D. Deamer, M. Akeson, and D. Branton, Three decades of nanopore sequencing, Nat Biotechnol, vol.34, pp.518-542, 2016.

J. J. Kasianowicz, E. Brandin, D. Branton, and D. W. Deamer, Characterization of individual polynucleotide molecules using a membrane channel, Proc Natl Acad Sci, vol.93, pp.13770-13773, 1996.

D. Stoddart, A. J. Heron, E. Mikhailova, G. Maglia, and H. Bayley, Single-nucleotide discrimination in immobilized DNA oligonucleotides with a biological nanopore, Proc Natl Acad Sci, vol.106, pp.7702-7709, 2009.

K. R. Lieberman, G. M. Cherf, M. J. Doody, F. Olasagasti, Y. Kolodji et al., Processive Replication of Single DNA Molecules in a Nanopore Catalyzed by phi29 DNA Polymerase, J Am Chem Soc, vol.132, pp.17961-72, 2010.

M. Akeson, D. Branton, J. J. Kasianowicz, E. Brandin, and D. W. Deamer, Microsecond Time-Scale Discrimination Among Polycytidylic Acid, Polyadenylic Acid, and Polyuridylic Acid as Homopolymers or as Segments Within Single RNA Molecules, Biophys J, vol.77, pp.3227-3260, 1999.

J. Eid, A. Fehr, J. Gray, K. Luong, J. Lyle et al., Real-time DNA sequencing from single polymerase molecules, Science, vol.323, pp.133-141, 2009.

A. M. Wenger, P. Peluso, W. J. Rowell, P. Chang, R. J. Hall et al., Accurate circular consensus long-read sequencing improves variant detection and assembly of a human genome, Nat Biotechnol, pp.1-8, 2019.

M. Loose, S. Malla, and M. Stout, Real-time selective sequencing using nanopore technology, Nat Methods, vol.13, pp.751-755, 2016.

M. W. Snyder, A. Adey, J. O. Kitzman, and J. Shendure, Haplotype-resolved genome sequencing: experimental methods and applications, Nat Rev Genet, vol.16, pp.344-58, 2015.

A. C. Rand, M. Jain, J. M. Eizenga, A. Musselman-brown, H. E. Olsen et al., Mapping DNA methylation with high-throughput nanopore sequencing, Nat Methods, vol.14, pp.411-414, 2017.

Q. Liu, L. Fang, G. Yu, D. Wang, C. Xiao et al., Detection of DNA base modifications by deep recurrent neural network on Oxford Nanopore sequencing data, Nat Commun, vol.10, p.2449, 2019.

F. Montel, Séquençage de l'ADN par nanopores. médecine/sciences, vol.34, pp.161-166, 2018.

B. E. Slatko, A. F. Gardner, and F. M. Ausubel, Overview of Next-Generation Sequencing Technologies, Curr Protoc Mol Biol, vol.122, p.59, 2018.

T. Gabrieli, H. Sharim, D. Fridman, N. Arbib, Y. Michaeli et al., Selective nanopore 88 sequencing of human BRCA1 by Cas9-assisted targeting of chromosome segments (CATCH), Nucleic Acids Res, vol.46, pp.87-87, 2018.

T. Gilpatrick, I. Lee, J. E. Graham, E. Raimondeau, R. Bowen et al., Targeted Nanopore Sequencing with Cas9 for studies of methylation, structural variants and mutations. bioRxiv, vol.604173, 2019.

J. Sone, S. Mitsuhashi, A. Fujita, T. Mizuguchi, K. Hamanaka et al., Long-read sequencing identifies GGC repeat expansions in NOTCH2NLC associated with neuronal intranuclear inclusion disease, Nat Genet, vol.1, 2019.

M. Jinek, K. Chylinski, I. Fonfara, M. Hauer, J. A. Doudna et al., A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity, Science, vol.337, pp.816-837, 2012.

K. A. Schaefer, W. Wu, D. F. Colgan, S. H. Tsang, A. G. Bassuk et al., Unexpected mutations after CRISPR-Cas9 editing in vivo, Nat Methods, vol.14, pp.547-555, 2017.

