1
|
Park SY, Feng Z, Choi SH, Zhang X, Tang Y, Gasser GN, Richart D, Yuan F, Qiu J, Engelhardt JF, Yan Z. Recombinant Adeno-Associated Virus Vector Mediated Gene Editing in Proliferating and Polarized Cultures of Human Airway Epithelial Cells. Hum Gene Ther 2025. [PMID: 40359132 DOI: 10.1089/hum.2024.260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. While CRISPR-based CFTR editing approaches have shown proof-of-concept for functional rescue in primary airway basal cells, induced pluripotent stem cells, and organoid cultures derived from patients with CF, their efficacy remains suboptimal. Here, we developed the CuFiCas9(Y66S)eGFP reporter system by integrating spCas9 and a non-fluorescent Y66S eGFP mutant into CuFi-8 cells, an immortalized human airway epithelial cell line derived from a patient with CF with homozygous F508del mutations. These cells retain the basal cell phenotype in proliferating cultures and can differentiate into polarized airway epithelium at an air-liquid interface (ALI), enabling both visualized detection of gene editing and electrophysiological assessment of CFTR functional restoration. Using this system, recombinant adeno-associated virus (rAAV)-mediated homology-directed repair (HDR) was evaluated in proliferating cultures. A correction rate of 13.5 ± 0.8% was achieved in a population where 82.3 ± 5.6% of cells were productively transduced by AAV.eGFP630g2-CMVmCh, an rAAV editing vector with an mCherry reporter. Dual-editing of F508del CFTR and Y66S eGFP was explored using AAV.HR-eGFP630-F508(g03) to deliver two templates and single guide RNAs. eGFP+ (Y66S-corrected) cells and eGFP- (non-corrected) cells were sorted via fluorescence-activated cell sorting and differentiated at an ALI to assess the recovery of CFTR function. Despite a low F508 correction rate of 2.8%, ALI cultures derived from the eGFP- population exhibited 25.2% of the CFTR-specific transepithelial Cl- transport observed in CuFi-ALI cultures treated with CFTR modulators. Next-generation sequencing revealed frequent co-editing at both genomic loci, with sixfold higher F508 correction rate in the eGFP+ cells than eGFP- cells. In both populations, non-homology end joining predominated over HDR. This reporter system provides a valuable platform for optimizing editing efficiencies in proliferating airway basal cells, particularly for development of strategies to enhance HDR through modulation of DNA repair pathways.
Collapse
Affiliation(s)
- Soo Yeun Park
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Zehua Feng
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Soon H Choi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Xiujuan Zhang
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Yinghua Tang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Grace N Gasser
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Donovan Richart
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Feng Yuan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
2
|
Keeler AM, Zhan W, Ram S, Fitzgerald KA, Gao G. The curious case of AAV immunology. Mol Ther 2025; 33:1946-1965. [PMID: 40156190 DOI: 10.1016/j.ymthe.2025.03.037] [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: 01/31/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Immune responses to adeno-associated virus (AAV) have long been perplexing, from its first discovery to the latest clinical trials of recombinant AAV (rAAV) therapy. Wild-type AAV (wtAAV) does not cause any known disease, making it an ideal vector for gene therapy, as viral vectors retain virus-like properties. Although AAV stimulates only a mild immune response compared with other viruses, it is still recognized by the innate immune system and induces adaptive immune responses. B cell responses against both wtAAV and rAAV are robust and can hinder gene therapy applications and prevent redosing. T cell responses can clear transduced cells or establish tolerance against gene therapy. Immune responses to AAV gene therapy are influenced by many factors. Most clinical immunotoxicities that develop in response to gene therapies have emerged as higher doses of AAV vectors have been utilized and were not properly modeled in preclinical animal studies. Thus, several strategies have been undertaken to reduce or mitigate immune responses to AAV. While we have learned a considerable amount about how the immune system responds to AAV gene therapy since the discovery of AAV virus, it still remains a curious case that requires more investigation to fully understand.
Collapse
Affiliation(s)
- Allison M Keeler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; NeroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sanjay Ram
- Division of Infectious Diseases and Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Katherine A Fitzgerald
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
3
|
Heiser BJ, Veyssi A, Ghosh D. Recent strategies for enhanced delivery of mRNA to the lungs. Nanomedicine (Lond) 2025; 20:1043-1069. [PMID: 40190037 PMCID: PMC12051540 DOI: 10.1080/17435889.2025.2485669] [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: 12/20/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
mRNA-based therapies have emerged as a transformative tool in modern medicine, gaining significant attention following their successful use in COVID-19 vaccines. Delivery to the lungs offers several compelling advantages for mRNA delivery. The lungs are one of the most vascularized organs in the body, which provides an extensive surface area that can facilitate efficient drug transport. Local delivery to the lungs bypasses gastrointestinal degradation, potentially enhancing therapeutic efficacy. In addition, the extensive capillary network of the lungs provides an ideal target for systemic delivery. However, developing effective mRNA therapies for the lungs presents significant challenges. The complex anatomy of the lungs and the body's immune response to foreign particles create barriers to delivery. This review discusses key approaches for overcoming these challenges and improving mRNA delivery to the lungs. It examines both local and systemic delivery strategies aimed at improving lung delivery while mitigating off-target effects. Although substantial progress has been made in lung-targeted mRNA therapies, challenges remain in optimizing cellular uptake and achieving therapeutic efficacy within pulmonary tissues. The continued refinement of delivery strategies that enhance lung-specific targeting while minimizing degradation is critical for the clinical success of mRNA-based pulmonary therapies.
Collapse
Affiliation(s)
- Brittany J. Heiser
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Arian Veyssi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
4
|
Zhang Y, Chen Z, Wang X, Yan R, Bao H, Chu X, Guo L, Wang X, Li Y, Mu Y, He Q, Zhang L, Zhang C, Zhou D, Ji D. Site-specific tethering nanobodies on recombinant adeno-associated virus vectors for retargeted gene therapy. Acta Biomater 2024; 187:304-315. [PMID: 39025389 DOI: 10.1016/j.actbio.2024.07.023] [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: 04/01/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Recombinant adeno-associated viruses (rAAVs) have been extensively studied for decades as carriers for delivering therapeutic genes. However, designing rAAV vectors with selective tropism for specific cell types and tissues has remained challenging. Here, we introduce a strategy for redirecting rAAV by attaching nanobodies with desired tropism at specific sites, effectively replacing the original tropism. To demonstrate this concept, we initially modified the genetic code of rAAV2 to introduce an azido-containing unnatural amino acid at a precise site within the capsid protein. Following a screening process, we identified a critical site (N587+1) where the introduction of unnatural amino acid eliminated the natural tropism of rAAV2. Subsequently, we successfully redirected rAAV2 by conjugating various nanobodies at the N587+1 site, using click and SpyTag-Spycatcher chemistries to form nanobody-AAV conjugates (NACs). By investigating the relationship between NACs quantity and effect and optimizing the linker between rAAV2 and the nanobody using a cathepsin B-susceptible valine-citrulline (VC) dipeptide, we significantly improved gene delivery efficiency both in vitro and in vivo. This enhancement can be attributed to the facilitated endosomal escape of rAAV2. Our method offers an exciting avenue for the rational modification of rAAV2 as a retargeting vehicle, providing a convenient platform for precisely engineering various rAAV2 vectors for both basic research and therapeutic applications. STATEMENT OF SIGNIFICANCE: AAVs hold great promise in the treatment of genetic diseases, but their clinical use has been limited by off-target transduction and efficiency. Here, we report a strategy to construct NACs by conjugating a nanobody or scFv to an rAAV capsid site, specifically via biorthogonal click chemistry and a spy-spycatcher reaction. We explored the structure-effect and quantity-effect relationships of NACs and then optimized the transduction efficiency by introducing a valine-citrulline peptide linker. This approach provides a biocompatible method for rational modification of rAAV as a retargeting platform without structural disruption of the virus or alteration of the binding capacity of the nanobody, with potential utility across a broad spectrum of applications in targeted imaging and gene delivery.
Collapse
Affiliation(s)
- Yuanjie Zhang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Zhiqian Chen
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xiaoyang Wang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Rongding Yan
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Han Bao
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xindang Chu
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Lingfeng Guo
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xinchen Wang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Yuanhao Li
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Yu Mu
- Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China.
| | - Qiuchen He
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Lihe Zhang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Chuanling Zhang
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Demin Zhou
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Shenzhen Bay Laboratory, Gaoke International Innovation Center, Shenzhen, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| | - Dezhong Ji
- Peking University-Yunnan Baiiyao International Medical Research Center, ChemicalBiology Center, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Peking University Ningbo Institute of Marine Medicines, Ningbo, China.
| |
Collapse
|
5
|
Excoffon KJDA, Smith MD, Falese L, Schulingkamp R, Lin S, Mahankali M, Narayan PKL, Glatfelter MR, Limberis MP, Yuen E, Kolbeck R. Inhalation of SP-101 Followed by Inhaled Doxorubicin Results in Robust and Durable hCFTRΔR Transgene Expression in the Airways of Wild-Type and Cystic Fibrosis Ferrets. Hum Gene Ther 2024; 35:710-725. [PMID: 39155828 DOI: 10.1089/hum.2024.064] [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] [Indexed: 08/20/2024] Open
Abstract
Cystic fibrosis (CF) is a serious genetic disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Approved small molecule therapies benefit the majority of people with CF (pwCF), but unfortunately not all. Gene addition offers a mutation agnostic treatment option for all pwCF. SP-101 is an adeno-associated virus gene therapy vector (AAV2.5T) that has been optimized for efficient human airway cell transduction, and that contains a functional and regulated shortened human CFTR minigene (hCFTRΔR) with a small synthetic promoter/enhancer. To understand SP-101 airway distribution, activity, and the associated immune response, in vivo studies were performed in wild-type and CF ferrets. After single dose inhaled delivery of SP-101, followed by single dose inhaled doxorubicin (an AAV transduction augmenter) or saline, SP-101 vector genomes were detected throughout the respiratory tract. hCFTRΔR mRNA expression was highest in ferrets also receiving doxorubicin and persisted for the duration of the study (13 weeks). Pre-existing mucus in the CF ferrets did not present a barrier to effective transduction. Binding and neutralizing antibodies to the AAV2.5T capsid were observed regardless of doxorubicin exposure. Only a portion of ferrets exhibited a weak T-cell response to AAV2.5T and no T-cell response was seen against hCFTRΔR. These data strongly support the continued development of inhaled SP-101, followed by inhaled doxorubicin, for the treatment of CF.
