W. Al-herz, A. Bousfiha, J. L. Casanova, T. Chatila, M. E. Conley et al., Primary immunodeficiency diseases: an update on the classification from the international union of immunological societies expert committee for primary immunodeficiency, Front Immunol, vol.5, p.162, 2014.

J. Heimall, J. Puck, R. Buckley, T. A. Fleisher, A. R. Gennery et al., Current Knowledge and Priorities for Future Research in Late Effects after Hematopoietic Stem Cell Transplantation (HCT) for Severe Combined Immunodeficiency Patients: A Consensus Statement from the Second Pediatric Blood and Marrow Transplant Consortium International Conference on Late Effects after Pediatric HCT, Biol Blood Marrow Transplant, vol.23, issue.3, pp.379-387, 2017.

A. Fischer, S. Hacein-bey-abina, and M. Cavazzana-calvo, Gene therapy for primary immunodeficiencies, Hematology/oncology clinics of North America, vol.25, issue.1, pp.89-100, 2011.

A. Aiuti, F. Cattaneo, S. Galimberti, U. Benninghoff, B. Cassani et al., Gene therapy for immunodeficiency due to adenosine deaminase deficiency, N Engl J Med, vol.360, issue.5, pp.447-58, 2009.

A. Aiuti, L. Biasco, S. Scaramuzza, F. Ferrua, M. P. Cicalese et al., Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome, Science, vol.341, issue.6148, p.1233151, 2013.

S. Mukherjee and A. J. Thrasher, Gene therapy for PIDs: progress, pitfalls and prospects

, Gene, vol.525, issue.2, pp.174-81, 2013.

A. Fischer, S. Hacein-bey-abina, and M. Cavazzana-calvo, Gene therapy of primary T cell immunodeficiencies, Gene, vol.525, issue.2, pp.170-173, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00817790

S. Hacein-bey-abina, H. B. Gaspar, J. Blondeau, L. Caccavelli, S. Charrier et al., Outcomes following gene therapy in patients with severe Wiskott-Aldrich syndrome, JAMA, vol.313, issue.15, pp.1550-63, 2015.

F. Pala, H. Morbach, M. C. Castiello, J. N. Schickel, S. Scaramuzza et al.,

, Lentiviral-mediated gene therapy restores B cell tolerance in Wiskott-Aldrich syndrome patients, J Clin Invest, vol.125, issue.10, pp.3941-51, 2015.

M. C. Castiello, S. Scaramuzza, F. Pala, F. Ferrua, P. Uva et al., B-cell reconstitution after lentiviral vector-mediated gene therapy in patients with Wiskott-Aldrich syndrome, J Allergy Clin Immunol, vol.136, issue.3, pp.692-702, 2015.

K. L. Shaw, E. Garabedian, S. Mishra, P. Barman, A. Davila et al., Clinical efficacy of gene-modified stem cells in adenosine deaminase-deficient immunodeficiency, J Clin Invest, vol.127, issue.5, pp.1689-99, 2017.

A. Aiuti, M. G. Roncarolo, and L. Naldini, Gene therapy for ADA-SCID, the first marketing approval of an ex vivo gene therapy in Europe: paving the road for the next generation of advanced therapy medicinal products, EMBO Mol Med, vol.9, issue.6, pp.737-777, 2017.

A. J. Thrasher, H. B. Gaspar, C. Baum, U. Modlich, A. Schambach et al., Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1, J Clin Invest, vol.443, issue.7109, pp.3132-3174, 2006.

S. J. Howe, M. R. Mansour, K. Schwarzwaelder, C. Bartholomae, M. Hubank et al., Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients, J Clin Invest, vol.118, issue.9, pp.3143-50, 2008.

S. Stein, M. G. Ott, S. Schultze-strasser, A. Jauch, B. Burwinkel et al., Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease, Nat Med, vol.16, issue.2, pp.198-204, 2010.

S. S. De-ravin, X. Wu, S. Moir, S. Anaya-o'brien, N. Kwatemaa et al., Lentiviral hematopoietic stem cell gene therapy for X-linked severe combined immunodeficiency, Sci Transl Med, vol.8, issue.335, pp.335-57, 2016.

E. Mamcarz, S. Zhou, T. Lockey, H. Abdelsamed, S. J. Cross et al., Lentiviral Gene Therapy Combined with Low-Dose Busulfan in Infants with SCID-X1, N Engl J Med, vol.380, issue.16, pp.1525-1559, 2019.

D. K. Shah and J. C. Zuniga-pflucker, An overview of the intrathymic intricacies of T cell development, J Immunol, vol.192, issue.9, pp.4017-4040, 2014.

