Review Article

Gene Therapy Cargoes Based on Viral Vector Delivery

Author(s): Kenneth Lundstrom*

Volume 23, Issue 2, 2023

Published on: 17 November, 2022

Page: [111 - 134] Pages: 24

DOI: 10.2174/1566523222666220921112753

Price: $65

Abstract

Viral vectors have been proven useful in a broad spectrum of gene therapy applications due to their possibility to accommodate foreign genetic material for both local and systemic delivery. The wide range of viral vectors has enabled gene therapy applications for both acute and chronic diseases. Cancer gene therapy has been addressed by the delivery of viral vectors expressing anti-tumor, toxic, and suicide genes for the destruction of tumors. Delivery of immunostimulatory genes such as cytokines and chemokines has also been applied for cancer therapy. Moreover, oncolytic viruses specifically replicating in and killing tumor cells have been used as such for tumor eradication or in combination with tumor killing or immunostimulatory genes. In a broad meaning, vaccines against infectious diseases and various cancers can be considered gene therapy, which has been highly successful, not the least for the development of effective COVID-19 vaccines. Viral vector-based gene therapy has also demonstrated encouraging and promising results for chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, and hemophilia. Preclinical gene therapy studies in animal models have demonstrated proof-of-concept for a wide range of disease indications. Clinical evaluation of drugs and vaccines in humans has showed high safety levels, good tolerance, and therapeutic efficacy. Several gene therapy drugs such as the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, lentivirus-based treatment of SCID-X1 disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease, and adenovirus-based vaccines against COVID-19 have been developed.

Keywords: Viral vectors, gene therapy, cancer therapy, preclinical studies, clinical trials, approved drugs/vaccines.

Next »
[1]
Lundstrom K. New era in gene therapy Novel Approaches and Strategies for Biologics, Vaccines and Cancer Therapies. Elsevier: San Diego, California 2015; pp. 15-37.
[2]
Martinez T, Wright N, López-Fraga M, et al. Silencing human genetic diseases with oligonucleotide-based therapies. Hum Genet 2013; 132: 481-93.
[3]
Bobbin ML, Rossi JJ. RNA interference (RNAi)-based therapeutics: delivering on the promise? Annu Rev Pharmacol Toxicol 2016; 56: 103-22.
[4]
Ramirez-Montagut T. Cancer vaccines Novel Approaches and Strategies for Biologics, Vaccines and Cancer Therapies. Elsevier: California 2015; pp. 365-88.
[5]
Sermer D, Brentjens R. CAR-T cell therapy: full speed ahead. Hematol Oncol 2019; 37: 95-100.
[6]
Lino CA, Harper JC, Carney JP, et al. Delivering CRISPR: a review of the challenges and approaches. Drug Deliv 2018; 25: 1234-57.
[7]
Hacein-Bey-Abina S, Garrigue A, Wang GP, et al. (2008) Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest 2008; 118: 3132-42.
[8]
Raper SE, Chirmule N, Lee FS, et al. Fatal systemic inflammatory response syndrome in an ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab 2003; 80: 148-58.
[9]
Fukuhara H, Ino Y, Todo T. Oncolytic virus therapy: A new era of cancer treatment at dawn. Cancer Sci 2016; 107: 1373-9.
[10]
Tatsis N, Ertl HCJ. Adenoviruses as vaccine vectors. Mol Ther 2004; 10: 616-29.
[11]
Wang F, Wang Z, Tian H, et al. Biodistribution and safety assessment of bladder cancer specific oncolytic adenovirus in subcutaneous xenografts tumor model in nude mice. Curr Gene Ther 2012; 12: 67-76.
[12]
Ricobaraza A, Gonzalez-Aparicio M, Mora-Jimenez L, et al. High-Capacity Adenovirus Vectors: Expanding the Scope of Gene Therapy. Int J Mol Sci 2020; 21: 3643.
[13]
Ehrke-Schulz E, Zhang W, Schiwon M, et al. Cloning and Large-Scale Production of High-Capacity Adenoviral Vectors Based on the Human Adenovirus Type 5. J Vis Exp 2016; 107: e52894.
[14]
Wei Q, Fan J, Liao J, et al. Engineering the rapid adenovirus production and amplification (RAPA) cell line to expedite the generation of recombinant adenoviruses. Cell Physiol Biochem 2017; 41: 2383-98.
[15]
Zhang WW, Li L, Li D, et al. The First Approved Gene Therapy Product for Cancer Ad-p53 (Gendicine): 12 Years in the Clinic. Hum Gene Ther 2018; 29: 160-79.
[16]
Lundstrom K. Viral vector-based vaccines against SARS-CoV-2. Explor Immunol 2021; 1: 295-308.
[17]
Samulski R, Muzycka N. AAV-mediated gene therapy for research and therapeutic purposes. Annu Rev Virol 2014; 1: 427-51.
[18]
Grieger C, Samulski RJ. Packaging capacity of adeno-associated virus serotypes: impact of larger genomes on infectivity and postentry steps. J Virol 2005; 79: 9933-44.
[19]
McClements ME, MacLaren RR. Adeno-associated virus (AAV) dual vector strategies for gene therapy encoding large transgenes. Yale J Biol Med 2017; 90: 611-23.
[20]
Park K, Kim WJ, Cho YH, et al. Cancer gene therapy using adeno-associated virus vectors. Front Biosci 2008; 13: 2653-9.
[21]
Nienhuis AW, Nathwani AC, Davidoff AM. Gene therapy for hemophilia. Mol Ther 2017; 25: 1163-7.
[22]
Moorehead T, Yong F, Neelakantan S, et al. Safety and tolerability of PF-06939926 in ambulatory boys with Duchenne muscular dystrophy: a phase 1b multicenter, open label, dose ascending study. Mol Ther 2020; 28: 272.
[23]
Mingozzi F, High KA. Immune responses to AAV vectors: overcoming barriers to successful gene therapy. Blood 2013; 122: 23-36.
[24]
Meliani A, Boisgerault F, Fitzpatrick Z, et al. Enhanced liver gene transfer and evasion of preexisting humoral immunity with exosome-enveloped AAV vectors. Blood Adv 2017; 1: 2019-31.
[25]
Epstein AL, Marconi P, Argnani R, et al. HSV-1 derived recombinant and amplicon vectors for gene transfer and gene therapy. Curr Gene Ther 2005; 5: 445-58.
[26]
Holmes KD, Cassam AK, Chan B, et al. A multi-mutant herpes simplex virus vector has minimal cytotoxic effects on the distribution of filamentous actin, alpha-actinin and a glutamate receptor in differentiated PC-12 cells. J Neurovirol 2000; 6: 33-45.
[27]
Li JM, Kao KC, Li LF. Micro-RNA-145 regulates oncolytic herpes simplex virus-1 for selective killing of human non-small lung cancer cells. Virol J 2013; 10: 241.