Collapse
Affiliation(s)
| | - Mark D Smith
- Spirovant Sciences, Inc, Philadelphia, Pennsylvania, USA
| | - Lillian Falese
- Spirovant Sciences, Inc, Philadelphia, Pennsylvania, USA
| | | | - Shen Lin
- Spirovant Sciences, Inc, Philadelphia, Pennsylvania, USA
| | | | | | | | | | - Eric Yuen
- Spirovant Sciences, Inc, Philadelphia, Pennsylvania, USA
| | - Roland Kolbeck
- Spirovant Sciences, Inc, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Excoffon KJDA, Lin S, Narayan PKL, Sitaraman S, Jimah AM, Fallon TT, James ML, Glatfelter MR, Limberis MP, Smith MD, Guffanti G, Kolbeck R. SP-101, A Novel Adeno-Associated Virus Gene Therapy for the Treatment of Cystic Fibrosis, Mediates Functional Correction of Primary Human Airway Epithelia From Donors with Cystic Fibrosis. Hum Gene Ther 2024; 35:695-709. [PMID: 39155805 DOI: 10.1089/hum.2024.063] [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: 08/20/2024] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the gene encoding the CF transmembrane conductance regulator (CFTR) protein. Although CF affects multiple organs, lung disease is the main cause of morbidity and mortality, and gene therapy is expected to provide a mutation-agnostic option for treatment. SP-101 is a recombinant adeno-associated virus (AAV) gene therapy vector carrying a human CFTR minigene, hCFTRΔR, and is being investigated as an inhalation treatment for people with CF. To further understand SP-101 activity, in vitro studies were performed in human airway epithelia (HAE) derived from multiple CF and non-CF donors. SP-101 restored CFTR-mediated chloride conductance, measured via Ussing chamber assay, at a multiplicity of infection (MOI) as low as 5E2 in the presence of doxorubicin, a small molecule known to augment AAV transduction. Functional correction of CF HAE increased with increasing MOI and doxorubicin concentration and correlated with increasing cell-associated vector genomes and hCFTRΔR mRNA expression. Tropism studies using a fluorescent reporter vector and single-cell mRNA sequencing of SP-101-mediated hCFTRΔR mRNA demonstrated broad expression in all cell types after apical transduction, including secretory, ciliated, and basal cells. In summary, SP-101, particularly in combination with doxorubicin, shows promise for a novel CF treatment strategy and strongly supports continued development.
Collapse
Affiliation(s)
| | - Shen Lin
- Spirovant Sciences, Inc., Philadelphia, Pennsylvania, USA
| | | | | | - Awal M Jimah
- Spirovant Sciences, Inc., Philadelphia, Pennsylvania, USA
| | - Tyler T Fallon
- Spirovant Sciences, Inc., Philadelphia, Pennsylvania, USA
| | - Melane L James
- Spirovant Sciences, Inc., Philadelphia, Pennsylvania, USA
| | | | | | - Mark D Smith
- Spirovant Sciences, Inc., Philadelphia, Pennsylvania, USA
| | | | - Roland Kolbeck
- Spirovant Sciences, Inc., Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Ciobanu C, Yanda M, Zeidan A, Izzi J, Guggino WB, Cebotaru L. Amelioration of airway and GI disease in G551D-CF ferrets by AAV1 and AAV6. Gene Ther 2024; 31:499-510. [PMID: 39069560 DOI: 10.1038/s41434-024-00469-7] [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: 03/04/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Gene therapy for CF has concentrated on targeting the lung. Here we took a different approach by injecting into the cephalic vein and spraying into the trachea of G551D, CF ferrets either AAV1 or 6 containing Δ27-264-CFTR, a truncated version of CFTR. Treatment with the potentiator VX-770 was halted for 7 days before instillation to induce a disease phenotype. Indeed, all ferrets were pancreas-insufficient when they entered the study. Four ferrets (three receiving AAV1 and one AAV6) were necropsied 48 days after vector delivery, and four (three receiving AAV6, one AAV1) were euthanized or died prior to the planned necropsy. AAV1 or AAV6 vector genomes, mRNA expression, and CFTR protein were detected in all tracheal and lung samples and in the liver, pancreas, and ileum of the treated ferrets. Surface and basal airway cells, pancreatic and bile ducts, and ileal crypts and villi were successfully transduced. Obstruction of the airways accompanied by pulmonary hemorrhaging, plugged pancreatic and bile ducts as well as mucous plugs in the ileum were noticed in untreated but absent from transduced ferrets necropsied at 48 days. Transduction of G551D ferrets suggests that a combination of systemic and airway application may be the preferred route of delivery for CF.
Collapse
Affiliation(s)
- Cristian Ciobanu
- Departments of Physiology and Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Murali Yanda
- Departments of Physiology and Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Adi Zeidan
- Departments of Physiology and Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jessica Izzi
- Departments of Physiology and Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - William B Guggino
- Departments of Physiology and Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Liudmila Cebotaru
- Departments of Physiology and Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
8
|
Słyk Ż, Stachowiak N, Małecki M. Recombinant Adeno-Associated Virus Vectors for Gene Therapy of the Central Nervous System: Delivery Routes and Clinical Aspects. Biomedicines 2024; 12:1523. [PMID: 39062095 PMCID: PMC11274884 DOI: 10.3390/biomedicines12071523] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The Central Nervous System (CNS) is vulnerable to a range of diseases, including neurodegenerative and oncological conditions, which present significant treatment challenges. The blood-brain barrier (BBB) restricts molecule penetration, complicating the achievement of therapeutic concentrations in the CNS following systemic administration. Gene therapy using recombinant adeno-associated virus (rAAV) vectors emerges as a promising strategy for treating CNS diseases, demonstrated by the registration of six gene therapy products in the past six years and 87 ongoing clinical trials. This review explores the implementation of rAAV vectors in CNS disease treatment, emphasizing AAV biology and vector engineering. Various administration methods-such as intravenous, intrathecal, and intraparenchymal routes-and experimental approaches like intranasal and intramuscular administration are evaluated, discussing their advantages and limitations in different CNS contexts. Additionally, the review underscores the importance of optimizing therapeutic efficacy through the pharmacokinetics (PK) and pharmacodynamics (PD) of rAAV vectors. A comprehensive analysis of clinical trials reveals successes and challenges, including barriers to commercialization. This review provides insights into therapeutic strategies using rAAV vectors in neurological diseases and identifies areas requiring further research, particularly in optimizing rAAV PK/PD.
Collapse
Affiliation(s)
- Żaneta Słyk
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Natalia Stachowiak
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
9
|
Cao D, Byrne BJ, de Jong YP, Terhorst C, Duan D, Herzog RW, Kumar SR. Innate Immune Sensing of Adeno-Associated Virus Vectors. Hum Gene Ther 2024; 35:451-463. [PMID: 38887999 PMCID: PMC11310564 DOI: 10.1089/hum.2024.040] [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: 03/13/2024] [Accepted: 05/04/2024] [Indexed: 06/20/2024] Open
Abstract
Adeno-associated virus (AAV) based viral vectors are widely used in human gene therapy and form the basis of approved treatments for several genetic diseases. Immune responses to vector and transgene products, however, substantially complicate these applications in clinical practice. The role of innate immune recognition of AAV vectors was initially unclear, given that inflammatory responses early after vector administration were typically mild in animal models. However, more recent research continues to identify innate immune pathways that are triggered by AAV vectors and that serve to provide activation signals for antigen-presenting cells and initiation of adaptive immune responses. Sensing of the AAV genome by the endosomal DNA receptor toll-like receptor 9 (TLR9) promotes early inflammatory response and interferon expression. Thus, activation of the TLR9>MyD88 pathway in plasmacytoid dendritic cells (pDCs) leads to the conditioning of antigen cross-presenting DCs through type I interferon (IFN-I) and ultimately CD8+ T cell activation. Alternatively, pDCs may also promote CD8+ T cell responses in a TLR9-independent manner by the production of IL-1 cytokines, thereby activating the IL-1R1>MyD88 signaling pathway. AAV can induce cytokine expression in monocyte-derived DCs, which in turn increases antibody formation. Binding of AAV capsid to complement components likely further elevates B cell activation. At high systemic vector doses in humans and in non-human primates, AAV vectors can trigger complement activation, with contributions by classical and alternative pathways, leading to severe toxicities. Finally, evidence for activation of TLR2 by the capsid and of additional innate receptors for nucleic acids has been presented. These observations show that AAV vectors can initiate several and likely redundant innate immune pathways resulting in an exaggerated adaptive immune response.
Collapse
Affiliation(s)
- Di Cao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA
| | - Barry J. Byrne
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Ype P. de Jong
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, New York, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Boston, Massachusetts, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Roland W. Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA
| | - Sandeep R.P. Kumar
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
10
|
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
|
11
|
Zhang J, Frabutt DA, Chrzanowski M, Li N, Miller LM, Tian J, Mulcrone PL, Lam AK, Draper BE, Jarrold MF, Herzog RW, Xiao W. A novel class of self-complementary AAV vectors with multiple advantages based on cceAAV lacking mutant ITR. Mol Ther Methods Clin Dev 2024; 32:101206. [PMID: 38390555 PMCID: PMC10881427 DOI: 10.1016/j.omtm.2024.101206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/30/2024] [Indexed: 02/24/2024]
Abstract
Self-complementary AAV vectors (scAAV) use a mutant inverted terminal repeat (mITR) for efficient packaging of complementary stranded DNA, enabling rapid transgene expression. However, inefficient resolution at the mITR leads to the packaging of monomeric or subgenomic AAV genomes. These noncanonical particles reduce transgene expression and may affect the safety of gene transfer. To address these issues, we have developed a novel class of scAAV vectors called covalently closed-end double-stranded AAV (cceAAV) that eliminate the mITR resolution step during production. Instead of using a mutant ITR, we used a 56-bp recognition sequence of protelomerase (TelN) to covalently join the top and bottom strands, allowing the vector to be generated with just a single ITR. To produce cceAAV vectors, the vector plasmid is initially digested with TelN, purified, and then subjected to a standard triple-plasmid transfection protocol followed by traditional AAV vector purification procedures. Such cceAAV vectors demonstrate yields comparable to scAAV vectors. Notably, we observed enhanced transgene expression as compared to traditional scAAV vectors. The treatment of mice with hemophilia B with cceAAV-FIX resulted in significantly enhanced long-term FIX expression. The cceAAV vectors hold several advantages over scAAV vectors, potentially leading to the development of improved human gene therapy drugs.