A. Nowrouzi, M. Penaud-budloo, C. Kaeppel, U. Appelt, L. Guiner et al., Integration frequency and intermolecular recombination of rAAV vectors in non-human primate skeletal muscle and liver, Mol Ther, vol.20, issue.6, pp.1177-86, 2012.

M. M. Davis and P. J. Bjorkman, T-cell antigen receptor genes and T-cell recognition, Nature, vol.334, issue.6181, pp.395-402, 1988.

C. Y. Wang, P. F. Yu, X. B. He, Y. X. Fang, W. Y. Cheng et al., alphabeta T-cell receptor bias in disease and therapy (Review), Int J Oncol, vol.48, issue.6, pp.2247-56, 2016.

R. Calcedo, S. Somanathan, Q. Qin, M. R. Betts, A. J. Rech et al., Class I-restricted T-cell responses to a polymorphic peptide in a gene therapy clinical trial for alpha-1-antitrypsin deficiency, Proc Natl Acad Sci, vol.114, issue.7, pp.1655-1664, 2017.

F. Mingozzi and K. A. High, Immune responses to AAV vectors: overcoming barriers to successful gene therapy, Blood, vol.122, issue.1, pp.23-36, 2013.

W. Xiao, N. Chirmule, M. A. Schnell, J. Tazelaar, J. V. Hughes et al., Route of administration determines induction of T-cell-independent humoral responses to adenoassociated virus vectors, Mol Ther, vol.1, issue.4, pp.323-332, 2000.

S. D. Mendoza, Y. El-shamayleh, and G. D. Horwitz, AAV-mediated delivery of optogenetic constructs to the macaque brain triggers humoral immune responses, J Neurophysiol, vol.117, issue.5, pp.2004-2017, 2017.

J. E. Walter, L. B. Rosen, K. Csomos, J. M. Rosenberg, D. Mathew et al., Broadspectrum antibodies against self-antigens and cytokines in RAG deficiency, J Clin Invest, vol.126, issue.11, p.4389, 2016.

C. D. Surh and J. Sprent, Regulation of mature T cell homeostasis, Semin Immunol, vol.17, issue.3, pp.183-91, 2005.

J. E. Cowan, S. M. Parnell, K. Nakamura, J. H. Caamano, P. J. Lane et al., The thymic medulla is required for Foxp3+ regulatory but not conventional CD4+ thymocyte development, J Exp Med, vol.210, issue.4, pp.675-81, 2013.

W. Van-ewijk, B. Wang, G. Hollander, H. Kawamoto, E. Spanopoulou et al., Thymic microenvironments, 3-D versus 2-D?, Semin Immunol, vol.11, issue.1, pp.57-64, 1999.

D. M. Su, S. Navarre, W. J. Oh, B. G. Condie, and N. R. Manley, A domain of Foxn1 required for crosstalk-dependent thymic epithelial cell differentiation, Nat Immunol, vol.4, issue.11, pp.1128-1163, 2003.

J. A. Dudakov and M. R. Van-den-brink, Greater than the sum of their parts: combination strategies for immune regeneration following allogeneic hematopoietic stem cell transplantation, Best Pract Res Clin Haematol, vol.24, issue.3, pp.467-76, 2011.

J. S. Sutherland, G. L. Goldberg, M. V. Hammett, A. P. Uldrich, S. P. Berzins et al., Activation of thymic regeneration in mice and humans following androgen blockade, J Immunol, vol.175, issue.4, pp.2741-53, 2005.

G. L. Goldberg, J. A. Dudakov, J. J. Reiseger, N. Seach, T. Ueno et al., Sex steroid ablation enhances immune reconstitution following cytotoxic antineoplastic therapy in young mice, J Immunol, vol.184, issue.11, pp.6014-6038, 2010.

E. Velardi, J. J. Tsai, A. M. Holland, T. Wertheimer, V. W. Yu et al., Sex steroid blockade enhances thymopoiesis by modulating Notch signaling, J Exp Med, vol.211, issue.12, pp.2341-2350, 2014.

D. Min, A. Panoskaltsis-mortari, O. M. Kuro, G. A. Hollander, B. R. Blazar et al., Sustained thymopoiesis and improvement in functional immunity induced by exogenous KGF administration in murine models of aging, Blood, vol.109, issue.6, pp.2529-2566, 2007.

S. W. Rossi, L. T. Jeker, T. Ueno, S. Kuse, M. P. Keller et al., Keratinocyte growth factor (KGF) enhances postnatal T-cell development via enhancements in proliferation and function of thymic epithelial cells, Blood, vol.109, issue.9, pp.3803-3814, 2007.

J. A. Dudakov, A. M. Hanash, R. R. Jenq, L. F. Young, A. Ghosh et al., Interleukin-22 drives endogenous thymic regeneration in mice, Science, vol.336, issue.6077, pp.91-96, 2012.

C. Reimann, E. Six, L. Dal-cortivo, A. Schiavo, K. Appourchaux et al.,

, Human T-lymphoid progenitors generated in a feeder-cell-free Delta-like-4 culture system promote T-cell reconstitution in NOD/SCID/gammac(-/-) mice, Stem Cells, vol.30, issue.8, pp.1771-80, 2012.

G. Awong, J. Singh, M. Mohtashami, M. Malm, L. Motte-mohs et al., Human proT-cells generated in vitro facilitate hematopoietic stem cell-derived T-lymphopoiesis in vivo and restore thymic architecture, Blood, vol.122, issue.26, pp.4210-4219, 2013.

J. A. Dudakov, A. M. Mertelsmann, M. H. O'connor, R. R. Jenq, E. Velardi et al., Loss of thymic innate lymphoid cells leads to impaired thymopoiesis in experimental graftversus-host disease, Blood, vol.130, issue.7, pp.933-975, 2017.

R. M. Kelly, E. M. Goren, P. A. Taylor, S. N. Mueller, H. E. Stefanski et al., Shortterm inhibition of p53 combined with keratinocyte growth factor improves thymic epithelial cell recovery and enhances T-cell reconstitution after murine bone marrow transplantation, Blood, vol.115, issue.5, pp.1088-97, 2010.

S. L. Zhang, X. Wang, S. Manna, D. A. Zlotoff, J. L. Bryson et al., Chemokine treatment rescues profound T-lineage progenitor homing defect after bone marrow transplant conditioning in mice, 2014.

S. Manna and A. Bhandoola, Intrathymic Injection, Methods Mol Biol, vol.1323, pp.203-212, 2016.

N. Maina, Z. Han, X. Li, Z. Hu, L. Zhong et al., Recombinant selfcomplementary adeno-associated virus serotype vector-mediated hematopoietic stem cell transduction and lineage-restricted, long-term transgene expression in a murine serial bone marrow transplantation model, Hum Gene Ther, vol.19, issue.4, pp.376-83, 2008.

C. Ling, K. Bhukhai, Z. Yin, M. Tan, M. C. Yoder et al., High-Efficiency Transduction of Primary Human Hematopoietic Stem/Progenitor Cells by AAV6 Vectors: Strategies for Overcoming Donor-Variation and Implications in Genome Editing, Scientific reports, vol.6, p.35495, 2016.

D. Huser, D. Khalid, T. Lutter, E. M. 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.91, issue.4, 2017.

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, issue.11, pp.1061-74, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01798967

F. F. Reichel, T. Peters, B. Wilhelm, M. Biel, M. Ueffing et al., Humoral Immune Response After Intravitreal But Not After Subretinal AAV8 in Primates and Patients, Invest Ophthalmol Vis Sci, vol.59, issue.5, pp.1910-1915, 2018.

F. F. Reichel, D. L. Dauletbekov, R. Klein, T. Peters, G. A. Ochakovski et al., AAV8 Can Induce Innate and Adaptive Immune Response in the Primate Eye, Mol Ther, vol.25, issue.12, pp.2648-60, 2017.

J. H. Mcintosh, M. Cochrane, S. Cobbold, H. Waldmann, S. A. Nathwani et al., Successful attenuation of humoral immunity to viral capsid and transgenic protein following AAV-mediated gene transfer with a non-depleting CD4 antibody and cyclosporine, Gene Ther, vol.19, issue.1, pp.78-85, 2012.

S. O. Han, S. Li, E. D. Brooks, E. Masat, C. Leborgne et al., Enhanced efficacy from gene therapy in Pompe disease using coreceptor blockade, Hum Gene Ther, vol.26, issue.1, pp.26-35, 2015.

M. Corti, M. Elder, D. Falk, L. Lawson, B. Smith et al., B-Cell Depletion is Protective Against Anti-AAV Capsid Immune Response: A Human Subject Case Study, Mol Ther Methods Clin Dev, vol.1, 2014.

D. L. Mack, K. Poulard, M. A. Goddard, V. Latournerie, J. M. Snyder et al.,

A. Systemic, Mediated Gene Therapy Drives Whole-Body Correction of Myotubular Myopathy in Dogs, Mol Ther, vol.25, issue.4, pp.839-54, 2017.

M. Elverman, M. A. Goddard, D. Mack, J. M. Snyder, M. W. Lawlor et al., Long-term effects of systemic gene therapy in a canine model of myotubular myopathy, Muscle Nerve, vol.56, issue.5, pp.943-53, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02179358

L. Guiner, C. Servais, L. Montus, M. Larcher, T. Fraysse et al., Long-term microdystrophin gene therapy is effective in a canine model of Duchenne muscular dystrophy, Nat Commun, vol.8, p.16105, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02179396

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, issue.12, pp.5310-5318, 2013.

G. Gernoux, J. M. Wilson, and C. Mueller, Regulatory and Exhausted T Cell Responses to AAV Capsid, Hum Gene Ther, vol.28, issue.4, pp.338-387, 2017.

D. L. Foss, E. Donskoy, and I. Goldschneider, The importation of hematogenous precursors by the thymus is a gated phenomenon in normal adult mice, J Exp Med, vol.193, issue.3, pp.365-74, 2001.

S. E. Prockop and H. T. Petrie, Regulation of thymus size by competition for stromal niches among early T cell progenitors, J Immunol, vol.173, issue.3, pp.1604-1615, 2004.

O. Adjali, R. R. Vicente, C. Ferrand, C. Jacquet, C. Mongellaz et al.,

, Intrathymic administration of hematopoietic progenitor cells enhances T cell reconstitution in ZAP-70 severe combined immunodeficiency, Proc Natl Acad Sci U S A, vol.102, issue.38, p.13586, 2005.

D. Grimm, M. A. Kay, and J. A. Kleinschmidt, Helper virus-free, optically controllable, and twoplasmid-based production of adeno-associated virus vectors of serotypes 1 to 6, Mol Ther, vol.7, issue.6, pp.839-50, 2003.

P. Chenuaud, T. Larcher, J. E. Rabinowitz, N. Provost, B. Joussemet et al., Optimal design of a single recombinant adeno-associated virus derived from serotypes 1 and 2 to achieve more tightly regulated transgene expression from nonhuman primate muscle, Mol Ther, vol.9, issue.3, pp.410-418, 2004.

A. Salvetti, S. Oreve, G. Chadeuf, D. Favre, Y. Cherel et al., Factors influencing recombinant adeno-associated virus production, Hum Gene Ther, vol.9, issue.5, pp.695-706, 1998.

R. Vicente, O. Adjali, C. Jacquet, V. S. Zimmermann, and N. Taylor, Intrathymic transplantation of bone marrow-derived progenitors provides long-term thymopoiesis, Blood, vol.115, issue.10, pp.1913-1933, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02193630

N. Lopes, H. Vachon, J. Marie, and M. Irla, Administration of RANKL boosts thymic regeneration upon bone marrow transplantation, EMBO Mol Med, vol.9, issue.6, pp.835-51, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01914636

A. Serge, A. L. Bailly, M. Aurrand-lions, B. A. Imhof, and M. Irla, For3D: Full organ reconstruction in 3D, an automatized tool for deciphering the complexity of lymphoid organs, Journal of immunological methods, vol.424, pp.32-42, 2015.

S. Firouzi, Y. Lopez, Y. Suzuki, K. Nakai, S. Sugano et al., Development and validation of a new high-throughput method to investigate the clonality of HTLV-1-infected cells based on provirus integration sites, Genome Med, vol.6, issue.6, p.46, 2014.

N. A. Gillet, N. Malani, A. Melamed, N. Gormley, R. Carter et al., The host genomic environment of the provirus determines the abundance of HTLV-1-infected T-cell clones, Blood, vol.117, issue.11, pp.3113-3135, 2011.

D. A. Bolotin, S. Poslavsky, I. Mitrophanov, M. Shugay, I. Z. Mamedov et al., MiXCR: software for comprehensive adaptive immunity profiling, Nat Methods, vol.12, issue.5, pp.380-381, 2015.

A. T. Martino, R. W. Herzog, I. Anegon, and O. Adjali, Measuring immune responses to recombinant AAV gene transfer, Methods Mol Biol, vol.807, pp.259-72, 2011.

Q. Z. Li, J. Zhou, A. E. Wandstrat, F. Carr-johnson, V. Branch et al., Protein array autoantibody profiles for insights into systemic lupus erythematosus and incomplete lupus syndromes, Clin Exp Immunol, vol.147, issue.1, pp.60-70, 2007.

V. Capo, M. C. Castiello, E. Fontana, S. Penna, M. Bosticardo et al., Efficacy of lentivirus-mediated gene therapy in an Omenn syndrome recombination-activating gene 2 mouse model is not hindered by inflammation and immune dysregulation, J Allergy Clin Immunol, vol.142, issue.3, pp.928-969, 2018.