S. Hacein-bey-abina, A. Garrigue, G. Wang, J. Soulier, A. Lim et al., Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1, Journal of Clinical Investigation, vol.118, issue.9, pp.3132-3174, 2008.
DOI : 10.1172/JCI35700DS1

S. Hacein-bey-abina, H. 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.
DOI : 10.1001/jama.2015.3253

S. Hacein-bey-abina, S. Pai, H. Gaspar, M. Armant, C. Berry et al., A Modified ??-Retrovirus Vector for X-Linked Severe Combined Immunodeficiency, New England Journal of Medicine, vol.371, issue.15, pp.1407-1424, 2009.
DOI : 10.1056/NEJMoa1404588

M. Cavazzana-calvo, E. Payen, O. Negre, G. Wang, K. Hehir et al., Transfusion independence and HMGA2 activation after gene therapy of human ??-thalassaemia, Nature, vol.6, issue.7313, pp.318-340, 2010.
DOI : 10.1093/nar/gkn125

URL : https://hal.archives-ouvertes.fr/cea-00905288

N. Cartier, S. Hacein-bey-abina, C. Bartholomae, G. Veres, M. Schmidt et al., Hematopoietic Stem Cell Gene Therapy with a Lentiviral Vector in X-Linked Adrenoleukodystrophy, Science, vol.116, issue.11, pp.818-841, 2009.
DOI : 10.1172/JCI28873

A. Aiuti, L. Biasco, S. Scaramuzza, F. Ferrua, M. Cicalese et al., Lentiviral Hematopoietic Stem Cell Gene Therapy in Patients with Wiskott-Aldrich Syndrome, Science, vol.177, issue.10, p.1233151, 2013.
DOI : 10.4049/jimmunol.177.10.7451

A. Biffi, E. Montini, L. Lorioli, M. Cesani, F. Fumagalli et al., Lentiviral Hematopoietic Stem Cell Gene Therapy Benefits Metachromatic Leukodystrophy, Science, vol.9, issue.3, p.1233158, 2013.
DOI : 10.1002/(SICI)1098-1004(1997)9:3??234::AID-HUMU4??3.0.CO;2-7

C. Manno, G. Pierce, V. Arruda, B. Glader, M. Ragni et al., Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response, Nat Med, 2006.

M. Dudley, J. Wunderlich, P. Robbins, J. Yang, P. Hwu et al., Cancer Regression and Autoimmunity in Patients After Clonal Repopulation with Antitumor Lymphocytes, Science, vol.298, issue.5594, pp.850-854, 2002.
DOI : 10.1126/science.1076514

L. Johnson, R. Morgan, M. Dudley, L. Cassard, J. Yang et al., Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen, Blood, vol.114, issue.3, pp.535-581, 2009.
DOI : 10.1182/blood-2009-03-211714

P. Robbins, S. Kassim, T. Tran, J. Crystal, R. Morgan et al., A Pilot Trial Using Lymphocytes Genetically Engineered with an NY-ESO-1-Reactive T-cell Receptor: Long-term Follow-up and Correlates with Response, Clinical Cancer Research, vol.21, issue.5, pp.1019-1046, 2015.
DOI : 10.1158/1078-0432.CCR-14-2708

S. Hacein-bey-abina, V. Kalle, C. Schmidt, M. Mccormack, M. Wulffraat et al., LMO2-Associated Clonal T Cell Proliferation in Two Patients after Gene Therapy for SCID-X1, Science, vol.302, issue.5644, pp.415-424, 2003.
DOI : 10.1126/science.1088547

A. Nathwani, U. Reiss, E. Tuddenham, C. Rosales, P. Chowdary et al., Long-Term Safety and Efficacy of Factor IX Gene Therapy in Hemophilia B, New England Journal of Medicine, vol.371, issue.21, pp.1994-2004, 2014.
DOI : 10.1056/NEJMoa1407309

A. Martino, E. Basner-tschakarjan, D. Markusic, J. Finn, C. Hinderer et al., Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-specific CD8+ T cells, Blood, vol.121, issue.12, pp.2224-2257, 2013.
DOI : 10.1182/blood-2012-10-460733

S. Maude, N. Frey, P. Shaw, R. Aplenc, D. Barrett et al., Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia, New England Journal of Medicine, vol.371, issue.16, pp.1507-1524, 2014.
DOI : 10.1056/NEJMoa1407222

D. Lee, J. Kochenderfer, M. Stetler-stevenson, Y. Cui, C. Delbrook et al., T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial, The Lancet, vol.385, issue.9967, pp.517-545, 2015.
DOI : 10.1016/S0140-6736(14)61403-3

P. Sharma and J. Allison, The future of immune checkpoint therapy, Science, vol.19, issue.5, pp.56-61, 2015.
DOI : 10.1158/1078-0432.CCR-12-1630

A. Lombardo, P. Genovese, C. Beausejour, S. Colleoni, Y. Lee et al., Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery, Nat Biotechnol, 2007.

P. Tebas, D. Stein, W. Tang, I. Frank, S. Wang et al., in Autologous CD4 T Cells of Persons Infected with HIV, New England Journal of Medicine, vol.370, issue.10, pp.901-911, 2014.
DOI : 10.1056/NEJMoa1300662

Q. Ding, Y. Lee, E. Schaefer, D. Peters, A. Veres et al., A TALEN Genome-Editing System for Generating Human Stem Cell-Based Disease Models, Cell Stem Cell, vol.12, issue.2, pp.238-51, 2013.
DOI : 10.1016/j.stem.2012.11.011

J. Xu, C. Peng, V. Sankaran, Z. Shao, E. Esrick et al., Correction of sickle cell disease in adult mice by interference with fetal hemoglobin silencing. Science, pp.993-999, 2011.

