide proarrhythmia in Andersen Tawil Syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.10.24318629. [PMID: 39711719 PMCID: PMC11661358 DOI: 10.1101/2024.12.10.24318629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Background Flecainide and other class-Ic antiarrhythmic drugs (AADs) are widely used in Andersen-Tawil syndrome type 1 (ATS1) patients. However, class-Ic drugs might be proarrhythmic in some cases. We investigated the molecular mechanisms of class-I AADs proarrhythmia and whether they might increase the risk of death in ATS1 patients with structurally normal hearts. Methods and Results Of 53 ATS1 patients reviewed from the literature, 54% responded partially to flecainide, with ventricular arrhythmia (VA) reduction in only 23%. Of the latter patients, VA persisted in 20-50%. Flecainide was ineffective in 23%, and surprisingly, 13.5% suffered a non-fatal cardiac arrest. In five cardiac-specific ATS1 mouse models (Kir2.1Δ314-315, Kir2.1C122Y, Kir2.1G215D and Kir2.1R67W and Kir2.1S136F), flecainide or propafenone (40 mg/Kg i.p.) differentially prolonged the P wave, and the PR, QRS and QTc intervals compared to Kir2.1WT; Kir2.1S136F had milder effects. Flecainide increased VA inducibility in all mutant mice except Kir2.1S136F, which exhibited significant VA reduction. At baseline, Kir2.1G215D cardiomyocytes had the lowest inward rectifier K+ channel (IK1) reduction, followed by Kir2.1C122Y, Kir2.1R67W and Kir2.1S136F. Kir2.1C122Y cardiomyocytes had a significant decrease in sodium inward current (INa). Flecainide (10 μM) slightly increased IK1 density in Kir2.1WT and Kir2.1S136F, while it decreased both IK1 and INa in Kir2.1C122Y and Kir2.1R67W, despite normal trafficking of mutant channels. Optical mapping in ATS1 patient-specific iPSC-CM monolayers expressing Kir2.1C122Y, Kir2.1G215D and Kir2.1R67W showed an increase in rotor incidence at baseline and under flecainide, confirming the drugś proarrhythmic effect. Lastly, in-silico molecular docking predicts that the Kir2.1-Cys311 pharmacophore-binding site is altered in Kir2.1C122Y heterotetramers, reducing flecainide accessibility and leading to channel closure and arrhythmias. Conclusions Class-Ic AADs are only partially effective and might be proarrhythmic in some ATS1 patients. Kir2.1 mutations impacting the resting membrane potential and cellular excitability create a substrate for life-threatening arrhythmias, raising significant concern about using these drugs in some ATS1 patients.
Collapse
Affiliation(s)
- Francisco M. Cruz
- Spanish National Centre for Cardiovascular Research (CNIC), Madrid, Spain
| | | | | | | | | | | | | | | | | | - Álvaro Macías
- Spanish National Centre for Cardiovascular Research (CNIC), Madrid, Spain
| | | | - Francisco J Bermúdez-Jiménez
- Spanish National Centre for Cardiovascular Research (CNIC), Madrid, Spain
- Cardiology Service, Virgen de las Nieves University Hospital, Granada, Spain
- Institute of Biosanitary Research of Granada (IBS), Spain
| | | | - Fernando Martínez de Benito
- Spanish National Centre for Cardiovascular Research (CNIC), Madrid, Spain
- Center for Biomedical Network Research on Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Aitana Braza-Boils
- Center for Biomedical Network Research on Cardiovascular Diseases (CIBERCV), Madrid, Spain
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | | | - CA Morillo
- Spanish National Centre for Cardiovascular Research (CNIC), Madrid, Spain
- Department of Cardiac Sciences, Libin CVI, University of Calgary, Canada
| | - Esther Zorio
- Center for Biomedical Network Research on Cardiovascular Diseases (CIBERCV), Madrid, Spain
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Inherited Cardiac Disease Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
- Medicine Department, University of Valencia, Spain
| | - Juan Jiménez-Jaimez
- Cardiology Service, Virgen de las Nieves University Hospital, Granada, Spain
- Institute of Biosanitary Research of Granada (IBS), Spain
| | - José Jalife
- Spanish National Centre for Cardiovascular Research (CNIC), Madrid, Spain
- Center for Biomedical Network Research on Cardiovascular Diseases (CIBERCV), Madrid, Spain
- Departments of Medicine and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
16
|
Lam P, Zygmunt DA, Ashbrook A, Yan C, Du H, Martin PT. Liver-directed AAV gene therapy normalizes disease symptoms and provides cross-correction in a model of lysosomal acid lipase deficiency. Mol Ther 2024; 32:4272-4284. [PMID: 39489913 PMCID: PMC11638878 DOI: 10.1016/j.ymthe.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Lysosomal acid lipase deficiency (LAL-D) is caused by mutations in the LIPA gene, which encodes the lysosomal enzyme that hydrolyzes triglycerides and cholesteryl esters to free fatty acids and free cholesterol. The objective of this study was to develop a curative single-treatment therapy for LAL-D using adeno-associated virus (AAV). Treatment at both early (1-2 days) and late (8-week) timepoints with rscAAVrh74.LP1.LIPA, a liver-directed AAV gene therapy, normalized many disease measures in Lipa-/- mice when measured at 24 weeks of age, including hepatosplenomegaly, serum transaminase activity, organ triglyceride and cholesterol levels, and biomarkers of liver inflammation and fibrosis. For most measures, liver-directed therapy was superior to therapy utilizing a constitutive tissue expression approach. rscAAVrh74.LP1.LIPA treatment elevated LAL enzyme activity above wild-type levels in all tissues tested, including liver, spleen, intestine, muscle, and brain, and treatment elicited minimal serum antibody responses to transgenic protein. AAV treatment at 8 weeks of age with 1 × 1013 vg/kg extended survival significantly, with all AAV-treated mice surviving beyond the maximal lifespan of untreated Lipa-/- mice. These results show that this liver-directed LIPA gene therapy has the potential to be a transformative treatment for LAL-D.
Collapse
Affiliation(s)
- Patricia Lam
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Deborah A Zygmunt
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Anna Ashbrook
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Cong Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hong Du
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paul T Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA; Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
17
|
Srinivasan P, Canova CT, Sha S, Nguyen TNT, Joseph J, Sangerman J, Maloney AJ, Katsikis G, Ou RW, Hong MS, Ng J, Yuan A, Antov D, Song S, Chen W, Neufeld C, Wolfrum JM, Barone PW, Sinskey AJ, Springs SL, Braatz RD. Multidose transient transfection of human embryonic kidney 293 cells modulates recombinant adeno-associated virus2/5 Rep protein expression and influences the enrichment fraction of filled capsids. Biotechnol Bioeng 2024; 121:3694-3714. [PMID: 39176568 DOI: 10.1002/bit.28828] [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: 02/02/2024] [Revised: 07/04/2024] [Accepted: 08/04/2024] [Indexed: 08/24/2024]
Abstract
Recombinant adeno-associated virus (rAAV) is a commonly used in vivo gene therapy vector because of its nonpathogenicity, long-term transgene expression, broad tropism, and ability to transduce both dividing and nondividing cells. However, rAAV vector production via transient transfection of mammalian cells typically yields a low fraction of filled-to-total capsids (~1%-30% of total capsids produced). Analysis of our previously developed mechanistic model for rAAV2/5 production attributed these low fill fractions to a poorly coordinated timeline between capsid synthesis and viral DNA replication and the repression of later phase capsid formation by Rep proteins. Here, we extend the model by quantifying the expression dynamics of total Rep proteins and their influence on the key steps of rAAV2/5 production using a multiple dosing transfection of human embryonic kidney 293 (HEK293) cells. We report that the availability of preformed empty capsids and viral DNA copies per cell are not limiting to the capsid-filling reaction. However, optimal expression of Rep proteins (<240 ± 13 ag per cell) enables enrichment of the filled capsid population (>12% of total capsids/cell) upstream. Our analysis suggests increased enrichment of filled capsids via regulating the expression of Rep proteins is possible but at the expense of per cell capsid titer in a triple plasmid transfection. Our study reveals an intrinsic limitation of scaling rAAV2/5 vector genome (vg) production and underscores the need for approaches that allow for regulating the expression of Rep proteins to maximize vg titer per cell upstream.
Collapse
Affiliation(s)
- Prasanna Srinivasan
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Christopher T Canova
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Sha Sha
- Ultragenyx Pharmaceutical Inc., Novato, Cambridge, USA
| | | | - John Joseph
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jose Sangerman
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | | - Rui Wen Ou
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Moo Sun Hong
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jaclyn Ng
- Stanford University School of Medicine, Stanford, California, USA
| | - Arella Yuan
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Daniel Antov
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Sally Song
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Wenyu Chen
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Caleb Neufeld
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jacqueline M Wolfrum
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Paul W Barone
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Anthony J Sinskey
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Stacy L Springs
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard D Braatz
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
18
|
Lkharrazi A, Tobler K, Marti S, Bratus-Neuenschwander A, Vogt B, Fraefel C. AAV2 can replicate its DNA by a rolling hairpin or rolling circle mechanism, depending on the helper virus. J Virol 2024; 98:e0128224. [PMID: 39382273 PMCID: PMC11575299 DOI: 10.1128/jvi.01282-24] [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: 07/26/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
Adeno-associated virus type 2 (AAV2) is a small, non-pathogenic, helper virus-dependent parvovirus with a single-stranded (ss) DNA genome of approximately 4.7 kb. AAV2 DNA replication requires the presence of a helper virus such as adenovirus type 5 (AdV5) or herpes simplex virus type 1 (HSV-1) and is generally assumed to occur as a strand-displacement rolling hairpin (RHR) mechanism initiated at the AAV2 3' inverted terminal repeat (ITR). We have recently shown that AAV2 replication supported by HSV-1 leads to the formation of double-stranded head-to-tail concatemers, which provides evidence for a rolling circle replication (RCR) mechanism. We have revisited AAV2 DNA replication and specifically compared the formation of AAV2 replication intermediates in the presence of either HSV-1 or AdV5 as the helper virus. The results confirmed that the AAV2 DNA replication mechanism is helper virus-dependent and follows a strand-displacement RHR mechanism when AdV5 is the helper virus and primarily an RCR mechanism when HSV-1 is the helper virus. We also demonstrate that recombination plays a negligible role in AAV2 genome replication. Interestingly, the formation of high-molecular-weight AAV2 DNA concatemers in the presence of HSV-1 as the helper virus was dependent on an intact HSV-1 DNA polymerase. IMPORTANCE AAV is a small helper virus-dependent, non-pathogenic parvovirus. The AAV genome replication mechanism was extensively studied in the presence of AdV as the helper virus and described to proceed using RHR. Surprisingly, HSV-1 co-infection facilitates RCR of the AAV2 DNA. We directly compared AdV5 and HSV-1 supported AAV2 DNA replication and showed that AAV2 can adapt its replication mechanism to the helper virus. A detailed understanding of the AAV replication mechanism expands our knowledge of virus biology and can contribute to increase gene therapy vector production.
