1
|
Cooney A, Loza L, Najdawi K, Brommel C, McCray P, Sinn P. High ionic strength vector formulations enhance gene transfer to airway epithelia. Nucleic Acids Res 2024; 52:9369-9383. [PMID: 39077931 PMCID: PMC11381324 DOI: 10.1093/nar/gkae640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024] Open
Abstract
A fundamental challenge for cystic fibrosis (CF) gene therapy is ensuring sufficient transduction of airway epithelia to achieve therapeutic correction. Hypertonic saline (HTS) is frequently administered to people with CF to enhance mucus clearance. HTS transiently disrupts epithelial cell tight junctions, but its ability to improve gene transfer has not been investigated. Here, we asked if increasing the concentration of NaCl enhances the transduction efficiency of three gene therapy vectors: adenovirus, AAV, and lentiviral vectors. Vectors formulated with 3-7% NaCl exhibited markedly increased transduction for all three platforms, leading to anion channel correction in primary cultures of human CF epithelial cells and enhanced gene transfer in mouse and pig airways in vivo. The mechanism of transduction enhancement involved tonicity but not osmolarity or pH. Formulating vectors with a high ionic strength solution is a simple strategy to greatly enhance efficacy and immediately improve preclinical or clinical applications.
Collapse
Affiliation(s)
- Ashley L Cooney
- University of Iowa, Stead Family Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Pappajohn Biomedical Institute; Iowa City, IA 52242, USA
| | - Laura Marquez Loza
- University of Iowa, Stead Family Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Pappajohn Biomedical Institute; Iowa City, IA 52242, USA
| | - Kenan Najdawi
- University of Iowa, Stead Family Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Pappajohn Biomedical Institute; Iowa City, IA 52242, USA
| | - Christian M Brommel
- University of Iowa, Stead Family Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Pappajohn Biomedical Institute; Iowa City, IA 52242, USA
| | - Paul B McCray
- University of Iowa, Stead Family Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Pappajohn Biomedical Institute; Iowa City, IA 52242, USA
- University of Iowa, Center for Gene Therapy; Iowa City, IA 52242, USA
| | - Patrick L Sinn
- University of Iowa, Stead Family Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Pappajohn Biomedical Institute; Iowa City, IA 52242, USA
- University of Iowa, Center for Gene Therapy; Iowa City, IA 52242, USA
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Pedro De-la-Torre, Wen H, Brower J, Martínez-Pérez K, Narui Y, Yeh F, Hale E, Ivanchenko MV, Corey DP, Sotomayor M, Indzhykulian AA. Elasticity and Thermal Stability are Key Determinants of Hearing Rescue by Mini-Protocadherin-15 Proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.16.599132. [PMID: 38948700 PMCID: PMC11212938 DOI: 10.1101/2024.06.16.599132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Protocadherin-15 is a core protein component of inner-ear hair-cell tip links pulling on transduction channels essential for hearing and balance. Protocadherin-15 defects can result in non-syndromic deafness or Usher syndrome type 1F (USH1F) with hearing loss, balance deficits, and progressive blindness. Three rationally engineered shortened versions of protocadherin-15 (mini-PCDH15s) amenable for gene therapy have been used to rescue function in USH1F mouse models. Two can successfully or partially rescue hearing, while another one fails. Here we show that despite varying levels of hearing rescue, all three mini-PCDH15 versions can rescue hair-cell mechanotransduction. Negative-stain electron microscopy shows that all three versions form dimers like the wild-type protein, while crystal structures of some engineered fragments show that these can properly fold and bind calcium ions essential for function. In contrast, simulations predict distinct elasticities and nano differential scanning fluorimetry shows differences in melting temperature measurements. Our data suggest that elasticity and thermal stability are key determinants of sustained hearing rescue by mini-PCDH15s.
Collapse
Affiliation(s)
- Pedro De-la-Torre
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Massachusetts Eye and Ear, 243 Charles St, Boston, MA, USA
| | - Haosheng Wen
- Department of Chemistry and Biochemistry, The Ohio State University, 484 W. 12th Avenue, Columbus, OH, USA
- Biophysics Program, The Ohio State University, 484 W. 12th Avenue, Columbus, OH, USA
| | - Joseph Brower
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Massachusetts Eye and Ear, 243 Charles St, Boston, MA, USA
| | - Karina Martínez-Pérez
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Massachusetts Eye and Ear, 243 Charles St, Boston, MA, USA
- Biology Program, Department of Basic Sciences, Universidad del Atlántico, Cra 30 # 8-49, Puerto Colombia, 081007, Colombia
| | - Yoshie Narui
- Center for Electron Microscopy and Analysis, The Ohio State University, 1275-1305 Kinnear Road, Columbus, OH, USA
| | - Frank Yeh
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Massachusetts Eye and Ear, 243 Charles St, Boston, MA, USA
| | - Evan Hale
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Massachusetts Eye and Ear, 243 Charles St, Boston, MA, USA
- Speech and Hearing Biosciences and Technology graduate program, Harvard University, Cambridge, MA, USA
| | - Maryna V. Ivanchenko
- Department of Neurobiology, Harvard Medical School, 200 Longwood Ave, Boston, MA, USA
| | - David P. Corey
- Department of Neurobiology, Harvard Medical School, 200 Longwood Ave, Boston, MA, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, 484 W. 12th Avenue, Columbus, OH, USA
- Biophysics Program, The Ohio State University, 484 W. 12th Avenue, Columbus, OH, USA
| | - Artur A. Indzhykulian
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Massachusetts Eye and Ear, 243 Charles St, Boston, MA, USA
- Speech and Hearing Biosciences and Technology graduate program, Harvard University, Cambridge, MA, USA
| |
Collapse
|
5
|
Tang Y, Ebadi M, Lei J, Feng Z, Fakhari S, Wu P, Smith MD, Limberis MP, Kolbeck R, Excoffon KJ, Yan Z, Engelhardt JF. Durable transgene expression and efficient re-administration after rAAV2.5T-mediated fCFTRΔR gene delivery to adult ferret lungs. Mol Ther Methods Clin Dev 2024; 32:101244. [PMID: 38638546 PMCID: PMC11024656 DOI: 10.1016/j.omtm.2024.101244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/26/2024] [Indexed: 04/20/2024]
Abstract
The dosing interval for effective recombinant adeno-associated virus (rAAV)-mediated gene therapy of cystic fibrosis lung disease remains unknown. Here, we assessed the durability of rAAV2.5T-fCFTRΔR-mediated transgene expression and neutralizing antibody (NAb) responses in lungs of adult wild-type ferrets. Within the first 3 months following rAAV2.5T-fCFTRΔR delivery to the lung, CFTRΔR transgene expression declined ∼5.6-fold and then remained stable to 5 months at ∼26% the level of endogenous CFTR. rAAV NAbs in the plasma and bronchoalveolar lavage fluid (BALF) peaked at 21 days, coinciding with peak ELISpot T cell responses to AAV capsid peptides, after which both responses declined and remained stable at 4-5 months post dosing. Administration of reporter vector rAAV2.5T-gLuc (gaussia luciferase) at 5 months following rAAV2.5T-fCFTRΔR dosing gave rise to similar levels of gLuc expression in the BALF as observed in age-matched reporter-only controls, demonstrating that residual BALF NAbs were functionally insignificant. Notably, the second vector administration led to a 2.6-fold greater ELISpot T cell response and ∼2.3-fold decline in fCFTRΔR mRNA and vector genomes derived from the initial rAAV2.5T-fCFTRΔR administration, suggesting selective destruction of transduced cells from the first vector dose. These findings provide insights into humoral and cellular immune response to rAAV that may be useful for optimizing gene therapy to the cystic fibrosis lung.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Mehrnoosh Ebadi
- 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
| | - Zehua Feng
- 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
| | - Peipei Wu
- 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
| | - John F. Engelhardt
- Department of Anatomy & Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
6
|
Cooney AL, Loza LM, Najdawi K, Brommel CM, McCray PB, Sinn PL. High ionic strength vector formulations enhance gene transfer to airway epithelia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576687. [PMID: 38328187 PMCID: PMC10849541 DOI: 10.1101/2024.01.22.576687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
A fundamental challenge for cystic fibrosis (CF) gene therapy is ensuring sufficient transduction of airway epithelia to achieve therapeutic correction. Hypertonic saline (HTS) is frequently administered to people with CF to enhance mucus clearance. HTS transiently disrupts epithelial cell tight junctions, but its ability to improve gene transfer has not been investigated. Here we asked if increasing the concentration of NaCl enhances the transduction efficiency of three gene therapy vectors: adenovirus, AAV, and lentiviral vectors. Vectors formulated with 3-7% NaCl exhibited markedly increased transduction for all three platforms, leading to anion channel correction in primary cultures of human CF epithelial cells and enhanced gene transfer in mouse and pig airways in vivo. The mechanism of transduction enhancement involved tonicity but not osmolarity or pH. Formulating vectors with a high ionic strength solution is a simple strategy to greatly enhance efficacy and immediately improve preclinical or clinical applications.