[28]
Bommareddy PK, Patel A, Hossain S, et al. Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma. J Clin Dermatol 2017; 18: 1-15.
[29]
Schambach A, Morgan M. Retroviral vectors for cancer gene therapy. Curr Strat Cancer Gene Ther 2016; 209: 17-35.
[30]
Hu WS, Pathak VK. Design of retroviral vectors and helper cells for gene therapy. Pharmacol Rev 2000; 52: 493-511.
[31]
Inoko K, Hiraoka K, Inagaki A, et al. Therapeutic activity of retroviral replicating vector-mediated prodrug activator gene therapy for pancreatic cancer. Cancer Gene Ther 2018; 25: 184-95.
[32]
Heinzerling L, Kunzi V, Oberholzer PA, et al. Oncolytic measles virus in cutaneous T-cell lymphomas mounts antitumor immune responses in vivo and targets interferon resistant tumor cells. Blood 2005; 106: 2287-94.
[33]
Hacein-Bey-Abina S, Pai SY, Gaspar HB, et al. A modified γ-retrovirus vector for X-linked severe combined immunodeficiency. N Engl J Med 2014; 371: 1407-17.
[34]
Vigna E, Naldini L. Lentiviral vectors: excellent tools for experimental gene transfer and promising candidates for gene therapy. J Gene Med 2000; 2: 308-16.
[35]
Kay MA, Glorioso JC, Naldini L. Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics. Nat Med 2001; 7: 33-40.
[36]
Ciuffi A. Mechanisms governing lentivirus integration site selection. Curr Gene Ther 2008; 8: 419-29.
[37]
Silvers RM, Smith JA, Schowalter M, et al. Modification of integration site preferences of an HIV-1-based vector by expression of a novel synthetic protein. Hum Gene Ther 2010; 21: 337-49.
[38]
Nakajima T, Nakamaru K, Ido E, et al. (2000) Development of novel simian immunodeficiency virus vectors carrying a dual gene expression system. Hum Gene Ther 2000; 11: 1863-74.
[39]
Hartmann K. Clinical aspects of feline retroviruses: a review. Viruses 2012; 4: 2684-710.
[40]
Olsen JC. Gene transfer vectors derived from equine infectious anemia virus. Gene Ther 1998; 5: 1481-7.
[41]
Ribel J-A, Hacein-Bey-Abina S, Paven E, et al. Gene therapy in a patient with sickle cell disease. N Engl J Med 2017; 376: 848-55.
[42]
Cartier N, Hacein-Bey-Abina S, Bartholomae CC, et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 2009; 326: 818-23.
[43]
Kohn DB, Hershfield MS, Puck JM, et al. Consensus approach for the management of severe combined immune deficiency caused by adenosine deaminase deficiency. J Allergy Clin Immunol 2019; 143: 852-63.
[44]
Strauss JH, Strauss EG. The alphaviruses; gene expression, replication and evolution. Microbiol Rev 1994; 58: 491-562.
[45]
Lundstrom K. Self-amplifying RNA viruses as RNA vaccines. Int J Mol Sci 2020; 2020: 5130.
[46]
Liljeström P, Garoff H. A new generation of animal cell expression vectors based on the Semliki Forest virus replicon. Biotechnology (N Y) 1991; 9: 1356-61.
[47]
Xiong C, Levis R, Shen P, et al. Sindbis virus: An efficient, broad host range vector for gene expression in animal cells. Science 1989; 243: 1188-91.
[48]
Davis NL, Willis LV, Smith JF, et al. In vitro synthesis of infectious Venezuelan equine encephalitis virus RNA from a cDNA clone: Analysis of a viable deletion mutant. Virology 1989; 171: 189-204.
[49]
Li M, Stollar V. Alphaviruses and apoptosis. Int Rev Immunol 2004; 23: 7-24.
[50]
Heikkilä JE, Vähä-Koskela MJ, Ruotsalainen JJ, et al. Intravenously administered alphavirus vector VA7 eradicates orthotopic human glioma xenografts in nude mice. PLoS One 2010; 5: e8603.
[51]
Zhang J, Liu Y, Tan J, et al. Necroptotic virotherapy of oncolytic alphavirus M1 cooperated with Doxorubicin displays promising therapeutic efficacy in TNBC. Oncogene 2021; 40: 4783-95.
[52]
Pijlman GP, Suhrbier A, Khromykh AA. Kunjin virus replicons: An RNA-based, non-cytopathic viral vector system for protein production, vaccine and gene therapy applications. Expert Opin Biol Ther 2006; 6: 134-45.
[53]
Scholle I, Girard YA, Zhao Q, et al. Trans-packaged West Nile virus-like particles: infectious properties in vitro and in infected mosquito vectors. J Virol 2004; 78: 11605-14.
[54]
Pang X, Zhang M, Dayton AI. Development of dengue virus type 2 replicons capable of prolonged expression in host cells. BMC Microbiol 2001; 1: 18.
[55]
Gherke R, Ecker M, Aberle SW, et al. Incorporation of tick-borne encephalitis virus replicons into virus-like particles by a packaging cell line. J Virol 2003; 77: 8924-33.
[56]
Jones CT, Patkar CG, Kuhn RJ. Construction and applications of yellow fever virus replicons. Virology 2005; 331: 247-59.
[57]
Khromykh AA, Varnavski AN, Westaway EG. Encapsidation of the flavivirus Kunjin replicon RNA by using a complementation system providing Kunjin virus structural proteins in trans. J Virol 1998; 72: 5967-77.
[58]
Apostolopoulos V. Vaccine delivery methods into the future. Vaccine 2016; 4: 9.
[59]
Lundstrom K. Self-replicating vehicles based on negative strand RNA viruses. Cancer Gene Ther 15: 1-14.
[60]
Singh M, Cattaneo R, Billeter MA. A recombinant measles virus expressing hepatitis B surface antigen induces humoral responses in genetically modified mice. J Virol 1999; 73: 4823-8.
[61]
Mateo M, Reynard S, Carnec X, et al. Vaccines inducing immunity to Lassa fever glycoprotein and nucleoprotein protect macaques after a single shot. Sci Transl Med 2019; 11: eaaw3163.
[62]
Grossardt C, Engeland CE, Bossow S, et al. Granulocyte-macrophage colony-stimulating factor-armed oncolytic measles virus is an effective therapeutic cancer vaccine. Hum Gene Ther 2013; 24: 644-54.
[63]
Msaouel P, Iankov ID, Allen C, et al. Engineered measles virus as a novel oncolytic therapy against prostate cancer. Prostate 2009; 69: 82-91.
[64]
Lange S, Lampe J, Bossow S, et al. A novel armed oncolytic measles vaccine virus for the treatment of cholangiocarcinoma. Hum Gene Ther 2013; 24: 554-64.
[65]
Finke S, Conzelmann KK. Recombinant rhabdoviruses: vectors for vaccine development and gene therapy. Curr Top Microbiol Immunol 2005; 292: 165-200.