Collapse
Affiliation(s)
- Junping Zhang
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Dylan A. Frabutt
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Ning Li
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Jiahe Tian
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Patrick L. Mulcrone
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anh K. Lam
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Martin F. Jarrold
- Chemistry Department, Indiana University, Bloomington, IN 47405, USA
| | - Roland W. Herzog
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Weidong Xiao
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
12
|
Lopez-Gordo E, Chamberlain K, Riyad JM, Kohlbrenner E, Weber T. Natural Adeno-Associated Virus Serotypes and Engineered Adeno-Associated Virus Capsid Variants: Tropism Differences and Mechanistic Insights. Viruses 2024; 16:442. [PMID: 38543807 PMCID: PMC10975205 DOI: 10.3390/v16030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024] Open
Abstract
Today, adeno-associated virus (AAV)-based vectors are arguably the most promising in vivo gene delivery vehicles for durable therapeutic gene expression. Advances in molecular engineering, high-throughput screening platforms, and computational techniques have resulted in a toolbox of capsid variants with enhanced performance over parental serotypes. Despite their considerable promise and emerging clinical success, there are still obstacles hindering their broader use, including limited transduction capabilities, tissue/cell type-specific tropism and penetration into tissues through anatomical barriers, off-target tissue biodistribution, intracellular degradation, immune recognition, and a lack of translatability from preclinical models to clinical settings. Here, we first describe the transduction mechanisms of natural AAV serotypes and explore the current understanding of the systemic and cellular hurdles to efficient transduction. We then outline progress in developing designer AAV capsid variants, highlighting the seminal discoveries of variants which can transduce the central nervous system upon systemic administration, and, to a lesser extent, discuss the targeting of the peripheral nervous system, eye, ear, lung, liver, heart, and skeletal muscle, emphasizing their tissue and cell specificity and translational promise. In particular, we dive deeper into the molecular mechanisms behind their enhanced properties, with a focus on their engagement with host cell receptors previously inaccessible to natural AAV serotypes. Finally, we summarize the main findings of our review and discuss future directions.
Collapse
|
13
|
Huang X, Wang X, Sun Y, Li L, Li A, Xu W, Xie X, Diao Y. Bleomycin promotes rAAV2 transduction via DNA-PKcs/Artemis-mediated DNA break repair pathways. Virology 2024; 590:109959. [PMID: 38100984 DOI: 10.1016/j.virol.2023.109959] [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: 08/07/2023] [Revised: 11/17/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Because it is safe and has a simple genome, recombinant adeno-associated virus (rAAV) is an extremely appealing vector for delivery in in vivo gene therapy. However, its low transduction efficiency for some cells, limits its further application in the field of gene therapy. Bleomycin is a chemotherapeutic agent approved by the FDA whose effect on rAAV transduction has not been studied. In this study, we systematically investigated the effect of Bleomycin on the second-strand synthesis and used CRISPR/CAS9 and RNAi methods to understand the effects of Bleomycin on rAAV vector transduction, particularly the effect of DNA repair enzymes. The results showed that Bleomycin could promote rAAV2 transduction both in vivo and in vitro. Increased transduction was discovered to be a direct result of decreased cytoplasmic rAAV particle degradation and increased second-strand synthesis. TDP1, PNKP, and SETMAR are required to repair the DNA damage gap caused by Bleomycin, TDP1, PNKP, and SETMAR promote rAAV second-strand synthesis. Bleomycin induced DNA-PKcs phosphorylation and phosphorylated DNA-PKcs and Artemis promoted second-strand synthesis. The current study identifies an effective method for increasing the capability and scope of in-vivo and in-vitro rAAV applications, which can amplify cell transduction at Bleomycin concentrations. It also supplies information on combining tumor gene therapy with chemotherapy.
Collapse
Affiliation(s)
- Xiaoping Huang
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China; Institute of Molecular Medicine, Huaqiao University, Quanzhou, China
| | - Xiao Wang
- Institute of Molecular Medicine, Huaqiao University, Quanzhou, China
| | - Yaqi Sun
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China
| | - Ling Li
- Institute of Molecular Medicine, Huaqiao University, Quanzhou, China
| | - Anna Li
- Institute of Molecular Medicine, Huaqiao University, Quanzhou, China
| | - Wentao Xu
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China
| | - Xiaolan Xie
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China.
| | - Yong Diao
- Institute of Molecular Medicine, Huaqiao University, Quanzhou, China.
| |
Collapse
|
14
|
Liu S, Chowdhury EA, Xu V, Jerez A, Mahmood L, Ly BQ, Le HK, Nguyen A, Rajwade A, Meno-Tetang G, Shah DK. Whole-Body Disposition and Physiologically Based Pharmacokinetic Modeling of Adeno-Associated Viruses and the Transgene Product. J Pharm Sci 2024; 113:141-157. [PMID: 37805073 DOI: 10.1016/j.xphs.2023.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
To facilitate model-informed drug development (MIDD) of adeno-associated virus (AAV) therapy, here we have developed a physiologically based pharmacokinetic (PBPK) model for AAVs following preclinical investigation in mice. After 2E11 Vg/mouse dose of AAV8 and AAV9 encoding a monoclonal antibody (mAb) gene, whole-body disposition of both the vector and the transgene mAb was evaluated over 3 weeks. At steady-state, the following tissue-to-blood (T/B) concentration ratios were found for AAV8/9: ∼50 for liver; ∼10 for heart and muscle; ∼2 for brain, lung, kidney, adipose, and spleen; ≤1 for bone, skin, and pancreas. T/B values for mAb were compared with the antibody biodistribution coefficients, and five different clusters of organs were identified based on their transgene expression profile. All the biodistribution data were used to develop a novel AAV PBPK model that incorporates: (i) whole-body distribution of the vector; (ii) binding, internalization, and intracellular processing of the vector; (iii) transgene expression and secretion; and (iv) whole-body disposition of the secreted transgene product. The model was able to capture systemic and tissue PK of the vector and the transgene-produced mAb reasonably well. Pathway analysis of the PBPK model suggested that liver, muscle, and heart are the main contributors for the secreted transgene mAb. Unprecedented PK data and the novel PBPK model developed here provide the foundation for quantitative systems pharmacology (QSP) investigations of AAV-mediated gene therapies. The PBPK model can also serve as a quantitative tool for preclinical study design and preclinical-to-clinical translation of AAV-based gene therapies.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Ekram Ahmed Chowdhury
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Vivian Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Anthony Jerez
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Leeha Mahmood
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Bao Quoc Ly
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Huyen Khanh Le
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Anne Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Aneesh Rajwade
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Guy Meno-Tetang
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
15
|
Ning K, Zhang X, Feng Z, Hao S, Kuz CA, Cheng F, Park SY, McFarlin S, Engelhardt JF, Yan Z, Qiu J. Inhibition of DNA-dependent protein kinase catalytic subunit boosts rAAV transduction of polarized human airway epithelium. Mol Ther Methods Clin Dev 2023; 31:101115. [PMID: 37841417 PMCID: PMC10568418 DOI: 10.1016/j.omtm.2023.101115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023]
Abstract
Adeno-associated virus 2.5T (AAV2.5T) was selected from the directed evolution of AAV capsid library in human airway epithelia. This study found that recombinant AAV2.5T (rAAV2.5T) transduction of well-differentiated primary human airway epithelia induced a DNA damage response (DDR) characterized by the phosphorylation of replication protein A32 (RPA32), histone variant H2AX (H2A histone family member X), and all three phosphatidylinositol 3-kinase-related kinases: ataxia telangiectasia mutated kinase, ataxia telangiectasia and Rad3-related kinase (ATR), and DNA-dependent protein kinase catalytic subunit (DNA-PKcs). While suppressing the expression of ATR by a specific pharmacological inhibitor or targeted gene silencing inhibited rAAV2.5T transduction, DNA-PKcs inhibition or targeted gene silencing significantly increased rAAV2.5T transgene expression. Notably, DNA-PKcs inhibitors worked as a "booster" to further increase rAAV2.5T transgene expression after treatment with doxorubicin and did not compromise epithelial integrity. Thus, our study provides evidence that DDR is associated with rAAV transduction in well-differentiated human airway epithelia, and DNA-PKcs inhibition has the potential to boost rAAV transduction. These findings highlight that the application of DDR inhibition-associated pharmacological interventions has the potential to increase rAAV transduction and thus to reduce the required vector dose.
Collapse
Affiliation(s)
- Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiujuan Zhang
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Zehua Feng
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Siyuan Hao
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Cagla Aksu Kuz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soo Yuen Park
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Shane McFarlin
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
16
|
Dogbey DM, Torres VES, Fajemisin E, Mpondo L, Ngwenya T, Akinrinmade OA, Perriman AW, Barth S. Technological advances in the use of viral and non-viral vectors for delivering genetic and non-genetic cargos for cancer therapy. Drug Deliv Transl Res 2023; 13:2719-2738. [PMID: 37301780 PMCID: PMC10257536 DOI: 10.1007/s13346-023-01362-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2023] [Indexed: 06/12/2023]
Abstract
The burden of cancer is increasing globally. Several challenges facing its mainstream treatment approaches have formed the basis for the development of targeted delivery systems to carry and distribute anti-cancer payloads to their defined targets. This site-specific delivery of drug molecules and gene payloads to selectively target druggable biomarkers aimed at inducing cell death while sparing normal cells is the principal goal for cancer therapy. An important advantage of a delivery vector either viral or non-viral is the cumulative ability to penetrate the haphazardly arranged and immunosuppressive tumour microenvironment of solid tumours and or withstand antibody-mediated immune response. Biotechnological approaches incorporating rational protein engineering for the development of targeted delivery systems which may serve as vehicles for packaging and distribution of anti-cancer agents to selectively target and kill cancer cells are highly desired. Over the years, these chemically and genetically modified delivery systems have aimed at distribution and selective accumulation of drug molecules at receptor sites resulting in constant maintenance of high drug bioavailability for effective anti-tumour activity. In this review, we highlighted the state-of-the art viral and non-viral drug and gene delivery systems and those under developments focusing on cancer therapy.
Collapse
Affiliation(s)
- Dennis Makafui Dogbey
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Emmanuel Fajemisin
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Liyabona Mpondo
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Takunda Ngwenya
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Olusiji Alex Akinrinmade
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, Bristol, UK
| | - Stefan Barth
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa.
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
17
|
Yanda MK, Zeidan A, Ciobanu C, Izzi J, Guggino WB, Cebotaru L. Transduction of Ferret Surface and Basal Cells of Airways, Lung, Liver, and Pancreas via Intratracheal or Intravenous Delivery of Adeno-Associated Virus 1 or 6. Hum Gene Ther 2023; 34:1135-1144. [PMID: 37650819 PMCID: PMC10659021 DOI: 10.1089/hum.2023.095] [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: 06/08/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
Cystic fibrosis (CF) is potentially treatable by gene therapy. Since the identification of the CF gene, preclinical and clinical trials have concentrated on achieving effective gene therapy targeting the lung. However, the lung has proven to be a formidable barrier to successful gene therapy especially for CF, and many clinical trials failed to achieve efficacy. Recent advances in vector design and adeno-associated virus (AAV) serotypes have increased the chances of success. Given that CF is a multi-organ disease, the goal of this study was to test whether a gene therapy approach involving AAV1 or AAV6 vector delivery via the systemic circulation would at the same time overcome the barrier of lung delivery and transduce organs commonly affected by CF. To accomplish this, we sprayed AAV1 containing green fluorescent protein (GFP) into the trachea or injected it intravenously (IV). We also tested AAV6 injected IV. No adverse events were noted. Ferrets were necropsied 30 days after vector delivery. AAV1 or AAV6 vector genomes, messenger RNA (mRNA) expression, and GFP were detected in all the tracheal and lung samples from the treated animals, whether AAV1 was sprayed into the trachea or injected IV or AAV6 was injected IV. Importantly, both surface epithelial and basal cells of the trachea and lung airways were successfully transduced, regardless of which route of delivery or vector serotype used for transduction. We detected also AAV1 and AAV6 vector genomes, mRNA expression, and GFP in the livers and pancreases, particularly in the acinar cells of the pancreatic duct. These data suggest that gene transfer is attainable in the airways, liver, and pancreas using either serotype, AAV1 or AAV6. Given that these same organs are affected in CF, systemic delivery of AAV may be the preferred route of delivery for a gene therapy for CF.
