, Détection de fractions mutées élevées (>1%)

, Le coefficient de corrélation R entre valeurs théoriques et observées, était optimal pour l'ADNg, un peu moins pour l'ADNc. Références

H. Dohner, E. Estey, and D. Grimwade, Blood, vol.129, issue.4, pp.424-448, 2017.

P. Voigt and D. Reinberg, Genomic and Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia The Cancer Genome Atlas Research Network, N Engl J Med, 2013.

M. Alikian, R. P. Gale, J. F. Apperley, L. Foroni, and M. Alikian, Molecular techniques for the personalised management of patients with chronic myeloid leukaemia. Biomol Detect Quantif, 2017.

H. Döhner, D. J. Weisdorf, and C. D. Bloomfield, Acute Myeloid Leukemia, N Engl J Med, 2015.

J. K. Box, N. Paquet, and M. N. Adams, Nucleophosmin: From structure and function to disease development, BMC Mol Biol, vol.17, issue.1, pp.1-12, 2016.

E. M. Heath, S. M. Chan, M. D. Minden, T. Murphy, L. I. Shlush et al., Biological and clinical consequences of NPM1 mutations in AML, Leukemia, vol.31, issue.4, pp.798-807, 2017.

B. Falini, N. Bolli, and A. Liso, Altered nucleophosmin transport in acute myeloid leukaemia with mutated NPM1: Molecular basis and clinical implications, Leukemia, 2009.
DOI : 10.1038/leu.2009.124

URL : http://www.nature.com/leu/journal/v23/n10/pdf/leu2009124a.pdf

I. Détection, K. Perez-toralla, and D. Pekin, PCR digitale en micro-compartiments, vol.31, pp.84-92, 2015.

E. Lee, K. J. Lee, and H. Park, Clinical Implications of Quantitative JAK2 V617F Analysis using Droplet Digital PCR in Myeloproliferative Neoplasms, Ann Lab Med, vol.38, issue.2, p.147, 2018.

N. Boissel, A. Renneville, and V. Biggio, Prevalence, clinical profile, and prognosis of NPM mutations in AML with normal karyotype, Blood, 2005.

B. Falini, C. Mecucci, and E. Tiacci, Cytoplasmic Nucleophosmin in Acute Myelogenous Leukemia with a Normal Karyotype, N Engl J Med, 2005.
DOI : 10.1056/nejmoa041974

URL : http://www.nejm.org/doi/pdf/10.1056/NEJMoa041974

D. Grimwade and S. D. Freeman, Defining minimal residual disease in acute myeloid leukemia: Which platforms are ready for "prime time, Blood, 2014.
DOI : 10.1182/asheducation-2014.1.222

URL : http://asheducationbook.hematologylibrary.org/content/2014/1/222.full.pdf

A. Karam and N. Aziz, Molecular landscape in acute myeloid leukemia: where do we stand in 2016, Cancer Biol Med, vol.13, issue.4, p.474, 2016.
DOI : 10.20892/j.issn.2095-3941.2016.0061

URL : http://www.cancerbiomed.org/index.php/cocr/article/download/985/1033

E. Papaemmanuil, M. Gerstung, and L. Bullinger, Genomic Classification and Prognosis in Acute Myeloid Leukemia, N Engl J Med, vol.374, issue.23, pp.2209-2221, 2016.

L. Bullinger, K. Döhner, and H. Dohner, Genomics of acute myeloid leukemia diagnosis and pathways, J Clin Oncol, vol.35, issue.9, pp.934-946, 2017.

R. L. Siegel, K. D. Miller, and A. Jemal, Cancer statistics, CA Cancer J Clin, vol.68, issue.1, pp.7-30, 2018.
DOI : 10.3322/caac.21208

A. Monnereau, F. Binder-foucard, and L. Remontet, Estimation nationale de l'incidence des cancers en France entre 1980 et 2012_Etude à partir des registres des, cancers du réseau Francim. InVS, 2013.

E. Estey, D. Grimwade, and S. Amadori, Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel, vol.129, pp.424-448, 2017.

