J. A. Gorzelany and M. P. De-souza, Protein replacement therapies for rare diseases: a breeze for regulatory approval?, Sci. Transl. Med, vol.5, pp.178-110, 2013.

D. Concolino, F. Deodato, and R. Parini, Enzyme replacement therapy: efficacy and limitations, Ital. J. Pediatr, vol.44, p.120, 2018.

H. Li, In vivo genome editing restores haemostasis in a mouse model of haemophilia, Nature, vol.475, pp.217-221, 2011.

C. Y. Park, Functional correction of large factor VIII gene chromosomal inversions in hemophilia A patient-derived iPSCs using CRISPR-Cas9, Cell Stem Cell, vol.17, pp.213-220, 2015.

A. Barzel, Promoterless gene targeting without nucleases ameliorates haemophilia B in mice, Nature, vol.517, pp.360-364, 2015.

R. Sharma, In vivo genome editing of the albumin locus as a platform for protein replacement therapy, Blood, vol.126, pp.1777-1784, 2015.

A. De-caneva, Coupling AAV-mediated promoterless gene targeting to SaCas9 nuclease to efficiently correct liver metabolic diseases, JCI Insight, vol.5, p.128863, 2019.

S. Boutin, 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, Hum. Gene Ther, vol.21, pp.704-712, 2010.

G. C. Pien, Capsid antigen presentation flags human hepatocytes for destruction after transduction by adeno-associated viral vectors, J. Clin. Invest, vol.119, pp.1688-1695, 2009.

C. T. Charlesworth, Identification of preexisting adaptive immunity to Cas9 proteins in humans, Nat. Med, vol.25, pp.249-254, 2019.

M. Hosel, Hepatitis B virus infection enhances susceptibility toward adeno-associated viral vector transduction in vitro and in vivo, Hepatology, vol.59, pp.2110-2120, 2014.

L. Sobrevals, AAV vectors transduce hepatocytes in vivo as efficiently in cirrhotic as in healthy rat livers, Gene Ther, vol.19, pp.411-417, 2012.

E. Dzierzak and S. Philipsen, Erythropoiesis: development and differentiation. Cold Spring Harb, Perspect. Med, vol.3, p.11601, 2013.

E. P. Vichinsky, Clinical manifestations of alpha-thalassemia, Cold Spring Harb. Perspect. Med, vol.3, p.11742, 2013.

A. H. Chang, M. T. Stephan, and M. Sadelain, Stem cell-derived erythroid cells mediate long-term systemic protein delivery, Nat. Biotechnol, vol.24, pp.1017-1021, 2006.

D. Wang, Reprogramming erythroid cells for lysosomal enzyme production leads to visceral and CNS cross-correction in mice with Hurler syndrome, Proc. Natl. Acad. Sci. USA, vol.106, 2009.

C. A. Montiel-equihua, The beta-globin locus control region in combination with the EF1alpha short promoter allows enhanced lentiviral vector-mediated erythroid gene expression with conserved multilineage activity, Mol. Ther, vol.20, pp.1400-1409, 2012.

R. Kurita, Establishment of immortalized human erythroid progenitor cell lines able to produce enucleated red blood cells, PLoS ONE, vol.8, p.59890, 2013.

J. Wang, Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors, Nat. Biotechnol, vol.33, pp.1256-1263, 2015.

M. Haeussler, Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR, Genome Biol, vol.17, p.148, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01346049

T. J. Cradick, P. Qiu, C. M. Lee, E. J. Fine, and G. Bao, COSMID: a web-based tool for identifying and validating CRISPR/Cas off-target sites, Mol. Ther. Nucleic Acids, vol.3, p.214, 2014.

E. K. Brinkman, T. Chen, M. Amendola, and B. Van-steensel, Easy quantitative assessment of genome editing by sequence trace decomposition, Nucleic Acids Res, vol.42, p.168, 2014.

X. Wang, Unbiased detection of off-target cleavage by CRISPR-Cas9 and TALENs using integrase-defective lentiviral vectors, Nat. Biotechnol, vol.33, pp.175-178, 2015.

R. Gabriel, An unbiased genome-wide analysis of zinc-finger nuclease specificity, Nat. Biotechnol, vol.29, pp.816-823, 2011.

V. Poletti, Preclinical development of a lentiviral vector for gene therapy of X-linked severe combined immunodeficiency, Mol. Ther. Methods Clin. Dev, vol.9, pp.257-269, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02178311

P. Simioni, X-linked thrombophilia with a mutant factor IX (factor IX Padua), N. Engl. J. Med, vol.361, pp.1671-1675, 2009.