Collapse
Affiliation(s)
- Murali K. Yanda
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Adi Zeidan
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cristian Ciobanu
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jessica Izzi
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William B. Guggino
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liudmila Cebotaru
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Keiser NW, Cant E, Sitaraman S, Shoemark A, Limberis MP. Restoring Ciliary Function: Gene Therapeutics for Primary Ciliary Dyskinesia. Hum Gene Ther 2023; 34:821-835. [PMID: 37624733 DOI: 10.1089/hum.2023.102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023] Open
Abstract
Primary ciliary dyskinesia (PCD) is a genetic disease characterized by defects in motile cilia, which play an important role in several organ systems. Lung disease is a hallmark of PCD, given the essential role of cilia in airway surface defense. Diagnosis of PCD is complicated due to its reliance on complex tests that are not utilized by every clinic and also its phenotypic overlap with several other respiratory diseases. Nonetheless, PCD is increasingly being recognized as more common than once thought. The disease is genetically complex, with several genes reported to be associated with PCD. There is no cure for PCD, but gene therapy remains a promising therapeutic strategy. In this review, we provide an overview of the clinical symptoms, diagnosis, genetics, and current treatment regimens for PCD. We also describe PCD model systems and discuss the therapeutic potential of different gene therapeutics for targeting the intended cellular target, the ciliated cells of the airway.
Collapse
Affiliation(s)
| | - Erin Cant
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | | | - Amelia Shoemark
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | | |
Collapse
|
19
|
Huang X, Wang X, Li L, Wang Q, Xu W, Wu W, Xie X, Diao Y. MiR133b-mediated inhibition of EGFR-PTK pathway promotes rAAV2 transduction by facilitating intracellular trafficking and augmenting second-strand synthesis. J Cell Mol Med 2023; 27:2714-2729. [PMID: 37469226 PMCID: PMC10494303 DOI: 10.1111/jcmm.17858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) is an extremely attractive vector in the in vivo delivery of gene therapy as it is safe and its genome is simple. However, challenges including low permissiveness to specific cells and restricted tissue specificity have hindered its clinical application. Based on the previous studies, epidermal growth factor receptor-protein tyrosine kinase (EGFR-PTK) negatively regulated rAAV transduction, and EGFR-positive cells were hardly permissive to rAAV transduction. We constructed a novel rAAV-miRNA133b vector, which co-expressed miRNA133b and transgene, and investigated its in vivo and in vitro transduction efficiency. Confocal microscopy, live-cell imaging, pharmacological reagents and labelled virion tracking were used to analyse the effect of miRNA133b on rAAV2 transduction and the underlying mechanisms. The results demonstrated that miRNA133b could promote rAAV2 transduction and the effects were limited to EGFR-positive cells. The increased transduction was found to be a direct result of decreased rAAV particles degradation in the cytoplasm and enhanced second-strand synthesis. ss-rAAV2-miRNA133b vector specifically increased rAAV2 transduction in EGFR-positive cells or tissues, while ss-rAAV2-Fluc-miRNA133b exerted an antitumor effect. rAAV-miRNA133b vector might emerge as a promising platform for delivering various transgene to treat EGFR-positive cell-related diseases, such as non-small-cell lung cancer.
Collapse
Affiliation(s)
- Xiaoping Huang
- College of Chemical Engineering and Materials SciencesQuanzhou Normal UniversityQuanzhouChina
- School of MedicineHuaqiao UniversityQuanzhouChina
| | - Xiao Wang
- School of MedicineHuaqiao UniversityQuanzhouChina
| | - Ling Li
- School of MedicineHuaqiao UniversityQuanzhouChina
| | - Qizhao Wang
- School of MedicineHuaqiao UniversityQuanzhouChina
| | - Wentao Xu
- College of Chemical Engineering and Materials SciencesQuanzhou Normal UniversityQuanzhouChina
| | - Wenlin Wu
- College of Chemical Engineering and Materials SciencesQuanzhou Normal UniversityQuanzhouChina
| | - Xiaolan Xie
- College of Chemical Engineering and Materials SciencesQuanzhou Normal UniversityQuanzhouChina
| | - Yong Diao
- School of MedicineHuaqiao UniversityQuanzhouChina
| |
Collapse
|
20
|
Srivastava A. Rationale and strategies for the development of safe and effective optimized AAV vectors for human gene therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:949-959. [PMID: 37293185 PMCID: PMC10244667 DOI: 10.1016/j.omtn.2023.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Recombinant adeno-associated virus (AAV) vectors have been, or are currently in use, in 332 phase I/II/III clinical trials in a number of human diseases, and in some cases, remarkable clinical efficacy has also been achieved. There are now three US Food and Drug Administration (FDA)-approved AAV "drugs," but it has become increasingly clear that the first generation of AAV vectors are not optimal. In addition, relatively large vector doses are needed to achieve clinical efficacy, which has been shown to provoke host immune responses culminating in serious adverse events and, more recently, in the deaths of 10 patients to date. Thus, there is an urgent need for the development of the next generation of AAV vectors that are (1) safe, (2) effective, and (3) human tropic. This review describes the strategies to potentially overcome each of the limitations of the first generation of AAV vectors and the rationale and approaches for the development of the next generation of AAV serotype vectors. These vectors promise to be efficacious at significant reduced doses, likely to achieve clinical efficacy, thereby increasing the safety as well as reducing vector production costs, ensuring translation to the clinic with higher probability of success, without the need for the use of immune suppression, for gene therapy of a wide variety of diseases in humans.
Collapse
Affiliation(s)
- Arun Srivastava
- Division of Cellular and Molecular Therapy, Departments of Pediatrics, Molecular Genetics and Microbiology, Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
21
|
Tang Y, Fakhari S, Huntemann ED, Feng Z, Wu P, Feng WY, Lei J, Yuan F, Excoffon KJ, Wang K, Limberis MP, Kolbeck R, Yan Z, Engelhardt JF. Immunosuppression reduces rAAV2.5T neutralizing antibodies that limit efficacy following repeat dosing to ferret lungs. Mol Ther Methods Clin Dev 2023; 29:70-80. [PMID: 36950451 PMCID: PMC10025970 DOI: 10.1016/j.omtm.2023.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
The efficacy of redosing the recombinant adeno-associated virus (rAAV) vector rAAV2.5T to ferret lung is limited by AAV neutralizing antibody (NAb) responses. While immunosuppression strategies have allowed for systemic rAAV repeat dosing, their utility for rAAV lung-directed gene therapy is largely unexplored. To this end, we evaluated two immunosuppression (IS) strategies to improve repeat dosing of rAAV2.5T to ferret lungs: (1) a combination of three IS drugs (Tri-IS) with broad coverage against cellular and humoral responses (methylprednisolone [MP], azathioprine, and cyclosporine) and (2) MP alone, which is typically used in systemic rAAV applications. Repeat dosing utilized AAV2.5T-SP183-fCFTRΔR (recombinant ferret CFTR transgene), followed 28 days later by AAV2.5T-SP183-gLuc (for quantification of transgene expression). Both the Tri-IS and MP strategies significantly improved transgene expression following repeat dosing and reduced AAV2.5T NAb responses in the bronchioalveolar lavage fluid (BALF) and plasma, while AAV2.5T binding antibody subtypes and cellular immune responses by ELISpot were largely unchanged by IS. One exception was the reduction in plasma AAV2.5T binding immunoglobulin G (IgG) in both IS groups. Only the Tri-IS strategy significantly suppressed splenocyte expression of IFNA (interferon α [IFN-α]) and IL4. Our studies suggest that IS strategies may be useful in clinical application of rAAV targeting lung genetic diseases such as cystic fibrosis.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shahab Fakhari
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Eric D. Huntemann
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zehua Feng
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Peipei Wu
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - William Y. Feng
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Junying Lei
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Feng Yuan
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - Kai Wang
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
| | | | | | - Ziying Yan
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - John F. Engelhardt
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
22
|
Arjomandnejad M, Dasgupta I, Flotte TR, Keeler AM. Immunogenicity of Recombinant Adeno-Associated Virus (AAV) Vectors for Gene Transfer. BioDrugs 2023; 37:311-329. [PMID: 36862289 PMCID: PMC9979149 DOI: 10.1007/s40259-023-00585-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/03/2023]
Abstract
Recombinant adeno-associated viruses (AAVs) have emerged as promising gene delivery vehicles resulting in three US Food and Drug Administration (FDA) and one European Medicines Agency (EMA)-approved AAV-based gene therapies. Despite being a leading platform for therapeutic gene transfer in several clinical trials, host immune responses against the AAV vector and transgene have hampered their widespread application. Multiple factors, including vector design, dose, and route of administration, contribute to the overall immunogenicity of AAVs. The immune responses against the AAV capsid and transgene involve an initial innate sensing. The innate immune response subsequently triggers an adaptive immune response to elicit a robust and specific response against the AAV vector. AAV gene therapy clinical trials and preclinical studies provide important information about the immune-mediated toxicities associated with AAV, yet studies suggest preclinical models fail to precisely predict the outcome of gene delivery in humans. This review discusses the contribution of the innate and adaptive immune response against AAVs, highlighting the challenges and potential strategies to mitigate these responses, thereby enhancing the therapeutic potential of AAV gene therapy.
Collapse
Affiliation(s)
- Motahareh Arjomandnejad
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 386 Plantation Street, Worcester, MA, 01605, USA
| | - Ishani Dasgupta
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 386 Plantation Street, Worcester, MA, 01605, USA
| | - Terence R Flotte
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 386 Plantation Street, Worcester, MA, 01605, USA
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Allison M Keeler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 386 Plantation Street, Worcester, MA, 01605, USA.