S. Gröschel, M. A. Sanders, and R. Hoogenboezem, A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in Leukemia, Cell, vol.157, issue.2, pp.369-381, 2014.

E. P. Nacheva, C. D. Grace, and D. Brazma, Does BCR/ABL1 positive Acute Myeloid Leukaemia Exist?, Br J Haematol, vol.161, issue.4, pp.541-550, 2013.

B. J. Wouters, B. Löwenberg, C. Erpelinck-verschueren, W. Van-putten, P. Valk et al., Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome, Blood, vol.113, issue.13, pp.3088-3091, 2009.

V. I. Gaidzik, V. Teleanu, and E. Papaemmanuil, RUNX1 mutations in acute myeloid leukemia are associated with distinct clinico-pathologic and genetic features
DOI : 10.1038/leu.2016.126

, Leukemia, vol.30, p.2282, 2016.

U. Bacher, S. Schnittger, and K. Macijewski, Multilineage dysplasia does not influence prognosis in CEBPA-mutated AML, supporting the WHO proposal to classify these patients as a unique entity, Blood, vol.119, issue.20, pp.4719-4722, 2012.

R. F. Schlenk, E. Taskesen, and Y. Van-norden, The value of allogeneic and autologous hematopoietic stem cell transplantation in prognostically favorable acute myeloid leukemia with double mutant CEBPA, Blood, vol.122, issue.9, pp.1576-1582, 2013.

C. Haferlach, C. Mecucci, and S. Schnittger, AML with mutated NPM1 carrying a normal or aberrant karyotype show overlapping biologic, pathologic, immunophenotypic, and prognostic features, Blood, vol.114, issue.14, pp.3024-3032, 2009.

B. Medsker, E. Forno, H. Simhan, and C. Juan, Sciences R. HHS Public Access, vol.70, issue.12, pp.773-779, 2016.

R. B. Walter, M. Othus, and A. K. Burnett, Signi fi cance of FAB subclassi fi cation of " acute myeloid leukemia , NOS " in the 2008 WHO classi fi cation : analysis of 5848 newly diagnosed patients, Bloodjournal.org, vol.121, issue.13, pp.2424-2432, 2015.

A. F. Yilmaz, G. Saydam, F. Sahin, and Y. Baran, Granulocytic sarcoma: a systematic review, Am J Blood Res, vol.3, issue.4, pp.265-270, 2013.

A. Roy, I. Roberts, and P. Vyas, Biology and management of transient abnormal myelopoiesis (TAM) in children with Down syndrome, Semin Fetal Neonatal Med, 2012.

K. Yoshida, T. Toki, and Y. Okuno, The landscape of somatic mutations in Down syndrome-related myeloid disorders, Nat Genet, 2013.

E. Matutes, W. F. Pickl, M. Veer, and . Van, outcome in 100 patients defined according to the WHO 2008 classification Mixed-phenotype acute leukemia : clinical and laboratory features and outcome in 100 patients defined according to the WHO 2008 classification, vol.117, pp.3163-3171, 2014.

W. Van-den-ancker, M. Terwijn, and T. M. Westers, Acute leukemias of ambiguous lineage: Diagnostic consequences of the WHO2008 classification, Leukemia, 2010.

C. Kawajiri, H. Tanaka, and S. Hashimoto, Successful treatment of Philadelphia chromosome-positive mixed phenotype acute leukemia by appropriate alternation of second-generation tyrosine kinase inhibitors according to BCR-ABL1 mutation status

, Int J Hematol, 2014.

J. M. Bennett, D. Catovsky, and M. T. Daniel, Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group, Br J Haematol, vol.33, issue.4, pp.451-458, 1976.

M. C. Béné, T. Nebe, and P. Bettelheim, Immunophenotyping of acute leukemia and lymphoproliferative disorders: A consensus proposal of the European LeukemiaNet Work Package 10, Leukemia, vol.25, issue.4, pp.567-574, 2011.