[66]
Ito N, Takayama-Ito M, Yamada K, et al. Improved recovery of rabies virus from cloned cDNA using a vaccinia virus-free reverse genetics system. Microbiol Immunol 2003; 47: 613-7.
[67]
Luo J, Zhao J, Tian Q, et al. A recombinant rabies virus carrying GFP between N and P affects viral transcription in vitro. Virus Genes 2016; 52: 379-87.
[68]
An H, Kim GN, Kang CY. Genetically modified VSV(NJ) vector is capable of accommodating a large foreign gene insert and allows high level gene expression. Virus Res 2013; 171: 168-77.
[69]
Pol JG, Zhang L, Bridle BW, et al. Maraba virus as a potent oncolytic vaccine vector. Mol Ther 2014; 22: 420-9.
[70]
Phase 2b/3 Trial of VSV-ΔG SARS-CoV-2 Vaccine (BRILIFE) Against Approved Comparator Vaccine. (BRILIFE002) www.clinicaltrials.gov/ct2/show/NCT04990466 (Accessed on January 27, 2022).
[71]
Ollmann Saphire E. A vaccine against Ebola virus. Cell 2020; 181: 6.
[72]
Ganar K, Das M, Sinha S, et al. Newcastle disease virus: current status and our understanding. Virus Res 2014; 184: 71-81.
[73]
Reichard K, Lorence RM, Cascino CJ, et al. Newcastle disease virus selectively kills human tumor cells. J Surg Res 1992; 52: 448-53.
[74]
Schirrmacher V, Griesbach A, Ahlert T. Antitumor effects of Newcastle disease virus in vivo: local versus systemic effects. Int J Oncol 2001; 18: 945-52.
[75]
Cheng X, Wang W, Xu Q, et al. Genetic modification of oncolytic Newcastle disease virus for cancer therapy. J Virol 2016; 90: 5343-52.
[76]
Kwak H, Honig H, Kaufman HL. (2003) Poxviruses as vectors for cancer immunotherapy. Curr Opin Drug Discov Devel 2003; 6: 161-8.
[77]
Lundstrom K. Application of Viruses for Gene Therapy and Vaccine Development The Biological Role of a Virus. Advances in Environmental Microbiology: Springer Nature, Switzerland 2022; pp. 285-341.
[78]
Zeh HJ, Bartlett DL. Development of a replication-selective oncolytic proxvirus for the treatment of human cancers. Cancer Gene Ther 2002; 9: 1001-12.
[79]
Pastoret P-P, Vanderplasschen A. Poxviruses as vaccine vectors. Comp Immunol Microbiol Infect Dis 2003; 26: 343-55.
[80]
Mastrangelo MJ, Lattime EC. Virotherapy clinical trials for regional disease: in situ immune modulation using recombinant poxvirus vectors. Cancer Gene Ther 2002; 9: 1013-21.
[81]
Madan RA, Arlen PM, Gullye JL. PANVAC-VF: Poxviral-based vaccine therapy targeting CEA and MUC1 in carcinoma. Expert Opin Biol Ther 2007; 7: 543-54.
[82]
Bradley S, Jakes AD, Harrington K, et al. Applications of coxsackievirus A21 in oncology. Oncolytic Virother 2014; 3: 47-55.
[83]
Kim D-S, Nam J-H. Application of attenuated coxsackievirus B3 as viral vector system for vaccines and gene therapy. Hum Vaccin 2011; 7: 410-6.
[84]
Jia Q, Liang F, Ohka S, et al. Expression of brain-derived neurotrophic factor in the central nervous system of mice using a poliovirus-based vector. J Neurovirol 2002; 8: 14-23.
[85]
Shafren DR, Au GG, Nguyen T, et al. Systemic therapy of malignant human melanoma tumors by a common cold-producing enterovirus, coxsackievirus a21. Clin Cancer Res 2014; 10: 53-60.
[86]
Skelding KA, Barry RD, Shafren DR. Enhanced oncolysis mediated by Coxsackievirus A21 in combination with doxorubicin hydrochloride. Invest New Drugs 2012; 30: 568-81.
[87]
Clements D, Helson E, Gujar SA, et al. Reovirus in cancer therapy: an evidence-based review. Oncol Virother 2014; 3: 69-82.
[88]
Gujar SA, Marcato P, Pan D, et al. Reovirus virotherapy overrides tumor antigen presentation evasion and promotes protective antitumor immunity. Mol Cancer Ther 2010; 9: 2924-33.
[89]
Kimchi-Sarfaty C, Gottesman MM. SV40 pseudovirions as highly efficient vectors for gene 1589 transfer and their potential application in cancer therapy. Curr Pharm Biotechnol 2004; 5: 451-8.
[90]
Toscano MG, van der Velden J, van der Werf S, et al. Generation of a Vero-based packaging cell line to produce SV40 gene delivery vectors for use in clinical gene therapy studies. Mol Ther Methods Clin Dev 2017; 6: 124-34.
[91]
Cordelier P, Bienvenu C, Lulka H, et al. Replication-deficient rSV40 mediate pancreatic gene transfer and long-term inhibition of tumor growth. Cancer Gene Ther 2007; 14: 19-29.
[92]
Liu Y, Deisseroth A. Tumor vascular targeting therapy with viral vectors. Blood 2006; 107: 3027-33.
[93]
Montaño-Samaniego M, Bravo-Estupiñan DM, Méndez-Guerrero O, et al. Strategies for Targeting Gene Therapy in Cancer Cells with Tumor-Specific Promoters. Front Oncol 2020; 10: 605380.
[94]
Eissa IR, Naoe Y, Bustos-Villalobos I, et al. Genomic signature of the natural oncolytic herpes simplex virus HF10 and its therapeutic role in preclinical and clinical trials. Front Oncol 2017; 7: 149.
[95]
Mostafa AA, Meyers DE, Thirukkumaran CM, et al. Oncolytic Reovirus and immune checkpoint inhibitor as a novel immunotherapeutic strategy for breast cancer. Cancers 2018; 10: 205.
[96]
Lin Y, Zhang H, Liang J, et al. Identification and characterization of alphavirus M1 as a selective oncolytic virus targeting ZAP-defective human cancers. Proc Natl Acad Sci USA 2014; 111: E4504-12.
[97]
Hu J, Cai XF, Yan G. Alphavirus M1 induces apoptosis of malignant glioma cells via downregulation and nucleolat translocation of p21WAF1/CIP1 protein. Cell Cycle 2009; 8: 3328-39.
[98]
Roche FP, Sheahan BJ, O’Mara SM, et al. Semliki Forest virus-mediated gene therapy of the RG2 rat glioma. Neuropathol Appl Neurobiol 2010; 36: 648-60.
[99]
Huang TT, Parab S, Burnett R, et al. Intravenous administration of retroviral replicating vector, Toca 511, demonstrates efficacy in orthotopic immune-competent mouse glioma model. Hum Gene Ther 2015; 26: 82-93.