Collapse
Affiliation(s)
- Anouk Lkharrazi
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Kurt Tobler
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Sara Marti
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | | | - Bernd Vogt
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Cornel Fraefel
- Institute of Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Dong C, Tan D, Sun H, Li Z, Zhang L, Zheng Y, Liu S, Zhang Y, He Q. Interleukin-12 Delivery Strategies and Advances in Tumor Immunotherapy. Curr Issues Mol Biol 2024; 46:11548-11579. [PMID: 39451566 PMCID: PMC11506767 DOI: 10.3390/cimb46100686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Interleukin-12 (IL-12) is considered to be a promising cytokine for enhancing an antitumor immune response; however, recombinant IL-12 has shown significant toxicity and limited efficacy in early clinical trials. Recently, many strategies for delivering IL-12 to tumor tissues have been developed, such as modifying IL-12, utilizing viral vectors, non-viral vectors, and cellular vectors. Previous studies have found that the fusion of IL-12 with extracellular matrix proteins, collagen, and immune factors is a way to enhance its therapeutic potential. In addition, studies have demonstrated that viral vectors are a good platform, and a variety of viruses such as oncolytic viruses, adenoviruses, and poxviruses have been used to deliver IL-12-with testing previously conducted in various cancer models. The local expression of IL-12 in tumors based on viral delivery avoids systemic toxicity while inducing effective antitumor immunity and acting synergistically with other therapies without compromising safety. In addition, lipid nanoparticles are currently considered to be the most mature drug delivery system. Moreover, cells are also considered to be drug carriers because they can effectively deliver therapeutic substances to tumors. In this article, we will systematically discuss the anti-tumor effects of IL-12 on its own or in combination with other therapies based on different delivery strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qing He
- State Key Laboratory of Drug Regulatory Sciences, National Institutes for Food and Drug Control, Beijing 102629, China; (C.D.); (D.T.); (H.S.); (Z.L.); (L.Z.); (Y.Z.); (S.L.); (Y.Z.)
| |
Collapse
|
20
|
Porter JM, Oswald MS, Busuttil K, Emmanuel SN, Bennett A, McKenna R, Smith JG. Mechanisms of AAV2 neutralization by human alpha-defensins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614754. [PMID: 39386661 PMCID: PMC11463608 DOI: 10.1101/2024.09.25.614754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Antiviral immunity compromises the efficacy of adeno-associated virus (AAV) vectors used for gene therapy. This is well understood for the adaptive immune response. However, innate immune effectors like alpha-defensin antimicrobial peptides also block AAV infection, although their mechanisms of action are unknown. To address this gap in knowledge, we investigated AAV2 neutralization by human neutrophil peptide 1 (HNP1), a myeloid alpha-defensin, and human defensin 5 (HD5), an enteric alpha-defensin. We found that both defensins bind to AAV2 and inhibit infection at low micromolar concentrations. While HD5 prevents AAV2 from binding to cells, HNP1 does not. However, AAV2 exposed to HD5 after binding to cells is still neutralized, indicating an additional block to infection. Accordingly, both HD5 and HNP1 inhibit externalization of the VP1 unique domain, which contains a phospholipase A 2 enzyme required for endosome escape and nuclear localization signals required for nuclear entry. Consequently, both defensins prevent AAV2 from reaching the nucleus. Disruption of intracellular trafficking of the viral genome to the nucleus is reminiscent of how alpha-defensins neutralize other non-enveloped viruses, suggesting a common mechanism of inhibition. These results will inform the development of vectors capable of overcoming these hurdles to improve the efficiency of gene therapy. Author Summary AAVs are commonly used as gene therapy vectors due to their broad tropism and lack of disease association; however, host innate immune factors, such as human alpha-defensin antimicrobial peptides, can hinder gene delivery. Although it is becoming increasingly evident that human alpha-defensins can block infection by a wide range of nonenveloped viruses, including AAVs, their mechanism of action remains poorly understood. In this study, we describe for the first time how two types of abundant human alpha-defensins neutralize a specific AAV serotype, AAV2. We found that one defensin prevents AAV2 from binding to cells, the first step in infection, while both defensins block a critical later step in AAV2 entry. Our findings support the emerging idea that defensins use a common strategy to block infection by DNA viruses that replicate in the nucleus. Through understanding how innate immune effectors interact with and impede AAV infection, vectors can be developed to bypass these interventions and allow more efficient gene delivery.
Collapse
|
21
|
Kato Y, Rice N, Pokrass M, Jeong J, Rodriguez R, Field JJ, Nowyhed H. Nonclinical characterization of ICVB-1042 as a selective oncolytic adenovirus for solid tumor treatment. Commun Biol 2024; 7:1132. [PMID: 39271928 PMCID: PMC11399272 DOI: 10.1038/s42003-024-06839-6] [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: 01/08/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
ICVB-1042 is an oncolytic adenovirus containing modifications to enhance replication, lysis, and viral spreading in tumor cells. The anti-tumor activity, immune activation, tropism, selectivity, and mechanism of action were evaluated in preparation for a first-in-human study. ICVB-1042 was at least 100-fold more cytotoxic in A549 cells than in normal primary cells tested, demonstrating its high tumor selectivity and a low likelihood of targeting primary tissues. ICVB-1042 administered to mice intravenously or intratumorally was effective in reducing tumor burden. Its intravenous administration also inhibited tumor growth in orthotopic models. ICVB-1042 was well tolerated in mice compared to HAdV-C5 (Wt Ad5), with reduced liver sequestration, supporting safety of the drug for systemic delivery. These preclinical data demonstrating the safety and potency of ICVB-1042 for treatment of various solid tumors support the ongoing clinical investigation (NCT05904236).
Collapse
Affiliation(s)
- Yu Kato
- IconOVir Bio, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Howe G, Bal M, Wasmuth M, Massaro G, Rahim AA, Ali S, Rivera M, Schofield DM, Omotosho A, Ward J, Keshavarz-Moore E, Mason C, Nesbeth DN. An autonucleolytic suspension HEK293F host cell line for high-titer serum-free AAV5 and AAV9 production with reduced levels of DNA impurity. Mol Ther Methods Clin Dev 2024; 32:101317. [PMID: 39257529 PMCID: PMC11385518 DOI: 10.1016/j.omtm.2024.101317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 08/07/2024] [Indexed: 09/12/2024]
Abstract
We sought to engineer mammalian cells to secrete nuclease activity as a step toward removing the need to purchase commercial nucleases as process additions in bioprocessing of AAV5 and AAV9 as gene therapy vectors. Engineering HeLa cells with a serratial nuclease transgene did not bring about nuclease activity in surrounding media whereas engineering serum-free, suspension-adapted HEK293F cells with a staphylococcal nuclease transgene did result in detectable nuclease activity in surrounding media of the resultant stable transfectant cell line, "NuPro-1S." When cultivated in serum-free media, NuPro-1S cells yielded 3.06 × 1010 AAV5 viral genomes (vg)/mL via transient transfection, compared with 3.85 × 109 vg/mL from the parental HEK293F cell line. AAV9 production, followed by purification by ultracentrifugation, yielded 1.8 × 1013 vg/mL from NuPro-1S cells compared with 7.35 × 1012 vg/mL from HEK293F cells. AAV9 from both HEK293F and NuPro-1S showed almost identical ability to transduce cells embedded in a scaffold tissue mimic or cells of mouse neonate brain tissue in vivo. Comparison of agarose gel data indicated that the DNA content of AAV5 and AAV9 process streams from NuPro-1S cells was reduced by approximately 60% compared with HEK293F cells. A similar reduction in HEK293F cells was only achievable with a 50 U/mL Benzonase treatment.