D. C. Linch, R. K. Hills, A. K. Burnett, A. Khwaja, and R. E. Gale, Impact of FLT3ITD mutant allele level on relapse risk in intermediate-risk acute myeloid leukemia, Blood, vol.124, issue.2, pp.273-276, 2014.

M. Pratcorona, S. Abbas, and M. A. Sanders, Acquired mutations in ASXL1 in acute myeloid leukemia: Prevalence and prognostic value, Haematologica, 2012.

F. G. Rücker, R. F. Schlenk, and L. Bullinger, TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome, Blood, 2012.

J. Saultz and R. Garzon, Acute Myeloid Leukemia: A Concise Review, J Clin Med, 2016.

S. C. Meyer and R. L. Levine, Translational implications of somatic genomics in acute myeloid leukaemia, Lancet Oncol, 2014.

M. Medinger, C. Lengerke, and J. Passweg, Novel Prognostic and Therapeutic Mutations in Acute Myeloid Leukemia, Cancer Genomics Proteomics, 2016.

R. E. Gale, C. Green, and C. Allen, The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia, Blood, 2008.

S. Kayser, R. F. Schlenk, and M. C. Londono, Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome, Blood, 2009.

C. Lu, P. S. Ward, and G. S. Kapoor, IDH mutation impairs histone demethylation and results in a block to cell differentiation, Nature, 2012.

J. P. Patel, M. Gönen, and M. E. Figueroa, Prognostic relevance of integrated genetic profiling in acute myeloid leukemia, N Engl J Med, 2012.

T. J. Ley, L. Ding, and M. J. Walter, DNMT3A mutations in acute myeloid leukemia, N Engl J Med, 2010.

R. Garzon, S. Volinia, and D. Papaioannou, Expression and prognostic impact of lncRNAs in acute myeloid leukemia, Proc Natl Acad Sci, 2014.

L. I. Shlush, S. Zandi, and A. Mitchell, Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia, Nature, 2014.

K. H. Metzeler, K. Maharry, and M. D. Radmacher, TET2 mutations improve the new European LeukemiaNet risk classification of acute myeloid leukemia: A cancer and leukemia group B study, J Clin Oncol, 2011.

W. C. Chou, S. C. Chou, and C. Y. Liu, TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics, Blood, 2011.

J. Tang, H. Hou, and C. Chen, AML1 / RUNX1 mutations in 470 adult patients with de novo acute myeloid leukemia : prognostic implication and interaction with other gene alterations, Blood, vol.114, issue.26, pp.5352-5361, 2009.

J. H. Mendler, K. Maharry, and M. D. Radmacher, RUNX1 mutations are associated with poor outcome in younger and older patients with cytogenetically normal acute myeloid leukemia and with distinct gene and MicroRNA expression signatures, J Clin Oncol, 2012.

S. Koschmieder, B. Halmos, E. Levantini, and D. G. Tenen, Dysregulation of the C/EBPalpha differentiation pathway in human cancer, J Clin Oncol, 2009.

A. Fasan, C. Haferlach, and T. Alpermann, The role of different genetic subtypes of CEBPA mutated AML, Leukemia, 2014.

K. H. Metzeler, H. Becker, and K. Maharry, ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal AML within the ELN Favorable genetic category, Blood, 2011.

T. Alpermann, C. Haferlach, and C. Eder, AML with gain of chromosome 8 as the sole chromosomal abnormality (+8sole) is associated with a specific molecular mutation pattern including ASXL1 mutations in 46.8% of the patients, Leuk Res, 2015.

P. Ernst, J. Wang, and S. J. Korsmeyer, The role of MLL in hematopoiesis and leukemia, Curr Opin Hematol, 2002.

T. M. Kadia, P. Jain, and F. Ravandi, TP53 mutations in newly diagnosed acute myeloid leukemia: Clinicomolecular characteristics, response to therapy, and outcomes. Cancer, 2016.

L. Yang, Y. Han, S. Saiz, F. Minden, and M. D. , A tumor suppressor and oncogene: The WT1 story, Leukemia, 2007.

C. Woehlecke, S. Wittig, C. Arndt, and B. Gruhn, Prognostic impact of WT1 expression prior to hematopoietic stem cell transplantation in children with malignant hematological diseases, J Cancer Res Clin Oncol, 2015.