Collapse
Affiliation(s)
- Ashley L. Cooney
- University of Iowa, Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Center for Cystic Fibrosis Gene Therapy; Iowa City, IA 52242, USA
| | - Laura Marquez Loza
- University of Iowa, Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Center for Cystic Fibrosis Gene Therapy; Iowa City, IA 52242, USA
| | - Kenan Najdawi
- University of Iowa, Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Center for Cystic Fibrosis Gene Therapy; Iowa City, IA 52242, USA
| | - Christian M. Brommel
- University of Iowa, Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Center for Cystic Fibrosis Gene Therapy; Iowa City, IA 52242, USA
| | - Paul B. McCray
- University of Iowa, Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Center for Cystic Fibrosis Gene Therapy; Iowa City, IA 52242, USA
- University of Iowa, Department of Microbiology and Immunology, Iowa City, IA 52242, USA
| | - Patrick L. Sinn
- University of Iowa, Department of Pediatrics; Iowa City, IA 52242, USA
- University of Iowa, Center for Cystic Fibrosis Gene Therapy; Iowa City, IA 52242, USA
- University of Iowa, Department of Microbiology and Immunology, Iowa City, IA 52242, USA
| |
Collapse
|
7
|
Cooney AL, Brommel CM, Traore S, Newby GA, Liu DR, McCray PB, Sinn PL. Reciprocal mutations of lung-tropic AAV capsids lead to improved transduction properties. Front Genome Ed 2023; 5:1271813. [PMID: 38077224 PMCID: PMC10702583 DOI: 10.3389/fgeed.2023.1271813] [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: 08/02/2023] [Accepted: 10/17/2023] [Indexed: 12/21/2023] Open
Abstract
Considerable effort has been devoted to developing adeno-associated virus (AAV)-based vectors for gene therapy in cystic fibrosis (CF). As a result of directed evolution and capsid shuffling technology, AAV capsids are available with widespread tropism for airway epithelial cells. For example, AAV2.5T and AAV6.2 are two evolved capsids with improved airway epithelial cell transduction properties over their parental serotypes. However, limited research has been focused on identifying their specific cellular tropism. Restoring cystic fibrosis transmembrane conductance regulator (CFTR) expression in surface columnar epithelial cells is necessary for the correction of the CF airway phenotype. Basal cells are a progenitor population of the conducting airways responsible for replenishing surface epithelial cells (including secretory cells and ionocytes), making correction of this cell population vital for a long-lived gene therapy strategy. In this study, we investigate the tropism of AAV capsids for three cell types in primary cultures of well-differentiated human airway epithelial (HAE) cells and primary human airway basal cells. We observed that AAV2.5T transduced surface epithelial cells better than AAV6.2, while AAV6.2 transduced airway basal cells better than AAV2.5T. We also investigated a recently developed capsid, AAV6.2FF, which has two surface tyrosines converted to phenylalanines. Next, we incorporated reciprocal mutations to create AAV capsids with further improved surface and basal cell transduction characteristics. Lastly, we successfully employed a split-intein approach using AAV to deliver an adenine base editor (ABE) to repair the CFTR R553X mutation. Our results suggest that rational incorporation of AAV capsid mutations improves AAV transduction of the airway surface and progenitor cells and may ultimately lead to improved pulmonary function in people with CF.
Collapse
Affiliation(s)
- Ashley L. Cooney
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| | - Christian M. Brommel
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| | - Soumba Traore
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| | - Gregory A. Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, United States
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, United States
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, United States
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, United States
| | - Paul B. McCray
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| | - Patrick L. Sinn
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
8
|
Cai Q, Luo M, Tang Y, Yu M, Yuan F, Gasser GN, Liu X, Engelhardt JF. Sonic Hedgehog Signaling Is Essential for Pulmonary Ionocyte Specification in Human and Ferret Airway Epithelia. Am J Respir Cell Mol Biol 2023; 69:295-309. [PMID: 37141531 PMCID: PMC10503308 DOI: 10.1165/rcmb.2022-0280oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 05/03/2023] [Indexed: 05/06/2023] Open
Abstract
Pulmonary ionocytes express high levels of cystic fibrosis transmembrane conductance regulator (CFTR), an anion channel that is critical for hydration of the airways and mucociliary clearance. However, the cellular mechanisms that govern ionocyte specification and function remain unclear. We observed that increased abundance of ionocytes in cystic fibrosis (CF) airway epithelium was associated with enhanced expression of Sonic Hedgehog (SHH) effectors. In this study, we evaluated whether the SHH pathway directly impacts ionocyte differentiation and CFTR function in airway epithelia. Pharmacological HPI1-mediated inhibition of SHH signaling component GLI1 significantly impaired human basal cell specification of ionocytes and ciliated cells but significantly enhanced specification of secretory cells. By contrast, activation of the SHH pathway effector smoothened (SMO) with the chemical agonist SAG significantly enhanced ionocyte specification. The abundance of CFTR+ BSND+ ionocytes under these conditions had a direct relationship with CFTR-mediated currents in differentiated air-liquid interface (ALI) airway cultures. These findings were corroborated in ferret ALI airway cultures generated from basal cells in which the genes encoding the SHH receptor PTCH1 or its intracellular effector SMO were genetically ablated using CRISPR-Cas9, causing aberrant activation or suppression of SHH signaling, respectively. These findings demonstrate that SHH signaling is directly involved in airway basal cell specification of CFTR-expressing pulmonary ionocytes and is likely responsible for enhanced ionocyte abundance in the CF proximal airways. Pharmacologic approaches to enhance ionocyte and reduce secretory cell specification after CFTR gene editing of basal cells may have utility in the treatment of CF.
Collapse
Affiliation(s)
- Qian Cai
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China; and
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China
| | - Meihui Luo
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Yinghua Tang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Miao Yu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China; and
| | - Feng Yuan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Grace N. Gasser
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Xiaoming Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
9
|
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: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|
10
|
Yip M, Chen J, Zhi Y, Tran NT, Namkung S, Pastor E, Gao G, Tai PWL. Querying Recombination Junctions of Replication-Competent Adeno-Associated Viruses in Gene Therapy Vector Preparations with Single Molecule, Real-Time Sequencing. Viruses 2023; 15:1228. [PMID: 37376529 DOI: 10.3390/v15061228] [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: 04/20/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 06/29/2023] Open
Abstract
Clinical-grade preparations of adeno-associated virus (AAV) vectors used for gene therapy typically undergo a series of diagnostics to determine titer, purity, homogeneity, and the presence of DNA contaminants. One type of contaminant that remains poorly investigated is replication-competent (rc)AAVs. rcAAVs form through recombination of DNA originating from production materials, yielding intact, replicative, and potentially infectious virus-like virions. They can be detected through the serial passaging of lysates from cells transduced by AAV vectors in the presence of wildtype adenovirus. Cellular lysates from the last passage are subjected to qPCR to detect the presence of the rep gene. Unfortunately, the method cannot be used to query the diversity of recombination events, nor can qPCR provide insights into how rcAAVs arise. Thus, the formation of rcAAVs through errant recombination events between ITR-flanked gene of interest (GOI) constructs and expression constructs carrying the rep-cap genes is poorly described. We have used single molecule, real-time sequencing (SMRT) to analyze virus-like genomes expanded from rcAAV-positive vector preparations. We present evidence that sequence-independent and non-homologous recombination between the ITR-bearing transgene and the rep/cap plasmid occurs under several events and rcAAVs spawn from diverse clones.