Collapse
Affiliation(s)
- Geoffrey Howe
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Mehtap Bal
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Matt Wasmuth
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Sadfer Ali
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Milena Rivera
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Desmond M Schofield
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Aminat Omotosho
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - John Ward
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Eli Keshavarz-Moore
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Chris Mason
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| | - Darren N Nesbeth
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, London WC1E 6BT, UK
| |
Collapse
|
23
|
Taylor NK, Guggenbiller MJ, Mistry PP, King OD, Harper SQ. A self-complementary AAV proviral plasmid that reduces cross-packaging and ITR promoter activity in AAV vector preparations. Mol Ther Methods Clin Dev 2024; 32:101295. [PMID: 39139628 PMCID: PMC11320455 DOI: 10.1016/j.omtm.2024.101295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/24/2024] [Indexed: 08/15/2024]
Abstract
Adeno-associated viral vectors (AAVs) are a leading delivery system for gene therapy in animal models and humans. With several Food and Drug Administration-approved AAV gene therapies on the market, issues related to vector manufacturing have become increasingly important. In this study, we focused on potentially toxic DNA contaminants that can arise from AAV proviral plasmids, the raw materials required for manufacturing recombinant AAV in eukaryotic cells. Typical AAV proviral plasmids are circular DNAs containing a therapeutic gene cassette flanked by natural AAV inverted terminal repeat (ITR) sequences, and a plasmid backbone carrying prokaryotic sequences required for plasmid replication and selection in bacteria. While the majority of AAV particles package the intended therapeutic payload, some capsids instead package the bacterial sequences located on the proviral plasmid backbone. Since ITR sequences also have promoter activity, potentially toxic bacterial open reading frames can be produced in vivo, thereby representing a safety risk. In this study, we describe a new AAV proviral plasmid for vector manufacturing that (1) significantly decreases cross-packaged bacterial sequences, (2) increases correctly packaged AAV payloads, and (3) blunts ITR-driven transcription of cross-packaged material to avoid expressing potentially toxic bacterial sequences. This system may help improve the safety of AAV vector products.
Collapse
Affiliation(s)
- Noah K. Taylor
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Matthew J. Guggenbiller
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Pranali P. Mistry
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Oliver D. King
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Scott Q. Harper
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
24
|
Patra AT, Tan E, Kok YJ, Ng SK, Bi X. Temporal insights into molecular and cellular responses during rAAV production in HEK293T cells. Mol Ther Methods Clin Dev 2024; 32:101278. [PMID: 39022743 PMCID: PMC11253160 DOI: 10.1016/j.omtm.2024.101278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/04/2024] [Indexed: 07/20/2024]
Abstract
The gene therapy field seeks cost-effective, large-scale production of recombinant adeno-associated virus (rAAV) vectors for high-dosage therapeutic applications. Although strategies like suspension cell culture and transfection optimization have shown moderate success, challenges persist for large-scale applications. To unravel molecular and cellular mechanisms influencing rAAV production, we conducted an SWATH-MS proteomic analysis of HEK293T cells transfected using standard, sub-optimal, and optimal conditions. Gene Ontology and pathway analysis revealed significant protein expression variations, particularly in processes related to cellular homeostasis, metabolic regulation, vesicular transport, ribosomal biogenesis, and cellular proliferation under optimal transfection conditions. This resulted in a 50% increase in rAAV titer compared with the standard protocol. Additionally, we identified modifications in host cell proteins crucial for AAV mRNA stability and gene translation, particularly regarding AAV capsid transcripts under optimal transfection conditions. Our study identified 124 host proteins associated with AAV replication and assembly, each exhibiting distinct expression pattern throughout rAAV production stages in optimal transfection condition. This investigation sheds light on the cellular mechanisms involved in rAAV production in HEK293T cells and proposes promising avenues for further enhancing rAAV titer during production.
Collapse
Affiliation(s)
- Alok Tanala Patra
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), Singapore 138668, Singapore
| | - Evan Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), Singapore 138668, Singapore
| | - Yee Jiun Kok
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), Singapore 138668, Singapore
| | - Say Kong Ng
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), Singapore 138668, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), Singapore 138668, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
- Food, Chemical and Biotechnology Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| |
Collapse
|
25
|
Wu G, Liu S, Hagenstein J, Alawi M, Hengel FE, Schaper M, Akyüz N, Liao Z, Wanner N, Tomas NM, Failla AV, Dierlamm J, Körbelin J, Lu S, Huber TB. Adeno-associated virus-based gene therapy treats inflammatory kidney disease in mice. J Clin Invest 2024; 134:e174722. [PMID: 39225099 PMCID: PMC11364381 DOI: 10.1172/jci174722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
Adeno-associated virus (AAV) is a promising in vivo gene delivery platform showing advantages in delivering therapeutic molecules to difficult or undruggable cells. However, natural AAV serotypes have insufficient transduction specificity and efficiency in kidney cells. Here, we developed an evolution-directed selection protocol for renal glomeruli and identified what we believe to be a new vector termed AAV2-GEC that specifically and efficiently targets the glomerular endothelial cells (GEC) after systemic administration and maintains robust GEC tropism in healthy and diseased rodents. AAV2-GEC-mediated delivery of IdeS, a bacterial antibody-cleaving proteinase, provided sustained clearance of kidney-bound antibodies and successfully treated antiglomerular basement membrane glomerulonephritis in mice. Taken together, this study showcases the potential of AAV as a gene delivery platform for challenging cell types. The development of AAV2-GEC and its successful application in the treatment of antibody-mediated kidney disease represents a significant step forward and opens up promising avenues for kidney medicine.
Collapse
Affiliation(s)
- Guochao Wu
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| | - Shuya Liu
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| | - Julia Hagenstein
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| | | | | | - Melanie Schaper
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| | - Nuray Akyüz
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, and
| | - Zhouning Liao
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| | - Nicola Wanner
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| | - Nicola M. Tomas
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| | | | - Judith Dierlamm
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, and
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, and
| | - Shun Lu
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| | - Tobias B. Huber
- III. Department of Medicine
- Hamburg Center for Kidney Health (HCKH)
| |
Collapse
|
26
|
Lam P, Zygmunt DA, Ashbrook A, Bennett M, Vetter TA, Martin PT. Dual FKRP/FST gene therapy normalizes ambulation, increases strength, decreases pathology, and amplifies gene expression in LGMDR9 mice. Mol Ther 2024; 32:2604-2623. [PMID: 38910327 PMCID: PMC11405156 DOI: 10.1016/j.ymthe.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 04/12/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
Recent clinical studies of single gene replacement therapy for neuromuscular disorders have shown they can slow or stop disease progression, but such therapies have had little impact on reversing muscle disease that was already present. To reverse disease in patients with muscular dystrophy, new muscle mass and strength must be rebuilt at the same time that gene replacement prevents subsequent disease. Here, we show that treatment of FKRPP448L mice with a dual FKRP/FST gene therapy packaged into a single adeno-associated virus (AAV) vector can build muscle strength and mass that exceed levels found in wild-type mice and can induce normal ambulation endurance in a 1-h walk test. Dual FKRP/FST therapy also showed more even increases in muscle mass and amplified muscle expression of both genes relative to either single gene therapy alone. These data suggest that treatment with single AAV-bearing dual FKRP/FST gene therapies can overcome loss of ambulation by improving muscle strength at the same time it prevents subsequent muscle damage. This design platform could be used to create therapies for other forms of muscular dystrophy that may improve patient outcomes.
Collapse
Affiliation(s)
- Patricia Lam
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Deborah A Zygmunt
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Anna Ashbrook
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Macey Bennett
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Tatyana A Vetter
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Paul T Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA; Department of Pediatrics, and Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
27
|
Lyle A, Stamatis C, Linke T, Hulley M, Schmelzer A, Turner R, Farid SS. Process economics evaluation and optimization of adeno-associated virus downstream processing. Biotechnol Bioeng 2024; 121:2435-2448. [PMID: 37086200 DOI: 10.1002/bit.28402] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/23/2023]
Abstract
Adeno-associated virus (AAV) manufacturing has traditionally focused upon lab-scale techniques to culture and purify vector products, leading to limitations in production capacity. The tool presented in this paper assesses the feasibility of using non-scalable technologies at high AAV demands and identifies optimal flowsheets at large-scale that meet both cost and purity targets. The decisional tool comprises (a) a detailed process economics model with the relevant mass balance, sizing, and costing equations for AAV upstream and downstream technologies, (b) a built-in Monte Carlo simulation to assess uncertainties, and (c) a brute-force optimization algorithm for rapid investigation into the optimal purification combinations. The results overall highlighted that switching to more scalable upstream and downstream processing alternatives is economically advantageous. The base case analysis showed the cost and robustness advantages of utilizing suspension cell culture over adherent, as well as a fully chromatographic purification platform over batch ultracentrifugation. Expanding the set of purification options available gave insights into the optimal combination to satisfy both cost and purity targets. As the purity target increased, the optimal polishing solution moved from the non-capsid purifying multimodal chromatography to anion-exchange chromatography or continuous ultracentrifugation.
Collapse
Affiliation(s)
- Annabel Lyle
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, UK
| | - Christos Stamatis
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, UK
| | - Thomas Linke
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Martyn Hulley
- Biopharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Albert Schmelzer
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Richard Turner
- Biopharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Suzanne S Farid
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, UK
| |
Collapse
|
28
|
Wang J, Gao G, Wang D. Developing AAV-delivered nonsense suppressor tRNAs for neurological disorders. Neurotherapeutics 2024; 21:e00391. [PMID: 38959711 PMCID: PMC11269797 DOI: 10.1016/j.neurot.2024.e00391] [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: 02/14/2024] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024] Open
Abstract
Adeno-associated virus (AAV)-based gene therapy is a clinical stage therapeutic modality for neurological disorders. A common genetic defect in myriad monogenic neurological disorders is nonsense mutations that account for about 11% of all human pathogenic mutations. Stop codon readthrough by suppressor transfer RNA (sup-tRNA) has long been sought as a potential gene therapy approach to target nonsense mutations, but hindered by inefficient in vivo delivery. The rapid advances in AAV delivery technology have not only powered gene therapy development but also enabled in vivo preclinical assessment of a range of nucleic acid therapeutics, such as sup-tRNA. Compared with conventional AAV gene therapy that delivers a transgene to produce therapeutic proteins, AAV-delivered sup-tRNA has several advantages, such as small gene sizes and operating within the endogenous gene expression regulation, which are important considerations for treating some neurological disorders. This review will first examine sup-tRNA designs and delivery by AAV vectors. We will then analyze how AAV-delivered sup-tRNA can potentially address some neurological disorders that are challenging to conventional gene therapy, followed by discussing available mouse models of neurological diseases for in vivo preclinical testing. Potential challenges for AAV-delivered sup-tRNA to achieve therapeutic efficacy and safety will also be discussed.