H. Ma, N. Marti-gutierrez, S. Park, J. Wu, Y. Lee et al., Correction of a pathogenic gene mutation in human embryos, Nature, vol.548, pp.413-422, 2017.

D. P. Dever, R. O. Bak, A. Reinisch, J. Camarena, G. Washington et al., CRISPR/Cas9 ?-globin gene targeting in human haematopoietic stem cells, Nature, vol.539, pp.384-393, 2016.

H. Morin, La naissance des « bébés Crispr » suscite une condamnation universelle. Le Monde, 2018.

P. D. Hsu, E. S. Lander, and F. Zhang, Development and Applications of CRISPR-Cas9 for Genome Engineering, Cell, vol.157, pp.1262-78, 2014.

A. Pickar-oliver and C. A. Gersbach, The next generation of CRISPR-Cas technologies and applications, Nat Rev Mol Cell Biol, vol.20, pp.490-507, 2019.

R. R. Wick, L. M. Judd, and K. E. Holt, Performance of neural network basecalling tools for Oxford Nanopore sequencing, Genome Biol, vol.20, p.129, 2019.

F. Pfeiffer, C. Gröber, M. Blank, K. Händler, M. Beyer et al., Systematic evaluation of error rates and causes in short samples in next-generation sequencing, Sci Rep, vol.8, p.10950, 2018.

M. Schirmer, D. 'amore, R. Ijaz, U. Z. Hall, N. Quince et al., Illumina error profiles: resolving finescale variation in metagenomic sequencing data, BMC Bioinformatics, vol.17, p.125, 2016.

M. A. Depristo, E. Banks, R. Poplin, K. Garimella, J. R. Maguire et al., A framework for variation discovery and genotyping using next-generation DNA sequencing data, Nat Genet, vol.43, pp.491-499, 2011.

, Association Nationale des Praticiens de Génétique Moléculaire, GT "Cahier des charges informatique, bio-analyse/bioinformatique, bases de données mutations

, Recommandations générales pour la gestion informatique des données et des analyses de séquençage à haut débit pour les laboratoires de diagnostic moléculaire de maladies génétiques

E. Afgan, D. Baker, B. Batut, M. Van-den-beek, D. Bouvier et al., The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2018 update, Nucleic Acids Res, vol.46, pp.537-581, 2018.

G. R. Wilkinson, Drug Metabolism and Variability among Patients in Drug Response, N Engl J Med, vol.352, pp.2211-2232, 2005.

M. Pirmohamed, Personalized pharmacogenomics: predicting efficacy and adverse drug reactions, Annu Rev Genomics Hum Genet, vol.15, pp.349-70, 2014.

L. Wang, H. L. Mcleod, and R. M. Weinshilboum, Genomics and drug response, N Engl J Med, vol.364, pp.1144-53, 2011.

D. Allorge and M. Loriot, Pharmacogenetics or the promise of a personalized medicine: variability in drug metabolism and transport

, Ann Biol Clin (Paris), vol.62, pp.499-511, 2004.

R. Weinshilboum, Inheritance and Drug Response. Guttmacher AE, Collins FS, N Engl J Med, vol.348, pp.529-566, 2003.

U. A. Meyer, Pharmacogenetics -five decades of therapeutic lessons from genetic diversity, Nat Rev Genet, vol.5, pp.669-76, 2004.

U. A. Meyer, U. M. Zanger, and M. Schwab, Omics and drug response, Annu Rev Pharmacol Toxicol, vol.53, pp.475-502, 2013.

D. M. Roden, H. L. Mcleod, M. Relling, M. S. Williams, G. A. Mensah et al., Pharmacogenomics. Lancet, vol.394, pp.521-553, 2019.

C. H. Wouden, M. H. Van-rhenen, W. Jama, M. Ingelman-sundberg, V. M. Lauschke et al., Development of the PG x-Passport: A Panel of Actionable Germline Genetic Variants for Pre-Emptive Pharmacogenetic Testing, Clin Pharmacol Ther, 2019.