Y. Shen, Y. M. Zhu, and X. Fan, Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia, Blood, 2011.

D. C. Liang, H. C. Liu, and C. P. Yang, Cooperating gene mutations in childhood acute myeloid leukemia with special reference on mutations of ASXL1, TET2, IDH1, IDH2, and DNMT3A, Blood, 2013.

D. T. Bowen, M. E. Frew, and R. Hills, RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years, Blood, 2005.

M. Sattler and R. Salgia, Targeting c-Kit mutations: Basic science to novel therapies, Leuk Res, 2004.
DOI : 10.1016/j.leukres.2003.10.004

P. Paschka, G. Marcucci, and A. S. Ruppert, Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study, J Clin Oncol, 2006.

N. Boissel, H. Leroy, and B. Brethon, Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia

. Leukemia, , 2006.

K. Lund, P. D. Adams, and M. Copland, EZH2 in normal and malignant hematopoiesis, Leukemia, 2014.
DOI : 10.1038/leu.2013.288

R. C. Lindsley, B. G. Mar, and E. Mazzola, Acute myeloid leukemia ontogeny is defined by distinct somatic mutations, Blood, 2015.
DOI : 10.1182/blood-2014-11-610543

URL : http://www.bloodjournal.org/content/125/9/1367.full.pdf

S. Thota, A. D. Viny, and H. Makishima, Genetic alterations of the cohesin complex genes in myeloid malignancies, Blood, 2014.

R. B. Walter, M. Othus, and G. Borthakur, Prediction of early death after induction therapy for newly diagnosed acute myeloid leukemia with pretreatment risk scores: A novel paradigm for treatment assignment, J Clin Oncol, 2011.

K. H. Metzeler, T. Herold, and M. Rothenberg-thurley, Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia, Blood, 2016.

D. Grimwade, A. Ivey, and B. Huntly, Molecular landscape of acute myeloid leukemia in younger adults and its clinical relevance, Blood, 2016.

V. I. Gaidzik, L. Bullinger, and R. F. Schlenk, RUNX1 mutations in acute myeloid leukemia: Results from a comprehensive genetic and clinical analysis from the AML study group

, J Clin Oncol, 2011.

S. Schnittger, C. Eder, and S. Jeromin, ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome, Leukemia, 2013.

M. Pratcorona, S. Brunet, and J. Nomdedéu, Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: Relevance to post-remission therapy, Blood, vol.121, issue.14, pp.2734-2738, 2013.

C. Allen, R. K. Hills, and K. Lamb, The importance of relative mutant level for evaluating impact on outcome of KIT, FLT3 and CBL mutations in core-binding factor acute myeloid leukemia, Leukemia, 2013.

J. E. Boissel, N. Chevret, and S. , Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia, Blood, 2013.

P. Paschka, J. Du, R. Schlenk, and V. Gaidzik, Secondary genetic lesions in acute myeloid leukemia with inv (16) or t (16;16): a study of the German-Austrian AML Study Group (AMLSG), Blood, vol.121, issue.1, pp.170-178, 2013.

N. Duployez, A. Marceau-renaut, and N. Boissel, Comprehensive mutational profiling of core binding factor acute myeloid leukemia, Blood, 2016.

Z. J. Faber, X. Chen, and A. L. Gedman, The genomic landscape of core-binding factor acute myeloid leukemias, Nat Genet, 2016.

S. Schnittger, W. Kern, and C. Tschulik, Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML, Blood, vol.114, issue.11, pp.2220-2231, 2009.

G. J. Schuurhuis, M. Heuser, and S. Freeman, Minimal/measurable residual disease in AML: consensus document from ELN MRD Working Party, Blood, vol.131, issue.12, 2018.

