781
Views
75
CrossRef citations to date
0
Altmetric
Reviews

Safety profile of recombinant adeno-associated viral vectors: focus on alipogene tiparvovec (Glybera®)

, &

References

  • Büning H. Gene therapy enters the pharma market: The short story of a long-journey. EMBO Mol. Med. 5, 1–3 (2013).
  • Santamarina-Fojo S. The familial chylomicronemia syndrome. Endocrinol. Metab. Clin. North Am. 27, 551–567 (1998).
  • Gaudet D, Methot J, Dery S et al. Efficacy and long-term safety of alipogenetiparvovec (AAV1-LPLS447X) gene therapy for lipoprotein lipase deficiency: an open-label trial. Gene Ther. 20, 361–369 (2013).
  • Carpentier AC, Frisch F, Labbé SM et al. Effect of alipogenetiparvovec (AAV1-LPL(S447X)) on postprandial chylomicron metabolism in lipoprotein lipase-deficient patients. J. Clin. Endocrinol. Metab. 97, 1635–1644 (2012).
  • Bryant LM, Christopher DM, Giles AR et al. Lessons learned from the clinical development and market authorization of glybera. Hum. Gene. Ther. Clin. Dev. 24, 55–64 (2013).
  • Tanne JH. US gene therapy trial is to restart, despite patient’s death. BMJ 335, 1172–1173 (2007).
  • Flotte TR, Conlon TJ, Poirier A, Campbell-Thompson M, Byrne BJ. Preclinical characterization of a recombinant adeno-associated virus type 1-pseudotyped vector demonstrates dose-dependent injection site inflammation and dissemination of vector genomes to distant sites. Hum. Gene. Ther. 18, 245–256 (2007).
  • Louboutin JP, Wang L, Wilson JM. Gene transfer into skeletal muscle using novel AAV serotypes. J. Gene. Med. 7, 442–451 (2005).
  • Riviere C, Danos O, DouarAM. Long-term expression and repeated administration of AAV type 1, 2 and 5 vectors in skeletal muscle of immunocompetent adult mice. Gene. Ther. 13, 1300–1308 (2006).
  • Lorain S, Gross DA, Goyenvalle A, Danos O, Davoust J, Garcia L. Transient immunomodulation allows repeated injections of AAV1 and correction of muscular dystrophy in multiple muscles. Mol. Ther. 16, 541–547 (2008).
  • Stroes ES, Nierman MC, Meulenberg JJ et al. Intramuscular administration of AAV1-lipoprotein lipase S447X lowers triglycerides in lipoprotein lipase-deficient patients. Arterioscler. Thromb. Vasc. Biol. 28, 303–304 (2008).
  • Mingozzi F, Meulenberg JJ, Hui DJ et al. AAV-1-mediated gene transfer to skeletal muscle in humans results in dose-dependent activation of capsid-specific T cells. Blood 114, 2077–2086 (2009).
  • Flotte TR, Trapnell BC, Humphries M et al. Phase 2 clinical trial of a recombinant adeno-associated viral vector expressing α1-antitrypsin: interim results. Hum. Gene. Ther. 22, 1239–1247 (2011).
  • Bowles DE, McPhee SW, Li C, Gray SJ, Samulski JJ, Camp AS et al. Phase 1 gene therapy for Duchenne muscular dystrophy using a translational optimized AAV vector. Mol. Ther. 20, 443–455 (2012).
  • Manno CS, Chew AJ, Hutchison S et al. AAV-mediated factor IX gene transfer to skeletal muscle in patients with severe hemophilia B. Blood 101, 2963–2972 (2003).
  • Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat. Med. 12, 342–347 (2006).
  • Nathwani AC, Tuddenham EG, Rangarajan S, Rosales C, McIntosh J, Linch DC et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N. Engl. J. Med. 365, 2357–2365 (2011).
  • Mingozzi F, High KA. Immune responses to AAV vectors: overcoming barriers to successful gene therapy. Blood 122, 23–36 (2013).
  • High KA, Aubourg P. rAAV human trial experience. Methods Mol. Biol. 807, 429–457 (2011).
  • Mays LE, Wilson JM. The complex and evolving story of T cell activation to AAV vector-encoded transgene products. Mol. Ther. 19, 16–27 (2011).
  • Bainbridge JW, Smith AJ, Barker SS et al. Effect of gene therapy on visual function in Leber's congenital amaurosis. N. Engl. J. Med. 358, 2231–2239 (2008).