Collapse
Affiliation(s)
- Mitchell Yip
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jing Chen
- Spirovant Sciences, Inc., Philadelphia, PA 19104, USA
| | - Yan Zhi
- Spirovant Sciences, Inc., Philadelphia, PA 19104, USA
| | - Ngoc Tam Tran
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Suk Namkung
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Eric Pastor
- Spirovant Sciences, Inc., Philadelphia, PA 19104, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
- Li Weibo Institute of Rare Diseases Research, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
- Li Weibo Institute of Rare Diseases Research, UMass Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
11
|
Lomunova MA, Gershovich PM. Gene Therapy for Cystic Fibrosis: Recent Advances and Future Prospects. Acta Naturae 2023; 15:20-31. [PMID: 37538805 PMCID: PMC10395777 DOI: 10.32607/actanaturae.11708] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/22/2023] [Indexed: 08/05/2023] Open
Abstract
Gene replacement therapies are novel therapeutic approaches that seek to tackle hereditary diseases caused by a congenital deficiency in a particular gene, when a functional copy of a gene can be delivered to the cells and tissues using various delivery systems. To do this, viral particles carrying a functional copy of the gene of interest and various nonviral gene delivery systems, including liposomes, nanoparticles, etc., can be used. In this review, we discuss the state of current knowledge regarding the molecular mechanisms and types of genetic mutations that lead to cystic fibrosis and highlight recent developments in gene therapy that can be leveraged to correct these mutations and to restore the physiological function of the carrier protein transporting sodium and chlorine ions in the airway epithelial cells. Restoration of carrier protein expression could lead to the normalization of ion and water transport across the membrane and induce a decrease in the viscosity of airway surface fluid, which is one of the pathological manifestations of this disease. This review also summarizes recently published preclinical and clinical data for various gene therapies to allow one to make some conclusions about future prospects for gene therapy in cystic fibrosis treatment.
Collapse
|
12
|
Shitik EM, Shalik IK, Yudkin DV. AAV- based vector improvements unrelated to capsid protein modification. Front Med (Lausanne) 2023; 10:1106085. [PMID: 36817775 PMCID: PMC9935841 DOI: 10.3389/fmed.2023.1106085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) is the leading platform for delivering genetic constructs in vivo. To date, three AAV-based gene therapeutic agents have been approved by the FDA and are used in clinical practice. Despite the distinct advantages of gene therapy development, it is clear that AAV vectors need to be improved. Enhancements in viral vectors are mainly associated with capsid protein modifications. However, there are other structures that significantly affect the AAV life cycle and transduction. The Rep proteins, in combination with inverted terminal repeats (ITRs), determine viral genome replication, encapsidation, etc. Moreover, transgene cassette expression in recombinant variants is directly related to AAV production and transduction efficiency. This review discusses the ways to improve AAV vectors by modifying ITRs, a transgene cassette, and the Rep proteins.
Collapse
|
13
|
Abstract
Cystic fibrosis (CF) is a multiorgan disease caused by a wide variety of mutations in the cystic fibrosis transmembrane conductance regulator gene. As treatment has progressed from symptom mitigation to targeting of specific molecular defects, genetics has played an important role in identifying the proper precision therapies for each individual. Novel therapeutic approaches are focused on expanding treatment to a greater number of individuals as well as working toward a cure. This review discusses the role of genetics in our understanding of CF with a particular emphasis on how genetics informs the exciting landscape of current and novel CF therapies.
Collapse
Affiliation(s)
- Anya T Joynt
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Garry R Cutting
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Neeraj Sharma
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
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: 9] [Impact Index Per Article: 3.0] [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
|
15
|
Mollard A, Peccate C, Forand A, Chassagne J, Julien L, Meunier P, Guesmia Z, Marais T, Bitoun M, Piétri-Rouxel F, Benkhelifa-Ziyyat S, Lorain S. Muscle regeneration affects Adeno Associated Virus 1 mediated transgene transcription. Sci Rep 2022; 12:9674. [PMID: 35690627 PMCID: PMC9188557 DOI: 10.1038/s41598-022-13405-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022] Open
Abstract
Duchenne muscular dystrophy is a severe neuromuscular disease causing a progressive muscle wasting due to mutations in the DMD gene that lead to the absence of dystrophin protein. Adeno-associated virus (AAV)-based therapies aiming to restore dystrophin in muscles, by either exon skipping or microdystrophin expression, are very promising. However, the absence of dystrophin induces cellular perturbations that hinder AAV therapy efficiency. We focused here on the impact of the necrosis-regeneration process leading to nuclear centralization in myofiber, a common feature of human myopathies, on AAV transduction efficiency. We generated centronucleated myofibers by cardiotoxin injection in wild-type muscles prior to AAV injection. Intramuscular injections of AAV1 vectors show that transgene expression was drastically reduced in regenerated muscles, even when the AAV injection occurred 10 months post-regeneration. We show also that AAV genomes were not lost from cardiotoxin regenerated muscle and were properly localised in the myofiber nuclei but were less transcribed leading to muscle transduction defect. A similar defect was observed in muscles of the DMD mouse model mdx. Therefore, the regeneration process per se could participate to the AAV-mediated transduction defect observed in dystrophic muscles which may limit AAV-based therapies.
Collapse
Affiliation(s)
- Amédée Mollard
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Cécile Peccate
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Anne Forand
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Julie Chassagne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Laura Julien
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Pierre Meunier
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Zoheir Guesmia
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Thibaut Marais
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Marc Bitoun
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - France Piétri-Rouxel
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Sofia Benkhelifa-Ziyyat
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France.
| | - Stéphanie Lorain
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France.,AFM-Téléthon, 1 rue de l'Internationale, BP59, 91002, Evry, France
| |
Collapse
|
16
|
Effective viral-mediated lung gene therapy: is airway surface preparation necessary? Gene Ther 2022:10.1038/s41434-022-00332-7. [DOI: 10.1038/s41434-022-00332-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022]
Abstract
AbstractGene-based therapeutics are actively being pursued for the treatment of lung diseases. While promising advances have been made over the last decades, the absence of clinically available lung-directed genetic therapies highlights the difficulties associated with this effort. Largely, progress has been hindered by the presence of inherent physical and physiological airway barriers that significantly reduce the efficacy of gene transfer. These barriers include surface mucus, mucociliary action, cell-to-cell tight junctions, and the basolateral cell membrane location of viral receptors for many commonly used gene vectors. Accordingly, airway surface preparation methods have been developed to disrupt these barriers, creating a more conducive environment for gene uptake into the target airway cells. The two major approaches have been chemical and physical methods. Both have proven effective for increasing viral-mediated gene transfer pre-clinically, although with variable effect depending on the specific strategy employed. While such methods have been explored extensively in experimental settings, they have not been used clinically. This review covers the airway surface preparation strategies reported in the literature, the advantages and disadvantages of each method, as well as a discussion about applying this concept in the clinic.
Collapse
|
17
|
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
|
18
|
Cooney AL, Wambach JA, Sinn PL, McCray PB. Gene Therapy Potential for Genetic Disorders of Surfactant Dysfunction. Front Genome Ed 2022; 3:785829. [PMID: 35098209 PMCID: PMC8798122 DOI: 10.3389/fgeed.2021.785829] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary surfactant is critically important to prevent atelectasis by lowering the surface tension of the alveolar lining liquid. While respiratory distress syndrome (RDS) is common in premature infants, severe RDS in term and late preterm infants suggests an underlying genetic etiology. Pathogenic variants in the genes encoding key components of pulmonary surfactant including surfactant protein B (SP-B, SFTPB gene), surfactant protein C (SP-C, SFTPC gene), and the ATP-Binding Cassette transporter A3 (ABCA3, ABCA3 gene) result in severe neonatal RDS or childhood interstitial lung disease (chILD). These proteins play essential roles in pulmonary surfactant biogenesis and are expressed in alveolar epithelial type II cells (AEC2), the progenitor cell of the alveolar epithelium. SP-B deficiency most commonly presents in the neonatal period with severe RDS and requires lung transplantation for survival. SFTPC mutations act in an autosomal dominant fashion and more commonly presents with chILD or idiopathic pulmonary fibrosis than neonatal RDS. ABCA3 deficiency often presents as neonatal RDS or chILD. Gene therapy is a promising option to treat monogenic lung diseases. Successes and challenges in developing gene therapies for genetic disorders of surfactant dysfunction include viral vector design and tropism for target cell types. In this review, we explore adeno-associated virus (AAV), lentiviral, and adenoviral (Ad)-based vectors as delivery vehicles. Both gene addition and gene editing strategies are compared to best design treatments for lung diseases resulting from pathogenic variants in the SFTPB, SFTPC, and ABCA3 genes.