M. Kozyra, M. Ingelman-sundberg, and V. M. Lauschke, Rare genetic variants in cellular transporters, metabolic enzymes, and nuclear receptors can be important determinants of interindividual differences in drug response, Genet Med, vol.19, pp.20-29, 2017.

G. Wright, B. Carleton, M. R. Hayden, and C. Ross, The global spectrum of protein-coding pharmacogenomic diversity, Pharmacogenomics J, vol.18, pp.187-95, 2018.

A. K. Daly, J. Brockmöller, F. Broly, M. Eichelbaum, W. E. Evans et al., Nomenclature for human CYP2D6 alleles, Pharmacogenetics, vol.6, pp.193-201, 1996.

A. Gaedigk, J. Dinh, H. Jeong, B. Prasad, and J. Leeder, Ten Years' Experience with the CYP2D6 Activity Score: A Perspective on Future Investigations to Improve Clinical Predictions for Precision Therapeutics, J Pers Med, vol.8, p.15, 2018.

M. Ingelman-sundberg, Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity, Pharmacogenomics J, vol.5, pp.6-13, 2005.

A. Llerena, M. Naranjo, F. Rodrigues-soares, E. M. Penas-lledó, H. Fariñas et al., Interethnic variability of CYP2D6 alleles and of predicted and measured metabolic phenotypes across world populations, Expert Opin Drug Metab Toxicol, vol.10, pp.1569-83, 2014.

A. Gaedigk, S. Simon, R. Pearce, L. Bradford, M. Kennedy et al., The CYP2D6 Activity Score: Translating Genotype Information into a Qualitative Measure of Phenotype, Clin Pharmacol Ther, vol.83, pp.234-276, 2008.

U. M. Zanger and M. Schwab, Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation, Pharmacol Ther, vol.138, pp.103-144, 2013.

M. M. Jukic, R. L. Smith, T. Haslemo, E. Molden, and M. Ingelman-sundberg, Effect of CYP2D6 genotype on exposure and efficacy of risperidone and aripiprazole: a retrospective, cohort study, The Lancet Psychiatry, vol.6, pp.418-444, 2019.

W. S. Oetting, B. Wu, D. P. Schladt, W. Guan, R. P. Remmel et al., Genome-wide association study identifies the common variants in CYP3A4 and CYP3A5 responsible for variation in tacrolimus trough concentration in Caucasian kidney transplant recipients, Pharmacogenomics J, vol.18, pp.501-506, 2018.

K. Birdwell, B. Decker, J. Barbarino, J. Peterson, C. Stein et al., Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for CYP3A5 Genotype and Tacrolimus Dosing, Clin Pharmacol Ther, vol.98, pp.19-24, 2015.

M. V. Relling, M. Schwab, M. Whirl-carrillo, G. Suarez-kurtz, C. Pui et al., Clinical Pharmacogenetics Implementation Consortium Guideline for Thiopurine Dosing Based on TPMT and NUDT 15 Genotypes, Clin Pharmacol Ther, vol.105, pp.1095-105, 2018.

T. Moriyama, R. Nishii, V. Perez-andreu, Y. W. Klussmann, F. A. Zhao et al., NUDT15 polymorphisms alter thiopurine metabolism and hematopoietic toxicity, Nat Genet, vol.48, pp.367-73, 2016.

G. J. Walker, J. W. Harrison, G. A. Heap, M. D. Voskuil, V. Andersen et al., Association of Genetic Variants in NUDT15 With Thiopurine-Induced Myelosuppression in Patients With Inflammatory Bowel Disease, JAMA, vol.321, p.773, 2019.

R. L. Roberts, M. C. Wallace, M. L. Seinen, A. A. Van-bodegraven, K. Krishnaprasad et al., Nonsynonymous Polymorphism in Guanine Monophosphate Synthetase Is a Risk Factor for Unfavorable Thiopurine Metabolite Ratios in Patients With Inflammatory Bowel Disease, Inflamm Bowel Dis, vol.24, pp.2606-2618, 2018.