[100]
Hoang-Le D, Smeenk L, Anraku I, et al. A Kunjin replicon vector encoding granulocyte macrophage colony-stimulating factor for intra-tumoral gene therapy. Gene Ther 2009; 16: 190-9.
[101]
Ying H, Zaks TZ, Wang R-F, et al. Cancer therapy using a self-replicating RNA vaccine. Nat Med 1999; 5: 823-7.
[102]
Niu Z, Bai F, Sun T, et al. Recombinant Newcastle disease virus expressing IL15 demonstrates promising antitumor efficiency in melanoma model. Technol Cancer Res Treat 2015; 14: 607-15.
[103]
Skelding KA, Barry RD, Shafren DR. Systemic targeting of metastatic human breast xenografts by Coxsackievirus A21. Breast Cancer Res Treat 2009; 113: 21-30.
[104]
Nagasato M, Rin Y, Yamamoto Y, et al. A tumor-targeting adenovirus with high gene transduction efficiency for primary pancreatic cancer and ascites cells. Anticancer Res 2017; 37: 3599-605.
[105]
Yamamoto Y, Nagasato M, Rin Y, et al. Strong antitumor efficacy of a pancreatic tumor targeting oncolytic adenovirus for neuroendocrine tumors. Cancer Med 2017; 6: 2385-97.
[106]
Etoh T, Himeno Y, Matsumoto T, et al. Oncolytic viral therapy for human pancreatic cancer cells by reovirus. Clin Cancer Res 2003; 9: 1218-23.
[107]
Petrulio CA, Kaufman HL. Development of the panvac-vf vaccine for pancreatic cancer. Expert Rev Vaccines 2006; 5: 9-19.
[108]
Ady JW, Johnsen C, Mojica K, et al. Oncolytic gene therapy with recombinant vaccinia virus strain GLV-2b372 efficiently kills hepatocellular carcinoma. Surgery 2015; 158: 331-8.
[109]
Mansfield DC, Kyula JN, Rosenfelder N, et al. Oncolytic vaccinia virus as a vector for 1688 therapeutic sodium iodide symporter gene therapy in prostate cancer. Gene Ther 2016; 23: 357-68.
[110]
Hirooka Y, Kasuya H, Ishikawa T, et al. A phase I clinical trial of EUS-guided intratumoral injection of the oncolytic virus, HF10 for unresectable locally advanced pancreatic cancer. BMC Cancer 2018; 18: 596.
[111]
Cloughesy TF, Landolfi J, Hogan DJ, et al. Phase I trial of vocimagine amiroretrorepvec and 5-fluorocytosine for recurrent high-grade glioma. Sci Transl Med 2016; 8: 341ra75.
[112]
Tocagen Reports Results of Toca 5 Phase 3 Trial in Recurrent Brain Cancer. Tocagen. Published September 12, 2019. https://bit.ly/2lPm19v Accessed March 2, 2022.
[113]
Galanis E, Hartmann LC, Cliby WA, et al. Phase I trial of intraperitoneal administration of an oncolytic measles virus strain engineered to express carcinoembryonic antigen for recurrent ovarian cancer. Cancer Res 2010; 70: 875-82.
[114]
Downs-Canner S, Guo ZS, Ravindranathan R, et al. Phase I study of intravenous oncolytic poxvirus (vvDD) in patients with advanced solid cancers. Mol Ther 2016; 24: 1492-501.
[115]
Pecora AL, Rizvi N, Cohen GI, et al. Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. J Clin Oncol 2002; 20: 2251-66.
[116]
Mohebtash M, Tsang KY, Madan RA, et al. A pilot study of MUC-1/CEA/TRICOM poxviral-based vaccine in patients with metastatic breast and ovarian cancer. Clin Cancer Res 2011; 17: 7164-73.
[117]
Akhter SA, Skaer CA, Kypson AP, et al. Restoration of beta-adrenergic signaling in failing cardiac ventricular myocytes via adenoviral-mediated gene transfer. Proc Natl Acad Sci USA 1997; 94: 12100-5.
[118]
Miyamoto MI, del Monte F, Schmidt U, et al. Adenoviral gene transfer of SERCa2a improves left-ventricular function in aortic-banded rats in transition to heart failure. Proc Natl Acad Sci USA 2000; 97: 793-8.
[119]
Sakata S, Lebeche D, Sakata Y, et al. Transcoronary transfer of SERCa2a increases coronary blood flow and decreases cardiomyocyte size in a type 2 diabetic rat model. Am J Physiol Heart Circ Physiol 2007; 292: H1204-7.
[120]
Zachary I, Morgan RD. Therapeutic angiogenesis for cardiovascular disease: biological context, challenges, prospects. Heart 2011; 97: 181-9.
[121]
Lazarous DF, Shou M, Stiber JA, et al. Adenoviral-mediated gene transfer induces sustained pericardial VEGF expression in dogs: effect on myocardial angiogenesis. Cardiovasc Res 1999; 44: 294-302.
[122]
Yang Z-J, Chen B, Shen Z, et al. Improvement of heart function in postinfarct heart failure swine models after hepatocyte growth factor transfer: comparison of low-, medium- and high-dose groups. Mol Biol Rep 2010; 37: 2075-81.
[123]
Igarashi T, Finet JE, Takeuchi A, et al. Connexin gene transfer preserves conduction velocity and prevents atrial fibrillation. Circulation 2012; 125: 216-25.
[124]
Amit G, Kikuchi K, Greener ID, et al. Selective molecular potassium channel blockade prevents atrial fibrillation. Circulation 2010; 121: 2263-70.
[125]
Fish KM, Ladage D, Kawase Y, et al. AAV9.I-1c delivered via direct coronary infusion in a porcine model of heart failure improves contractility and mitigates adverse remodeling. Clin Heart Fail 2013; 6: 310-7.
[126]
Wang S, Li Y, Xu Y, et al. AAV Gene Therapy Prevents and Reverses Heart Failure in a Murine Knockout Model of Barth Syndrome. Circ Res 2020; 126: 1024-39.
[127]
Hadri L, Bobe R, Kawase Y, et al. SERCA2a gene transfer enhances eNOS expression and activity in endothelial cells. Mol Ther 2010; 18: 1284-92.
[128]
Raake PWJ, Schlegel P, Ksienzyk J, et al. AAV6.βARKct cardiac gene therapy ameliorates cardiac function and normalizes the catecholaminergic axis in a clinically relevant large animal heart failure model. Eur Heart J 2013; 34: 1437-47.
[129]
Qian L, Huang Y, Spencer CI, et al. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature 2012; 485: 593-8.
[130]
Niwano K, Arai M, Koitabashi N, et al. Lentiviral vector–mediated SERCA2 gene transfer protects against heart failure and left ventricular remodeling after myocardial infarction in rats. Mol Ther 2008; 16: 1026-32.