U. Platzbecker, G. Avvisati, and L. Cicconi, Improved outcomes with retinoic acid and arsenic trioxide compared with retinoic acid and chemotherapy in non-high-risk acute promyelocytic leukemia: Final results of the randomized Italian-German APL0406 trial

, J Clin Oncol, 2017.

D. Grimwade, J. V. Jovanovic, and R. K. Hills, Prospective minimal residual disease monitoring to predict relapse of acute promyelocytic leukemia and to direct pre-emptive arsenic trioxide therapy, J Clin Oncol, 2009.

C. Willekens, O. Blanchet, and A. Renneville, Prospective long-term minimal residual disease monitoring using RQ-PCR in RUNX1-RUNX1T1-positive acute myeloid leukemia: Results of the French CBF-2006 trial, Haematologica, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01817446

J. A. Liu-yin, O. 'brien, M. A. Hills, R. K. Daly, S. B. Wheatley et al., Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: Results of the United Kingdom MRC AML-15 trial, Blood, 2012.

H. Sugiyama, Wilms' tumor gene wt1: Its oncogenic function and clinical application, Int J Hematol, vol.73, issue.2, pp.177-187, 2001.

J. A. Liu-yin, O. 'brien, M. A. Hills, R. K. Daly, S. B. Wheatley et al., Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: Results of the United Kingdom MRC AML-15 trial, Blood, vol.120, issue.14, pp.2826-2835, 2012.

D. A. Arber, A. Orazi, and R. Hasserjian, The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia, Blood, 2016.

L. Federici and B. Falini, Nucleophosmin mutations in acute myeloid leukemia: A tale of protein unfolding and mislocalization, Protein Sci, 2013.

W. Wang, A. Budhu, M. Forgues, and X. W. Wang, Temporal and spatial control of nucleophosmin by the Ran-Crm1 complex in centrosome duplication, Nat Cell Biol, 2005.

Y. Yu, L. B. Maggi, and S. N. Brady, Nucleophosmin Is Essential for Ribosomal Protein L5 Nuclear Export, Mol Cell Biol, 2006.

W. Y. Chan, Q. R. Liu, and J. Borjigin, Characterization of the cDNA encoding human nucleophosmin and studies of its role in normal and abnormal growth, Biochemistry, 1989.

K. Hingorani and M. O. Olson, Mapping the functional domains of nucleolar protein B23, J Biol Chem, 2000.

Y. Nishimura, T. Ohkubo, Y. Furuichi, and H. Umekawa, Tryptophans 286 and 288 in the Cterminal region of protein B23.1 are important for its nucleolar localization, Biosci Biotechnol Biochem, 2002.

N. Bolli, I. Nicoletti, D. Marco, and M. F. , Born to be exported: COOH-terminal nuclear export signals of different strength ensure cytoplasmic accumulation of nucleophosmin leukemic mutants, Cancer Res, 2007.

Y. Jian, Z. Gao, and J. Sun, RNA aptamers interfering with nucleophosmin oligomerization induce apoptosis of cancer cells, Oncogene, 2009.

E. Colombo, J. C. Marine, D. Danovi, B. Falini, and P. G. Pelicci, Nucleophosmin regulates the stability and transcriptional activity of p53, Nat Cell Biol, 2002.

S. Kurki, K. Peltonen, and L. Latonen, Nucleolar protein NPM interacts with HDM2 and protects tumor suppressor protein p53 from HDM2-mediated degradation. Cancer Cell, 2004.

J. D. Weber, L. J. Taylor, M. F. Roussel, C. J. Sherr, and D. Bar-sagi, Nucleolar Arf sequesters Mdm2 and activates p53, Nat Cell Biol, 1999.

M. L. Kuo, W. Den-besten, D. Bertwistle, M. F. Roussel, and C. J. Sherr, N-terminal polyubiquitination and degradation of the Arf tumor suppressor, Genes Dev, 2004.

B. Falini, M. P. Martelli, and N. Bolli, Immunohistochemistry predicts nucleophosmin (NPM) mutations in acute myeloid leukemia, Blood, 2006.

K. Döhner, R. F. Schlenk, and M. Habdank, Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: Interaction with other gene mutations, Blood, 2005.