A. Wilson, L. E. Jansen, R. V. Rose, J. C. Gregor, T. Ponich et al., HLA-DQA1-HLA-DRB1 polymorphism is a major predictor of azathioprine-induced pancreatitis in patients with inflammatory bowel disease, Aliment Pharmacol Ther, vol.47, pp.615-635, 2018.

G. A. Heap, M. N. Weedon, C. M. Bewshea, A. Singh, M. Chen et al., HLA-DQA1-HLA-DRB1 variants confer susceptibility to pancreatitis induced by thiopurine immunosuppressants, Nat Genet, vol.46, pp.1131-1135, 2014.

C. Lunenburg, L. M. Henricks, A. Van-kuilenburg, R. Mathijssen, J. Schellens et al., Diagnostic and Therapeutic Strategies for Fluoropyrimidine Treatment of Patients Carrying Multiple DPYD Variants, Genes (Basel), vol.9, p.585, 2018.

M. Lindqvist, K. Skoglund, A. Karlgren, P. Söderkvist, C. Peterson et al., Explaining TPMT genotype/phenotype discrepancy by haplotyping of TPMT*3A and identification of a novel sequence variant, TPMT*23, Pharmacogenet Genomics, vol.17, pp.891-896, 2007.

, Recherche de déficit en dihydropyrimidine déshydrogénase en vue de prévenir certaines toxicités sévères survenant sous traitement comportant des fluoropyrimidines (5-fluorouracile), Recommandations et référentiels, INCa, HAS. Décembre, 2018.

L. M. Henricks, C. Lunenburg, F. M. De-man, D. Meulendijks, G. Frederix et al., DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis, Lancet Oncol, vol.19, pp.1459-67, 2018.

M. Loriot, J. Ciccolini, F. Thomas, C. Barin-le-guellec, B. Royer et al., Dihydropyrimidine déhydrogenase (DPD) deficiency screening and securing of fluoropyrimidine-based chemotherapies: Update and recommendations of the French GPCO-Unicancer and

, Bull Cancer, vol.105, pp.397-407, 2018.

L. M. Henricks, F. L. Opdam, J. H. Beijnen, A. Cats, and J. Schellens, DPYD genotype-guided dose individualization to improve patient safety of fluoropyrimidine therapy: call for a drug label update, Ann Oncol, vol.28, pp.2915-2937, 2017.

, A global reference for human genetic variation, Nature, vol.526, pp.68-74, 2015.

J. Boyer, E. Thomas, F. Quaranta, S. Picard, N. Loriot et al., Bull Cancer, vol.101, pp.533-53, 2014.

J. Woillard, L. Chouchana, N. Picard, and M. Loriot, Pharmacogenetics of immunosuppressants: State of the art and clinical implementation -recommendations from the French National Network of Pharmacogenetics (RNPGx), Therapies, vol.72, pp.285-99, 2017.

S. E. Levy and R. M. Myers, Advancements in Next-Generation Sequencing, Annu Rev Genomics Hum Genet, vol.17, pp.95-115, 2016.

D. C. Koboldt, K. M. Steinberg, D. E. Larson, R. K. Wilson, and E. R. Mardis, The next-generation sequencing revolution and its impact on genomics, Cell, vol.155, pp.27-38, 2013.

D. Aird, M. G. Ross, W. Chen, M. Danielsson, T. Fennell et al., Analyzing and minimizing PCR amplification bias in Illumina sequencing libraries, Genome Biol, vol.12, p.18, 2011.

V. M. Lauschke, L. Milani, and M. Ingelman-sundberg, Pharmacogenomic Biomarkers for Improved Drug Therapy-Recent Progress and Future Developments, AAPS J, vol.20, p.4, 2018.

T. Charalampous, G. L. Kay, H. Richardson, A. Aydin, R. Baldan et al., Nanopore metagenomics enables rapid clinical diagnosis of bacterial lower respiratory infection, Nat Biotechnol, vol.37, pp.783-792, 2019.