Collapse
Affiliation(s)
- Ashley L. Cooney
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
- *Correspondence: Ashley L. Cooney,
| | - Jennifer A. Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Patrick L. Sinn
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
| | - Paul B. McCray
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
| |
Collapse
|
19
|
Lopez-Gordo E, Orlowski A, Wang A, Weinberg A, Sahoo S, Weber T. Hydroxylation of N-acetylneuraminic Acid Influences the in vivo Tropism of N-linked Sialic Acid-Binding Adeno-Associated Viruses AAV1, AAV5, and AAV6. Front Med (Lausanne) 2021; 8:732095. [PMID: 35036407 PMCID: PMC8757481 DOI: 10.3389/fmed.2021.732095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated virus (AAV) vectors are promising candidates for gene therapy. However, a number of recent preclinical large animal studies failed to translate into the clinic. This illustrates the formidable challenge of choosing the animal models that promise the best chance of a successful translation into the clinic. Several of the most common AAV serotypes use sialic acid (SIA) as their primary receptor. However, in contrast to most mammals, humans lack the enzyme CMAH, which hydroxylates cytidine monophosphate-N-acetylneuraminic acid (CMP-Neu5Ac) into cytidine monophosphate-N-glycolylneuraminic acid (CMP-Neu5Gc). As a result, human glycans only contain Neu5Ac and not Neu5Gc. Here, we investigate the tropism of AAV1, 5, 6 and 9 in wild-type C57BL/6J (WT) and CMAH knock-out (CMAH−/−) mice. All N-linked SIA-binding serotypes (AAV1, 5 and 6) showed significantly lower transduction of the heart in CMAH−/− when compared to WT mice (5–5.8-fold) and, strikingly, skeletal muscle transduction by AAV5 was almost 30-fold higher in CMAH−/− compared to WT mice. Importantly, the AAV tropism or distribution of expression among different organs was also affected. For AAV1, AAV5 and AAV6, expression in the heart compared to the liver was 4.6–8-fold higher in WT than in CMAH−/− mice, and for AAV5 the expression in the heart compared to the skeletal muscle was 57.3-fold higher in WT than in CMAH−/− mice. These data thus strongly suggest that the relative abundance of Neu5Ac and Neu5Gc plays a role in AAV tropism, and that results obtained in commonly used animal models might not translate into the clinic.
Collapse
Affiliation(s)
- Estrella Lopez-Gordo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Alejandro Orlowski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Arthur Wang
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Alan Weinberg
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Thomas Weber
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- *Correspondence: Thomas Weber
| |
Collapse
|
20
|
Mnyandu N, Limani SW, Arbuthnot P, Maepa MB. Advances in designing Adeno-associated viral vectors for development of anti-HBV gene therapeutics. Virol J 2021; 18:247. [PMID: 34903258 PMCID: PMC8670254 DOI: 10.1186/s12985-021-01715-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/26/2021] [Indexed: 12/25/2022] Open
Abstract
Despite the five decades having passed since discovery of the hepatitis B virus (HBV), together with development of an effective anti-HBV vaccine, infection with the virus remains a serious public health problem and results in nearly 900,000 annual deaths worldwide. Current therapies do not eliminate the virus and viral replication typically reactivates after treatment withdrawal. Hence, current endeavours are aimed at developing novel therapies to achieve a functional cure. Nucleic acid-based therapeutic approaches are promising, with several candidates showing excellent potencies in preclinical and early stages of clinical development. However, this class of therapeutics is yet to become part of standard anti-HBV treatment regimens. Obstacles delaying development of gene-based therapies include lack of clinically relevant delivery methods and a paucity of good animal models for preclinical characterisation. Recent studies have demonstrated safety and efficiency of Adeno-associated viral vectors (AAVs) in gene therapy. However, AAVs do have flaws and this has prompted research aimed at improving design of novel and artificially synthesised AAVs. Main goals are to improve liver transduction efficiencies and avoiding immune clearance. Application of AAVs to model HBV replication in vivo is also useful for characterising anti-HBV gene therapeutics. This review summarises recent advances in AAV engineering and their contributions to progress with anti-HBV gene therapy development.
Collapse
Affiliation(s)
- Njabulo Mnyandu
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shonisani Wendy Limani
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mohube Betty Maepa
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
21
|
Croze RH, Kotterman M, Burns CH, Schmitt CE, Quezada M, Schaffer D, Kirn D, Francis P. Viral Vector Technologies and Strategies: Improving on Nature. Int Ophthalmol Clin 2021; 61:59-89. [PMID: 34196318 PMCID: PMC8253506 DOI: 10.1097/iio.0000000000000361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
22
|
Tissue and cell-type-specific transduction using rAAV vectors in lung diseases. J Mol Med (Berl) 2021; 99:1057-1071. [PMID: 34021360 DOI: 10.1007/s00109-021-02086-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Gene therapy of genetically determined diseases, including some pathologies of the respiratory system, requires an efficient method for transgene delivery. Recombinant adeno-associated viral (rAAV) vectors are well studied and employed in gene therapy, as they are relatively simple and low immunogenic and able to efficiently transduce eukaryotic cells. To date, many natural and artificial (with modified capsids) AAV serotypes have been isolated, demonstrating preferential tropism toward different tissues and cells in accordance with the prevalent receptors on the cell surface. However, rAAV-mediated delivery is not strictly specific due to wide tropism of some viral serotypes. Thus, the development of the methods allowing modulating specificity of these vectors could be beneficial in some cases. This review describes various approaches for retargeting rAAV to respiratory cells, for example, using different types of capsid modifications and regulation of a transgene expression by tissue-specific promoters. Part of the review is devoted to the issues of transduction of stem and progenitor lung cells using AAV, which is a complicated task today.
Collapse
|
23
|
|
24
|
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: 2] [Impact Index Per Article: 0.4] [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
|
25
|
Choi SH, Reeves RE, Romano Ibarra GS, Lynch TJ, Shahin WS, Feng Z, Gasser GN, Winter MC, Evans TIA, Liu X, Luo M, Zhang Y, Stoltz DA, Devor EJ, Yan Z, Engelhardt JF. Detargeting Lentiviral-Mediated CFTR Expression in Airway Basal Cells Using miR-106b. Genes (Basel) 2020; 11:E1169. [PMID: 33036232 PMCID: PMC7601932 DOI: 10.3390/genes11101169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Lentiviral-mediated integration of a CFTR transgene cassette into airway basal cells is a strategy being considered for cystic fibrosis (CF) cell-based therapies. However, CFTR expression is highly regulated in differentiated airway cell types and a subset of intermediate basal cells destined to differentiate. Since basal stem cells typically do not express CFTR, suppressing the CFTR expression from the lentiviral vector in airway basal cells may be beneficial for maintaining their proliferative capacity and multipotency. We identified miR-106b as highly expressed in proliferating airway basal cells and extinguished in differentiated columnar cells. Herein, we developed lentiviral vectors with the miR-106b-target sequence (miRT) to both study miR-106b regulation during basal cell differentiation and detarget CFTR expression in basal cells. Given that miR-106b is expressed in the 293T cells used for viral production, obstacles of viral genome integrity and titers were overcome by creating a 293T-B2 cell line that inducibly expresses the RNAi suppressor B2 protein from flock house virus. While miR-106b vectors effectively detargeted reporter gene expression in proliferating basal cells and following differentiation in the air-liquid interface and organoid cultures, the CFTR-miRT vector produced significantly less CFTR-mediated current than the non-miR-targeted CFTR vector following transduction and differentiation of CF basal cells. These findings suggest that miR-106b is expressed in certain airway cell types that contribute to the majority of CFTR anion transport in airway epithelium.
Collapse
Affiliation(s)
- Soon H. Choi
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - Rosie E. Reeves
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | | | - Thomas J. Lynch
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - Weam S. Shahin
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - Zehua Feng
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - Grace N. Gasser
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - Michael C. Winter
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - T. Idil Apak Evans
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - Xiaoming Liu
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - Meihui Luo
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - Yulong Zhang
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - David A. Stoltz
- Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA;
| | - Eric J. Devor
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA;
| | - Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (S.H.C.); (R.E.R.); (T.J.L.); (W.S.S.); (Z.F.); (G.N.G.); (M.C.W.); (T.I.A.E.); (X.L.); (M.L.); (Y.Z.); (Z.Y.)
| |
Collapse
|
26
|
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
|
27
|
Vu A, McCray PB. New Directions in Pulmonary Gene Therapy. Hum Gene Ther 2020; 31:921-939. [PMID: 32814451 PMCID: PMC7495918 DOI: 10.1089/hum.2020.166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
The lung has long been a target for gene therapy, yet efficient delivery and phenotypic disease correction has remained challenging. Although there have been significant advancements in gene therapies of other organs, including the development of several ex vivo therapies, in vivo therapeutics of the lung have been slower to transition to the clinic. Within the past few years, the field has witnessed an explosion in the development of new gene addition and gene editing strategies for the treatment of monogenic disorders. In this review, we will summarize current developments in gene therapy for cystic fibrosis, alpha-1 antitrypsin deficiency, and surfactant protein deficiencies. We will explore the different gene addition and gene editing strategies under investigation and review the challenges of delivery to the lung.