Collapse
Affiliation(s)
- Jiaming Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Dan Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
29
|
Catalán-Tatjer D, Tzimou K, Nielsen LK, Lavado-García J. Unravelling the essential elements for recombinant adeno-associated virus (rAAV) production in animal cell-based platforms. Biotechnol Adv 2024; 73:108370. [PMID: 38692443 DOI: 10.1016/j.biotechadv.2024.108370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/05/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
Recombinant adeno-associated viruses (rAAVs) stand at the forefront of gene therapy applications, holding immense significance for their safe and efficient gene delivery capabilities. The constantly increasing and unmet demand for rAAVs underscores the need for a more comprehensive understanding of AAV biology and its impact on rAAV production. In this literature review, we delved into AAV biology and rAAV manufacturing bioprocesses, unravelling the functions and essentiality of proteins involved in rAAV production. We discuss the interconnections between these proteins and how they affect the choice of rAAV production platform. By addressing existing inconsistencies, literature gaps and limitations, this review aims to define a minimal set of genes that are essential for rAAV production, providing the potential to advance rAAV biomanufacturing, with a focus on minimizing the genetic load within rAAV-producing cells.
Collapse
Affiliation(s)
- David Catalán-Tatjer
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Konstantina Tzimou
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Lars K Nielsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark; Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Australia
| | - Jesús Lavado-García
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark.
| |
Collapse
|
30
|
Liu H, Zhang Y, Yip M, Ren L, Liang J, Chen X, Liu N, Du A, Wang J, Chang H, Oh H, Zhou C, Xing R, Xu M, Guo P, Gessler D, Xie J, Tai PW, Gao G, Wang D. Producing high-quantity and high-quality recombinant adeno-associated virus by low-cis triple transfection. Mol Ther Methods Clin Dev 2024; 32:101230. [PMID: 38558570 PMCID: PMC10979107 DOI: 10.1016/j.omtm.2024.101230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
Recombinant adeno-associated virus (rAAV)-based gene therapy is entering clinical and commercial stages at an unprecedented pace. Triple transfection of HEK293 cells is currently the most widely used platform for rAAV manufacturing. Here, we develop low-cis triple transfection that decreases transgene plasmid use by 10- to 100-fold and overcomes several major limitations associated with standard triple transfection. This new method improves packaging of yield-inhibiting transgenes by up to 10-fold, and generates rAAV batches with reduced plasmid backbone contamination that otherwise cannot be eliminated in downstream processing. When tested in mice and compared with rAAV produced by standard triple transfection, low-cis rAAV shows comparable or superior potency and results in diminished plasmid backbone DNA and RNA persistence in tissue. Mechanistically, low-cis triple transfection relies on the extensive replication of transgene cassette (i.e., inverted terminal repeat-flanked vector DNA) in HEK293 cells during production phase. This cost-effective method can be easily implemented and is widely applicable to producing rAAV of high quantity, purity, and potency.
Collapse
Affiliation(s)
- Hao Liu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Yue Zhang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Mitchell Yip
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lingzhi Ren
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jialing Liang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Xiupeng Chen
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Nan Liu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ailing Du
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jiaming Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Hao Chang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Hyejin Oh
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Chen Zhou
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ruxiao Xing
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Mengyao Xu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Peiyi Guo
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dominic Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Phillip W.L. Tai
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dan Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
31
|
Hersch SJ, Chandrasekaran S, Lam J, Nafissi N, Slavcev RA. Manufacturing DNA in E. coli yields higher-fidelity DNA than in vitro enzymatic synthesis. Mol Ther Methods Clin Dev 2024; 32:101227. [PMID: 38516691 PMCID: PMC10951457 DOI: 10.1016/j.omtm.2024.101227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
Biotechnologies such as gene therapy have brought DNA vectors to the forefront of pharmaceuticals. The quality of starting material plays a pivotal role in determining final product quality. Here, we examined the fidelity of DNA replication using enzymatic methods (in vitro) compared to plasmid DNA produced in vivo in E. coli. Next-generation sequencing approaches rely on in vitro polymerases, which have inherent limitations in sensitivity. To address this challenge, we introduce a novel assay based on loss-of-function (LOF) mutations in the conditionally toxic sacB gene. Our findings show that DNA production in E. coli results in significantly fewer LOF mutations (80- to 3,000-fold less) compared to enzymatic DNA replication methods such as polymerase chain reaction (PCR) and rolling circle amplification (RCA). These results suggest that using DNA produced by PCR or RCA may introduce a substantial number of mutation impurities, potentially affecting the quality and yield of final pharmaceutical products. Our study underscores that DNA synthesized in vitro has a significantly higher mutation rate than DNA produced traditionally in E. coli. Therefore, utilizing in vitro enzymatically produced DNA in biotechnology and biomanufacturing may entail considerable fidelity-related risks, while using DNA starting material derived from E. coli substantially mitigates this risk.
Collapse
Affiliation(s)
| | | | - Jamie Lam
- Mediphage Bioceuticals, Inc, Toronto, ON, Canada
| | - Nafiseh Nafissi
- Mediphage Bioceuticals, Inc, Toronto, ON, Canada
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Roderick A. Slavcev
- Mediphage Bioceuticals, Inc, Toronto, ON, Canada
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
- Centre for Eye and Vision Research, HKSTP, Ma Liu Shui, Hong Kong
| |
Collapse
|
32
|
Xue W, Fulco C, Sha S, Alden N, Panteli J, Hossler P, Warren J. Adeno-associated virus perfusion enhanced expression: A commercially scalable, high titer, high quality producer cell line process. Mol Ther Methods Clin Dev 2024; 32:101266. [PMID: 38868441 PMCID: PMC11166877 DOI: 10.1016/j.omtm.2024.101266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/10/2024] [Indexed: 06/14/2024]
Abstract
With safety and efficacy demonstrated over hundreds of clinical trials in the last 30 years, along with at least six recent global marketing authorizations achieved since 2017, recombinant adeno-associated viruses (rAAVs) have been established as the leading therapeutic gene transfer vector for rare, monogenic diseases. Significant advances in manufacturing technology have been made in the last few decades to address challenges with GMP production of rAAV products, although yield, cost, scalability, and quality remain a challenge. With transient transfection processes established as a manufacturing platform for multiple commercial AAV products, there remains significant yield, cost, robustness, and scalability constraints that need to be resolved to enable a reliable supply of rAAV products for global patient access. The development of stable producer cell lines for rAAV products has enabled scalability and, in some cases, improvements in productivity. Herein we describe a novel AAV perfusion-enhanced expression (APEX) process, resulting in higher maximum cell densities in the production bioreactor with a 3- to 6-fold increase in volumetric productivity. This process has been successfully demonstrated across multiple serotypes in large scale cell culture with titers approaching 1 × 1012 GC/mL. The APEX production platform marks a significant leap forward in the efficient and effective manufacturing of rAAV vector products.
Collapse
Affiliation(s)
- Wei Xue
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Cameron Fulco
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Sha Sha
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Nick Alden
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Jan Panteli
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Patrick Hossler
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - James Warren
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| |
Collapse
|
33
|
Johari YB, Pohle TH, Whitehead J, Scarrott JM, Liu P, Mayer A, James DC. Molecular design of controllable recombinant adeno-associated virus (AAV) expression systems for enhanced vector production. Biotechnol J 2024; 19:e2300685. [PMID: 38900035 DOI: 10.1002/biot.202300685] [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: 12/03/2023] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024]
Abstract
Recombinant adeno-associated virus (rAAV) is the leading vector for the delivery of gene therapies. However, low viral genome (VG) titers are common and the proportion of "full" capsids containing the therapeutic gene payload can be highly variable. The coordinated molecular design of plasmids encoding viral components and Helper functions remains a major challenge for rAAV manufacturing. Here we present the design of improved Rep/Cap and Helper plasmids for rAAV2/8 production, (i) a Rep/Cap expression vector harboring independently controllable rep and cap genes and (ii) an improved Helper plasmid harboring E4 gene deletion variants. First, an optimized Rep/Cap vector utilized a truncated p5 promoter, a p5 cis-regulatory element at the 3' end in combination with a heterologous promoter to drive Cap expression and an additional copy of the rep52/40 gene to overexpress short Rep proteins. We demonstrate that Rep78 is essential for efficient rAAV2/8 production in HEK293 cells, and a higher ratio of short Rep to long Rep proteins enhances genome packaging. Second, we identified regulators and open reading frames within the Helper plasmid that contribute to increased rAAV2/8 production. While L4-33k/22k is integral to optimal production, the use of E4orf6-6/7 subset significantly enhanced VG titer. Together, an optimal combination of engineered Rep/Cap and Helper plasmid variants increased VG titer by 3.1-fold. This study demonstrates that configuring and controlling the expression of the different AAV genetic elements contributes toward high rAAV production and product quality (full/empty capsid ratio).