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
23
|
Huang X, Wang X, Ren Y, Gao P, Xu W, Xie X, Diao Y. Reactive oxygen species enhance rAAV transduction by promoting its escape from late endosomes. Virol J 2023; 20:2. [PMID: 36611172 PMCID: PMC9825130 DOI: 10.1186/s12985-023-01964-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/04/2023] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Recent seminal studies have revealed that endosomal reactive oxygen species (ROS) promote rather than inhibit viral infection. Some ROS generators, including shikonin and H2O2, have the potential to enhance recombinant adeno-associated virus (rAAV) transduction. However, the impact of ROS on rAAV intracellular trafficking remains unclear. METHODS To understand the effects of ROS on the transduction of rAAV vectors, especially the rAAV subcellular distribution profiles, this study systematically explored the effect of ROS on each step of rAAV intracellular trafficking pathway using fluorescently-labeled rAAV and qPCR quantification determination. RESULTS The results showed promoted in-vivo and in-vitro rAAV transduction by ROS exposure, regardless of vector serotype or cell type. ROS treatment directed rAAV intracellular trafficking towards a more productive pathway by upregulating the expression of cathepsins B and L, accelerating the rAAV transit in late endosomes, and increasing the rAAV nucleus entry. CONCLUSIONS These data support that ROS generative drugs, such as shikonin, have the potential to promote rAAV vector transduction by promoting rAAV's escape from late endosomes, and enhancing its productive trafficking to the nucleus.
Collapse
Affiliation(s)
- Xiaoping Huang
- grid.449406.b0000 0004 1757 7252College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China
| | - Xiao Wang
- grid.411404.40000 0000 8895 903XSchool of Medicine, Huaqiao University, Quanzhou, China
| | - Yanxuan Ren
- grid.411404.40000 0000 8895 903XSchool of Medicine, Huaqiao University, Quanzhou, China
| | - Pingzhang Gao
- grid.449406.b0000 0004 1757 7252College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China
| | - Wentao Xu
- grid.449406.b0000 0004 1757 7252College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China
| | - Xiaolan Xie
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China.
| | - Yong Diao
- School of Medicine, Huaqiao University, Quanzhou, China.
| |
Collapse
|
24
|
High KA. THE JEREMIAH METZGER LECTURE: TURNING GENES INTO MEDICINES: HIGHLIGHTS AND HURDLES IN THE DEVELOPMENT OF GENE THERAPY FOR GENETIC DISEASE. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2023; 133:204-233. [PMID: 37701622 PMCID: PMC10493758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
The journey from in vitro transfer of genes into mammalian cells to approved gene therapy products has spanned decades. This manuscript summarizes hurdles encountered and obstacles overcome in the development of successful adeno-associated viral (AAV) vectors for hemophilia B and for an inherited retinal dystrophy caused by mutations in the RPE65 gene. In the case of hemophilia B, careful analysis of the first unsuccessful attempts led to the realization that the human immune response to AAV vectors was preventing durable expression; elucidation of the response to the recombinant virion led to strategies that enabled successful long-lasting gene transfer. For RPE65 deficiency, a key to success was development and validation of a novel clinical endpoint for a disease that previously lacked a pharmacologic treatment.
Collapse
|
25
|
Shiraishi Y, Adachi T, Cacicedo JM, Ido Y. Development of a high-yield, high-quality purification process for adeno-associated virus vectors that can be used in vivo without ultracentrifugation: Application to a lung endothelial cell-targeted adeno-associated virus. FASEB J 2022; 36:e22653. [PMID: 36374251 DOI: 10.1096/fj.202200840rr] [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: 06/03/2022] [Revised: 10/15/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Recombinant adeno-associated viruses (rAAVs) are useful vectors for expressing genes of interest in vivo because of their low immunogenicity and long-term gene expression. Various mutations have been introduced in recent years and have enabled high-efficacy, stabilized, and organ-oriented transduction. Our purpose for using rAAV is to express our target gene in the mouse lung to investigate pulmonary artery hypertension. We constructed a self-complementary AAV having mutant capsids with the ESGHGYF insert, which directs the vectors to lung endothelial cells. However, when this mutant virus was purified from the producing cells by the conventional method using an ultracentrifuge, it resulted in a low yield. In addition, the purification method using an ultracentrifuge is tedious and labor-intensive. Therefore, we aimed to develop a simple, high-quality method for obtaining enough lung-targeted rAAV. First, we modified amino acids (T491V and Y730F) of the capsid to stabilize the rAAV from degradation, and we optimized culture conditions. Next, we noticed that many rAAVs were released from the cells into the culture medium. We, therefore, improved our purification method by purifying from the culture medium without the ultracentrifugation step. Purification without ultracentrifugation had the problem that impurities were mixed in, causing inflammation. However, by performing PEG precipitation and chloroform extraction twice, we were able to purify rAAV that caused only as little inflammation as that obtained by the ultracentrifuge method. Sufficient rAAV was obtained and can now be administered to a rat as well as mice from a single dish: 1.50 × 1013 ± 3.58 × 1012 vector genome from one φ150 mm dish (mean ± SEM).
Collapse
Affiliation(s)
- Yasunaga Shiraishi
- Division of Environmental Medicine, National Defense Medical College Research Institute, National Defense Medical College, Saitama, Japan.,Division of Cardiovascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Takeshi Adachi
- Division of Cardiovascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Jose M Cacicedo
- Department of Research and Development, ALPCO Diagnostics, Salem, New Hampshire, USA
| | - Yasuo Ido
- Division of Cardiovascular Medicine, Department of Internal Medicine, National Defense Medical College, Saitama, Japan.,Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Sui H, Xu X, Su Y, Gong Z, Yao M, Liu X, Zhang T, Jiang Z, Bai T, Wang J, Zhang J, Xu C, Luo M. Gene therapy for cystic fibrosis: Challenges and prospects. Front Pharmacol 2022; 13:1015926. [PMID: 36304167 PMCID: PMC9592762 DOI: 10.3389/fphar.2022.1015926] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/29/2022] [Indexed: 11/25/2022] Open
Abstract
Cystic fibrosis (CF) is a life-threatening autosomal-recessive disease caused by mutations in a single gene encoding cystic fibrosis transmembrane conductance regulator (CFTR). CF effects multiple organs, and lung disease is the primary cause of mortality. The median age at death from CF is in the early forties. CF was one of the first diseases to be considered for gene therapy, and efforts focused on treating CF lung disease began shortly after the CFTR gene was identified in 1989. However, despite the quickly established proof-of-concept for CFTR gene transfer in vitro and in clinical trials in 1990s, to date, 36 CF gene therapy clinical trials involving ∼600 patients with CF have yet to achieve their desired outcomes. The long journey to pursue gene therapy as a cure for CF encountered more difficulties than originally anticipated, but immense progress has been made in the past decade in the developments of next generation airway transduction viral vectors and CF animal models that reproduced human CF disease phenotypes. In this review, we look back at the history for the lessons learned from previous clinical trials and summarize the recent advances in the research for CF gene therapy, including the emerging CRISPR-based gene editing strategies. We also discuss the airway transduction vectors, large animal CF models, the complexity of CF pathogenesis and heterogeneity of CFTR expression in airway epithelium, which are the major challenges to the implementation of a successful CF gene therapy, and highlight the future opportunities and prospects.
Collapse
Affiliation(s)
- Hongshu Sui
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| | - Xinghua Xu
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Yanping Su
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Zhaoqing Gong
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Minhua Yao
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Xiaocui Liu
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Ting Zhang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Ziyao Jiang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Tianhao Bai
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Junzuo Wang
- The Affiliated Tai’an City Central Hospital of Qingdao University, Tai’an, Shandong, China
| | - Jingjun Zhang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Changlong Xu
- The Reproductive Medical Center of Nanning Second People’s Hospital, Nanning, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| | - Mingjiu Luo
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| |
Collapse
|
27
|
Li X, Wei X, Lin J, Ou L. A versatile toolkit for overcoming AAV immunity. Front Immunol 2022; 13:991832. [PMID: 36119036 PMCID: PMC9479010 DOI: 10.3389/fimmu.2022.991832] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 12/12/2022] Open
Abstract
Recombinant adeno-associated virus (AAV) is a promising delivery vehicle for in vivo gene therapy and has been widely used in >200 clinical trials globally. There are already several approved gene therapy products, e.g., Luxturna and Zolgensma, highlighting the remarkable potential of AAV delivery. In the past, AAV has been seen as a relatively non-immunogenic vector associated with low risk of toxicity. However, an increasing number of recent studies indicate that immune responses against AAV and transgene products could be the bottleneck of AAV gene therapy. In clinical studies, pre-existing antibodies against AAV capsids exclude many patients from receiving the treatment as there is high prevalence of antibodies among humans. Moreover, immune response could lead to loss of efficacy over time and severe toxicity, manifested as liver enzyme elevations, kidney injury, and thrombocytopenia, resulting in deaths of non-human primates and patients. Therefore, extensive efforts have been attempted to address these issues, including capsid engineering, plasmapheresis, IgG proteases, CpG depletion, empty capsid decoy, exosome encapsulation, capsid variant switch, induction of regulatory T cells, and immunosuppressants. This review will discuss these methods in detail and highlight important milestones along the way.
Collapse
Affiliation(s)
- Xuefeng Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaoli Wei
- Guangzhou Dezheng Biotechnology Co., Ltd., Guangzhou, China
| | - Jinduan Lin
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Li Ou
- Genemagic Biosciences, Philadelphia, PA, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Li Ou,
| |
Collapse
|
28
|
Jäschke N, Büning H. Adeno-Associated Virus Vector Design-Moving the Adeno-Associated Virus to a Bioengineered Therapeutic Nanoparticle. Hematol Oncol Clin North Am 2022; 36:667-685. [PMID: 35778330 DOI: 10.1016/j.hoc.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Although the number of market-approved gene therapies is still low, this new class of therapeutics has become an integral part of modern medicine. The success and safety of gene therapy depend on the vectors used to deliver the therapeutic material. Adeno-associated virus (AAV) vectors have emerged as the most frequently used delivery system for in vivo gene therapy. This success was achieved with first-generation vectors, using capsids derived from natural AAV serotypes. Their broad tropism, the high seroprevalence for many of the AAV serotypes in the human population, and the high vector doses needed to transduce a sufficient number of therapy-relevant target cells are challenges that are addressed by engineering the capsid and the vector genome, improving the efficacy of these biological nanoparticles.
Collapse
Affiliation(s)
- Nico Jäschke
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str.1, Hannover 30625, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Str.1, Hannover 30625, Germany; REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str.1, Hannover 30625, Germany; German Center for Infection Research, Partner Site Hannover-Braunschweig.
| |
Collapse
|
29
|
Wang Y, Yang C, Hu H, Chen C, Yan M, Ling F, Wang KC, Wang X, Deng Z, Zhou X, Zhang F, Lin S, Du Z, Zhao K, Xiao X. Directed evolution of adeno-associated virus 5 capsid enables specific liver tropism. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:293-306. [PMID: 35474733 PMCID: PMC9010518 DOI: 10.1016/j.omtn.2022.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/18/2022] [Indexed: 02/07/2023]
Abstract
Impressive achievements in clinical trials to treat hemophilia establish a milestone in the development of gene therapy. It highlights the significance of AAV-mediated gene delivery to liver. AAV5 is a unique serotype featured by low neutralizing antibody prevalence. Nevertheless, its liver infectivity is relatively weak. Consequently, it is vital to exploit novel AAV5 capsid mutants with robust liver tropism. To this aim, we performed AAV5-NNK library and barcode screening in mice, from which we identified one capsid variant, called AAVzk2. AAVzk2 displayed a similar yield but divergent post-translational modification sites compared with wild-type serotypes. Mice intravenously injected with AAVzk2 demonstrated a stronger liver transduction than AAV5, roughly comparable with AAV8 and AAV9, with undetectable transduction of other tissues or organs such as heart, lung, spleen, kidney, brain, and skeletal muscle, indicating a liver-specific tropism. Further studies showed a superior human hepatocellular transduction of AAVzk2 to AAV5, AAV8 and AAV9, whereas the seroreactivity of AAVzk2 was as low as AAV5. Overall, we provide a novel AAV serotype that facilitates a robust and specific liver gene delivery to a large population, especially those unable to be treated by AAV8 and AAV9.