A. Miccio, In vivo selection of genetically modified erythroblastic progenitors leads to long-term correction of beta-thalassemia, Proc. Natl. Acad. Sci. USA 105, pp.10547-10552, 2008.

Z. Romero, Editing the sickle cell disease mutation in human hematopoietic stem cells: comparison of endonucleases and homologous donor templates, Mol. Ther, vol.27, pp.1389-1406, 2019.

G. Schiroli, Precise gene editing preserves hematopoietic stem cell function following transient p53-mediated DNA damage response, Cell Stem Cell, vol.24, p.558, 2019.

A. H. Futerman and G. Van-meer, The cell biology of lysosomal storage disorders, Nat. Rev. Mol. Cell Biol, vol.5, pp.554-565, 2004.

F. Ishikawa, Development of functional human blood and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice, Blood, vol.106, pp.1565-1573, 2005.

Z. Hu, N. Van-rooijen, and Y. G. Yang, Macrophages prevent human red blood cell reconstitution in immunodeficient mice, Blood, vol.118, pp.5938-5946, 2011.

C. A. Vakulskas, A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells, Nat. Med, vol.24, pp.1216-1224, 2018.

F. A. Ran, Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity, Cell, vol.154, pp.1380-1389, 2013.

G. Cullot, CRISPR-Cas9 genome editing induces megabase-scale chromosomal truncations, Nat. Commun, vol.10, p.1136, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02395971

T. Braulke and J. S. Bonifacino, Sorting of lysosomal proteins, Biochim. Biophys. Acta, vol.1793, pp.605-614, 2009.

S. P. Mcdermott, K. Eppert, E. R. Lechman, M. Doedens, and J. E. Dick, Comparison of human cord blood engraftment between immunocompromised mouse strains, Blood, vol.116, pp.193-200, 2010.

H. A. Wallace, Manipulating the mouse genome to engineer precise functional syntenic replacements with human sequence, Cell, vol.128, pp.197-209, 2007.

J. E. Wraith, Limitations of enzyme replacement therapy: current and future, J. Inherit. Metab. Dis, vol.29, pp.442-447, 2006.

J. Wang, Neutralizing antibodies to therapeutic enzymes: considerations for testing, prevention and treatment, Nat. Biotechnol, vol.26, pp.901-908, 2008.

R. W. Herzog, Complexity of immune responses to AAV transgene products -Example of factor IX, Cell Immunol, vol.342, p.103658, 2019.

N. Cartier, Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy, Science, vol.326, pp.818-823, 2009.

A. Biffi, Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy, Science, vol.341, p.1233158, 2013.

Y. Chen, J. A. Schroeder, E. L. Kuether, G. Zhang, and Q. Shi, Platelet gene therapy by lentiviral gene delivery to hematopoietic stem cells restores hemostasis and induces humoral immune tolerance in FIX(null) mice, Mol. Ther, vol.22, pp.169-177, 2014.

A. Ramezani, L. A. Zweier-renn, and R. G. Hawley, Factor VIII delivered by haematopoietic stem cell-derived B cells corrects the phenotype of haemophilia A mice, Thromb. Haemost, vol.105, pp.676-687, 2011.

Q. Shi, Platelet gene therapy corrects the hemophilic phenotype in immunocompromised hemophilia A mice transplanted with genetically manipulated human cord blood stem cells, Blood, vol.123, pp.395-403, 2014.

A. H. Chang, M. T. Stephan, L. Lisowski, and M. Sadelain, Erythroid-specific human factor IX delivery from in vivo selected hematopoietic stem cells following nonmyeloablative conditioning in hemophilia B mice, Mol. Ther, vol.16, pp.1745-1752, 2008.

D. Wang, Engineering a lysosomal enzyme with a derivative of receptorbinding domain of apoE enables delivery across the blood-brain barrier, Proc. Natl. Acad. Sci. USA, vol.110, pp.2999-3004, 2013.

A. J. Grimm, S. Kontos, G. Diaceri, X. Quaglia-thermes, and J. A. Hubbell, Memory of tolerance and induction of regulatory T cells by erythrocytetargeted antigens, Sci. Rep, vol.5, p.15907, 2015.

S. Kontos, I. C. Kourtis, K. Y. Dane, and J. A. Hubbell, Engineering antigens for in situ erythrocyte binding induces T-cell deletion, Proc. Natl. Acad. Sci. USA, vol.110, pp.60-68, 2013.