Collapse
Affiliation(s)
- Yusuf B Johari
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Thilo H Pohle
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
- Syngensys Ltd., Sheffield, UK
| | - Jared Whitehead
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Joseph M Scarrott
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Ping Liu
- Cell Line Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Ayda Mayer
- Cell Line Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
- Syngensys Ltd., Sheffield, UK
| |
Collapse
|
34
|
Li L, Qin R, Liu Y, Tseng YS, Zhang W, Yu L, Mietzsch M, Zou X, Liu H, Lu G, Hu H, Mckenna R, Yang J, Wei Y, Agbandje-Mckenna M, Hu J, Yang L. Dissecting positive selection events and immunological drives during the evolution of adeno-associated virus lineages. PLoS Pathog 2024; 20:e1012260. [PMID: 38885242 PMCID: PMC11182496 DOI: 10.1371/journal.ppat.1012260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Adeno-associated virus (AAV) serotypes from primates are being developed and clinically used as vectors for human gene therapy. However, the evolutionary mechanism of AAV variants is far from being understood, except that genetic recombination plays an important role. Furthermore, little is known about the interaction between AAV and its natural hosts, human and nonhuman primates. In this study, natural AAV capsid genes were subjected to systemic evolutionary analysis with a focus on selection drives during the diversification of AAV lineages. A number of positively selected sites were identified from these AAV lineages with functional relevance implied by their localization on the AAV structures. The selection drives of the two AAV2 capsid sites were further investigated in a series of biological experiments. These observations did not support the evolution of the site 410 of the AAV2 capsid driven by selection pressure from the human CD4+ T-cell response. However, positive selection on site 548 of the AAV2 capsid was directly related to host humoral immunity because of the profound effects of mutations at this site on the immune evasion of AAV variants from human neutralizing antibodies at both the individual and population levels. Overall, this work provides a novel interpretation of the genetic diversity and evolution of AAV lineages in their natural hosts, which may contribute to their further engineering and application in human gene therapy.
Collapse
Affiliation(s)
- Lirong Li
- Department of Cardiology and Laboratory of Gene Therapy for Heart Diseases, State Key Laboratory of Biotherapy, and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Runkuan Qin
- Department of Cardiology and Laboratory of Gene Therapy for Heart Diseases, State Key Laboratory of Biotherapy, and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunbo Liu
- Department of Cardiology and Laboratory of Gene Therapy for Heart Diseases, State Key Laboratory of Biotherapy, and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Shan Tseng
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, Florida, United States of America
| | - Weihan Zhang
- General Surgery Department, Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Yu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mario Mietzsch
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, Florida, United States of America
| | - Xinkai Zou
- Center for Immunology and Hematology, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haizhou Liu
- Computational Virology Group, Center for Bacteria and Viruses Resources and Bioinformation, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Guangwen Lu
- Department of Emergency Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongbo Hu
- Center for Immunology and Hematology, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Robert Mckenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, Florida, United States of America
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mavis Agbandje-Mckenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, Florida, United States of America
| | - Jiankun Hu
- General Surgery Department, Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Yang
- Department of Cardiology and Laboratory of Gene Therapy for Heart Diseases, State Key Laboratory of Biotherapy, and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Garbutt TA, Wang Z, Wang H, Ma H, Ruan H, Dong Y, Xie Y, Tan L, Phookan R, Stouffer J, Vedantham V, Yang Y, Qian L, Liu J. Epigenetic Regulation of Cardiomyocyte Maturation by Arginine Methyltransferase CARM1. Circulation 2024; 149:1501-1515. [PMID: 38223978 PMCID: PMC11073921 DOI: 10.1161/circulationaha.121.055738] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/19/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND During the neonatal stage, the cardiomyocyte undergoes a constellation of molecular, cytoarchitectural, and functional changes known collectively as cardiomyocyte maturation to increase myocardial contractility and cardiac output. Despite the importance of cardiomyocyte maturation, the molecular mechanisms governing this critical process remain largely unexplored. METHODS We leveraged an in vivo mosaic knockout system to characterize the role of Carm1, the founding member of protein arginine methyltransferase, in cardiomyocyte maturation. Using a battery of assays, including immunohistochemistry, immuno-electron microscopy imaging, and action potential recording, we assessed the effect of loss of Carm1 function on cardiomyocyte cell growth, myofibril expansion, T-tubule formation, and electrophysiological maturation. Genome-wide transcriptome profiling, H3R17me2a chromatin immunoprecipitation followed by sequencing, and assay for transposase-accessible chromatin with high-throughput sequencing were used to investigate the mechanisms by which CARM1 (coactivator-associated arginine methyltransferase 1) regulates cardiomyocyte maturation. Finally, we interrogated the human syntenic region to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks for single-nucleotide polymorphisms associated with human heart diseases. RESULTS We report that mosaic ablation of Carm1 disrupts multiple aspects of cardiomyocyte maturation cell autonomously, leading to reduced cardiomyocyte size and sarcomere thickness, severe loss and disorganization of T tubules, and compromised electrophysiological maturation. Genomics study demonstrates that CARM1 directly activates genes that underlie cardiomyocyte cytoarchitectural and electrophysiological maturation. Moreover, our study reveals significant enrichment of human heart disease-associated single-nucleotide polymorphisms in the human genomic region syntenic to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks. CONCLUSIONS This study establishes a critical and multifaceted role for CARM1 in regulating cardiomyocyte maturation and demonstrates that deregulation of CARM1-dependent cardiomyocyte maturation gene expression may contribute to human heart diseases.
Collapse
Affiliation(s)
- Tiffany A. Garbutt
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zhenhua Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cardiovascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Ma
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Present address: Department of Cardiology, 2 Affiliated Hospital, School of Medicine, Zhejiang University. Hangzhou 310009, China
| | - Hongmei Ruan
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yanhan Dong
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yifang Xie
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Lianmei Tan
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ranan Phookan
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joy Stouffer
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Vasanth Vedantham
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
36
|
Moreno-Manuel AI, Macías Á, Cruz FM, Gutiérrez LK, Martínez F, González-Guerra A, Martínez Carrascoso I, Bermúdez-Jimenez FJ, Sánchez-Pérez P, Vera-Pedrosa ML, Ruiz-Robles JM, Bernal JA, Jalife J. The Kir2.1E299V mutation increases atrial fibrillation vulnerability while protecting the ventricles against arrhythmias in a mouse model of short QT syndrome type 3. Cardiovasc Res 2024; 120:490-505. [PMID: 38261726 PMCID: PMC11060485 DOI: 10.1093/cvr/cvae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/24/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
AIMS Short QT syndrome type 3 (SQTS3) is a rare arrhythmogenic disease caused by gain-of-function mutations in KCNJ2, the gene coding the inward rectifier potassium channel Kir2.1. We used a multidisciplinary approach and investigated arrhythmogenic mechanisms in an in-vivo model of de-novo mutation Kir2.1E299V identified in a patient presenting an extremely abbreviated QT interval and paroxysmal atrial fibrillation. METHODS AND RESULTS We used intravenous adeno-associated virus-mediated gene transfer to generate mouse models, and confirmed cardiac-specific expression of Kir2.1WT or Kir2.1E299V. On ECG, the Kir2.1E299V mouse recapitulated the QT interval shortening and the atrial-specific arrhythmia of the patient. The PR interval was also significantly shorter in Kir2.1E299V mice. Patch-clamping showed extremely abbreviated action potentials in both atrial and ventricular Kir2.1E299V cardiomyocytes due to a lack of inward-going rectification and increased IK1 at voltages positive to -80 mV. Relative to Kir2.1WT, atrial Kir2.1E299V cardiomyocytes had a significantly reduced slope conductance at voltages negative to -80 mV. After confirming a higher proportion of heterotetrameric Kir2.x channels containing Kir2.2 subunits in the atria, in-silico 3D simulations predicted an atrial-specific impairment of polyamine block and reduced pore diameter in the Kir2.1E299V-Kir2.2WT channel. In ventricular cardiomyocytes, the mutation increased excitability by shifting INa activation and inactivation in the hyperpolarizing direction, which protected the ventricle against arrhythmia. Moreover, Purkinje myocytes from Kir2.1E299V mice manifested substantially higher INa density than Kir2.1WT, explaining the abbreviation in the PR interval. CONCLUSION The first in-vivo mouse model of cardiac-specific SQTS3 recapitulates the electrophysiological phenotype of a patient with the Kir2.1E299V mutation. Kir2.1E299V eliminates rectification in both cardiac chambers but protects against ventricular arrhythmias by increasing excitability in both Purkinje-fiber network and ventricles. Consequently, the predominant arrhythmias are supraventricular likely due to the lack of inward rectification and atrial-specific reduced pore diameter of the Kir2.1E299V-Kir2.2WT heterotetramer.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Action Potentials
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/metabolism
- Atrial Fibrillation/genetics
- Atrial Fibrillation/physiopathology
- Atrial Fibrillation/metabolism
- Disease Models, Animal
- Genetic Predisposition to Disease
- Heart Rate/genetics
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phenotype
- Potassium Channels, Inwardly Rectifying/genetics
- Potassium Channels, Inwardly Rectifying/metabolism
Collapse
Affiliation(s)
- Ana I Moreno-Manuel
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Francisco M Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Lilian K Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Fernando Martínez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Andrés González-Guerra
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Isabel Martínez Carrascoso
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Francisco José Bermúdez-Jimenez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Department of Cardiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
| | - Patricia Sánchez-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | | | - Juan Manuel Ruiz-Robles
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Juan A Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departments of Internal Medicine and Molecular and Integrative Physiology, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 4810, USA
| |
Collapse
|
37
|
De Carluccio G, Fusco V, di Bernardo D. Engineering a synthetic gene circuit for high-performance inducible expression in mammalian systems. Nat Commun 2024; 15:3311. [PMID: 38632224 PMCID: PMC11024104 DOI: 10.1038/s41467-024-47592-y] [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: 09/28/2023] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
Inducible gene expression systems can be used to control the expression of a gene of interest by means of a small-molecule. One of the most common designs involves engineering a small-molecule responsive transcription factor (TF) and its cognate promoter, which often results in a compromise between minimal uninduced background expression (leakiness) and maximal induced expression. Here, we focus on an alternative strategy using quantitative synthetic biology to mitigate leakiness while maintaining high expression, without modifying neither the TF nor the promoter. Through mathematical modelling and experimental validations, we design the CASwitch, a mammalian synthetic gene circuit based on combining two well-known network motifs: the Coherent Feed-Forward Loop (CFFL) and the Mutual Inhibition (MI). The CASwitch combines the CRISPR-Cas endoribonuclease CasRx with the state-of-the-art Tet-On3G inducible gene system to achieve high performances. To demonstrate the potentialities of the CASwitch, we apply it to three different scenarios: enhancing a whole-cell biosensor, controlling expression of a toxic gene and inducible production of Adeno-Associated Virus (AAV) vectors.