J. Quick, N. J. Loman, S. Duraffour, J. T. Simpson, E. Severi et al., Real-time, portable genome sequencing for Ebola surveillance, Nature, vol.530, pp.228-260, 2016.

C. Belser, B. Istace, E. Denis, M. Dubarry, F. Baurens et al., Chromosome-scale assemblies of plant genomes using nanopore long reads and optical maps, Nat Plants, vol.4, pp.879-87, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01977046

R. Bowden, R. W. Davies, A. Heger, A. T. Pagnamenta, M. De-cesare et al., Sequencing of human genomes with nanopore technology, Nat Commun, vol.10, p.1869, 2019.

M. Jain, S. Koren, K. H. Miga, J. Quick, A. C. Rand et al., Nanopore sequencing and assembly of a human genome with ultra-long reads, Nat Biotechnol, vol.36, pp.338-383, 2018.

Y. Liau, S. Maggo, A. L. Miller, J. F. Pearson, M. A. Kennedy et al., Nanopore sequencing of the pharmacogene CYP2D6 allows simultaneous haplotyping and detection of duplications. bioRxiv, vol.576280, 2019.

P. Orsini, C. F. Minervini, C. Cumbo, L. Anelli, A. Zagaria et al., Design and MinION testing of a nanopore targeted gene sequencing panel for chronic lymphocytic leukemia, Sci Rep, vol.8, p.11798, 2018.

Y. Zhang, Y. Zhang, J. M. Burke, K. Gleitsman, S. M. Friedrich et al., A Simple Thermoplastic Substrate Containing Hierarchical Silica Lamellae for High-MolecularWeight DNA Extraction, Adv Mater, vol.28, pp.10630-10636, 2016.

K. Labun, T. G. Montague, J. A. Gagnon, S. B. Thyme, and E. Valen, CHOPCHOP v2: a web tool for the next generation of CRISPR genome engineering, Nucleic Acids Res, vol.44, pp.272-278, 2016.

H. Li, Minimap2: pairwise alignment for nucleotide sequences. Birol I, editor, Bioinformatics, vol.34, pp.3094-100, 2018.

H. Li, A statistical framework for SNP calling, mutation discovery, association mapping and population genetical parameter estimation from sequencing data, Bioinformatics, vol.27, pp.2987-93, 2011.

E. Garrison and G. Marth, Haplotype-based variant detection from short-read sequencing, 2012.

J. T. Simpson, R. E. Workman, P. C. Zuzarte, M. David, L. J. Dursi et al., Detecting DNA cytosine methylation using nanopore sequencing, Nat Methods, vol.14, pp.407-417, 2017.

M. Martin, M. Patterson, S. Garg, S. O. Fischer, N. Pisanti et al., WhatsHap: fast and accurate read-based phasing, bioRxiv, vol.085050, 2016.

B. Grüning, R. Dale, A. Sjödin, B. A. Chapman, J. Rowe et al., Bioconda: sustainable and comprehensive software distribution for the life sciences, Nat Methods, vol.15, pp.475-481, 2018.

J. T. Robinson, H. Thorvaldsdóttir, W. Winckler, M. Guttman, E. S. Lander et al., Integrative genomics viewer, Nat Biotechnol, vol.29, pp.24-30, 2011.

D. Coster, W. , D. Hert, S. Schultz, D. T. Cruts et al., NanoPack: visualizing and processing long-read sequencing data, Bioinformatics, vol.34, pp.2666-2675, 2018.

J. Koster and S. Rahmann, Snakemake--a scalable bioinformatics workflow engine, Bioinformatics, vol.28, pp.2520-2522, 2012.

H. Li and R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinformatics, vol.25, pp.1754-60, 2009.

D. C. Koboldt, Q. Zhang, D. E. Larson, D. Shen, M. D. Mclellan et al., VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing, Genome Res, vol.22, pp.568-76, 2012.