  • Brantly ML, Chulay JD, Wang L et al. Sustained transgene expression despite T lymphocyte responses in a clinical trial of rAAV1-AAT gene therapy. Proc. Natl Acad. Sci. U.S.A. 106, 16363–16368 (2009).
  • Cideciyan AV, Aleman TS, Boye SL et al. Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics. Proc. Natl Acad. Sci. USA 105, 15112–15117 (2008).
  • 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. 15, 171–181 (2009).
  • Kaplitt MG, Feigin A, Tang C et al. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial. Lancet 369, 2097–2105 (2007).
  • Kay MA, Manno CS, Ragni MV et al. Evidence for gene transfer and expression of factor IX in haemophilia B patients treated with an AAV vector. Nat. Genet. 24, 257–261 (2000).
  • Maguire AM, Simonelli F, Pierce EA et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N. Engl. J. Med. 358, 2240–2248 (2008).
  • Maguire AM, High KA, Auricchio A et al. Age-dependent effects of RPE65 gene therapy for Leber's congenital amaurosis: a phase 1 dose-escalation trial. Lancet 374, 1597–1605 (2009).
  • Mendell JR, Rodino-Klapac LR et al. Limb-girdle muscular dystrophy type 2D gene therapy restores alpha-sarcoglycan and associated proteins. Ann. Neurol. 66, 290–297 (2009).
  • Jacobson SG, Cideciyan AV, Ratnakaram R et al. Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years. Arch. Ophthalmol. 130, 9–24 (2012).
  • McPhee SW, Janson CG, Li C et al. Immune responses to AAV in a phase I study for Canavan disease. J. Gene. Med. 8, 577–588 (2006).
  • Mingozzi F, Maus MV, Hui DJ et al. CD8(+) T-cell responses to adeno-associated virus capsid in humans. Nat. Med. 13, 419–422 (2007).
  • Velazquez VM, Bowen DG, Walker CM. Silencing of T lymphocytes by antigen-driven programmed death in recombinant adeno-associated virus vector-mediated gene therapy. Blood 113(3), 538–545 (2009).
  • Lin SW, Hensley SE, Tatsis N, Lasaro MO, Ertl HC. Recombinant adeno-associated virus vectors induce functionally impaired transgene product-specific CD8+ T cells in mice. J. Clin. Invest. 117(12), 3958–3970 (2007).
  • Asokan A, Schaffer DV, Samulski RJ. The AAV vector toolkit: poised at the clinical crossroads. Mol. Ther. 20, 699–708 (2012).
  • Rabinowitz JE, Rolling F, Li C et al. Cross-packaging of a single adeno-associated virus (AAV) type 2 vector genome into multiple AAV serotypes enables transduction with broad specificity. J. Virol. 76, 791–801 (2002).
  • Davidoff AM, Gray JT, Ng CY et al. Comparison of the ability of adeno-associated viral vectors pseudotyped with serotype 2, 5, and 8 capsid proteins to mediate efficient transduction of the liver in murine and nonhuman primate models. Mol. Ther. 11, 875–88 (2005).
  • Nathwani AC, Gray JT, Ng CY et al. Self-complementary adeno-associated virus vectors containing a novel liver-specific human factor IX expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood 107, 2653–2661 (2006).
  • Paneda A, Vanrell L, Mauleon I et al. Effect of adeno-associated virus serotype and genomic structure on liver transduction and biodistribution in mice of both genders. Hum. Gene. Ther. 20, 908–917 (2009).
  • Nathwani AC, Gray JT, Ng CY et al. Self-complementary adeno-associated virus vectors containing a novel liver-specific human factor IX expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood 107, 2653–2661 (2006).
  • Nathwani AC, Rosales C, McIntosh J et al. Long-term safety and efficacy following systemic administration of a self-complementary AAV vector encoding human FIX pseudotyped with serotype 5 and 8 capsid proteins. Mol. Ther. 19, 876–885 (2011).
  • Qiao C, Zhang W, Yuan Z et al. Adeno-associated virus serotype 6 capsid tyrosine-to-phenylalanine mutations improve gene transfer to skeletal muscle. Hum. Gene. Ther. 21, 1343–1348 (2010).
  • Pulicherla N, Shen S, Yadav S et al. Engineering liver-detargeted AAV9 vectors for cardiac and musculoskeletal gene transfer. Mol. Ther. 19, 1070–1078 (2011).
  • Zhong L, Li B, Mah CS, Govindasamy L et al. Next generation of adeno-associated virus 2 vectors: point mutations in tyrosines lead to high-efficiency transduction at lower doses. Proc. Natl Acad. Sci. USA 105(22), 7827–7832 (2008).