Collapse
Affiliation(s)
- Giuliano De Carluccio
- Telethon Institute of Genetics and Medicine, Naples, Italy
- University of Naples Federico II, Department of Chemical Materials and Industrial Engineering, Naples, Italy
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
| | - Virginia Fusco
- Telethon Institute of Genetics and Medicine, Naples, Italy
- University of Naples Federico II, Department of Chemical Materials and Industrial Engineering, Naples, Italy
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine, Naples, Italy.
- University of Naples Federico II, Department of Chemical Materials and Industrial Engineering, Naples, Italy.
| |
Collapse
|
38
|
Cruz FM, Macías Á, Moreno-Manuel AI, Gutiérrez LK, Vera-Pedrosa ML, Martínez-Carrascoso I, Pérez PS, Robles JMR, Bermúdez-Jiménez FJ, Díaz-Agustín A, de Benito FM, Arias-Santiago S, Braza-Boils A, Martín-Martínez M, Gutierrez-Rodríguez M, Bernal JA, Zorio E, Jiménez-Jaimez J, Jalife J. Extracellular Kir2.1 C122Y Mutant Upsets Kir2.1-PIP 2 Bonds and Is Arrhythmogenic in Andersen-Tawil Syndrome. Circ Res 2024; 134:e52-e71. [PMID: 38497220 PMCID: PMC11009053 DOI: 10.1161/circresaha.123.323895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Andersen-Tawil syndrome type 1 is a rare heritable disease caused by mutations in the gene coding the strong inwardly rectifying K+ channel Kir2.1. The extracellular Cys (cysteine)122-to-Cys154 disulfide bond in the channel structure is crucial for proper folding but has not been associated with correct channel function at the membrane. We evaluated whether a human mutation at the Cys122-to-Cys154 disulfide bridge leads to Kir2.1 channel dysfunction and arrhythmias by reorganizing the overall Kir2.1 channel structure and destabilizing its open state. METHODS We identified a Kir2.1 loss-of-function mutation (c.366 A>T; p.Cys122Tyr) in an ATS1 family. To investigate its pathophysiological implications, we generated an AAV9-mediated cardiac-specific mouse model expressing the Kir2.1C122Y variant. We employed a multidisciplinary approach, integrating patch clamping and intracardiac stimulation, molecular biology techniques, molecular dynamics, and bioluminescence resonance energy transfer experiments. RESULTS Kir2.1C122Y mice recapitulated the ECG features of ATS1 independently of sex, including corrected QT prolongation, conduction defects, and increased arrhythmia susceptibility. Isolated Kir2.1C122Y cardiomyocytes showed significantly reduced inwardly rectifier K+ (IK1) and inward Na+ (INa) current densities independently of normal trafficking. Molecular dynamics predicted that the C122Y mutation provoked a conformational change over the 2000-ns simulation, characterized by a greater loss of hydrogen bonds between Kir2.1 and phosphatidylinositol 4,5-bisphosphate than wild type (WT). Therefore, the phosphatidylinositol 4,5-bisphosphate-binding pocket was destabilized, resulting in a lower conductance state compared with WT. Accordingly, on inside-out patch clamping, the C122Y mutation significantly blunted Kir2.1 sensitivity to increasing phosphatidylinositol 4,5-bisphosphate concentrations. In addition, the Kir2.1C122Y mutation resulted in channelosome degradation, demonstrating temporal instability of both Kir2.1 and NaV1.5 proteins. CONCLUSIONS The extracellular Cys122-to-Cys154 disulfide bond in the tridimensional Kir2.1 channel structure is essential for the channel function. We demonstrate that breaking disulfide bonds in the extracellular domain disrupts phosphatidylinositol 4,5-bisphosphate-dependent regulation, leading to channel dysfunction and defects in Kir2.1 energetic stability. The mutation also alters functional expression of the NaV1.5 channel and ultimately leads to conduction disturbances and life-threatening arrhythmia characteristic of Andersen-Tawil syndrome type 1.
Collapse
Affiliation(s)
- Francisco M. Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | | | - Lilian K. Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | | | | | | | | | - Francisco J Bermúdez-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - Aitor Díaz-Agustín
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Fernando Martínez de Benito
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Salvador Arias-Santiago
- Servicio de Dermatología Hospital Universitario Virgen de las Nieves
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - Aitana Braza-Boils
- Unit of Inherited Cardiomyopathies and Sudden Death (CAFAMUSME), Health Research Institute La Fe, La Fe Hospital, Valencia, Spain
- Cardiology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Mercedes Martín-Martínez
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Marta Gutierrez-Rodríguez
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Juan A. Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Esther Zorio
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Unit of Inherited Cardiomyopathies and Sudden Death (CAFAMUSME), Health Research Institute La Fe, La Fe Hospital, Valencia, Spain
- Cardiology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Juan Jiménez-Jaimez
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departments of Medicine and Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
39
|
Körbelin J, Arrulo A, Schwaninger M. Gene therapy targeting the blood-brain barrier. VITAMINS AND HORMONES 2024; 126:191-217. [PMID: 39029973 DOI: 10.1016/bs.vh.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Endothelial cells are the building blocks of vessels in the central nervous system (CNS) and form the blood-brain barrier (BBB). An intact BBB limits permeation of large hydrophilic molecules into the CNS. Thus, the healthy BBB is a major obstacle for the treatment of CNS disorders with antibodies, recombinant proteins or viral vectors. Several strategies have been devised to overcome the barrier. A key principle often consists in attaching the therapeutic compound to a ligand of receptors expressed on the BBB, for example, the transferrin receptor (TfR). The fusion molecule will bind to TfR on the luminal side of brain endothelial cells, pass the endothelial layer by transcytosis and be delivered to the brain parenchyma. However, attempts to endow therapeutic compounds with the ability to cross the BBB can be difficult to implement. An alternative and possibly more straight-forward approach is to produce therapeutic proteins in the endothelial cells that form the barrier. These cells are accessible from blood circulation and have a large interface with the brain parenchyma. They may be an ideal production site for therapeutic protein and afford direct supply to the CNS.
Collapse
Affiliation(s)
- Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, UKE Hamburg-Eppendorf, Hamburg, Germany
| | - Adriana Arrulo
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany; DZHK (German Research Centre for Cardiovascular Research), Hamburg-Lübeck-Kiel, Germany.
| |
Collapse
|
40
|
Wang JH, Gessler DJ, Zhan W, Gallagher TL, Gao G. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct Target Ther 2024; 9:78. [PMID: 38565561 PMCID: PMC10987683 DOI: 10.1038/s41392-024-01780-w] [Citation(s) in RCA: 180] [Impact Index Per Article: 180.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Adeno-associated virus (AAV) has emerged as a pivotal delivery tool in clinical gene therapy owing to its minimal pathogenicity and ability to establish long-term gene expression in different tissues. Recombinant AAV (rAAV) has been engineered for enhanced specificity and developed as a tool for treating various diseases. However, as rAAV is being more widely used as a therapy, the increased demand has created challenges for the existing manufacturing methods. Seven rAAV-based gene therapy products have received regulatory approval, but there continue to be concerns about safely using high-dose viral therapies in humans, including immune responses and adverse effects such as genotoxicity, hepatotoxicity, thrombotic microangiopathy, and neurotoxicity. In this review, we explore AAV biology with an emphasis on current vector engineering strategies and manufacturing technologies. We discuss how rAAVs are being employed in ongoing clinical trials for ocular, neurological, metabolic, hematological, neuromuscular, and cardiovascular diseases as well as cancers. We outline immune responses triggered by rAAV, address associated side effects, and discuss strategies to mitigate these reactions. We hope that discussing recent advancements and current challenges in the field will be a helpful guide for researchers and clinicians navigating the ever-evolving landscape of rAAV-based gene therapy.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurological Surgery, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Thomas L Gallagher
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
41
|
Todorov-Völgyi K, González-Gallego J, Müller SA, Beaufort N, Malik R, Schifferer M, Todorov MI, Crusius D, Robinson S, Schmidt A, Körbelin J, Bareyre F, Ertürk A, Haass C, Simons M, Paquet D, Lichtenthaler SF, Dichgans M. Proteomics of mouse brain endothelium uncovers dysregulation of vesicular transport pathways during aging. NATURE AGING 2024; 4:595-612. [PMID: 38519806 DOI: 10.1038/s43587-024-00598-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
Age-related decline in brain endothelial cell (BEC) function contributes critically to neurological disease. Comprehensive atlases of the BEC transcriptome have become available, but results from proteomic profiling are lacking. To gain insights into endothelial pathways affected by aging, we developed a magnetic-activated cell sorting-based mouse BEC enrichment protocol compatible with proteomics and resolved the profiles of protein abundance changes during aging. Unsupervised cluster analysis revealed a segregation of age-related protein dynamics with biological functions, including a downregulation of vesicle-mediated transport. We found a dysregulation of key regulators of endocytosis and receptor recycling (most prominently Arf6), macropinocytosis and lysosomal degradation. In gene deletion and overexpression experiments, Arf6 affected endocytosis pathways in endothelial cells. Our approach uncovered changes not picked up by transcriptomic studies, such as accumulation of vesicle cargo and receptor ligands, including Apoe. Proteomic analysis of BECs from Apoe-deficient mice revealed a signature of accelerated aging. Our findings provide a resource for analysing BEC function during aging.
Collapse
Affiliation(s)
- Katalin Todorov-Völgyi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Judit González-Gallego
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Graduate School of Systemic Neuroscience (GSN), University Hospital, LMU Munich, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nathalie Beaufort
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mihail Ivilinov Todorov
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Dennis Crusius
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Sophie Robinson
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Graduate School of Systemic Neuroscience (GSN), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Andree Schmidt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florence Bareyre
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center Munich (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Ali Ertürk
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Division of Metabolic Biochemistry, Biomedical Center Munich (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
42
|
Drouyer M, Merjane J, Nazareth D, Knight M, Scott S, Liao SHY, Ginn SL, Zhu E, Alexander IE, Lisowski L. Development of CNS tropic AAV1-like variants with reduced liver-targeting following systemic administration in mice. Mol Ther 2024; 32:818-836. [PMID: 38297833 PMCID: PMC10928139 DOI: 10.1016/j.ymthe.2024.01.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/27/2023] [Accepted: 01/18/2024] [Indexed: 02/02/2024] Open
Abstract
Directed evolution of natural AAV9 using peptide display libraries have been widely used in the search for an optimal recombinant AAV (rAAV) for transgene delivery across the blood-brain barrier (BBB) to the CNS following intravenous ( IV) injection. In this study, we used a different approach by creating a shuffled rAAV capsid library based on parental AAV serotypes 1 through 12. Following selection in mice, 3 novel variants closely related to AAV1, AAV-BBB6, AAV-BBB28, and AAV-BBB31, emerged as top candidates. In direct comparisons with AAV9, our novel variants demonstrated an over 270-fold improvement in CNS transduction and exhibited a clear bias toward neuronal cells. Intriguingly, our AAV-BBB variants relied on the LY6A cellular receptor for CNS entry, similar to AAV9 peptide variants AAV-PHP.eB and AAV.CAP-B10, despite the different bioengineering methods used and parental backgrounds. The variants also showed reduced transduction of both mouse liver and human primary hepatocytes in vivo. To increase clinical translatability, we enhanced the immune escape properties of our new variants by introducing additional modifications based on rational design. Overall, our study highlights the potential of AAV1-like vectors for efficient CNS transduction with reduced liver tropism, offering promising prospects for CNS gene therapies.
Collapse
Affiliation(s)
- Matthieu Drouyer
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Jessica Merjane
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Deborah Nazareth
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Maddison Knight
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Suzanne Scott
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Sophia H Y Liao
- Gene Therapy Research Unit, Children's Medical Research Institute and Sydney Children's Hospitals Network, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Samantha L Ginn
- Gene Therapy Research Unit, Children's Medical Research Institute and Sydney Children's Hospitals Network, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Erhua Zhu
- Gene Therapy Research Unit, Children's Medical Research Institute and Sydney Children's Hospitals Network, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute and Sydney Children's Hospitals Network, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia; Discipline of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Leszek Lisowski
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia; Australian Genome Therapeutics Centre, Children's Medical Research Institute and Sydney Children's Hospitals Network, Westmead, NSW, Australia; Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warsaw, Poland.
| |
Collapse
|
43
|
Wang Y, Fu Q, Park SY, Lee YS, Park SY, Lee DY, Yoon S. Decoding cellular mechanism of recombinant adeno-associated virus (rAAV) and engineering host-cell factories toward intensified viral vector manufacturing. Biotechnol Adv 2024; 71:108322. [PMID: 38336188 DOI: 10.1016/j.biotechadv.2024.108322] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Recombinant adeno-associated virus (rAAV) is one of the prominent gene delivery vehicles that has opened promising opportunities for novel gene therapeutic approaches. However, the current major viral vector production platform, triple transfection in mammalian cells, may not meet the increasing demand. Thus, it is highly required to understand production bottlenecks from the host cell perspective and engineer the cells to be more favorable and tolerant to viral vector production, thereby effectively enhancing rAAV manufacturing. In this review, we provided a comprehensive exploration of the intricate cellular process involved in rAAV production, encompassing various stages such as plasmid entry to the cytoplasm, plasmid trafficking and nuclear delivery, rAAV structural/non-structural protein expression, viral capsid assembly, genome replication, genome packaging, and rAAV release/secretion. The knowledge in the fundamental biology of host cells supporting viral replication as manufacturing factories or exhibiting defending behaviors against viral production is summarized for each stage. The control strategies from the perspectives of host cell and materials (e.g., AAV plasmids) are proposed as our insights based on the characterization of molecular features and our existing knowledge of the AAV viral life cycle, rAAV and other viral vector production in the Human embryonic kidney (HEK) cells.
Collapse
Affiliation(s)
- Yongdan Wang
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, United States of America
| | - Qiang Fu
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, Lowell, MA 01854, United States of America
| | - So Young Park
- Department of Pharmaceutical Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States of America
| | - Yong Suk Lee
- Department of Pharmaceutical Sciences, University of Massachusetts Lowell, Lowell, MA 01854, United States of America
| | - Seo-Young Park
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Dong-Yup Lee
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, United States of America.
| |
Collapse
|
44
|
Merten OW. Development of Stable Packaging and Producer Cell Lines for the Production of AAV Vectors. Microorganisms 2024; 12:384. [PMID: 38399788 PMCID: PMC10892526 DOI: 10.3390/microorganisms12020384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/22/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Today, recombinant adeno-associated virus (rAAV) vectors represent the vector systems which are mostly used for in vivo gene therapy for the treatment of rare and less-rare diseases. Although most of the past developments have been performed by using a transfection-based method and more than half of the authorized rAAV-based treatments are based on transfection process, the tendency is towards the use of stable inducible packaging and producer cell lines because their use is much more straightforward and leads in parallel to reduction in the overall manufacturing costs. This article presents the development of HeLa cell-based packaging/producer cell lines up to their use for large-scale rAAV vector production, the more recent development of HEK293-based packaging and producer cell lines, as well as of packaging cell lines based on the use of Sf9 cells. The production features are presented in brief (where available), including vector titer, specific productivity, and full-to-empty particle ratio.
Collapse
|
45
|
Hadi M, Qutaiba B Allela O, Jabari M, Jasoor AM, Naderloo O, Yasamineh S, Gholizadeh O, Kalantari L. Recent advances in various adeno-associated viruses (AAVs) as gene therapy agents in hepatocellular carcinoma. Virol J 2024; 21:17. [PMID: 38216938 PMCID: PMC10785434 DOI: 10.1186/s12985-024-02286-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024] Open
Abstract
Primary liver cancer, which is scientifically referred to as hepatocellular carcinoma (HCC), is a significant concern in the field of global health. It has been demonstrated that conventional chemotherapy, chemo-hormonal therapy, and conformal radiotherapy are ineffective against HCC. New therapeutic approaches are thus urgently required. Identifying single or multiple mutations in genes associated with invasion, metastasis, apoptosis, and growth regulation has resulted in a more comprehensive comprehension of the molecular genetic underpinnings of malignant transformation, tumor advancement, and host interaction. This enhanced comprehension has notably propelled the development of novel therapeutic agents. Therefore, gene therapy (GT) holds great promise for addressing the urgent need for innovative treatments in HCC. However, the complexity of HCC demands precise and effective therapeutic approaches. The adeno-associated virus (AAV) distinctive life cycle and ability to persistently infect dividing and nondividing cells have rendered it an alluring vector. Another appealing characteristic of the wild-type virus is its evident absence of pathogenicity. As a result, AAV, a vector that lacks an envelope and can be modified to transport DNA to specific cells, has garnered considerable interest in the scientific community, particularly in experimental therapeutic strategies that are still in the clinical stage. AAV vectors emerge as promising tools for HCC therapy due to their non-immunogenic nature, efficient cell entry, and prolonged gene expression. While AAV-mediated GT demonstrates promise across diverse diseases, the current absence of ongoing clinical trials targeting HCC underscores untapped potential in this context. Furthermore, gene transfer through hepatic AAV vectors is frequently facilitated by GT research, which has been propelled by several congenital anomalies affecting the liver. Notwithstanding the enthusiasm associated with this notion, recent discoveries that expose the integration of the AAV vector genome at double-strand breaks give rise to apprehensions regarding their enduring safety and effectiveness. This review explores the potential of AAV vectors as versatile tools for targeted GT in HCC. In summation, we encapsulate the multifaceted exploration of AAV vectors in HCC GT, underlining their transformative potential within the landscape of oncology and human health.
Collapse
Affiliation(s)
- Meead Hadi
- Department of Microbiology, Faculty of Basic Science, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | - Mansoureh Jabari
- Medical Campus, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Asna Mahyazadeh Jasoor
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Omid Naderloo
- Department of Laboratory Sciences, Faculty of Medicine, Islamic Azad University of Gorgan Breanch, Gorgan, Iran
| | | | | | - Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
46
|
Schiller L, Ko C, Kosinska AD, Grimm D, Protzer U. Production and Purification of Adeno-Associated Viral Vectors for the Development of Immune-Competent Mouse Models of Persistent Hepatitis B Virus Replication. Methods Mol Biol 2024; 2837:207-218. [PMID: 39044087 DOI: 10.1007/978-1-0716-4027-2_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Mice infected with a recombinant adeno-associated virus carrying a replication-competent hepatitis B virus genome (rAAV-HBV) via the intravenous route establish a persistent HBV replication in hepatocytes and develop immune tolerance. They serve as models to evaluate antiviral immunity and to assess potential therapeutic approaches for chronic HBV infection. Combining selected HBV variants and different mouse genotypes allows for addressing a broad spectrum of research questions. This chapter describes the basic principles of the rAAV-HBV mouse model, rAAV-HBV production and purification methods, and finally, the in vivo application.