[131]
Rincon MY, VandenDriessche T, Chuah MK. Gene therapy for cardiovascular disease: advances in vector development, targeting, and delivery for clinical translation. Cardiovasc Res 2015; 108: 4-20.
[132]
Stewart DJ, Hilton JD, Arnold JMO, et al. Angiogenic gene therapy in patients with nonrevascularizable ischemic heart disease: a phase 2 randomized, controlled trial of AdVEGF(121) (AdVEGF121) versus maximum medical treatment. Gene Ther 2006; 13: 1503-11.
[133]
Leikas AJ, Hassinen I, Hedman A, et al. Long-term safety and efficacy of intramyocardial adenovirus-mediated VEGF-DΔNΔC gene therapy eight-year follow-up of phase I KAT301 study. Gene Ther 2021.
[http://dx.doi.org/10.1038/s41434-021-00295-1]
[134]
Grines CL, Watkins MW, Helmer G, et al. Angiogenic Gene Therapy (AGENT) trial in patients with stable angina pectoris. Circulation 2002; 105: 1291-7.
[135]
Grines CL, Watkins MW, Mahmarian JJ, et al. A randomized, double-blind, placebo-controlled trial of Ad5FGF-4 gene therapy and its effect on myocardial perfusion in patients with stable angina. J Am Coll Cardiol 2003; 42: 1339-47.
[136]
Henry TD, Grines CL, Watkins MW, et al. Effects of Ad5FGF-4 in patients with angina: an analysis of pooled data from the AGENT-3 and AGENT-4 trials. J Am Coll Cardiol 2007; 50: 1038-46.
[137]
Jaski BE, Jessup ML, Mancini DM, et al. Calcium Up-Regulation by Percutaneous Administration of Gene Therapy In Cardiac Disease (CUPID) Trial Investigators. Calcium upregulation by percutaneous administration of gene therapy in cardiac disease (CUPID Trial), a first-in-human phase 1/2 clinical trial. J Card Fail 2009; 15: 171-81.
[138]
Jessup M, Greenberg B, Mancini D, et al. Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID) Investigators. Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID): a phase 2 trial of intracoronary gene therapy of sarcoplasmic reticulum Ca2+-ATPase in patients with advanced heart failure. Circulation 2011; 124: 304-13.
[139]
Zsebo K, Yaroshinsky A, Rudy JJ, et al. Long-term effects of AAV1/SERCA2a gene transfer in patients with severe heart failure: analysis of recurrent cardiovascular events and mortality. Circ Res 2014; 114: 101-8.
[140]
Alexander IE, Cunningham SC, Logan GJ, et al. Potential of AAV vectors in the treatment of metabolic disease. Gene Ther 2008; 15: 831-9.
[141]
Watson GL, Sayles JN, Chen C, et al. Treatment of lysosomal storage disease in MPS VII mice using a recombinant adeno-associated virus. Gene Ther 1998; 5: 1642-9.
[142]
Moscioni D, Morizono H, McCarter RJ, et al. Long-term correction of ammonia metabolism and prolonged survival in ornithine transcarbamylase-deficient mice following liver-directed treatment with adeno-associated viral vectors. Mol Ther 2006; 14: 25-33.
[143]
Lebherz C, Gao G, Louboutin JP, et al. Gene therapy with novel adeno-associated virus vectors substantially diminishes atherosclerosis in a murine model of familial hypercholesterolemia. J Gene Med 2004; 6: 663-72.
[144]
Jimenez V, Jambrina C, Casana E, et al. FGF21 gene therapy as treatment for obesity and insulin resistance. EMBO Mol Med 2018; 10: e8791.
[145]
Xu J, Lu Y, Ding F, et al. Reversal of diabetes in mice by intrahepatic injection of bone-derived GFP-murine mesenchymal stem cells infected with the recombinant retrovirus-carrying human insulin gene. World J Surg 2007; 31: 1872-82.
[146]
Hegde V, Na H-N, Dubuisson O. An adenovirus-derived protein: a novel candidate for anti-diabetic drug development. Biochemie 2016; 121: 140-50.
[147]
D’Avola D, López-Franco E, Sangro B, et al. Phase I open label liver-directed gene therapy clinical trial for acute intermittent porphyria. J Hepatol 2016; 65: 776-83.
[148]
Brantly ML, Chulay JD, Wang L, et al. Proc Natl Acad Sci USA 2009; 106: 16363-8.
[149]
Flotte TR, Trapnell BC, Humphries M, et al. Phase 2 clinical trial of a recombinant adeno-associated viral vector expressing alpha1- antitrypsin: interim results. Hum Gene Ther 2011; 22: 1239-47.
[150]
Balagué C, Zhou J, Dai Y, et al. Sustained high-level expression of full-length human factor VIII and a restoration of clotting activity in hemophilic mice using a minimal adenovirus vector. Blood 2000; 95: 820-8.
[151]
Dai Y, Schwarz EM, Gu D, et al. Cellular and humoral immune responses to adenoviral vectors containing factor IX gene: tolerization of factor IX and vector antigens allows long-term expression. Proc Natl Acad Sci USA 1995; 92: 1401-5.
[152]
Kay MA, Landen CN, Rothenberg SR, et al. In vivo hepatic gene therapy: complete albeit transient correction of factor IX deficiency in hemophilia B dogs. Proc Natl Acad Sci USA 1994; 91: 2353-7.
[153]
Fang B, Eisensmith RC, Wang H, et al. Gene therapy for hemophilia B: host immunosuppression prolongs the therapeutic effect of adenovirus-mediated factor IX expression. Hum Gene Ther 1995; 6: 1039-44.
[154]
Crudele JM, Finn JD, Siner JI, et al. AAV liver expression of FIX-Padua prevents and eradicates FIX inhibitor without increasing thrombogenicity in hemophilia B dogs and mice. Blood 2015; 125: 1553-61.
[155]
Doering CB, Denning G, Shields JE, et al. Preclinical development of a hematopoietic stem and progenitor cell bioengineered factor VIII lentiviral vector gene therapy of hemophilia A. Hum Gene Ther 2018; 29: 1183-201.
[156]
Marcos-Contreras OA, Smith SM, Bellinger DA, et al. Sustained correction of FVII deficiency in dogs using AAV-mediated expression of zymogen FVII. Blood 2016; 127: 565-71.
[157]
Garcia-Gomez M, Calabria A, Garcia-Bravo M, et al. Safe and efficient gene therapy for pyruvate kinase deficiency. Mol Ther 2016; 24: 1187-98.
[158]
Huang NF, Niiyama H, Peter C, et al. Embryonic stem cell-derived endothelial cells engraft into the ischemic hindlimb and restore perfusion. Arterioscler Thromb Vasc Biol 2010; 30: 984-91.
[159]
Saqib A, Prasad K-MR, Katwal AB, et al. Adeno-associated virus serotype 9-mediated overexpression of extracellular superoxide dismutase improves recovery from surgical hind limb ischemia in BALB/c mice. J Vasc Surg 2011; 54: 810-8.