R. Poplin, P. Chang, A. D. Schwartz, S. Colthurst, T. Ku et al., A universal SNP and small-indel variant caller using deep neural networks, Nat Biotechnol, vol.36, pp.983-990, 2018.

A. Mckenna, M. Hanna, E. Banks, A. Sivachenko, K. Cibulskis et al., The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data, Genome Res, vol.20, pp.1297-303, 2010.

C. Chiang, R. M. Layer, G. G. Faust, M. R. Lindberg, D. B. Rose et al., SpeedSeq: ultra-fast personal genome analysis and interpretation, Nat Methods, vol.12, pp.966-974, 2015.

A. Supernat, O. V. Vidarsson, V. M. Steen, and T. Stokowy, Comparison of three variant callers for human whole genome sequencing, Sci Rep, vol.8, p.17851, 2018.

R. Luo, F. J. Sedlazeck, T. Lam, and M. C. Schatz, A multi-task convolutional deep neural network for variant calling in single molecule sequencing, Nat Commun, vol.10, p.998, 2019.

M. Kumaran, U. Subramanian, and B. Devarajan, Performance assessment of variant calling pipelines using human whole exome sequencing and simulated data, BMC Bioinformatics, vol.20, p.342, 2019.

D. R. Greig, C. Jenkins, S. Gharbia, and T. J. Dallman, Comparison of single-nucleotide variants identified by Illumina and Oxford Nanopore technologies in the context of a potential outbreak of Shiga toxin-producing Escherichia coli, Gigascience, vol.8, 2019.

Y. Liau, S. L. Cree, S. Maggo, A. L. Miller, J. F. Pearson et al., A Multiplex Pharmacogenetics Assay using the MinION Nanopore Sequencing Device

, , vol.563262

R. C. Stevens, J. L. Steele, W. R. Glover, J. F. Sanchez-garcia, S. D. Simpson et al., A novel CRISPR/Cas9 associated technology for sequence-specific nucleic acid enrichment, PLoS One, vol.14, p.215441, 2019.

M. Leija-salazar, F. J. Sedlazeck, M. Toffoli, S. Mullin, K. Mokretar et al., Evaluation of the detection of GBA missense mutations and other variants using the Oxford Nanopore MinION, Mol Genet Genomic Med, vol.7, p.564, 2019.

Ó. Mortensen, L. N. Lydersen, K. D. Apol, G. Andorsdóttir, B. Á. Steig et al., Using dried blood spot samples from a trio for linked-read whole-exome sequencing, Eur J Hum Genet, vol.27, pp.980-988, 2019.

G. Zheng, B. T. Lau, M. Schnall-levin, M. Jarosz, J. M. Bell et al., Haplotyping germline and cancer genomes with high-throughput linked-read sequencing, Nat Biotechnol, vol.34, pp.303-314, 2016.

M. Ivanov, M. Kacevska, and M. Ingelman-sundberg, Epigenomics and Interindividual Differences in Drug Response, Clin Pharmacol Ther, vol.92, pp.727-763, 2012.

J. E. Pool, I. Hellmann, J. D. Jensen, and R. Nielsen, Population genetic inference from genomic sequence variation, Genome Res, vol.20, pp.291-300, 2010.

E. Ahn and T. Park, Analysis of population-specific pharmacogenomic variants using nextgeneration sequencing data, Sci Rep, vol.7, p.8416, 2017.

S. Hwang, E. Kim, I. Lee, and E. M. Marcotte, Systematic comparison of variant calling pipelines using gold standard personal exome variants, Sci Rep, vol.5, p.17875, 2015.

M. T. Noakes, H. Brinkerhoff, A. H. Laszlo, I. M. Derrington, K. W. Langford et al., Increasing the accuracy of nanopore DNA sequencing using a time-varying cross membrane voltage, Nat Biotechnol, vol.37, pp.651-657, 2019.

F. J. Rang, W. P. Kloosterman, and J. De-ridder, From squiggle to basepair: computational approaches for improving nanopore sequencing read accuracy, Genome Biol, vol.19, p.90, 2018.