Collapse
Affiliation(s)
- Amber Vu
- Stead Family Department of Pediatrics, Center for Gene Therapy, The University of Iowa, Iowa City, Iowa, USA
| | - Paul B. McCray
- Stead Family Department of Pediatrics, Center for Gene Therapy, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
28
|
Fry LE, Peddle CF, Stevanovic M, Barnard AR, McClements ME, MacLaren RE. Promoter Orientation within an AAV-CRISPR Vector Affects Cas9 Expression and Gene Editing Efficiency. CRISPR J 2020; 3:276-283. [PMID: 32833533 PMCID: PMC7469699 DOI: 10.1089/crispr.2020.0021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Adeno-associated virus (AAV) vectors have been widely adopted for delivery of CRISPR-Cas components, especially for therapeutic gene editing. For a single vector system, both the Cas9 and guide RNA (gRNA) are encoded within a single transgene, usually from separate promoters. Careful design of this bi-cistronic construct is required due to the minimal packaging capacity of AAV. We investigated how placement of the U6 promoter expressing the gRNA on the reverse strand to SaCas9 driven by a cytomegalovirus promoter affected gene editing rates compared to placement on the forward strand. We show that orientation in the reverse direction reduces editing rates from an AAV vector due to reduced transcription of both SaCas9 and guide RNA. This effect was observed only following AAV transduction; it was not seen following plasmid transfection. These results have implications for the design of AAV-CRISPR vectors, and suggest that results from optimizing plasmid transgenes may not translate when delivered via AAV.
Collapse
Affiliation(s)
- Lewis E. Fry
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Caroline F. Peddle
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Marta Stevanovic
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Alun R. Barnard
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Michelle E. McClements
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Robert E. MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
29
|
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
|
30
|
Engineering adeno-associated virus vectors for gene therapy. Nat Rev Genet 2020; 21:255-272. [DOI: 10.1038/s41576-019-0205-4] [Citation(s) in RCA: 342] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
|
31
|
Tornabene P, Trapani I. Can Adeno-Associated Viral Vectors Deliver Effectively Large Genes? Hum Gene Ther 2020; 31:47-56. [DOI: 10.1089/hum.2019.220] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Patrizia Tornabene
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy
| | - Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy
| |
Collapse
|
32
|
Cooney AL, Thornell IM, Singh BK, Shah VS, Stoltz DA, McCray PB, Zabner J, Sinn PL. A Novel AAV-mediated Gene Delivery System Corrects CFTR Function in Pigs. Am J Respir Cell Mol Biol 2019; 61:747-754. [PMID: 31184507 PMCID: PMC6890402 DOI: 10.1165/rcmb.2019-0006oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/05/2019] [Indexed: 11/24/2022] Open
Abstract
Cystic fibrosis is an autosomal-recessive disease that is caused by a mutant CFTR (cystic fibrosis transmembrane conductance regulator) gene and is characterized by chronic bacterial lung infections and inflammation. Complementation with functional CFTR normalizes anion transport across the airway surface. Adeno-associated virus (AAV) is a useful vector for gene therapy because of its low immunogenicity and ability to persist for months to years. However, because its episomal expression may decrease after cell division, readministration of the AAV vector may be required. To overcome this, we designed an integrating AAV-based CFTR-expressing vector, termed piggyBac (PB)/AAV, carrying CFTR flanked by the terminal repeats of the piggyBac transposon. With codelivery of the piggyBac transposase, PB/AAV can integrate into the host genome. Because of the packaging constraints of AAV, careful consideration was required to ensure that the vector would package and express its CFTR cDNA cargo. In this short-term study, PB/AAV-CFTR was aerosolized to the airways of CF pigs in the absence of the transposase. Two weeks later, transepithelial Cl- current was restored in freshly excised tracheal and bronchial tissue. Additionally, we observed an increase in tracheal airway surface liquid pH and bacterial killing in comparison with untreated CF pigs. Airway surface liquid from primary airway cells cultured from treated CF pigs exhibited increased pH correlating with decreased viscosity. Together, these results show that complementing CFTR in CF pigs with PB/AAV rescues the anion transport defect in a large-animal CF model. Delivery of this integrating viral vector system to airway progenitor cells could lead to persistent, life-long expression in vivo.
Collapse
Affiliation(s)
- Ashley L. Cooney
- Stead Family Department of Pediatrics
- Pappajohn Biomedical Institute
- Center for Gene Therapy, and
| | - Ian M. Thornell
- Pappajohn Biomedical Institute
- Center for Gene Therapy, and
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Brajesh K. Singh
- Stead Family Department of Pediatrics
- Pappajohn Biomedical Institute
- Center for Gene Therapy, and
| | - Viral S. Shah
- Pappajohn Biomedical Institute
- Center for Gene Therapy, and
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - David A. Stoltz
- Pappajohn Biomedical Institute
- Center for Gene Therapy, and
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Paul B. McCray
- Stead Family Department of Pediatrics
- Pappajohn Biomedical Institute
- Center for Gene Therapy, and
| | - Joseph Zabner
- Pappajohn Biomedical Institute
- Center for Gene Therapy, and
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Patrick L. Sinn
- Stead Family Department of Pediatrics
- Pappajohn Biomedical Institute
- Center for Gene Therapy, and
| |
Collapse
|
33
|
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: 70] [Impact Index Per Article: 11.7] [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
|
34
|
Keeler AM, Flotte TR. Recombinant Adeno-Associated Virus Gene Therapy in Light of Luxturna (and Zolgensma and Glybera): Where Are We, and How Did We Get Here? Annu Rev Virol 2019; 6:601-621. [PMID: 31283441 PMCID: PMC7123914 DOI: 10.1146/annurev-virology-092818-015530] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The recent market approvals of recombinant adeno-associated virus (rAAV) gene therapies in Europe and the United States are landmark achievements in the history of modern science. These approvals are also anticipated to herald the emergence of a new class of therapies for monogenic disorders, which had hitherto been considered untreatable. These events can be viewed as stemming from the convergence of several important historical trends: the study of basic virology, the development of genomic technologies, the imperative for translational impact of National Institutes of Health-funded research, and the development of economic models for commercialization of rare disease therapies. In this review, these historical trends are described and the key developments that have enabled clinical rAAV gene therapies are discussed, along with an overview of the current state of the field and future directions.
Collapse
Affiliation(s)
- Allison M Keeler
- Horae Gene Therapy Center and Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA;
| | - Terence R Flotte
- Horae Gene Therapy Center and Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA;
| |
Collapse
|
35
|
Dudek AM, Porteus MH. AAV6 Is Superior to Clade F AAVs in Stimulating Homologous Recombination-Based Genome Editing in Human HSPCs. Mol Ther 2019; 27:1701-1705. [PMID: 31537456 DOI: 10.1016/j.ymthe.2019.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Amanda M Dudek
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
36
|
Cooney AL, Singh BK, Loza LM, Thornell IM, Hippee CE, Powers LS, Ostedgaard LS, Meyerholz DK, Wohlford-Lenane C, Stoltz DA, B McCray P, Sinn PL. Widespread airway distribution and short-term phenotypic correction of cystic fibrosis pigs following aerosol delivery of piggyBac/adenovirus. Nucleic Acids Res 2019; 46:9591-9600. [PMID: 30165523 PMCID: PMC6182177 DOI: 10.1093/nar/gky773] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/22/2018] [Indexed: 11/14/2022] Open
Abstract
Cystic fibrosis (CF) is a common genetic disease caused by mutations in the gene coding for cystic fibrosis transmembrane conductance regulator (CFTR). Although CF affects multiple organ systems, chronic bacterial infections and inflammation in the lung are the leading causes of morbidity and mortality in people with CF. Complementation with a functional CFTR gene repairs this defect, regardless of the disease-causing mutation. In this study, we used a gene delivery system termed piggyBac/adenovirus (Ad), which combines the delivery efficiency of an adenoviral-based vector with the persistent expression of a DNA transposon-based vector. We aerosolized piggyBac/Ad to the airways of pigs and observed widespread pulmonary distribution of vector. We quantified the regional distribution in the airways and observed transduction of large and small airway epithelial cells of non-CF pigs, with ∼30–50% of surface epithelial cells positive for GFP. We transduced multiple cell types including ciliated, non-ciliated, basal, and submucosal gland cells. In addition, we phenotypically corrected CF pigs following delivery of piggyBac/Ad expressing CFTR as measured by anion channel activity, airway surface liquid pH, and bacterial killing ability. Combining an integrating DNA transposon with adenoviral vector delivery is an efficient method for achieving functional CFTR correction from a single vector administration.