[160]
Chapin JC, Monahan PE. Gene therapy for hemophilia. BioDrugs 2018; 32: 9-25.
[161]
Leebeek FW, Miesbach W. Gene therapy for hemophilia: a review on clinical benefit, limitations, and remaining issues. Blood 2021; 138: 923-31.
[162]
Spencer HT, Riley BE, Doering CB. State of the art: gene therapy of haemophilia. Haemophilia 2016; 22 (Suppl. 5): 66-71.
[163]
Nathwani AC. Gene therapy for hemophilia. Hematology (Am Soc Hematol Educ Program) 2019; 2019: 1-8.
[164]
Nathwani AC, Reiss UM, Tuddenham EG, et al. Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N Engl J Med 2014; 371: 1994-2004.
[165]
Kim J, Yoon YS, Lee H, et al. AAV-GAD gene for rat models of neuropathic pain and Parkinson’s disease. Acta Neurochir 2008; (Suppl. 101)99-105.
[166]
Fan D, Shen Y, Kang D, et al. Adeno-associated virus vector-mediated triple gene transfer of dopamine synthetic enzymes. Chin Med J 2001; 114: 1276-9.
[167]
Björklund A, Kirik D, Rosenblad C, et al. Towards a neuroprotective gene therapy for Parkinson’s disease: use of adenovirus, AAV and lentivirus vectors for gene transfer of GDNF to the nigrostriatal system in the rat Parkinson model. Brain Res 2000; 886: 82-98.
[168]
Kordower JH, Emborg ME, Bloch JH, et al. Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate model of Parkinson’s disease. Science 2000; 290: 767-73.
[169]
1 Wu K, Meyer EM, Bennett JA, et al. AAV2/5-mediated NGF gene delivery protects septal cholinergic neurons following axotomy. Brain Res 2005; 1061: 107–13.
[170]
Fol R, Bradeau J, Ludewig S, et al. Viral gene transfer of APPsα rescues synaptic failure in an Alzheimer’s disease model. Acta Neuropathol 2016; 131: 247-66.
[171]
Revilla S, Ursulet S, Alvarez-Lopez JM, et al. Lenti-GDNF gene therapy protects against Alzheimer’s disease-like neuropathology in 3xTg-AD mice and MC65 cells. CNS Neurosci Ther 2014; 20: 961-72.
[172]
Zeng C-Y, Yang T-T, Zhou H-J, et al. Lentiviral vector-mediated overexpression of klotho in the brain improves Alzheimer’s disease-like pathology and cognitive deficits in mice. Neurobiol Aging 2019; 78: 18-28.
[173]
Rosenberg JB, Kaplitt MG, De BP, et al. AAVrh.10-mediated APOE2 central nervous system gene therapy for APOE4-associated Alzheimer’s disease. Hum Gene Ther Clin Dev 2018; 29: 24-47.
[174]
Miniarikova J, Zimmer V, Martier R, et al. AAV5-miHTT gene therapy demonstrates suppression of huntingtin aggregation and neuronal dysfunction in a rat model of Huntington’s disease. Gene Ther 2017; 24: 630-9.
[175]
Evers MM, Miniarikova J, Juhas S, et al. AAV5-miHTT gene therapy demonstrates broad distribution and strong human mutant huntingtin lowering in Huntington’s disease minipig model. Mol Ther 2018; 26: 2163-77.
[176]
Sinnett SE, Gray SJ. Recent endeavors in MECP2 gene transfer for gene therapy of Rett syndrome. Discov Med 2017; 24: 153-9.
[177]
Eberling JL, Jagust WJ, Christine CW, et al. Results from a phase I safety trial of hAADC gene therapy for Parkinson disease. Neurology 2008; 70: 1980-3.
[178]
Palfi S, Gurruchaga JM, Scott Ralph G, et al. Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson’s disease: a dose escalation, open-label, phase 1/2 trial. Lancet 2014; 383: 1138-46.
[179]
Rafii MS, Tuszynski MH, Thomas RG, et al. Adeno-associated viral vector (Serotype 2)–nerve growth factor for patients with Alzheimer Disease. JAMA Neurol 2018; 75: 834-41.
[180]
Pattali R, Mou Y, Li X-J. AAV9 vector: a novel modality in gene therapy for spinal muscular atrophy. Gene Ther 2019; 26: 287-95.
[181]
Mendell JR, Al-Zaidy S, Schell R, et al. Single-dose gene-replacement therapy for spinal muscular atrophy. N Engl J Med 2017; 377: 1713-22.
[182]
Martier R, Konstantinova P. Gene Therapy for Neurodegenerative Diseases: Slowing Down the Ticking Clock. Front Neurosci 2020; 14: 580179.
[183]
Gao W, Soloff AC, Lu X, et al. Protection of mice and poultry from lethal HSN1 avian influenza virus through adenovirus-based immunization. J Virol 2006; 80: 1959-64.
[184]
Schultz-Cherry S, Dybing JK, Davis NL, et al. Influenza virus (a/HK/156/97) hemagglutinin expressed by an alphavirus replicon system protects against lethal infection with Hong Kong origin H5N1 viruses. Virology 2000; 278: 55-9.
[185]
Fleeton MN, Chen M, Berglund P, et al. Self-replicative RNA vaccines elicit protection against influenza a virus, respiratory syncytial virus, and a tickborne encephalitis virus. J Infect Dis 2001; 183: 1395-8.
[186]
Chattopadhyay A, Aquila PV, Bopp NE, et al. A recombinant virus vaccine that protects both against Chikungunya and Zika virus infections. Vaccine 2018; 36: 3894-900.
[187]
Lin J, Calcedo R, Vanderberghe LH, et al. Impact of preexisting vector immunity on the efficacy of adeno-associated virus-based HIV-1 gag vaccines. Hum Gene Ther 2008; 19: 663-9.
[188]
Rodr GD, Rodr GJR, Llorente M, et al. A human immunodeficiency virus type 1 Env-granulocyte-macrophage-colony-stimulating factor fusion protein enhances the cellular immune response to Env in a vaccinia virus-based vaccine. J Gen Virol 1999; 80: 217-23.
[189]
McGettigan JP, Foley HD, Belyakov IM, et al. Rabies virus-based vectors expressing human immunodeficiency virus type 1 (HIV-1) envelope protein induce a strong, cross-reactive cytotoxic T-lymphocyte response against envelope proteins from different HIV-1 isolates. J Virol 2001; 75: 4430-4.
[190]
Ajbani SP, Velhal SM, Kadam RB, et al. Immunogenicity of virus-like Semliki Forest virus replicon particles expressing Indian HIV-1C gag, env and pol RT genes. Immunol Lett 2017; 190: 221-32.
[191]
Safronetz D, Mire C, Rosenke K, et al. A recombinant vesicular stomatitis virus-based Lassa fever vaccine protects guinea pigs and macaques against challenge with geographically and genetically distinct Lassa viruses. PLoS Negl Trop Dis 2015; 9: e0003736.