  • Markusic DM, Herzog RW, Aslanidi GV et al. High-efficiency transduction and correction of murine hemophilia B using AAV2 vectors devoid of multiple surface-exposed tyrosines. Mol. Ther. 18(12), 2048–2056 (2010).
  • Aslanidi GV, Rivers AE, Ortiz L et al. Optimization of the capsid ofrecombinantadeno-associated virus 2 (AAV2) vectors: the final threshold? PLoS ONE 8(3), e59142 (2013).
  • Wang L, Louboutin JP, Bell P et al. Muscle-directed gene therapy for hemophilia B with more efficient and less immunogenic AAV vectors. J. Thromb. Haemost. 9, 2009–2019 (2011).
  • Varadi K, Michelfelder S, Korff T et al. Novel random peptide libraries displayed on AAV serotype 9 for selection of endothelial cell-directed gene transfer vectors. Gene. Ther. 19, 800–809 (2012).
  • Hackett NR, El Sawy T, Lee LY et al. Use of quantitative TaqMan real-time PCR to track the time-dependent distribution of gene transfer vectors in vivo. Mol. Ther. 2, 649–656 (2000).
  • Gonin P, Gaillard C. Gene transfer vector biodistribution: pivotal safety studies in clinical gene therapy development. Gene. Ther. 11( Suppl. 11), S98-S108 (2004).
  • Brandon EF, Hermsen HP, van Eijkeren JC, Tiesjema B. Effect of administration route on the biodistribution and shedding of replication-deficient AAV2: a qualitative modelling approach. Curr. Gene. Ther. 10, 91–106 (2010).
  • Le Guiner C, Moullier P, Arruda VR. Biodistribution and shedding of AAV vectors. Methods Mol. Biol. 807, 339–359 (2011).
  • Favre D, Provost N, Blouin V A et al. Immediate and long-term safety of recombinant adeno-associated virus injection into the nonhuman primate muscle. Mol. Ther. 4, 559–566 (2001).
  • Levak-Frank S, Radner H, Walsh A et al. Muscle-specific overexpression of lipoprotein lipase causes a severe myopathy characterized by proliferation of mitochondria and peroxisomes in transgenic mice. J. Clin. Invest. 96, 976–986 (1995).
  • Hoefler G, Noehammer C, Levak-Frank S et al. Muscle-specific overexpression of human lipoprotein lipase in mice causes increased intracellular free fatty acids and induction of peroxisomal enzymes. Biochimie 79, 163–168 (1997).
  • Koike T, Wang X, Unoki H et al. Increased expression of lipoprotein lipase in transgenic rabbits does not lead to abnormalities in skeletal and heart muscles. Muscle Nerve 26, 823–827 (2002).
  • Sonnemans MA, van Amersfoort ES, Jardine L et al. Prenatal development and longitudinal transmission studies with alipogenetiparvovec, an AAV1-based gene therapy vector. AAPS J. 13(S1), W3025 (2011).
  • MacLachlan TK, McIntyre M, Mitrophanous K, Miskin J, Jolly DJ, Cavagnaro JA. Not reinventing the wheel: applying the 3Rs concepts to viral vector gene therapy biodistribution studies. Hum. Gene. Ther. Clin. Dev. 24, 1–4 (2013).
  • Schnepp BC, Clark KR, Klemanski DL, Pacak CA, Johnson PR. Genetic fate of recombinant adeno-associated virus vector genomes in muscle. J. Virol. 77, 3495–3504 (2003).
  • Kotin RM, Linden RM, Berns KI. Characterization of a preferred site on human chromosome 19q for integration of adeno-associated virus DNA by non-homologous recombination. EMBO J. 11, 5071–5078 (1992).
  • Im DS, Muzyczka N. The AAV origin binding protein Rep68 is an ATP-dependent site-specific endonuclease with DNA helicase activity. Cell 61, 447–457 (1990).
  • Hüser D, Gogol-Döring A, Lutter T et al. Integration preferences of wildtype AAV-2 for consensus rep-binding sites at numerous loci in the human genome. PLoS Pathog. 6, e1000985 (2010).
  • Russell DW, Miller AD, Alexander IE. Adeno-associated virus vectors preferentially transduce cells in S phase. Proc. Natl Acad. Sci. U.S.A. 91, 8915–8919 (1994).
  • McCarty DM, Young SM Jr, Samulski RJ. Integration of adeno-associated virus (AAV) and recombinant AAV vectors. Annu. Rev. Genet. 38, 819–845 (2004).
  • Gao G, Alvira MR, Somanathan S, et al. Adeno-associated viruses undergo substantial evolution in primates during natural infections. Proc. Natl Acad. Sci. U.S.A. 100, 6081–6086 (2003).
  • Sun X, Lu Y, Bish LT, Calcedo R, Wilson JM, Gao G. Molecular analysis of vector genome structures after liver transduction by conventional and self-complementary adeno-associated viral serotype vectors in murine and non human primate models. Hum. Gene. Ther. 21, 750–761 (2010).
  • McLaughlin SK, Collis P, Hermonat PL, Muzyczka N. Adeno-associated virus general transduction vectors: analysis of proviral structures. J. Virol. 62, 1963–1973 (1988).
  • Samulski RJ, Chang LS, Shenk T. Helper-free stocks of recombinant adeno-associated viruses: normal integration does not require viral gene expression. J. Virol. 63, 3822–3828 (1989).
  • Ponnazhagan S, Mukherjee P, Wang XS. et al. Adeno-associated virus type 2-mediated transduction in primary human bone marrow-derived CD34+ hematopoietic progenitor cells: donor variation and correlation of transgene expression with cellular differentiation. J. Virol. 71, 8262–8267 (1997).
  • Rutledge EA, Russell DW. Adeno-associated virus vector integration junctions. J. Virol. 71, 8429–8436 (1997).
  • Miller DG, Rutledge EA, Russell DW. Chromosomal effects of adeno-associated virus vector integration. Nat. Genet. 30, 147–148 (2002).
  • Miller DG, Trobridge GD, Petek LM, Jacobs MA, Kaul R, Russell DW. Large-scale analysis of adeno-associated virus vector integration sites in normal human cells. J. Virol. 79, 11434–11442 (2005).
  • Nakai H, Iwaki Y, Kay MA, Couto LB. Isolation of recombinant adeno-associated virus vector-cellular DNA junctions from mouse liver. J. Virol. 73, 5438–5447 (1999).
  • Nakai H, Montini E, Fuess S, Storm TA, Grompe M, Kay MA. AAV serotype 2 vectors preferentially integrate into active genes in mice. Nat. Genet. 34, 297–302 (2003).
  • Nakai H, Wu X, Fuess S. et al. Large-scale molecular characterization of adeno-associated virus vector integration in mouse liver. J. Virol. 79, 3606–3614 (2005).
  • Inagaki K, Lewis SM, Wu X et al. DNA palindromes with a modest arm length of greater, similar 20 base pairs are a significant target for recombinant adeno-associated virus vector integration in the liver, muscles, and heart in mice. J. Virol. 81, 11290–11303 (2007).
  • Han Z, Zhong L, Maina N et al. Stable integration of recombinant adeno-associated virus vector genomes after transduction of murine hematopoietic stem cells. Hum. Gene. Ther. 19, 267–278 (2008).
  • Pachori AS, Melo LG, Zhang L, Loda M, Pratt RE, Dzau VJ. Potential for germ line transmission after intramyocardial gene delivery by adeno-associated virus. Biochem. Biophys. Res. Commun. 313, 528–533 (2004).
  • Wu P, Phillips MI, Bui J, Terwilliger EF. Adeno-associated virus vector-mediated transgene integration into neurons and other nondividing cell targets. J. Virol. 72, 5919–5926 (1998).
  • Inagaki K, Piao C, Kotchey NM, Wu X, Nakai H. Frequency and spectrum of genomic integration of recombinant adeno-associated virus serotype 8 vector in neonatal mouse liver. J. Virol. 82, 9513–9524 (2008).
  • Zhong L. Malani N, Li M et al. Recombinant adeno-associated virus integration sites in murine liver after ornithine transcarbamylase gene correction. Hum. Gen. Ther. 24, 520–525 (2013).
  • Li H, Malani N, Hamilton SR, Schlachterman A et al. Assessing the potential for AAV vector genotoxicity in a murine model. Blood 117, 3311–3319 (2011).
  • Bell P, Wang L, Lebherz C et al. No evidence for tumorigenesis of AAV vectors in a large-scale study in mice. Mol. Ther. 12, 299–306 (2005).
  • Wilson JM, Shchelochkov OA, Gallagher RC, Batshaw ML. Hepatocellular carcinoma in a research subject with ornithine transcarbamylase deficiency. Mol. Genet. Metab. 105, 263–265 (2012).
  • Penaud-Budloo M, Le Guiner C, Nowrouzi A et al. Adeno-associated virus vector genomes persist as episomal chromatin in primate muscle. J. Virol. 82, 7875–7885 (2008).
  • Flageul M, Aubert D, Pichard V et al. Transient expression of genes delivered to newborn rat liver using recombinant adeno-associated virus 2/8 vectors. J. Gene. Med. 11, 689–696 (2009).
  • Schmidt M, Schwarzwaelder K, Bartholomae C et al. High-resolution insertion-site analysis by linear amplification-mediated PCR (LAM-PCR). Nat. Methods 4, 1051–1057 (2007).
  • Ott MG, Schmidt M, Schwarzwaelder K et al. Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat. Med. 12, 401–409 (2006).
  • Deichmann A, Hacein-Bey-Abina S, Schmidt M et al. Vector integration is nonrandom and clustered and influences the fate of lymphopoiesis in SCID-X1 gene therapy. J. Clin. Invest. 117, 2225–2232 (2007).
  • Hacein-Bey-Abina S, Von Kalle C, Schmidt M et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302, 415–419 (2003).
  • Gabriel R, Eckenberg R, Paruzynski A et al. Comprehensive genomic access to vector integration in clinical gene therapy. Nat. Med. 15, 1431–1436 (2009).
  • Montini E, Cesana D, Schmidt M et al. Hematopoietic stem cell gene transfer in a tumor-prone mouse model uncovers low genotoxicity of lentiviral vector integration. Nat. Biotechnol. 24, 687–696 (2006).
  • Schwarzwaelder K, Howe SJ, Schmidt M et al. Gammaretrovirus-mediated correction of SCID-X1 is associated with skewed vector integration site distribution in vivo. J. Clin. Invest. 117, 2241–2249 (2007).
  • Paruzynski A, Arens A, Gabriel R et al. Genome-wide high-throughput integrome analyses by nrLAM-PCR and next-generation sequencing. Nat. Protocols 5, 1379–1395 (2010).
  • Kaeppel C, Beattie SG, Fronza R et al. A largely random AAV integration profile after LPLD gene therapy. Nat. Med. 19, 889–910 (2013).
  • Chen SJ, Tazelaar J, Moscioni AD, Wilson JM. In vivo selection of hepatocytes transduced with adeno-associated viral vectors. Mol. Ther. 1, 414–422 (2000).
  • Donsante A, Vogler C, Muzyczka N et al. Observed incidence of tumorigenesis in long-term rodent studies of rAAV vectors. Gene. Ther. 8, 1343–1346 (2001).
  • Daly TM, Okuyama T, Vogler C, Haskins ME, Muzyczka N, Sands MS. Neonatal intramuscular injection with recombinant adeno-associated virus results in prolonged beta-glucuronidase expression in situ and correction of liver pathology in mucopolysaccharidosis type VII mice. Hum. Gene. Ther. 10, 85–94 (1999).
  • Donsante A, Miller DG, Li Y et al. AAV vector integration sites in mouse hepatocellular carcinoma. Science 317, 477 (2007).
  • Naf D, Krupke DM, Sundberg JP, Eppig JT, Bult CJ. The Mouse Tumor Biology Database: a public resource for cancer genetics and pathology of the mouse. Cancer Res. 62, 1235–1240 (2002).
  • 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. 14, 25–33 (2006).
  • Bell P, Moscioni AD, McCarter RJ, Wu D, Gao G, Hoang A et al. Analysis of tumors arising in male B6C3F1 mice with and without AAV vector delivery to liver. Mol. Ther. 14, 34–44 (2006).
  • Buchmann A, Bauer-Hofmann R, Mahr J, Drinkwater NR, Luz A, Schwarz M. Mutational activation of the c-Ha-ras gene in liver tumors of different rodent strains: correlation with susceptibility to hepatocarcinogenesis. Proc. Natl Acad. Sci. U.S.A. 88, 911–915 (1991).
  • Leakey JE, Seng JE, Latendresse JR, Hussain N, Allen LJ, Allaben WT. Dietary controlled carcinogenicity study of chloral hydrate in male B6C3F1 mice. Toxicol. Appl. Pharmacol. 193, 266–280 (2003).
  • Koeberl DD. Vector-related tumorigenesis not found in ornithine transcarbamylase-deficient mice. Mol. Ther. 14, 1–2 (2006).
  • Niemeyer GP, Herzog RW, Mount J et al. Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy. Blood 113, 797–806 (2009).
  • Nathwani AC, Gray JT, McIntosh J et al. Safe and efficient transduction of the liver after peripheral vein infusion of self-complementary AAV vector results in stable therapeutic expression of human FIX in nonhuman primates. Blood 109, 1414–1421 (2007).
  • Rogers S, Pfuderer P. Use of viruses as carriers of added genetic information. Nature 219 (5155), 749–51 (1968).

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.