Collapse
Affiliation(s)
- Ashley L Cooney
- Department of Microbiology, The University of Iowa, Iowa City, IA 52242, USA
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| | - Brajesh K Singh
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| | - Laura Marquez Loza
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Molecular Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Ian M Thornell
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Camilla E Hippee
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| | - Linda S Powers
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Lynda S Ostedgaard
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - David K Meyerholz
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, The University of Iowa, Iowa City, IA 52242, USA
| | - Chris Wohlford-Lenane
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| | - David A Stoltz
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, The University of Iowa, Iowa City, IA 52242, USA
| | - Paul B McCray
- Department of Microbiology, The University of Iowa, Iowa City, IA 52242, USA
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
- Department of Molecular Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Patrick L Sinn
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy for Cystic Fibrosis, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
37
|
Hamilton BA, Li X, Pezzulo AA, Abou Alaiwa MH, Zabner J. Polarized AAVR expression determines infectivity by AAV gene therapy vectors. Gene Ther 2019; 26:240-249. [PMID: 30962536 PMCID: PMC6588428 DOI: 10.1038/s41434-019-0078-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/04/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022]
Abstract
Adeno-associated virus (AAV) has been investigated to transfer the cystic fibrosis transmembrane conductance regulator (CFTR) to airways. Inhaled AAV2-CFTR in people with cystic fibrosis (CF) is safe, but inefficient. In vitro, AAV2 transduction of human airway epithelia on the apical (luminal) side is inefficient, but efficient basolaterally. We previously selected AAV2.5T, a novel capsid that apically transduces CF human airway epithelia and efficiently restores CFTR function. We hypothesize the AAV receptor (AAVR) is basolaterally localized, and that AAV2.5T utilizes an alternative apical receptor. We found AAVR in human airway epithelia by western blot and RNA-Seq analyses. Using immunocytochemistry we did not find endogenous AAVR at membranes but overexpression localized AAVR to the basolateral membrane, where it preferentially increased transduction. Anti-AAVR antibodies blocked transduction by AAV2 from the basolateral side but not AAV2.5T from the apical side, suggesting a unique apical receptor. Finally, we found infection by AAV2 but not AAV2.5T was blocked by CRISPR knockout of AAVR in cell lines. Our data suggest the absence of apical AAVR is rate limiting for AAV2, and efficient transduction by AAV2.5T is accomplished using an AAVR independent pathway. Our findings inform the development of gene therapy for CF, and AAV vectors in general.
Collapse
Affiliation(s)
- Bradley A Hamilton
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
- Molecular Medicine Program, The University of Iowa, Iowa City, IA, USA
| | - Xiaopeng Li
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
| | - Alejandro A Pezzulo
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
| | - Mahmoud H Abou Alaiwa
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, The University of Iowa, Iowa City, IA, USA
| | - Joseph Zabner
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, Pappajohn Biomedical Institute, The University of Iowa, Iowa City, IA, USA.
- Molecular Medicine Program, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
38
|
Trapani I. Adeno-Associated Viral Vectors as a Tool for Large Gene Delivery to the Retina. Genes (Basel) 2019; 10:genes10040287. [PMID: 30970639 PMCID: PMC6523333 DOI: 10.3390/genes10040287] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/26/2019] [Accepted: 04/05/2019] [Indexed: 12/26/2022] Open
Abstract
Gene therapy using adeno-associated viral (AAV) vectors currently represents the most promising approach for the treatment of many inherited retinal diseases (IRDs), given AAV's ability to efficiently deliver therapeutic genes to both photoreceptors and retinal pigment epithelium, and their excellent safety and efficacy profiles in humans. However, one of the main obstacles to widespread AAV application is their limited packaging capacity, which precludes their use from the treatment of IRDs which are caused by mutations in genes whose coding sequence exceeds 5 kb. Therefore, in recent years, considerable effort has been made to identify strategies to increase the transfer capacity of AAV vectors. This review will discuss these new developed strategies, highlighting the advancements as well as the limitations that the field has still to overcome to finally expand the applicability of AAV vectors to IRDs due to mutations in large genes.
Collapse
Affiliation(s)
- Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy.
- Medical Genetics, Department of Translational Medicine, Federico II University, 80131 Naples, Italy.
| |
Collapse
|
39
|
Abstract
Adeno-associated virus (AAV)-mediated gene therapy has evolved from bench to bedside, and now is the therapy of choice for certain inherited diseases. However, the small packaging capacity of AAV vectors prevents this technique from treating genetic diseases with mutations of large genes. Multiple strategies, including split AAV gene delivery and oversized AAV gene delivery, have been explored to deliver large gene expression cassettes. These strategies have gained some success in animal experiments. In this chapter, we review the progress of AAV-mediated delivery of large expression cassettes. We also review using AAV to deliver multiple transgenes.
Collapse
Affiliation(s)
- Aman Patel
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Department of Bioengineering, University of Missouri, Columbia, MO, USA
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
40
|
Zhang XP, Zhang WT, Qiu Y, Ju MJ, Tu GW, Luo Z. Understanding Gene Therapy in Acute Respiratory Distress Syndrome. Curr Gene Ther 2019; 19:93-99. [PMID: 31267871 DOI: 10.2174/1566523219666190702154817] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/07/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
Acute Respiratory Distress Syndrome (ARDS) and its complications remain lifethreatening conditions for critically ill patients. The present therapeutic strategies such as prone positioning ventilation strategies, nitric oxide inhalation, restrictive intravenous fluid management, and extracorporeal membrane oxygenation (ECMO) do not contribute much to improving the mortality of ARDS. The advanced understanding of the pathophysiology of acute respiratory distress syndrome suggests that gene-based therapy may be an innovative method for this disease. Many scientists have made beneficial attempts to regulate the immune response genes of ARDS, maintain the normal functions of alveolar epithelial cells and endothelial cells, and inhibit the fibrosis and proliferation of ARDS. Limitations to effective pulmonary gene therapy still exist, including the security of viral vectors and the pulmonary defense mechanisms against inhaled particles. Here, we summarize and review the mechanism of gene therapy for acute respiratory distress syndrome and its application.
Collapse
Affiliation(s)
- Xue-Peng Zhang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Wei-Tao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, No. 179 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Yue Qiu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Min-Jie Ju
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Guo-Wei Tu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Zhe Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
- Department of Critical Care Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, No. 668 Jinghu Road, Huli District, Xiamen 361015, China
| |
Collapse
|
41
|
Dong ZQ, Hu ZG, Li HQ, Jiang YM, Cao MY, Chen P, Lu C, Pan MH. Construction and characterization of a synthetic Baculovirus-inducible 39K promoter. J Biol Eng 2018; 12:30. [PMID: 30534200 PMCID: PMC6280533 DOI: 10.1186/s13036-018-0121-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/19/2018] [Indexed: 01/27/2023] Open
Abstract
Background Silkworm genetic engineering is widely used in gene function, silk engineering and disease-resistant engineering in most of Asia. Some of the earliest promoter elements are used to control the development of silkworm transgenic expression and gene therapy. However, the low expression and specificity of natural promoters limit the applications of genetic engineering. To construct a highly efficient synthetic inducible promoter in the Bombyx mori (Lepidoptera), we analyzed the regulatory elements and functional regions of the B. mori nucleopolyhedrovirus 39 K promoter. Results Truncated mutation analysis of the 39 K promoter showed that the transcriptional regulatory region spanning positions - 573 to - 274 and + 1 to + 62 are essential for virus-inducible promoter activity. Further investigations using the electrophoretic mobility shift assay revealed that the baculovirus IE-1 protein binds to the 39 K promoter at the - 310 to - 355 region, and transcription activates the expression of 39 K promoter assay. Finally, we successfully constructed a synthetic inducible promoter that increased the virus-inducing activity of other promoters using the baculovirus-inducible transcriptional activation region that binds to specific core elements of 39 K (i.e., spanning the region - 310 to - 355). Conclusions In summary, we constructed a novel, synthetic, and highly efficient biological tool, namely, a virus-inducible 39 K promoter, which provides endless possibilities for future research on gene function, gene therapy, and pest control in genetic engineering.
Collapse
Affiliation(s)
- Zhan-Qi Dong
- 1State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716 China
| | - Zhi-Gang Hu
- 1State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716 China
| | - Hai-Qing Li
- 1State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716 China
| | - Ya-Ming Jiang
- 1State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716 China
| | - Ming-Ya Cao
- 3Joint National Laboratory for Antibody Drug Engineering, Institute of Immunology, Henan University School of Medicine, Kaifeng, 475004 China
| | - Peng Chen
- 1State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716 China
| | - Cheng Lu
- 1State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716 China.,2Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing, 400716 China
| | - Min-Hui Pan
- 1State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716 China.,2Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry, Southwest University, Chongqing, 400716 China
| |
Collapse
|
42
|
Cooney AL, McCray PB, Sinn PL. Cystic Fibrosis Gene Therapy: Looking Back, Looking Forward. Genes (Basel) 2018; 9:genes9110538. [PMID: 30405068 PMCID: PMC6266271 DOI: 10.3390/genes9110538] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 01/02/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that encodes a cAMP-regulated anion channel. Although CF is a multi-organ system disease, most people with CF die of progressive lung disease that begins early in childhood and is characterized by chronic bacterial infection and inflammation. Nearly 90% of people with CF have at least one copy of the ΔF508 mutation, but there are hundreds of CFTR mutations that result in a range of disease severities. A CFTR gene replacement approach would be efficacious regardless of the disease-causing mutation. After the discovery of the CFTR gene in 1989, the in vitro proof-of-concept for gene therapy for CF was quickly established in 1990. In 1993, the first of many gene therapy clinical trials attempted to rescue the CF defect in airway epithelia. Despite the initial enthusiasm, there is still no FDA-approved gene therapy for CF. Here we discuss the history of CF gene therapy, from the discovery of the CFTR gene to current state-of-the-art gene delivery vector designs. While implementation of CF gene therapy has proven more challenging than initially envisioned; thanks to continued innovation, it may yet become a reality.
Collapse
Affiliation(s)
- Ashley L Cooney
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Paul B McCray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Patrick L Sinn
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
43
|
Montoro DT, Haber AL, Biton M, Vinarsky V, Lin B, Birket SE, Yuan F, Chen S, Leung HM, Villoria J, Rogel N, Burgin G, Tsankov AM, Waghray A, Slyper M, Waldman J, Nguyen L, Dionne D, Rozenblatt-Rosen O, Tata PR, Mou H, Shivaraju M, Bihler H, Mense M, Tearney GJ, Rowe SM, Engelhardt JF, Regev A, Rajagopal J. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 2018; 560:319-324. [PMID: 30069044 PMCID: PMC6295155 DOI: 10.1038/s41586-018-0393-7] [Citation(s) in RCA: 819] [Impact Index Per Article: 117.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 06/21/2018] [Indexed: 12/16/2022]
Abstract
The airways of the lung are the primary sites of disease in asthma and cystic fibrosis. Here we study the cellular composition and hierarchy of the mouse tracheal epithelium by single-cell RNA-sequencing (scRNA-seq) and in vivo lineage tracing. We identify a rare cell type, the Foxi1+ pulmonary ionocyte; functional variations in club cells based on their location; a distinct cell type in high turnover squamous epithelial structures that we term 'hillocks'; and disease-relevant subsets of tuft and goblet cells. We developed 'pulse-seq', combining scRNA-seq and lineage tracing, to show that tuft, neuroendocrine and ionocyte cells are continually and directly replenished by basal progenitor cells. Ionocytes are the major source of transcripts of the cystic fibrosis transmembrane conductance regulator in both mouse (Cftr) and human (CFTR). Knockout of Foxi1 in mouse ionocytes causes loss of Cftr expression and disrupts airway fluid and mucus physiology, phenotypes that are characteristic of cystic fibrosis. By associating cell-type-specific expression programs with key disease genes, we establish a new cellular narrative for airways disease.
Collapse
Affiliation(s)
- Daniel T Montoro
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adam L Haber
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Moshe Biton
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Vladimir Vinarsky
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Brian Lin
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Susan E Birket
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, AL, USA
| | - Feng Yuan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sijia Chen
- Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Hui Min Leung
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jorge Villoria
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Noga Rogel
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Grace Burgin
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexander M Tsankov
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Avinash Waghray
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michal Slyper
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Waldman
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lan Nguyen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Purushothama Rao Tata
- Department of Cell Biology, Duke University, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
- Division of Pulmonary Critical Care, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Regeneration Next, Duke University, Durham, NC, USA
| | - Hongmei Mou
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Manjunatha Shivaraju
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Hermann Bihler
- CFFT Lab, Cystic Fibrosis Foundation, Lexington, MA, USA
| | - Martin Mense
- CFFT Lab, Cystic Fibrosis Foundation, Lexington, MA, USA
| | - Guillermo J Tearney
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Steven M Rowe
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, AL, USA
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute and Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
44
|
Assembly and Functional Analysis of an S/MAR Based Episome with the Cystic Fibrosis Transmembrane Conductance Regulator Gene. Int J Mol Sci 2018; 19:ijms19041220. [PMID: 29673202 PMCID: PMC5979583 DOI: 10.3390/ijms19041220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/24/2022] Open
Abstract
Improving the efficacy of gene therapy vectors is still an important goal toward the development of safe and efficient gene therapy treatments. S/MAR (scaffold/matrix attached region)-based vectors are maintained extra-chromosomally in numerous cell types, which is similar to viral-based vectors. Additionally, when established as an episome, they show a very high mitotic stability. In the present study we tested the idea that addition of an S/MAR element to a CFTR (cystic fibrosis transmembrane conductance regulator) expression vector, may allow the establishment of a CFTR episome in bronchial epithelial cells. Starting from the observation that the S/MAR vector pEPI-EGFP (enhanced green fluorescence protein) is maintained as an episome in human bronchial epithelial cells, we assembled the CFTR vector pBQ-S/MAR. This vector, transfected in bronchial epithelial cells with mutated CFTR, supported long term wt CFTR expression and activity, which in turn positively impacted on the assembly of tight junctions in polarized epithelial cells. Additionally, the recovery of intact pBQ-S/MAR, but not the parental vector lacking the S/MAR element, from transfected cells after extensive proliferation, strongly suggested that pBQ-S/MAR was established as an episome. These results add a new element, the S/MAR, that can be considered to improve the persistence and safety of gene therapy vectors for cystic fibrosis pulmonary disease.
Collapse
|
45
|
Julien L, Chassagne J, Peccate C, Lorain S, Piétri-Rouxel F, Danos O, Benkhelifa-Ziyyat S. RFX1 and RFX3 Transcription Factors Interact with the D Sequence of Adeno-Associated Virus Inverted Terminal Repeat and Regulate AAV Transduction. Sci Rep 2018; 8:210. [PMID: 29317724 PMCID: PMC5760533 DOI: 10.1038/s41598-017-18604-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/13/2017] [Indexed: 12/31/2022] Open
Abstract
Adeno-associated virus (AAV) transduction efficiency depends on the way in which cellular proteins process viral genomes in the nucleus. In this study, we have investigated the binding of nuclear proteins to the double stranded D (dsD) sequence of the AAV inverted terminal repeat (ITRs) by electromobility shift assay. We present here several lines of evidence that transcription factors belonging to the RFX protein family bind specifically and selectively to AAV2 and AAV1 dsD sequences. Using supershift experiments, we characterize complexes containing RFX1 homodimers and RFX1/RFX3 heterodimers. Following transduction of HEK-293 cells, the AAV genome can be pulled-down by RFX1 and RFX3 antibodies. Moreover, our data suggest that RFX proteins which interact with transcriptional enhancers of several mammalian DNA viruses, can act as regulators of AAV mediated transgene expression.