[192]
Maruyama J, Mateer EJ, Manning JT, et al. Adenoviral vector-based vaccine is fully protective against lethal Lassa fever challenge in Hartley guinea pigs. Vaccine 2019; 37: 6824-31.
[193]
Fischer RJ, Purushotham JN, van Doremalen N, et al. ChAdOx1-vectored Lassa fever vaccine elicits a robust cellular and humoral immune response and protects guinea pigs against lethal Lassa virus challenge. npj. Vaccines 2020; 6: 32.
[194]
Wilson JA, Hart MK. Protection from Ebola virus mediated by cytotoxic T-lymphocytes specific for the viral nucleoprotein. J Virol 2001; 75: 2660-4.
[195]
Marzi A, Robertson SJ, Haddock E, et al. Ebola vaccine. VSV-EBOV rapidly protects macaques against infection with the 2014/2015 Ebola virus outbreak strain. Science 2015; 349: 739-42.
[196]
Lundstrom K. The Current Status of COVID-19 Vaccines. Front Genome Edit 2020; 2: 579297.
[197]
Lundstrom K. Viral vectors for COVID-19 vaccine development. Viruses 2021; 13: 317.
[198]
Faber M, Lamirande EW, Roberts A, et al. A single immunization with a rhabdovirus-based vector expressing severe acute respiratory syndrome coronavirus (SARS-CoV) S protein results in the production of high levels of SARS-CoV-neutralizing antibodies. J Gen Virol 2005; 86: 1435-40.
[199]
Deming D, Sheahan T, Heise M, et al. Vaccine efficacy in senescent mice challenged with recombinant SARS-CoV bearing epidemic and zoonotic spike variants. PLoS Med 2006; 3: e525.
[200]
Haagmans BL, van den Brand JM, Raj VS, et al. An orthopoxvirus-based vaccine reduces virus excretion after MERS-CoV infection in dromedary camels. Science 2016; 351: 77-81.
[201]
van Doremalen N, Lambe T, Spencer A, et al. ChAdOx1 nCov-19 vaccination prevents SARS-CoV-2 pneumonia in rhesus macaques. Nature 2020; 586: 578-82.
[202]
Wu S, Zhong G, Zhang J, et al. A single dose of an adenovirus-vectored vaccine provides protection against SARS-CoV-2 challenge. Nat Commun 2020; 11: 4081.
[203]
Feng L, Wang Q, Shan C, et al. An adenovirus-vectored COVID-19 vaccine confers protection from SARS-CoV-2 challenge in rhesus macaques. Nat Commun 2020; 11: 4207.
[204]
Balakrishnan VVS. The arrival of Sputnik V. Lancet Comm 2020; 20: 1128.
[205]
Tostanoski LH, Wegmann F, Martinot J, et al. Ad26 vaccine protects against SARS-CoV-2 severe clinical disease in hamsters. Nat Med 2020; 26: 1694-700.
[206]
Mercado NB, Zahn R, Wegmann F, Loos C, et al. Single-shot Ad26 vaccine protects against SARS-CoV-2 in rhesus macaques. Nature 2020; 586: 583-8.
[207]
Erasmus JH, Khandhar AP, O’Connor MA, et al. An Alphavirus-derived replicon RNA vaccine induces SARS-CoV-2 neutralizing antibody and T cell responses in mice and nonhuman primates. Sci Transl Med 2020; 12: eabc9396.
[208]
Purushotham J, Lambe T, Gilbert SC. Vaccine platforms for the prevention of Lassa fever. Immunol Lett 2019; 215: 1-11.
[209]
Henao-Restrepo AM, Longini IM, Egger M, et al. Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: interim results from the Guinea ring vaccination cluster-randomised trial. Lancet 2015; 386: 857-66.
[210]
Henao-Restrepo AM, Camacho A, Longini IM, et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ca Suffit!). Lancet 2017; 389: 505-18.
[211]
Folegatti PM, Ewer KJ, Aley PK, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2 single-blind, randomised controlled trial. Lancet 2020; 396: 467-78.
[212]
Voysey M, Costa Clemens SA, Madhi SA, et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 2021; 397: 99-111.
[213]
Zhu FC, Guan XH, Li YH, et al. Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2020; 396: 479-88.
[214]
Wu S, Huang J, Zhang Z, et al. Safety, tolerability, and immunogenicity of an aerosolised adenovirus type-5 vector-based COVID-19 vaccine (Ad5-nCoV) in adults: preliminary report of an open-label and randomised phase 1 clinical trial. Lancet Infect Dis 2021; 21: 1654-64.
[215]
Logunov DY, Dolzhikova IV, Zubkova OV, et al. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia. Lancet 2020; 396: 887-97.
[216]
Logunov DY, Dolzhikova IV, Shcheblyakov DV, et al. Safety and efficacy of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19: an interim analysis of a randomised controlled phase 3 in Russia. Lancet 2021; 397: 671-81.
[217]
Sadoff J, Le Gars M, Shukarev G, et al. Interim results of a phase 1-2a trial of Ad26.COV.S Covid-19 vaccine. N Engl J Med 2021; 384: 1824-35.
[218]
Stephenson KE, Le Gars M, Sadoff J, et al. Immunogenicity of the Ad26.COV2.S vaccine for COVID-19. JAMA 2021; 325: 1535-44.
[219]
Callaway E. Russia’s fast-track coronavirus vaccine draws outrage over safety. Nature 2020; 584: 334-5.
[220]
Martin KRG, Quigley HA, Zack DJ, et al. Gene therapy with brain-derived neurotrophic factor as a protection: retinal ganglion cells in a rat glaucoma model. Invest Ophthalmol Vis Sci 2003; 44: 4357-65.
[221]
O’Callaghan J, Crosbie DE, Cassidy PS, et al. Therapeutic potential of AAV-mediated MMP-3 secretion from corneal endothelium in treating glaucoma. Hum Mol Genet 2017; 26: 1230-46.
[222]
Bush RA, Zeng Y, Colosi P, et al. Preclinical dose-escalation study of intravitreal AAV-RS1 gene therapy in a mouse model of X-linked retinoschisis: dose-dependent expression and improved retinal structure and function. Hum Gene Ther 2016; 27: 376-89.
[223]
Feathers KL, Jia L, Dayanthi Perera N, et al. Development of a gene therapy vector for RDH12-associated retinal dystrophy. Hum Gene Ther 2019; 30: 1325-35.
[224]
Hassall MM, Barnard AR, MacLaren RE. Gene Therapy for Color Blindness. Yale J Biol Med 2017; 90: 543-51.
[225]
Alexander JJ, Umino Y, Everhart D, et al. Restoration of cone vision in a mouse model of achromatopsia. Nat Med 2007; 13: 685-7.
[226]
Komáromy AM, Alexander JJ, Rowlan JS, et al. Gene therapy rescues cone function in congenital achromatopsia. Hum Mol Genet 2010; 19: 2581-93.