Collapse
Affiliation(s)
- Yuqiu Wang
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Chen Yang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Hanyang Hu
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Chen Chen
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Mengdi Yan
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Feixiang Ling
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Kathy Cheng Wang
- Department of Biology, New York University, 24 Waverly Pl, New York, NY 10003, USA
| | - Xintao Wang
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zhe Deng
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xinyue Zhou
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Feixu Zhang
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Sen Lin
- Department of Ophthalmology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing 400042, China
| | - Zengmin Du
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Kai Zhao
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Corresponding author Kai Zhao, School of Bioengineering and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| | - Xiao Xiao
- School of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Corresponding author Xiao Xiao, School of Bioengineering and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
30
|
Carraro G, Stripp BR. Insights gained in the pathology of lung disease through single cell transcriptomics. J Pathol 2022; 257:494-500. [PMID: 35608561 DOI: 10.1002/path.5971] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/17/2022] [Indexed: 11/07/2022]
Abstract
The human lung is a relatively quiescent organ in the normal healthy state but contains stem/progenitor cells that contribute to normal tissue maintenance and either repair or remodeling in response to injury and disease. Maintenance or repair lead to proper restoration of functional lung tissue and maintenance of physiological functions, with remodeling resulting in altered structure and function that is typically associated with disease. Knowledge of cell types contributing to lung tissue maintenance and repair/remodeling have largely relied on mouse models of injury-repair and lineage tracing of local progenitors. Therefore, many of the functional alterations underlying remodeling in human lung disease, have remained poorly defined. However, the advent of advanced genomics approaches to define the molecular phenotype of lung cells at single cell resolution has paved the way for rapid advances in our understanding of cell types present within the normal human lung and changes that accompany disease. Here we summarize recent advances in our understanding of disease-related changes in the molecular phenotype of human lung epithelium that have emerged from single-cell transcriptomic studies. We focus attention on emerging concepts of epithelial transitional states that characterize the pathological remodeling that accompanies chronic lung diseases, including idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, cystic fibrosis, and asthma. Concepts arising from these studies are actively evolving and require corroborative studies to improve our understanding of disease mechanisms. Whenever possible we highlight opportunities for providing a unified nomenclature in this rapidly advancing field of research. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Gianni Carraro
- Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Barry R Stripp
- Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
31
|
Barillà C, Suzuki S, Rab A, Sorscher EJ, Davis BR. Targeted Gene Insertion for Functional CFTR Restoration in Airway Epithelium. Front Genome Ed 2022; 4:847645. [PMID: 35330693 PMCID: PMC8940244 DOI: 10.3389/fgeed.2022.847645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
Cystic Fibrosis (CF) is caused by a diverse set of mutations distributed across the approximately 250 thousand base pairs of the CFTR gene locus, of which at least 382 are disease-causing (CFTR2.org). Although a variety of editing tools are now available for correction of individual mutations, a strong justification can be made for a more universal gene insertion approach, in principle capable of correcting virtually all CFTR mutations. Provided that such a methodology is capable of efficiently correcting relevant stem cells of the airway epithelium, this could potentially provide life-long correction for the lung. In this Perspective we highlight several requirements for efficient gene insertion into airway epithelial stem cells. In addition, we focus on specific features of the transgene construct and the endogenous CFTR locus that influence whether the inserted gene sequences will give rise to robust and physiologically relevant levels of CFTR function in airway epithelium. Finally, we consider how in vitro gene insertion methodologies may be adapted for direct in vivo editing.
Collapse
Affiliation(s)
- Cristina Barillà
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Shingo Suzuki
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Andras Rab
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Eric J Sorscher
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Brian R Davis
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
32
|
Yanda MK, Tomar V, Cebotaru CV, Guggino WB, Cebotaru L. Short-Term Steroid Treatment of Rhesus Macaque Increases Transduction. Hum Gene Ther 2022; 33:131-147. [PMID: 34806411 PMCID: PMC8885436 DOI: 10.1089/hum.2021.239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Repeat dosing poses a major hurdle for the development of an adeno-associated virus (AAV)-based gene therapy for cystic fibrosis, in part because of the potential for development of an immune reaction to the AAV1 capsid proteins. Here, to dampen the immune response to AAV1, we treated Rhesus monkeys with methylprednisolone before and after the instillation of two doses of AAV1Δ27-264-CFTR into their airways at 0 and 30 days, followed by a single dose of AAV1-GFP on day 60. Animals were euthanized on day 90, except for one monkey that was sacrificed at 1 year. No adverse events occurred, indicating that the two AAV1 vectors are safe. rAAV1-CFTR and AAV1-GFP vector genomes and mRNA transcripts were detectable in all lung sections and in the liver and pancreas at day 90 and after 1 year at levels comparable with animals necropsied at 90 days. The numbers of vector genomes for cystic fibrosis transmembrane regulator (CFTR) and green fluorescent protein (GFP) detected here were higher than those found in the monkeys infected without methylprednisolone treatment that we tested previously.1 Also, lung surface and keratin 5-positive basal cells showed higher CFTR and GFP staining than did the cells from the uninfected monkey control. Positive immunostaining, also detected in the liver and pancreas, remained stable for at least a year. All animals seroconverted for anticapsid antibodies by 90 days post-treatment. The neutralizing antibody titer declined in the animal necropsied at 1 year. Conclusion: AAV1 safely and effectively transduces monkey airway and basal cells. Both the presence of vector genomes and transduction from AAV1-CFTR and AAV1-GFP virus seen in the monkeys 4 months to 1 year after the first instillation suggest that repeat dosing with AAV1-based vectors is achievable, particularly after methylprednisolone treatment.
Collapse
Affiliation(s)
- Murali K. Yanda
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vartika Tomar
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cristina Valeria Cebotaru
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William B. Guggino
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liudmila Cebotaru
- Department of Medicine and Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Correspondence: Dr. Liudmila Cebotaru, Departments of Medicine and Physiology, Johns Hopkins University School of Medicine, Hunterian 415, 725 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
33
|
Ashby F, Heldermon C. The significance of triple-capsid-mutant AAV8 for treatment of Sanfilippo Syndrome Type B. ARCHIVES OF STEM CELL AND THERAPY 2022; 3:11-17. [PMID: 38596745 PMCID: PMC11003760 DOI: 10.46439/stemcell.3.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Sanfilippo Syndrome Type-B remains an untreatable childhood neurodegenerative disease with great burden for both patient and caregiver. Very few clinical trials have been undertaken to treat the disease, and none of these have yet yielded clinically obtainable products for patients. Caused by a simple enzyme function deficiency, Sanfilippo Syndrome Type-B has been considered a great prospect for gene-therapy interventions. Adeno-associated virus (AAV) remains a major choice for therapeutic gene delivery due to its relatively low-immunogenicity, versatility and tissue tropism. However, many clinical trials with AAV continue to use wild-type capsids, which in many cases are not able to reach stable transgene expression for long enough to be clinically effective in most cases. Previous research in AAV gene-therapy has created a litany of novel AAV capsids that can improve overall transduction efficiency far above that of wild-type AAV capsids. One such example is the triple-capsid mutant AAV8 (TCM8), which has been shown to exhibit transgene expression far superior to other capsids in Sanfilippo mouse models, specifically. Originally designed to bypass capsid ubiquitination intracellularly, mouse studies suggest this TCM8 vector outperforms both AAV5 and AAV9 when delivered to the central nervous system. This implies it as an ideal contender for an effective gene-therapy clinical trial candidate and has the potential to advance the progress of Sanfilippo Syndrome treatment. Here we provide commentary on the TCM8 vector and its context in the field of Sanfilippo Syndrome Type-B research.
Collapse
Affiliation(s)
- Frederick Ashby
- University of Florida, College of Medicine, Gainesville, FL 32610-0278, USA
| | - Coy Heldermon
- University of Florida, College of Medicine, Gainesville, FL 32610-0278, USA
| |
Collapse
|
34
|
Fakhiri J, Grimm D. Best of most possible worlds: Hybrid gene therapy vectors based on parvoviruses and heterologous viruses. Mol Ther 2021; 29:3359-3382. [PMID: 33831556 PMCID: PMC8636155 DOI: 10.1016/j.ymthe.2021.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Parvoviruses and especially the adeno-associated virus (AAV) species provide an exciting and versatile platform for the rational design or molecular evolution of human gene-therapy vectors, documented by literature from over half a century, hundreds of clinical trials, and the recent commercialization of multiple AAV gene therapeutics. For the last three decades, the power of these vectors has been further potentiated through various types of hybrid vectors created by intra- or inter-genus juxtaposition of viral DNA and protein cis elements or by synergistic complementation of parvoviral features with those of heterologous, prokaryotic, or eukaryotic viruses. Here, we provide an overview of the history and promise of this rapidly expanding field of hybrid parvoviral gene-therapy vectors, starting with early generations of chimeric particles composed of a recombinant AAV genome encapsidated in shells of synthetic AAVs or of adeno-, herpes-, baculo-, or protoparvoviruses. We then dedicate our attention to two newer, highly promising types of hybrid vectors created via (1) pseudotyping of AAV genomes with bocaviral serotypes and capsid mutants or (2) packaging of AAV DNA into, or tethering of entire vector particles to, bacteriophages. Finally, we conclude with an outlook summarizing critical requirements and improvements toward clinical translation of these original concepts.
Collapse
Affiliation(s)
- Julia Fakhiri
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Heidelberg, Germany; BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Heidelberg, Germany; BioQuant, University of Heidelberg, Heidelberg, Germany; German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), Partner site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
35
|
Viral Membrane Fusion Proteins and RNA Sorting Mechanisms for the Molecular Delivery by Exosomes. Cells 2021; 10:cells10113043. [PMID: 34831268 PMCID: PMC8622164 DOI: 10.3390/cells10113043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022] Open
Abstract
The advancement of precision medicine critically depends on the robustness and specificity of the carriers used for the targeted delivery of effector molecules in the human body. Numerous nanocarriers have been explored in vivo, to ensure the precise delivery of molecular cargos via tissue-specific targeting, including the endocrine part of the pancreas, thyroid, and adrenal glands. However, even after reaching the target organ, the cargo-carrying vehicle needs to enter the cell and then escape lysosomal destruction. Most artificial nanocarriers suffer from intrinsic limitations that prevent them from completing the specific delivery of the cargo. In this respect, extracellular vesicles (EVs) seem to be the natural tool for payload delivery due to their versatility and low toxicity. However, EV-mediated delivery is not selective and is usually short-ranged. By inserting the viral membrane fusion proteins into exosomes, it is possible to increase the efficiency of membrane recognition and also ease the process of membrane fusion. This review describes the molecular details of the viral-assisted interaction between the target cell and EVs. We also discuss the question of the usability of viral fusion proteins in developing extracellular vesicle-based nanocarriers with a higher efficacy of payload delivery. Finally, this review specifically highlights the role of Gag and RNA binding proteins in RNA sorting into EVs.