N. Pishesha, Engineered erythrocytes covalently linked to antigenic peptides can protect against autoimmune disease, Proc. Natl. Acad. Sci. USA, vol.114, pp.3157-3162, 2017.

N. J. Huang, Genetically engineered red cells expressing single domain camelid antibodies confer long-term protection against botulinum neurotoxin, Nat. Commun, vol.8, p.423, 2017.

S. Marktel, Intrabone hematopoietic stem cell gene therapy for adult and pediatric patients affected by transfusion-dependent ss-thalassemia, Nat. Med, vol.25, pp.234-241, 2019.

E. Abadir, C. Bryant, S. Larsen, and G. J. Clark, Targeting the niche: depleting haemopoietic stem cells with targeted therapy, Bone Marrow Transpl, vol.54, pp.961-968, 2019.

M. J. Domingues, S. K. Nilsson, and B. Cao, New agents in HSC mobilization, Int J. Hematol, vol.105, pp.141-152, 2017.

A. Lombardo, Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery, Nat. Biotechnol, vol.25, pp.1298-1306, 2007.

I. Theurl, On-demand erythrocyte disposal and iron recycling requires transient macrophages in the liver, Nat. Med, vol.22, pp.945-951, 2016.

H. Wang, High-level protein production in erythroid cells derived from in vivo transduced hematopoietic stem cells, Blood Adv, vol.3, pp.2883-2894, 2019.

B. Oller-salvia, M. Sanchez-navarro, E. Giralt, and M. Teixido, Blood-brain barrier shuttle peptides: an emerging paradigm for brain delivery, Chem. Soc. Rev, vol.45, pp.4690-4707, 2016.

B. J. Ebbink, Classic infantile Pompe patients approaching adulthood: a cohort study on consequences for the brain, Dev. Med. Child Neurol, vol.60, pp.579-586, 2018.

A. M. Kabadi, D. G. Ousterout, I. B. Hilton, and C. A. Gersbach, Multiplex CRISPR/Cas9-based genome engineering from a single lentiviral vector, Nucleic Acids Res, vol.42, p.147, 2014.

S. Karnan, Improved methods of AAV-mediated gene targeting for human cell lines using ribosome-skipping 2A peptide, Nucleic Acids Res, vol.44, p.54, 2016.

J. H. Kim, High cleavage efficiency of a 2A peptide derived from porcine teschovirus-1 in human cell lines, zebrafish and mice, PLoS ONE, vol.6, p.18556, 2011.

A. Follenzi, L. E. Ailles, S. Bakovic, M. Geuna, and L. Naldini, Gene transfer by lentiviral vectors is limited by nuclear translocation and rescued by HIV-1 pol sequences, Nat. Genet, vol.25, pp.217-222, 2000.

A. Lattanzi, Optimization of CRISPR/Cas9 delivery to human hematopoietic stem and progenitor cells for therapeutic genomic rearrangements, Mol. Ther, vol.27, pp.137-150, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02880803

E. Ayuso, F. Mingozzi, and F. Bosch, Production, purification and characterization of adeno-associated vectors, Curr. Gene Ther, vol.10, pp.423-436, 2010.

U. P. Rohr, Fast and reliable titration of recombinant adeno-associated virus type-2 using quantitative real-time PCR, J. Virol. Methods, vol.106, pp.81-88, 2002.

M. C. Canver, BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis, Nature, vol.527, pp.192-197, 2015.

S. Menoret, Homology-directed repair in rodent zygotes using Cas9 and TALEN engineered proteins, Sci. Rep, vol.5, p.14410, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01271745

J. Wen, W. Tao, S. Hao, and Y. Zu, Cellular function reinstitution of offspring red blood cells cloned from the sickle cell disease patient blood post CRISPR genome editing, J. Hematol. Oncol, vol.10, p.119, 2017.

A. Meliani, Enhanced liver gene transfer and evasion of preexisting humoral immunity with exosome-enveloped AAV vectors, Blood Adv, vol.1, pp.2019-2031, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02377508

F. Aguisanda, Neural stem cells for disease modeling of Wolman disease and evaluation of therapeutics, Orphanet. J. Rare Dis, vol.12, p.120, 2017.

J. Hamilton, I. Jones, R. Srivastava, and P. Galloway, A new method for the measurement of lysosomal acid lipase in dried blood spots using the inhibitor Lalistat 2, Clin. Chim. Acta, vol.413, pp.1207-1210, 2012.

C. A. Schneider, W. S. Rasband, and K. W. Eliceiri, NIH Image to ImageJ: 25 years of image analysis, Nat. Methods, vol.9, pp.671-675, 2012.