[227]
Heier JS, Kherani S, Desai S, et al. Intravitreous injection of AAV2-sFLT01 in patients with advanced neovascular age-related macular degeneration: a phase I, open-label trial. Lancet 2017; 390: 50-61.
[228]
Constable IJ, Pierce CM, Lai C-M, et al. Phase 2a randomized clinical trial: safety and post hoc analysis of subretinal rAAV.sFLT-1 for wet age-related macular degeneration. EBioMedicine 2016; 14: 168-75.
[229]
Ratican SE, Osborne A, Martin KR. Progress in gene therapy to prevent retinal ganglion cell loss in glaucoma and Leber’s hereditary optic neuropathy. Neural Plast 2018; 2018: 7108948.
[230]
Wan X, Pei H, Zhao M-J, et al. Efficacy and safety of rAAV2-ND4 treatment for Leber’s hereditary optic neuropathy. Sci Rep 2016; 6: 2016.
[231]
Guy J, Feuer WJ, Davis JL, et al. Gene therapy for Leder hereditary optic neuropathy: low and medium-dose visual results. Ophthalmology 2017; 124: 1621-34.
[232]
Vignal C, Uretsky S, Fitoussi S, et al. Safety of rAAV2/2-ND4 gene therapy. Ophthalmology 2018; 125: 945-7.
[233]
Russell S, Bennett J, Wellman JA, et al. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial. Lancet 2017; 390: 849-60.
[234]
Chamberlain JR, Chamberlain JS. Progress toward gene therapy for Duchenne muscular dystrophy. Mol Ther 2017; 25: 1125-31.
[235]
Yuasa K, Miaygoe Y, Yamamoto K, et al. Effective restoration of dystrophin-associated proteins in vivo by adenovirus-mediated transfer of truncated dystrophin cDNAs. FEBS Lett 1998; 425: 329-36.
[236]
Sakamoto M, Yuasa K, Yoshimura M, et al. Micro-dystrophin cDNA ameliorates dystrophic phenotypes when introduced into mdx mice as a transgene. Biochem Biophys Res Commun 2002; 293: 1265-72.
[237]
Gregorevic P, Allen JM, Minami E, et al. rAAV6-microdystrophin preserves muscle function and extends lifespan in severely dystrophic mice. Nat Med 2006; 12: 787-9.
[238]
Wang Z, Storb R, Halbert CL, et al. Successful regional delivery and long-term expression of 1958 a dystrophin gene in canine muscular dystrophy: a preclinical model for human therapies. Mol Ther 2012; 20: 1501-7.
[239]
Malerba A, Klein P, Bachtarzi H, et al. PABPN1 gene therapy for oculopharyngeal muscular dystrophy. Nat Commun 2017; 8: 14848.
[240]
Valori CF, Ning K, Wyles M, et al. Systemic delivery of scAAV9 expressing SMN prolongs survival in a model of spinal muscular atrophy. Sci Transl Med 2010; 235: 35ra42.
[241]
Duque SI, Arnold WD, Odermatt P, et al. A large animal model of spinal muscular atrophy and correction of phenotype. Ann Neurol 2015; 77: 399-414.
[242]
Mendell JR, Sahenk Z, Lehman K, et al. (2020) Assessment of systemic delivery of rAAVrh74. MHCK7.micro-dystrophin in children with Duchenne muscular dystrophy. JAMA Neurol 2020; 77: 1122-31.
[243]
Flotte TR, Afione SA, Conrad C, et al. Stable in vivo expression of the cystic fibrosis transmembrane conductance regulator with an adeno-associated virus vector. Proc Natl Acad Sci USA 1993; 90: 10613-7.
[244]
O’Sullivan BP, Freedman SD. Cystic fibrosis. Lancet 2009; 373(9678): 1891-904.
[245]
Conrad CK, Allen SS, Afione SA, et al. Safety of single-dose administration of an adeno1348 associated virus (AAV)-CFTR vector in the primate lung. Gene Ther 1996; 3: 658-68.
[246]
Cao H, Ouyang H, Grasemann H, et al. Transducing Airway Basal Cells with a Helper-Dependent Adenoviral Vector for Lung Gene Therapy. Hum Gene Ther 2018; 29: 643-52.
[247]
Marquez Loza LI, Yuen EC, McCray PB Jr. Lentiviral vectors for the treatment and prevention of cystic fibrosis lung disease. Genes 2019; 10: 218.
[248]
Limberis M, Anson DS, Fuller M, et al. Recovery of airway cystic fibrosis transmembrane conductance regulator function in mice with cystic fibrosis after single-dose lentivirus-mediated gene transfer. Hum Gene Ther 2002; 13: 1961-70.
[249]
Farrow N, Miller D, Cmielewski P, et al. Airway gene transfer in a non-human primate: lentiviral gene expression in marmoset lungs. Sci Rep 2013; 3: 1287.
[250]
Mitomo K, Griesenbach U, Inoue M, et al. Toward gene therapy for cystic fibrosis using a lentivirus pseudotyped with Sendai virus envelopes. Mol Ther 2010; 18: 1173-82.
[251]
Cooney AL, Abou Alaiwa MH, Shah VS, et al. Lentiviral-mediated phenotypic correction of cystic fibrosis pigs. JCI Insight 2016; 1: e88730.
[252]
Alton EWFW, Beekman JM, Boyd AC, et al. Preparation for a first-in-man lentivirus trial in patients with cystic fibrosis. Thorax 2017; 72: 137-47.
[253]
Poletti V, Charrier S, Corre G, et al. Preclinical Development of a Lentiviral Vector for Gene Therapy of X-Linked Severe Combined Immunodeficiency. Mol Ther Methods Clin Dev 2018; 9: 257-69.
[254]
Poletti V, Charrier S, Martin S, et al. Preclinical Development of Gene Therapy for X-linked Severe Combined Immunodeficiency (SCID-X1). Blood 2016; 128: 4705.
[255]
Cavazzana-Calvo M, Hacein-Bey S, de Saint BG, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 2000; 288: 669-72.
[256]
Fischer A, Hacein-Bey-Abina S. Gene therapy for severe combined immunodeficiencies and beyond. J Exp Med 2020; 217: e20190607.
[257]
Gaspar HB, Parsley KL, Howe S, et al. Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector. Lancet 2004; 364: 2181-7.
[258]
Howe SJ, Mansour MR, Schwarzwaelder K, et al. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest 2008; 118: 3143-50.
[259]
Cuvelier GDE, Logan BR, Prockop SE, et al. Outcomes following treatment of ADA-deficient severe combined immune deficiency: a report from the PIDTC. Blood 2022; 140: 685-705.
[260]
Kohn DB, Booth C, Shaw KL, et al. Autologous Ex Vivo Lentiviral Gene Therapy for Adenosine Deaminase Deficiency. N Engl J Med 2021; 384: 2002-13.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy