A. M. Maguire, K. A. High, A. Auricchio, J. F. Wright, E. A. Pierce et al., Age-dependent effects of RPE65 gene therapy for Leber's congenital amaurosis: a phase 1 dose-escalation trial, Lancet, vol.374, pp.61836-61841, 2009.

J. Bainbridge, M. S. Mehat, V. Sundaram, S. J. Robbie, S. E. Barker et al., Long-term effect of gene therapy on Leber's congenital amaurosis, N Engl J Med, vol.372, pp.1887-97, 2015.

A. C. Nathwani, U. M. Reiss, E. Tuddenham, C. Rosales, P. Chowdary et al., Long-term safety and efficacy of factor IX gene therapy in hemophilia B, N Engl J Med, vol.371, pp.1994-2004, 2014.

C. Mueller, J. D. Chulay, B. C. Trapnell, M. Humphries, B. Carey et al., Human Treg responses allow sustained recombinant adeno-associated virus-mediated transgene expression, J Clin Invest, vol.123, pp.5310-5318, 2013.

V. Ferreira, J. Twisk, K. Kwikkers, E. Aronica, D. Brisson et al., Immune responses to intramuscular administration of alipogene tiparvovec (AAV1-LPL(S447X)) in a phase II clinical trial of lipoprotein lipase deficiency gene therapy, Hum Gene Ther, vol.25, pp.180-188, 2014.

D. Melchiorri, L. Pani, P. Gasparini, G. Cossu, J. Ancans et al., Regulatory evaluation of Glybera in Europe -two committees, one mission, Nat Rev Drug Discov, vol.12, p.719, 2013.

S. Russell, J. Bennett, J. A. Wellman, D. C. Chung, Z. Yu 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, vol.390, issue.17, pp.31868-31876, 2017.

S. M. Hoy, Onasemnogene abeparvovec: first global approval, Drugs, vol.79, pp.1255-62, 2019.

C. S. Manno, F. Glenn, V. R. Arruda, G. F. Pierce, B. Glader et al., Successful transduction of liver in hemophilia by AAV-Factor, I. X, and limitations imposed by the host immune response, Nat Med, vol.12, pp.342-349, 2006.

F. Mingozzi, M. V. Maus, D. J. Hui, D. E. Sabatino, S. L. Murphy et al., CD8+ T-cell responses to adeno-associated virus capsid in humans, Nat Med, vol.13, pp.419-441, 2007.

A. C. Nathwani, E. Tuddenham, S. Rangarajan, C. Rosales, J. Mcintosh et al., Adenovirus-associated virus vector-mediated gene transfer in hemophilia B, N Engl J Med, vol.365, pp.2357-65, 2011.

G. C. Pien, E. Basner-tschakarjan, D. J. Hui, A. N. Mentlik, J. D. Finn et al., Capsid antigen presentation flags human hepatocytes for destruction after transduction by adeno-associated viral vectors, J Clin Invest, vol.119, pp.1688-95, 2009.

C. Alanio, F. Lemaitre, H. Law, M. Hasan, and M. L. Albert, Enumeration of human antigen-specific naive CD8+ T cells reveals conserved precursor frequencies, Blood, vol.115, pp.3718-3743, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-01402100

F. Legoux, E. Debeaupuis, K. Echasserieau, H. De-la-salle, X. Saulquin et al., Impact of TCR reactivity and HLA phenotype on naive CD8 T cell frequency in humans, J Immunol, vol.184, pp.6731-6739, 2010.

B. Rodenko, M. Toebes, S. R. Hadrup, W. Van-esch, A. M. Molenaar et al., Generation of peptide-MHC class I complexes through UV-mediated ligand exchange, Nat Protoc, vol.1, pp.1120-1152, 2006.

N. Chirmule, K. Propert, S. Magosin, Y. Qian, R. Qian et al., Immune responses to adenovirus and adeno-associated virus in humans, Gene Ther, vol.6, pp.1574-83, 1999.

H. Li, M. O. Lasaro, B. Jia, S. W. Lin, L. H. Haut et al., Capsid-specific Tcell responses to natural infections with adeno-associated viruses in humans differ from those of nonhuman primates, Mol Ther, vol.19, pp.2021-2051, 2011.

P. Veron, C. Leborgne, V. Monteilhet, S. Boutin, S. Martin et al., Humoral and cellular capsid-specific immune responses to adeno-associated virus type 1 in randomized healthy donors, J Immunol, vol.188, pp.6418-6442, 2012.

R. Calcedo, L. H. Vandenberghe, G. Gao, J. Lin, and J. M. Wilson, Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses, J Infect Dis, vol.199, pp.381-90, 2009.

L. Jeune, V. Joergensen, J. A. Hajjar, R. J. Weber, and T. , Pre-existing anti-adenoassociated virus antibodies as a challenge in AAV gene therapy, Hum Gene Ther Methods, vol.24, pp.59-67, 2013.

B. Balakrishnan and G. Jayandharan, Basic biology of adeno-associated virus (AAV) vectors used in gene therapy, Current Gene Therapy, vol.14, pp.86-100, 2014.

B. D. Evavold, J. Sloan-lancaster, K. J. Wilson, J. B. Rothbard, A. et al., Specific T cell recognition of minimally homologous peptides: evidence for multiple endogenous ligands, Immunity, vol.2, pp.90010-90011, 1995.

J. M. Eberhard, F. Ahmad, H. S. Hong, N. Bhatnagar, P. Keudel et al., Partial recovery of senescence and differentiation disturbances in CD8+ T cell effector-memory cells in HIV-1 infection after initiation of anti-retroviral treatment, Clin Exp Immunol, vol.186, pp.227-265, 2016.

T. Shen, J. Zheng, C. Xu, J. Liu, W. Zhang et al., Pd-1 expression on peripheral Cd8+ tem/temra subsets closely correlated with Hcv viral load in chronic hepatitis C patients, Virol J, vol.7, p.310, 2010.

M. Weltevrede, R. Eilers, H. E. De-melker, and D. Van-baarle, Cytomegalovirus persistence and T-cell immunosenescence in people aged fifty and older: a systematic review, Exp Gerontol, vol.77, pp.87-95, 2016.

J. Geginat, A. Lanzavecchia, and F. Sallusto, Proliferation and differentiation potential of human CD8+ memory T-cell subsets in response to antigen or homeostatic cytokines, Blood, vol.101, pp.4260-4266, 2003.

M. J. Pittet, D. Valmori, P. R. Dunbar, D. E. Speiser, D. Liénard et al., High frequencies of naive Melan-A/MART-1-specific CD8(+) T cells in a large proportion of human histocompatibility leukocyte antigen (HLA)-A2 individuals, J Exp Med, vol.190, pp.705-720, 1999.

V. Voelter, N. Rufer, R. S. Greub, G. Brookes, R. Guillaume et al., Characterization of Melan-A reactive memory CD8+ T cells in a healthy donor, Int Immunol, vol.20, pp.1087-96, 2008.

V. Bhardwaj, V. Kumar, H. M. Geysen, and E. E. Sercarz, Degenerate recognition of a dissimilar antigenic peptide by myelin basic protein-reactive T cells. Implications for thymic education and autoimmunity, J Immunol, vol.151, pp.5000-5010, 1993.

D. T. Hagerty, A. , and P. M. , Intramolecular mimicry. Identification and analysis of two cross-reactive T cell epitopes within a single protein, J Immunol, vol.155, pp.2993-3001, 1995.

D. J. Loftus, C. Castelli, T. M. Clay, P. Squarcina, F. M. Marincola et al., Identification of epitope mimics recognized by CTL reactive to the melanoma/melanocyte-derived peptide MART-1(27-35), J Exp Med, vol.184, pp.647-57, 1996.

J. L. Grogan, A. Kramer, A. Nogai, L. Dong, M. Ohde et al., Cross-reactivity of myelin basic protein-specific T cells with multiple microbial peptides: experimental autoimmune encephalomyelitis induction in TCR transgenic mice, J Immunol, vol.163, pp.3764-70, 1999.

B. Hemmer, B. T. Fleckenstein, M. Vergelli, G. Jung, H. Mcfarland et al., Identification of high potency microbial and self ligands for a human autoreactive class II-restricted T cell clone, J Exp Med, vol.185, pp.1651-1660, 1997.

I. S. Misko, S. M. Cross, R. Khanna, S. L. Elliott, C. Schmidt et al., Crossreactive recognition of viral, self, and bacterial peptide ligands by human class I-restricted cytotoxic T lymphocyte clonotypes: implications for molecular mimicry in autoimmune disease, Proc Natl Acad Sci, vol.96, pp.2279-84, 1999.

M. A. Brehm, A. K. Pinto, K. A. Daniels, J. P. Schneck, R. M. Welsh et al., T cell immunodominance and maintenance of memory regulated by unexpectedly cross-reactive pathogens, Nat Immunol, vol.3, pp.627-661, 2002.

E. Barnes, S. M. Ward, V. O. Kasprowicz, G. Dusheiko, P. Klenerman et al., Ultra-sensitive class I tetramer analysis reveals previously undetectable populations of antiviral CD8+ T cells, Eur J Immunol, vol.34, pp.1570-1577, 2004.

M. A. Lima, A. Marzocchetti, P. Autissier, T. Tompkins, Y. Chen et al., Frequency and phenotype of JC virus-specific CD8+ T lymphocytes in the peripheral blood of patients with progressive multifocal leukoencephalopathy, J Virol, vol.81, pp.3361-3369, 2007.

C. Alanio, F. Nicoli, P. Sultanik, T. Flecken, B. Perot et al., Bystander hyperactivation of preimmune CD8+ T cells in chronic HCV patients, ELife, vol.4, p.7916, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01306801

D. J. Hui, S. C. Edmonson, G. M. Podsakoff, G. C. Pien, L. Ivanciu et al., AAV capsid CD8+ T-cell epitopes are highly conserved across AAV serotypes

, Mol Ther Methods Clin Dev, vol.2, p.15029, 2015.

F. Mingozzi and K. A. High, Immune responses to AAV vectors: overcoming barriers to successful gene therapy, Blood, vol.122, pp.23-36, 2013.

M. D'asaro, F. Dieli, N. Caccamo, M. Musso, F. Porretto et al., Increase of CCR7-CD45RA+ CD8 T cells (T(EMRA)) in chronic graft-versus-host disease, Leukemia, vol.20, pp.545-552, 2006.

L. Barbarash, I. Kudryavtsev, N. Rutkovskaya, and A. Golovkin, T cell response in patients with implanted biological and mechanical prosthetic heart valves, Mediators Inflamm, 2016.

K. Erles, P. Sebökovà, and J. R. Schlehofer, Update on the prevalence of serum antibodies (IgG and IgM) to adeno-associated virus (AAV), J Med Virol, vol.59, pp.406-417, 1999.

S. L. Murphy, H. Li, F. Mingozzi, D. Sabatino, D. Hui et al., Diverse IgG subclass responses to adeno-associated virus infection and vector administration, J Med Virol, vol.81, pp.65-74, 2009.

Y. J. Hernandez, J. Wang, W. G. Kearns, S. Loiler, A. Poirier et al., Latent adeno-associated virus infection elicits humoral but not cell-mediated immune responses in a nonhuman primate model, J Virol, vol.73, pp.8549-58, 1999.

A. Salvetti, S. Orève, G. Chadeuf, D. Favre, Y. Cherel et al., Factors influencing recombinant adeno-associated virus production, Human Gene Therapy, vol.9, pp.695-706, 1998.

B. C. Schnepp, R. L. Jensen, K. R. Clark, J. , and P. R. , Infectious molecular clones of adeno-associated virus isolated directly from human tissues, J Virol, vol.83, pp.1456-64, 2009.

G. Gao, L. H. Vandenberghe, M. R. Alvira, Y. Lu, R. Calcedo et al., Clades of adeno-associated viruses are widely disseminated in human tissues, J Virol, vol.78, pp.6381-6389, 2004.

B. C. Schnepp, R. L. Jensen, C. Chen, P. R. Johnson, and K. R. Clark, Characterization of adeno-associated virus genomes isolated from human tissues, J Virol, vol.79, pp.14793-803, 2005.

V. Rohde, K. Erles, H. P. Sattler, H. Derouet, B. Wullich et al., Detection of adeno-associated virus in human semen: does viral infection play a role in the pathogenesis of male infertility?, Fertil Steril, vol.72, pp.814-820, 1999.

D. Hüser, D. Khalid, T. Lutter, E. Hammer, S. Weger et al., High prevalence of infectious adeno-associated virus (AAV) in human peripheral blood mononuclear cells indicative of T lymphocytes as sites of AAV persistence, J Virol, vol.941, 2016.

T. Hussain and K. M. Quinn, Similar but different: virtual memory CD8 T cells as a memory-like cell population, Immunol Cell Biol, vol.97, pp.675-84, 2019.

F. Jacomet, E. Cayssials, S. Basbous, A. Levescot, N. Piccirilli et al., Evidence for eomesodermin-expressing innate-like CD8(+) KIR/NKG2A(+)

, T cells in human adults and cord blood samples, Eur J Immunol, vol.45, pp.1926-1959, 2015.

S. Boutin, V. Monteilhet, P. Veron, C. Leborgne, O. Benveniste et al., Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors, Human Gene Therapy, vol.21, pp.704-716, 2010.

S. Stanford, R. Pink, D. Creagh, A. Clark, G. Lowe et al., Adenovirusassociated antibodies in UK cohort of hemophilia patients: a seroprevalence study of the presence of adenovirus-associated virus vector-serotypes AAV5 and AAV8 neutralizing activity and antibodies in patients with hemophilia A, Res Pract Thromb Haemost, vol.3, pp.261-268, 2019.

K. Kuranda, P. Jean-alphonse, C. Leborgne, R. Hardet, F. Collaud et al., Exposure to wild-type AAV drives distinct capsid immunity profiles in humans, J Clin Invest, vol.128, pp.5267-79, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01977027

J. W. Northfield, C. P. Loo, J. D. Barbour, G. Spotts, F. M. Hecht et al., Human immunodeficiency virus type 1 (HIV-1)-specific CD8+

, T(EMRA) cells in early infection are linked to control of HIV-1 viremia and predict the subsequent viral load set point, J Virol, vol.81, pp.5759-65, 2007.

C. Vandamme, O. Adjali, and F. Mingozzi, Unraveling the complex story of immune responses to AAV vectors trial after trial, Hum Gene Ther, vol.28, pp.1061-74, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01798967