Collapse
Affiliation(s)
- Lisa Schiller
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany
| | - Chunkyu Ko
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Anna D Kosinska
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner sites, Munich and Heidelberg, Germany
| | - Dirk Grimm
- German Center for Infection Research (DZIF), partner sites, Munich and Heidelberg, Germany
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany.
- German Center for Infection Research (DZIF), partner sites, Munich and Heidelberg, Germany.
| |
Collapse
|
47
|
Chuecos MA, Lagor WR. Liver directed adeno-associated viral vectors to treat metabolic disease. J Inherit Metab Dis 2024; 47:22-40. [PMID: 37254440 PMCID: PMC10687323 DOI: 10.1002/jimd.12637] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/05/2023] [Accepted: 05/25/2023] [Indexed: 06/01/2023]
Abstract
The liver is the metabolic center of the body and an ideal target for gene therapy of inherited metabolic disorders (IMDs). Adeno-associated viral (AAV) vectors can deliver transgenes to the liver with high efficiency and specificity and a favorable safety profile. Recombinant AAV vectors contain only the transgene cassette, and their payload is converted to non-integrating circular double-stranded DNA episomes, which can provide stable expression from months to years. Insights from cellular studies and preclinical animal models have provided valuable information about AAV capsid serotypes with a high liver tropism. These vectors have been applied successfully in the clinic, particularly in trials for hemophilia, resulting in the first approved liver-directed gene therapy. Lessons from ongoing clinical trials have identified key factors affecting efficacy and safety that were not readily apparent in animal models. Circumventing pre-existing neutralizing antibodies to the AAV capsid, and mitigating adaptive immune responses to transduced cells are critical to achieving therapeutic benefit. Combining the high efficiency of AAV delivery with genome editing is a promising path to achieve more precise control of gene expression. The primary safety concern for liver gene therapy with AAV continues to be the small risk of tumorigenesis from rare vector integrations. Hepatotoxicity is a key consideration in the safety of neuromuscular gene therapies which are applied at substantially higher doses. The current knowledge base and toolkit for AAV is well developed, and poised to correct some of the most severe IMDs with liver-directed gene therapy.
Collapse
Affiliation(s)
- Marcel A. Chuecos
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX USA
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX USA
| | - William R. Lagor
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
48
|
Adsero A, Chestnut B, Shahnejat-Bushehri S, Sasnoor L, McMurphy T, Swenor M, Pasquino R, Pradhan A, Hernandez V, Padegimas L, Dismuke D. A Novel Role for the Adenovirus L4 Region 22K and 33K Proteins in Adeno-Associated Virus Production. Hum Gene Ther 2024; 35:59-69. [PMID: 38062776 PMCID: PMC10818037 DOI: 10.1089/hum.2023.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/26/2023] [Indexed: 01/18/2024] Open
Abstract
Despite decades of research in adeno-associated virus (AAV) and the role of adenovirus in production, the interplay of AAV and adenovirus is not fully understood. Specific regions of the adenoviral genome containing E1, E2a, E4 open reading frame (ORF), and VA RNA have been demonstrated as necessary for AAV production; however, incorporating these regions into either a producer cell line or subcloning into an Ad helper plasmid may lead to inclusion of neighboring adenoviral sequence or ORFs with unknown function. Because AAV is frequently used in gene therapies, removing excessive adenovirus sequences improves the Ad helper plasmid size and manufacturability, and may lead to safer vectors for patients. Furthermore, deepening our understanding of the helper virus genes required for recombinant AAV (rAAV) production has the potential to increase yields and manufacturability of rAAV for clinical and commercial applications. One region continuously included in various Ad helper plasmid iterations is the adenoviral E2a promoter region that appears to be necessary for E2a expression. Due to the compact nature of viral genomes, the E2a promoter region overlaps with the Hexon Assembly/100K protein and the L4 region. The L4 region, which contains the coding sequences for 22K and 33K proteins, had not been thought to be necessary for AAV production. Through molecular techniques, this study demonstrates that the adenoviral 22K protein is essential for rAAV production in HEK293 cells by triple transfection and that the 33K protein synergistically increases rAAV yield.
Collapse
Affiliation(s)
- Angela Adsero
- Molecular Development, Forge Biologics, Grove City, Ohio, USA
| | | | | | - Lalita Sasnoor
- Molecular Development, Forge Biologics, Grove City, Ohio, USA
| | - Travis McMurphy
- Molecular Development, Forge Biologics, Grove City, Ohio, USA
| | - Mike Swenor
- Molecular Development, Forge Biologics, Grove City, Ohio, USA
- Process Development, Forge Biologics, Grove City, Ohio, USA
| | - Ryan Pasquino
- Molecular Development, Forge Biologics, Grove City, Ohio, USA
| | - Arun Pradhan
- Molecular Development, Forge Biologics, Grove City, Ohio, USA
| | | | - Linas Padegimas
- Molecular Development, Forge Biologics, Grove City, Ohio, USA
| | - David Dismuke
- Chief Technical Officer, Forge Biologics, Grove City, Ohio, USA
| |
Collapse
|
49
|
Luo LL, Xu J, Wang BQ, Chen C, Chen X, Hu QM, Wang YQ, Zhang WY, Jiang WX, Li XT, Zhou H, Xiao X, Zhao K, Lin S. A novel capsid-XL32-derived adeno-associated virus serotype prompts retinal tropism and ameliorates choroidal neovascularization. Biomaterials 2024; 304:122403. [PMID: 38016335 DOI: 10.1016/j.biomaterials.2023.122403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/24/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023]
Abstract
Gene therapy has been adapted, from the laboratory to the clinic, to treat retinopathies. In contrast to subretinal route, intravitreal delivery of AAV vectors displays the advantage of bypassing surgical injuries, but the viral particles are more prone to be nullified by the host neutralizing factors. To minimize such suppression of therapeutic effect, especially in terms of AAV2 and its derivatives, we introduced three serine-to-glycine mutations, based on the phosphorylation sites identified by mass spectrum analysis, to the XL32 capsid to generate a novel serotype named AAVYC5. Via intravitreal administration, AAVYC5 was transduced more effectively into multiple retinal layers compared with AAV2 and XL32. AAVYC5 also enabled successful delivery of anti-angiogenic molecules to rescue laser-induced choroidal neovascularization and astrogliosis in mice and non-human primates. Furthermore, we detected fewer neutralizing antibodies and binding IgG in human sera against AAVYC5 than those specific for AAV2 and XL32. Our results thus implicate this capsid-optimized AAVYC5 as a promising vector suitable for a wide population, particularly those with undesirable AAV2 seroreactivity.
Collapse
Affiliation(s)
- Lin-Lin Luo
- Department of Ophthalmology, Army Medical Center of PLA, Army Medical University, Chongqing, 400042, China
| | - Jie Xu
- Department of Ophthalmology, Army Medical Center of PLA, Army Medical University, Chongqing, 400042, China
| | - Bing-Qiao Wang
- Department of Neurology, The Second Affiliated Hospital, Army Medical University, Chongqing, 400042, China
| | - Chen Chen
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China; Belief BioMed Co., Ltd, Shanghai, China
| | - Xi Chen
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China
| | - Qiu-Mei Hu
- Department of Ophthalmology, Army Medical Center of PLA, Army Medical University, Chongqing, 400042, China
| | - Yu-Qiu Wang
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China; Analytical Research Center for Organic and Biological Molecules, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wan-Yun Zhang
- Department of Neurology, The Second Affiliated Hospital, Army Medical University, Chongqing, 400042, China
| | - Wan-Xiang Jiang
- Sichuan Greentech Bioscience Co,. Ltd, Bencao Avenue, New Economic Development Zone, Meishan, Sichuan, 620010, China
| | - Xin-Ting Li
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Hu Zhou
- Analytical Research Center for Organic and Biological Molecules, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao Xiao
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China; Belief BioMed Co., Ltd, Shanghai, China.
| | - Kai Zhao
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China; Belief BioMed Co., Ltd, Shanghai, China.
| | - Sen Lin
- Department of Neurology, The Second Affiliated Hospital, Army Medical University, Chongqing, 400042, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China.
| |
Collapse
|
50
|
Walker S, Rose J, Hayes BC, Mulvania T. Generation of Complex Protein Structures by Coinfection with High-Quality Recombinant Baculovirus. Methods Mol Biol 2024; 2829:159-173. [PMID: 38951332 DOI: 10.1007/978-1-0716-3961-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
The baculovirus expression vector system (BEVS) is a powerful platform for protein expression in insect cells. A prevalent application is the expression of complex protein structures consisting of multiple, interacting proteins. Coinfection with multiple baculoviruses allows for production of complex structures, facilitating structure-function studies, allowing augmentation of insect cell functionality, and production of clinically relevant products such as virus-like particles (VLPs) and adeno-associated viral vectors (AAV). Successful coinfections require the generation of robust and well-quantified recombinant baculovirus stocks. Virus production through homologous recombination, combined with rigorous quantification of viral titers, allows for synchronous coinfections producing high end-product titers. In this chapter, we describe the streamlined workflow for generation and quantification of high-quality recombinant baculovirus stocks and successful coinfection as defined by a preponderance of dually infected cells in the insect cell culture.
Collapse
Affiliation(s)
- Scott Walker
- Expression Systems, LLC, an Advancion Company, Davis, CA, USA
| | - Jacqueline Rose
- Expression Systems, LLC, an Advancion Company, Davis, CA, USA
| | - Brooks C Hayes
- Expression Systems, LLC, an Advancion Company, Davis, CA, USA
| | - Thera Mulvania
- Expression Systems, LLC, an Advancion Company, Davis, CA, USA.
| |
Collapse
|