Collapse
Affiliation(s)
- Laura Julien
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Julie Chassagne
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Cécile Peccate
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Stéphanie Lorain
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - France Piétri-Rouxel
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Olivier Danos
- REGENXBIO, 9600 Blackwell Rd, Rockville, MD, 20850, USA
| | - Sofia Benkhelifa-Ziyyat
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France.
| |
Collapse
|
46
|
Carlon MS, Vidović D, Birket S. Roadmap for an early gene therapy for cystic fibrosis airway disease. Prenat Diagn 2017; 37:1181-1190. [DOI: 10.1002/pd.5164] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/12/2017] [Accepted: 09/28/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Marianne S. Carlon
- Molecular Virology and Gene Therapy; Department of Pharmaceutical and Pharmacological Sciences; KU Leuven Flanders Belgium
| | - Dragana Vidović
- Molecular Virology and Gene Therapy; Department of Pharmaceutical and Pharmacological Sciences; KU Leuven Flanders Belgium
- Current affiliation: Cellular Protein Chemistry, Faculty of Science; Utrecht University; The Netherlands
| | - Susan Birket
- Department of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| |
Collapse
|
47
|
Guggino WB, Cebotaru L. Adeno-Associated Virus (AAV) gene therapy for cystic fibrosis: current barriers and recent developments. Expert Opin Biol Ther 2017; 17:1265-1273. [PMID: 28657358 DOI: 10.1080/14712598.2017.1347630] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Since the cystic fibrosis (CF) gene was discovered in 1989, researchers have worked to develop a gene therapy. One of the most promising and enduring vectors is the AAV, which has been shown to be safe. In particular, several clinical trials have been conducted with AAV serotype 2. All of them detected viral genomes, but identification of mRNA transduction was not consistent; clinical outcomes in Phase II studies were also inconsistent. The lack of a positive outcome has been attributed to a less-than-efficient viral infection by AAV2, a weak transgene promoter and the host immune response to the vector. Areas covered: Herein, the authors focus on AAV gene therapy for CF, evaluating past experience with this approach and identifying ways forward, based on the progress that has already been made in identifying and overcoming the limitations of AAV gene therapy. Expert opinion: Such progress makes it clear that this is an opportune time to push forward toward the development of a gene therapy for CF. Drugs to treat the basic defect in CF represent a remarkable advance but cannot treat a significant cohort of patients with rare mutations. Thus, there is a critical need to develop a gene therapy for those individuals.
Collapse
Affiliation(s)
- William B Guggino
- a Departments of Medicine and Physiology , Johns Hopkins University , Baltimore , MD , USA
| | - Liudmila Cebotaru
- a Departments of Medicine and Physiology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
48
|
Baruteau J, Waddington SN, Alexander IE, Gissen P. Gene therapy for monogenic liver diseases: clinical successes, current challenges and future prospects. J Inherit Metab Dis 2017; 40:497-517. [PMID: 28567541 PMCID: PMC5500673 DOI: 10.1007/s10545-017-0053-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 02/08/2023]
Abstract
Over the last decade, pioneering liver-directed gene therapy trials for haemophilia B have achieved sustained clinical improvement after a single systemic injection of adeno-associated virus (AAV) derived vectors encoding the human factor IX cDNA. These trials demonstrate the potential of AAV technology to provide long-lasting clinical benefit in the treatment of monogenic liver disorders. Indeed, with more than ten ongoing or planned clinical trials for haemophilia A and B and dozens of trials planned for other inherited genetic/metabolic liver diseases, clinical translation is expanding rapidly. Gene therapy is likely to become an option for routine care of a subset of severe inherited genetic/metabolic liver diseases in the relatively near term. In this review, we aim to summarise the milestones in the development of gene therapy, present the different vector tools and their clinical applications for liver-directed gene therapy. AAV-derived vectors are emerging as the leading candidates for clinical translation of gene delivery to the liver. Therefore, we focus on clinical applications of AAV vectors in providing the most recent update on clinical outcomes of completed and ongoing gene therapy trials and comment on the current challenges that the field is facing for large-scale clinical translation. There is clearly an urgent need for more efficient therapies in many severe monogenic liver disorders, which will require careful risk-benefit analysis for each indication, especially in paediatrics.
Collapse
Affiliation(s)
- Julien Baruteau
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, UK.
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, UK.
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ian E Alexander
- Gene Therapy Research Unit, The Children's Hospital at Westmead and Children's Medical Research Institute, Westmead, Australia
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia
| | - Paul Gissen
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
49
|
Yan Z, Feng Z, Sun X, Zhang Y, Zou W, Wang Z, Jensen-Cody C, Liang B, Park SY, Qiu J, Engelhardt JF. Human Bocavirus Type-1 Capsid Facilitates the Transduction of Ferret Airways by Adeno-Associated Virus Genomes. Hum Gene Ther 2017; 28:612-625. [PMID: 28490200 DOI: 10.1089/hum.2017.060] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Human bocavirus type-1 (HBoV1) has a high tropism for the apical membrane of human airway epithelia. The packaging of a recombinant adeno-associated virus 2 (rAAV2) genome into HBoV1 capsid produces a chimeric vector (rAAV2/HBoV1) that also efficiently transduces human airway epithelia. As such, this vector is attractive for use in gene therapies to treat lung diseases such as cystic fibrosis. However, preclinical development of rAAV2/HBoV1 vectors has been hindered by the fact that humans are the only known host for HBoV1 infection. This study reports that rAAV2/HBoV1 vector is capable of efficiently transducing the lungs of both newborn (3- to 7-day-old) and juvenile (29-day-old) ferrets, predominantly in the distal airways. Analyses of in vivo, ex vivo, and in vitro models of the ferret proximal airway demonstrate that infection of this particular region is less effective than it is in humans. Studies of vector binding and endocytosis in polarized ferret proximal airway epithelial cultures revealed that a lack of effective vector endocytosis is the main cause of inefficient transduction in vitro. While transgene expression declined proportionally with growth of the ferrets following infection at 7 days of age, reinfection of ferrets with rAAV2/HBoV1 at 29 days gave rise to approximately 5-fold higher levels of transduction than observed in naive infected 29-day-old animals. The findings presented here lay the foundation for clinical development of HBoV1 capsid-based vectors for lung gene therapy in cystic fibrosis using ferret models.
Collapse
Affiliation(s)
- Ziying Yan
- 1 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa.,2 Center for Gene Therapy, University of Iowa , Iowa City, Iowa
| | - Zehua Feng
- 1 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - Xingshen Sun
- 1 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - Yulong Zhang
- 1 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - Wei Zou
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center , Kansas City, Kansas
| | - Zekun Wang
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center , Kansas City, Kansas
| | | | - Bo Liang
- 1 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - Soo-Yeun Park
- 1 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa
| | - Jianming Qiu
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center , Kansas City, Kansas
| | - John F Engelhardt
- 1 Department of Anatomy and Cell Biology, University of Iowa , Iowa City, Iowa.,2 Center for Gene Therapy, University of Iowa , Iowa City, Iowa
| |
Collapse
|
50
|
Steines B, Dickey DD, Bergen J, Excoffon KJ, Weinstein JR, Li X, Yan Z, Abou Alaiwa MH, Shah VS, Bouzek DC, Powers LS, Gansemer ND, Ostedgaard LS, Engelhardt JF, Stoltz DA, Welsh MJ, Sinn PL, Schaffer DV, Zabner J. CFTR gene transfer with AAV improves early cystic fibrosis pig phenotypes. JCI Insight 2016; 1:e88728. [PMID: 27699238 DOI: 10.1172/jci.insight.88728] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The physiological components that contribute to cystic fibrosis (CF) lung disease are steadily being elucidated. Gene therapy could potentially correct these defects. CFTR-null pigs provide a relevant model to test gene therapy vectors. Using an in vivo selection strategy that amplifies successful capsids by replicating their genomes with helper adenovirus coinfection, we selected an adeno-associated virus (AAV) with tropism for pig airway epithelia. The evolved capsid, termed AAV2H22, is based on AAV2 with 5 point mutations that result in a 240-fold increased infection efficiency. In contrast to AAV2, AAV2H22 binds specifically to pig airway epithelia and is less reliant on heparan sulfate for transduction. We administer AAV2H22-CFTR expressing the CF transmembrane conductance regulator (CFTR) cDNA to the airways of CF pigs. The transduced airways expressed CFTR on ciliated and nonciliated cells, induced anion transport, and improved the airway surface liquid pH and bacterial killing. Most gene therapy studies to date focus solely on Cl- transport as the primary metric of phenotypic correction. Here, we describe a gene therapy experiment where we not only correct defective anion transport, but also restore bacterial killing in CFTR-null pig airways.
Collapse
Affiliation(s)
- Benjamin Steines
- Department of Internal Medicine.,Molecular and Cellular Biology Program, and.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - David D Dickey
- Department of Internal Medicine.,Molecular and Cellular Biology Program, and
| | - Jamie Bergen
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, California, USA
| | | | - John R Weinstein
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, California, USA
| | - Xiaopeng Li
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | | | - Mahmoud H Abou Alaiwa
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Viral S Shah
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | | | | | | | - Lynda S Ostedgaard
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | | | - David A Stoltz
- Department of Internal Medicine.,Molecular and Cellular Biology Program, and.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Michael J Welsh
- Department of Internal Medicine.,Molecular and Cellular Biology Program, and.,Molecular Physiology and Biophysics
| | - Patrick L Sinn
- Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA.,Howard Hughes Medical Institute, and
| | - David V Schaffer
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, California, USA
| | - Joseph Zabner
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|