D. Yang, M. Scavuzzo, J. Chmielowiec, R. Sharp, A. Bajic et al., Enrichment of G2/M cell cycle phase in human pluripotent stem cells enhances HDR-mediated gene repair with customizable endonucleases, Scientific Reports, vol.339, issue.1, p.21264, 2016.
DOI : 10.1126/science.1231143

M. Porteus and D. Baltimore, Chimeric Nucleases Stimulate Gene Targeting in Human Cells, Science, vol.300, issue.5620, p.763, 2003.
DOI : 10.1126/science.1078395

M. Jinek, K. Chylinski, I. Fonfara, M. Hauer, J. Doudna et al., A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity, Science, vol.274, issue.45, pp.816-837, 2012.
DOI : 10.1074/jbc.274.45.31896

D. Cox, R. Platt, and F. Zhang, Therapeutic genome editing: prospects and challenges, Nature Medicine, vol.122, issue.2, pp.121-152
DOI : 10.1126/science.332.6033.1031

F. Ran, P. Hsu, J. Wright, V. Agarwala, D. Scott et al., Genome engineering using the CRISPR-Cas9 system. Nature Protocols, pp.2281-308, 2013.

F. Ran, P. Hsu, C. Lin, J. Gootenberg, S. Konermann et al., Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell, pp.1380-1389, 2013.

S. Choi, Y. Kim, J. Shim, J. Park, R. Wang et al., Efficient drug screening and gene correction for treating liver disease using patient-specific stem cells, Hepatology, vol.40, issue.6, pp.2458-68, 2013.
DOI : 10.1093/nar/gks518

M. Osborn, C. Starker, A. Mcelroy, B. Webber, M. Riddle et al., TALEN-based Gene Correction for Epidermolysis Bullosa, Molecular Therapy, vol.21, issue.6, p.2013
DOI : 10.1038/mt.2013.56

N. Ma, B. Liao, H. Zhang, L. Wang, Y. Shan et al., Transcription Activator-like Effector Nuclease (TALEN)-mediated Gene Correction in Integration-free ??-Thalassemia Induced Pluripotent Stem Cells, Journal of Biological Chemistry, vol.258, issue.48, pp.34671-34680, 2013.
DOI : 10.1038/nbt.1940

X. Huang, Y. Wang, Y. W. Smith, C. Ye, Z. Wang et al., Production of Gene-Corrected Adult Beta Globin Protein in Human Erythrocytes Differentiated from Patient iPSCs After Genome Editing of the Sickle Point Mutation, STEM CELLS, vol.3, issue.5, pp.1470-1479
DOI : 10.5966/sctm.2013-0054

G. Romeo and E. Levin, UROPORPHYRINOGEN III COSYNTHETASE IN HUMAN CONGENITAL ERYTHROPOIETIC PORPHYRIA, Proceedings of the National Academy of Sciences, vol.63, issue.3, 1969.
DOI : 10.1073/pnas.63.3.856

J. Frank, X. Wang, H. Lam, V. Aita, F. Jugert et al., C73R is a hotspot mutation in the uroporphyrinogen III synthase gene in congenital erythropoietic porphyria, Annals of Human Genetics, vol.62, issue.3, pp.225-255, 1998.
DOI : 10.1046/j.1469-1809.1998.6230225.x

J. Deybach, H. De-verneuil, S. Boulechfar, B. Grandchamp, and Y. Nordmann, Point mutations in the uroporphyrinogen III synthase gene in congenital erythropoietic porphyria (Günther's disease). Blood, pp.1763-1768, 1990.

C. Warner, H. Yoo, A. Roberts, and R. Desnick, Congenital erythropoietic porphyria: identification and expression of exonic mutations in the uroporphyrinogen III synthase gene., Journal of Clinical Investigation, vol.89, issue.2, pp.693-700, 1992.
DOI : 10.1172/JCI115637

A. Erwin, M. Balwani, R. Desnick, R. Pagon, M. Adam et al., Porphyrias Consortium of the NIH- Sponsored Rare Diseases Clinical Research Network. Congenital Erythropoietic Porphyria, GeneReviews(®) [Internet]

P. May and M. E. , Protéine p53 : de l'interaction avec les protéines virales à la pathogénie des cancers humains, Virologie, vol.2, pp.347-54, 1998.

T. Wang, J. Wei, D. Sabatini, and E. Lander, Genetic screens in human cells using the CRISPR-Cas9 system. Science, pp.80-84, 2014.

H. Li, N. Fujimoto, N. Sasakawa, S. Shirai, T. Ohkame et al., Precise Correction of the Dystrophin Gene in Duchenne Muscular Dystrophy Patient Induced Pluripotent Stem Cells by TALEN and CRISPR-Cas9, Stem Cell Reports, vol.4, issue.1, pp.143-54, 2015.
DOI : 10.1016/j.stemcr.2014.10.013

B. Shen, W. Zhang, J. Zhang, J. Zhou, J. Wang et al., Efficient genome modification by CRISPR-Cas9 nickase with minimal off-target effects, Nature Methods, vol.11, issue.4, pp.399-402
DOI : 10.1093/bioinformatics/btq033

I. Slaymaker, L. Gao, B. Zetsche, D. Scott, W. Yan et al., Rationally engineered Cas9 nucleases with improved specificity, Science, vol.520, issue.25, pp.84-92, 2016.
DOI : 10.1038/nature14299

J. Ribeil, S. Hacein-bey-abina, E. Payen, A. Magnani, M. Semeraro et al., Gene Therapy in a Patient with Sickle Cell Disease, New England Journal of Medicine, vol.376, issue.9, pp.848-55, 201702.
DOI : 10.1056/NEJMoa1609677