Collapse
|
36
|
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked, muscle wasting disease that affects 1 in 5000 males. Affected individuals become wheelchair bound by the age of twelve and eventually die in their third decade due to respiratory and cardiac complications. The disease is caused by mutations in the DMD gene that codes for dystrophin. Dystrophin is a structural protein that maintains the integrity of muscle fibres and protects them from contraction-induced damage. The absence of dystrophin compromises the stability and function of the muscle fibres, eventually leading to muscle degeneration. So far, there is no effective treatment for deteriorating muscle function in DMD patients. A promising approach for treating this life-threatening disease is gene transfer to restore dystrophin expression using a safe, non-pathogenic viral vector called adeno-associated viral (AAV) vector. Whilst microdystrophin gene transfer using AAV vectors shows extremely impressive therapeutic success so far in large animal models of DMD, translating this advanced therapy medicinal product from bench to bedside still offers scope for many optimization steps. In this paper, the authors review the current progress of AAV-microdystrophin gene therapy for DMD and other treatment strategies that may apply to a subset of DMD patients depending on the mutations they carry.
Collapse
Affiliation(s)
- Nertiyan Elangkovan
- Centres for Gene & Cell Therapy and Biomedical Sciences, Department of Biological Sciences, School of Life & Environmental Sciences, Royal Holloway - University of London, Surrey, TW20 0EX, UK
| | - George Dickson
- Centres for Gene & Cell Therapy and Biomedical Sciences, Department of Biological Sciences, School of Life & Environmental Sciences, Royal Holloway - University of London, Surrey, TW20 0EX, UK
| |
Collapse
|
37
|
Gilkes JA, Judkins BL, Herrera BN, Mandel RJ, Boye SL, Boye SE, Srivastava A, Heldermon CD. Site-specific modifications to AAV8 capsid yields enhanced brain transduction in the neonatal MPS IIIB mouse. Gene Ther 2021; 28:447-455. [PMID: 33244179 PMCID: PMC8149485 DOI: 10.1038/s41434-020-00206-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/03/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022]
Abstract
Mucopolysaccharidosis type IIIB (MPS IIIB) is an autosomal recessive lysosomal disease caused by defective production of the enzyme α-N-acetylglucosaminidase. It is characterized by severe and complex central nervous system degeneration. Effective therapies will likely target early onset disease and overcome the blood-brain barrier. Modifications of adeno-associated viral (AAV) vector capsids that enhance transduction efficiency have been described in the retina. Herein, we describe for the first time, a transduction assessment of two intracranially administered adeno-associated virus serotype 8 variants, in which specific surface-exposed tyrosine (Y) and threonine (T) residues were substituted with phenylalanine (F) and valine (V) residues, respectively. A double-mutant (Y444 + 733F) and a triple-mutant (Y444 + 733F + T494V) AAV8 were evaluated for their efficacy for the potential treatment of MPS IIIB in a neonatal setting. We evaluated biodistribution and transduction profiles of both variants compared to the unmodified parental AAV8, and assessed whether the method of vector administration would modulate their utility. Vectors were administered through four intracranial routes: six sites (IC6), thalamic (T), intracerebroventricular, and ventral tegmental area into neonatal mice. Overall, we conclude that the IC6 method resulted in the widest biodistribution within the brain. Noteworthy, we demonstrate that GFP intensity was significantly more robust with AAV8 (double Y-F + T-V) compared to AAV8 (double Y-F). This provides proof of concept for the enhanced utility of IC6 administration of the capsid modified AAV8 (double Y-F + T-V) as a valid therapeutic approach for the treatment of MPS IIIB, with further implications for other monogenic diseases.
Collapse
Affiliation(s)
- Janine A Gilkes
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Benjamin L Judkins
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Brontie N Herrera
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ronald J Mandel
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
| | - Sanford L Boye
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Shannon E Boye
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida College of Medicine, Gainesville, FL, USA
| | - Arun Srivastava
- Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida College of Medicine, Gainesville, FL, USA
| | - Coy D Heldermon
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
38
|
|
39
|
Tang Y, Yan Z, Lin S, Huntemann ED, Feng Z, Park SY, Sun X, Yuen E, Engelhardt JF. Repeat Dosing of AAV2.5T to Ferret Lungs Elicits an Antibody Response That Diminishes Transduction in an Age-Dependent Manner. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:186-200. [PMID: 33209961 PMCID: PMC7648090 DOI: 10.1016/j.omtm.2020.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/11/2020] [Indexed: 11/12/2022]
Abstract
Readministration of recombinant adeno-associated virus (rAAV) may be necessary to treat cystic fibrosis (CF) lung disease using gene therapy. However, little is known about rAAV-mediated immune responses in the lung. Here, we demonstrate the suitability of the ferret for testing AAV2.5T-mediated CFTR delivery to the lung and characterization of neutralizing-antibody (NAb) responses. AAV2.5T-SP183-hCFTRΔR efficiently transduced both human and ferret airway epithelial cultures and complemented CFTR Cl– currents in CF airway cultures. Delivery of AAV2.5T-hCFTRΔR to neonatal and juvenile ferret lungs produced hCFTR mRNA at 200%–300% greater levels than endogenous fCFTR. Single-dose (AAV2.5T-SP183-gLuc) or repeat dosing (AAV2.5T-SP183-fCFTRΔR followed by AAV2.5T-SP183-gLuc) of AAV2.5T was performed in neonatal and juvenile ferrets. Repeat dosing significantly reduced transgene expression (11-fold) and increased bronchoalveolar lavage fluid (BALF) NAbs only in juvenile, but not neonatal, ferrets, despite near-equivalent plasma NAb responses in both age groups. Notably, both age groups demonstrated a reduction in BALF anti-capsid binding immunoglobulin (Ig) G, IgM, and IgA antibodies after repeat dosing. Unique to juvenile ferrets was a suppression of plasma anti-capsid-binding IgM after the second vector administration. Thus, age-dependent immune system maturation and isotype switching may affect the development of high-affinity lung NAbs after repeat dosing of AAV2.5T and may provide a path to blunt AAV-neutralizing responses in the lung.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Ziying Yan
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shen Lin
- Spirovant Science Inc., Philadelphia, PA 19104, USA
| | - Eric D Huntemann
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zehua Feng
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Soo-Yeun Park
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Xingshen Sun
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Eric Yuen
- Spirovant Science Inc., Philadelphia, PA 19104, USA
| | - John F Engelhardt
- Department of Anatomy & Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
40
|
Journey to the Center of the Cell: Tracing the Path of AAV Transduction. Trends Mol Med 2020; 27:172-184. [PMID: 33071047 DOI: 10.1016/j.molmed.2020.09.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 02/08/2023]
Abstract
As adeno-associated virus (AAV)-based gene therapies are being increasingly approved for use in humans, it is important that we understand vector-host interactions in detail. With the advances in genome-wide genetic screening tools, a clear picture of AAV-host interactions is beginning to emerge. Understanding these interactions can provide insights into the viral life cycle. Accordingly, novel strategies to circumvent the current limitations of AAV-based vectors may be explored. Here, we summarize our current understanding of the various stages in the journey of the vector from the cell surface to the nucleus and contextualize the roles of recently identified host factors.
Collapse
|
41
|
Breaking the sound barrier: Towards next-generation AAV vectors for gene therapy of hearing disorders. Hear Res 2020; 413:108092. [PMID: 33268240 DOI: 10.1016/j.heares.2020.108092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/14/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022]
Abstract
Owing to the advances in transgenic animal technology and the advent of the next-generation sequencing era, over 120 genes causing hereditary hearing loss have been identified by now. In parallel, the field of human gene therapy continues to make exciting and rapid progress, culminating in the recent approval of several ex vivo and in vivo applications. Despite these encouraging developments and the growing interest in causative treatments for hearing disorders, gene therapeutic interventions in the inner ear remain in their infancy and await clinical translation. This review focuses on the adeno-associated virus (AAV), which nowadays represents one of the safest and most promising vectors in gene therapy. We first provide an overview of AAV biology and outline the principles of therapeutic gene transfer with recombinant AAV vectors, before pointing out major challenges and solutions for clinical translation including vector manufacturing and species translatability. Finally, we highlight seminal technologies for engineering and selection of next-generation "designer" AAV capsids, and illustrate their power and potential with recent examples of their application for inner ear gene transfer in animals.
Collapse
|
42
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
43
|
Rambhai HK, Ashby FJ, Qing K, Srivastava A. Role of Essential Metal Ions in AAV Vector-Mediated Transduction. Mol Ther Methods Clin Dev 2020; 18:159-166. [PMID: 32637447 PMCID: PMC7321778 DOI: 10.1016/j.omtm.2020.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/19/2020] [Indexed: 12/30/2022]
Abstract
Metal elements are essential components of approximately half of all cellular proteins, and approximately one-third of all known enzymes thus far are metalloenzymes. Several cellular proteins and enzymes undoubtedly impact the transduction efficiency of recombinant adeno-associated virus (AAV) vectors, but the precise role of metal ions in this process has not been studied in detail. In the present studies, we systematically evaluated the effects of all 10 essential metal ions (calcium, cobalt, copper, iron, magnesium, manganese, molybdenum, potassium, sodium, and zinc) on the transduction efficiency of AAV vectors. We report herein that five essential metal ions (iron, magnesium, manganese, molybdenum, and sodium) had little to no effect, and calcium strongly inhibited the transduction efficiency of AAV2 vectors. Whereas copper and potassium increased the transduction efficiency by ∼5-fold and ∼2-fold, respectively, at low concentrations, both essential metals were strongly inhibitory at higher concentrations. Calcium also inhibited the transduction efficiency by ∼3-fold. Two metal ions (cobalt and zinc) increased the transduction efficiency up to ∼10-fold in a dose-dependent manner. The combined use of cobalt and zinc resulted in more than an additive effect on AAV2 vector transduction efficiency (∼30-fold). The transduction efficiency of AAV serotypes 1 through 6 (AAV1-AAV6) vectors was also augmented by zinc. Similarly, the transduction of both single-stranded (ss) and self-complementary (sc) AAV3 vectors was enhanced by zinc. Zinc treatment also led to a dose-dependent increase in expression of a therapeutic protein, the human clotting factor IX (hF.IX), mediated by scAAV3 vectors in a human hepatic cell line. This simple strategy of essential metal ion-mediated enhancement may be useful to lower the dose of AAV vectors for their optimal use in human gene therapy.
Collapse
Affiliation(s)
- Himanshu K. Rambhai
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611-3633, USA
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Frederick J. Ashby
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611-3633, USA
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Keyun Qing
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611-3633, USA
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611-3633, USA
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
44
|
Carneiro A, Lee H, Lin L, van Haasteren J, Schaffer DV. Novel Lung Tropic Adeno-Associated Virus Capsids for Therapeutic Gene Delivery. Hum Gene Ther 2020; 31:996-1009. [PMID: 32799685 DOI: 10.1089/hum.2020.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Efforts to identify mutations that underlie inherited genetic diseases combined with strides in the development of gene therapy vectors over the last three decades have culminated in the approval of several adeno-associated virus (AAV)-based gene therapies. Genetic diseases that manifest in the lung such as cystic fibrosis (CF) and surfactant deficiencies, however, have so far proven to be elusive targets. Early clinical trials in CF using AAV serotype 2 (AAV2) achieved safety, but not efficacy endpoints; however, importantly, these studies provided critical information on barriers that need to be surmounted to translate AAV lung gene therapy toward clinical success. Bolstered with an improved understanding of AAV biology and more clinically relevant lung models, next-generation molecular biology and bioinformatics approaches have given rise to novel AAV capsid variants that offer improvements in transduction efficiency, immunological profile, and the ability to circumvent physical barriers in the lung such as mucus. This review discusses the principal limiting barriers to clinical success in lung gene therapy and focuses on novel engineered AAV capsid variants that have been developed to overcome those challenges.
Collapse
Affiliation(s)
- Ana Carneiro
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA
| | - Hyuncheol Lee
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California, USA
| | - Li Lin
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA
| | - Joost van Haasteren
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California, USA.,Department of Bioengineering, University of California, Berkeley, California, USA.,Department of Molecular and Cell Biology, University of California, Berkeley, California, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA.,Innovative Genomics Institute (IGI), University of California, Berkeley, California, USA
| |
Collapse
|
45
|
King NE, Suzuki S, Barillà C, Hawkins FJ, Randell SH, Reynolds SD, Stripp BR, Davis BR. Correction of Airway Stem Cells: Genome Editing Approaches for the Treatment of Cystic Fibrosis. Hum Gene Ther 2020; 31:956-972. [PMID: 32741223 PMCID: PMC7495916 DOI: 10.1089/hum.2020.160] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/30/2020] [Indexed: 12/26/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease caused by variations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Although CF affects multiple organs, the primary cause of mortality is respiratory failure resulting from poor clearance of hyperviscous secretions and subsequent airway infection. Recently developed CFTR modulators provide significant therapeutic benefit to the majority of CF individuals. However, treatments directed at the underlying cause are needed for the ∼7% of CF patients who are not expected to be responsive to these modulators. Genome editing can restore the native CFTR genetic sequence and function to mutant cells, representing an approach to establish durable physiologic CFTR correction. Although editing the CFTR gene in various airway cell types may transiently restore CFTR activity, effort is focused on editing airway basal stem/progenitor cells, since their correction would allow appropriate and durable expression of CFTR in stem cell-derived epithelial cell types. Substantial progress has been made to directly correct airway basal cells in vitro, theoretically enabling transplantation of autologous corrected cells to regenerate an airway with CFTR functional cells. Another approach to create autologous, gene-edited airway basal cells is derivation of CF donor-specific induced pluripotent stem cells, correction of the CFTR gene, and subsequent directed differentiation to airway basal cells. Further work is needed to translate these advances by developing effective transplantation methods. Alternatively, gene editing in vivo may enable CFTR correction. However, this approach will require robust delivery methods ensuring that basal cells are efficiently targeted and corrected. Recent advances in gene editing-based therapies provide hope that the genetic underpinning of CF can be durably corrected in airway epithelial stem cells, thereby preventing or treating lung disease in all people with CF.
Collapse
Affiliation(s)
- Nicholas E. King
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shingo Suzuki
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Cristina Barillà
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Finn J. Hawkins
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Scott H. Randell
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Susan D. Reynolds
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Barry R. Stripp
- Lung and Regenerative Medicine Institutes, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Brian R. Davis
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
46
|
Reisinger E. Dual-AAV delivery of large gene sequences to the inner ear. Hear Res 2020; 394:107857. [DOI: 10.1016/j.heares.2019.107857] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/22/2019] [Indexed: 01/06/2023]
|
47
|
Tang Y, Yan Z, Engelhardt JF. Viral Vectors, Animal Models, and Cellular Targets for Gene Therapy of Cystic Fibrosis Lung Disease. Hum Gene Ther 2020; 31:524-537. [PMID: 32138545 PMCID: PMC7232698 DOI: 10.1089/hum.2020.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
After more than two decades since clinical trials tested the first use of recombinant adeno-associated virus (rAAV) to treat cystic fibrosis (CF) lung disease, gene therapy for this disorder has undergone a tremendous resurgence. Fueling this enthusiasm has been an enhanced understanding of rAAV transduction biology and cellular processes that limit transduction of airway epithelia, the development of new rAAV serotypes and other vector systems with high-level tropism for airway epithelial cells, an improved understanding of CF lung pathogenesis and the cellular targets for gene therapy, and the development of new animal models that reproduce the human CF disease phenotype. These advances have created a preclinical path for both assessing the efficacy of gene therapies in the CF lung and interrogating the target cell types in the lung required for complementation of the CF disease state. Lessons learned from early gene therapy attempts with rAAV in the CF lung have guided thinking for the testing of next-generation vector systems. Although unknown questions still remain regarding the cellular targets in the lung that are required or sufficient to complement CF lung disease, the field is now well positioned to tackle these challenges. This review will highlight the role that next-generation CF animal models are playing in the preclinical development of gene therapies for CF lung disease and the knowledge gaps in disease pathophysiology that these models are attempting to fill.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
48
|
Shirley JL, Keeler GD, Sherman A, Zolotukhin I, Markusic DM, Hoffman BE, Morel LM, Wallet MA, Terhorst C, Herzog RW. Type I IFN Sensing by cDCs and CD4 + T Cell Help Are Both Requisite for Cross-Priming of AAV Capsid-Specific CD8 + T Cells. Mol Ther 2020; 28:758-770. [PMID: 31780366 PMCID: PMC7054715 DOI: 10.1016/j.ymthe.2019.11.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/23/2022] Open
Abstract
Adeno-associated virus (AAV) vectors are widely used in clinical gene therapy to correct genetic disease by in vivo gene transfer. Although the vectors are useful, in part because of their limited immunogenicity, immune responses directed at vector components have complicated applications in humans. These include, for instance, innate immune sensing of vector components by plasmacytoid dendritic cells (pDCs), which sense the vector DNA genome via Toll-like receptor 9. Adaptive immune responses employ antigen presentation by conventional dendritic cells (cDCs), which leads to cross-priming of capsid-specific CD8+ T cells. In this study, we sought to determine the mechanisms that promote licensing of cDCs, which is requisite for CD8+ T cell activation. Blockage of type 1 interferon (T1 IFN) signaling by monoclonal antibody therapy prevented cross-priming. Furthermore, experiments in cell-type-restricted knockout mice showed a specific requirement for the receptor for T1 IFN (IFNaR) in cDCs. In contrast, natural killer (NK) cells are not needed, indicating a direct rather than indirect effect of T1 IFN on cDCs. In addition, co-stimulation by CD4+ T cells via CD40-CD40L was required for cross-priming, and blockage of co-stimulation but not of T1 IFN additionally reduced antibody formation against capsid. These mechanistic insights inform the development of targeted immune interventions.
Collapse
Affiliation(s)
- Jamie L Shirley
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | | | | | - Irene Zolotukhin
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | - David M Markusic
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brad E Hoffman
- Department Pediatrics, University of Florida, Gainesville, FL, USA
| | - Laurence M Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Mark A Wallet
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Roland W Herzog
- Department Pediatrics, University of Florida, Gainesville, FL, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
49
|
Nidetz NF, McGee MC, Tse LV, Li C, Cong L, Li Y, Huang W. Adeno-associated viral vector-mediated immune responses: Understanding barriers to gene delivery. Pharmacol Ther 2019; 207:107453. [PMID: 31836454 DOI: 10.1016/j.pharmthera.2019.107453] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023]
Abstract
Adeno-associated viral (AAV) vectors have emerged as the leading gene delivery platform for gene therapy and vaccination. Three AAV-based gene therapy drugs, Glybera, LUXTURNA, and ZOLGENSMA were approved between 2012 and 2019 by the European Medicines Agency and the United States Food and Drug Administration as treatments for genetic diseases hereditary lipoprotein lipase deficiency (LPLD), inherited retinal disease (IRD), and spinal muscular atrophy (SMA), respectively. Despite these therapeutic successes, clinical trials have demonstrated that host anti-viral immune responses can prevent the long-term gene expression of AAV vector-encoded genes. Therefore, it is critical that we understand the complex relationship between AAV vectors and the host immune response. This knowledge could allow for the rational design of optimized gene transfer vectors capable of either subverting host immune responses in the context of gene therapy applications, or stimulating desirable immune responses that generate protective immunity in vaccine applications to AAV vector-encoded antigens. This review provides an overview of our current understanding of the AAV-induced immune response and discusses potential strategies by which these responses can be manipulated to improve AAV vector-mediated gene transfer.
Collapse
Affiliation(s)
- Natalie F Nidetz
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michael C McGee
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Longping V Tse
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Le Cong
- Department of Pathology and Department of Genetics, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Yunxing Li
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
50
|
Yan Z, McCray Jr PB, Engelhardt JF. Advances in gene therapy for cystic fibrosis lung disease. Hum Mol Genet 2019; 28:R88-R94. [PMID: 31332440 PMCID: PMC6796993 DOI: 10.1093/hmg/ddz139] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/27/2022] Open
Abstract
Cystic fibrosis (CF) is a multiorgan recessive genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Gene therapy efforts have focused on treating the lung, since it manifests the most significant life-threatening disease. Over two decades have past since the first CF lung gene therapy trials and significant advances in the therapeutic implementation of pharmacologic CFTR modulators have renewed the field's focus on developing gene therapies for the 10% of CF patients these modulators cannot help. This review summarizes recent progress made in developing vectors for airway transduction and CF animal models required for understanding the relevant cellular targets in the lung and testing the efficacy of gene therapy approaches. We also highlight future opportunities in emerging gene editing strategies that may offer advantages for treating diseases like CF where the gene target is highly regulated at the cellular level. The outcomes of CF lung gene therapy trials will likely inform productive paths toward gene therapy for other complex genetic disorders, while also advancing treatments for all CF patients.
Collapse
Affiliation(s)
- Ziying Yan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Paul B McCray Jr
- Department of Pediatrics, Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|