C. Thiede, S. Koch, and E. Creutzig, Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML), Blood, 2006.

P. Kunchala, S. Kuravi, R. Jensen, J. Mcguirk, and R. Balusu, When the good go bad: Mutant NPM1 in acute myeloid leukemia, Blood Rev, vol.32, issue.3, pp.167-183, 2018.

T. Suzuki, H. Kiyoi, and K. Ozeki, Clinical characteristics and prognostic implications of NPM1 mutations in acute myeloid leukemia, Blood, 2005.

S. Grisendi, R. Bernardi, and M. Rossi, Role of nucleophosmin in embryonic development and tumorigenesis, Nature, 2005.

E. Colombo, P. Martinelli, and R. Zamponi, Delocalization and destabilization of the Arf tumor suppressor by the leukemia-associated NPM mutant, Cancer Res, 2006.

B. Falini, N. Bolli, and J. Shan, Both carboxy-terminus NES motif and mutated tryptophan(s) are crucial for aberrant nuclear export of nucleophosmin leukemic mutants in NPMc+AML, Blood, vol.107, issue.11, pp.4514-4523, 2006.

P. Sportoletti, S. Grisendi, and S. M. Majid, Npm1 is a haploinsufficient suppressor of myeloid and lymphoid malignancies in the mouse, Blood, 2008.

W. Den-besten, M. L. Kuo, R. T. Williams, and C. J. Sherr, Myeloid leukemia-associated nucleophosmin mutants perturb p53-dependent and independent activities of the Arf tumor suppressor protein. Cell Cycle, 2005.

S. M. Leong, B. X. Tan, and B. B. Ahmad, Mutant nucleophosmin deregulates cell death and myeloid differentiation through excessive caspase-6 and-8 inhibition, Blood, vol.116, issue.17, pp.3286-3296, 2010.

L. Pasqualucci, A. Liso, and M. P. Martelli, Mutated nucleophosmin detects clonal multilineage involvement in acute myeloid leukemia: Impact on WHO classification, Blood, 2006.

S. Schnittger, C. Schoch, and W. Kern, Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype, Blood, 2005.

M. P. Martelli, N. Manes, and V. Pettirossi, Absence of nucleophosmin leukaemic mutants in B and T cells from AML with NPM1 mutations: Implications for the cell of origin of NPMc+ AML, vol.4

. Leukemia, , 2008.

J. Caudill, A. J. Sternberg, C. Y. Li, A. Tefferi, T. L. Lasho et al., C-terminal nucleophosmin mutations are uncommon in chronic myeloid disorders, Br J Haematol, 2006.

W. C. Chou, J. L. Tang, and L. I. Lin, Nucleophosmin mutations in de novo acute myeloid leukemia: The age-dependent incidences and the stability during disease evolution, Cancer Res, 2006.

M. Alcalay, E. Tiacci, and R. Bergomas, Acute myeloid leukemia bearing cytoplasmic nucleophosmin (NPMc+AML) shows a distinct gene expression profile characterized by up-regulation of genes involved in stem-cell maintenance, Blood, 2005.

M. S. De-propris, S. Raponi, and D. Diverio, High CD33 expression levels in acute myeloid leukemia cells carrying the nucleophosmin (NPM1) mutation. Haematologica, 2011.

C. Haferlach, C. Mecucci, and S. Schnittger, AML with mutated NPM1 carrying a normal or aberrant karyotype show overlapping biologic, pathologic, immunophenotypic, and prognostic features, Blood, 2009.
DOI : 10.1182/blood-2009-01-197871

URL : http://www.bloodjournal.org/content/bloodjournal/114/14/3024.full.pdf

A. Dufour, F. Schneider, and K. H. Metzeler, Acute myeloid leukemia with biallelic CEBPA gene mutations and normal karyotype represents a distinct genetic entity associated with a favorable clinical outcome, J Clin Oncol, 2010.

C. L. Green, K. K. Koo, R. K. Hills, A. K. Burnett, D. C. Linch et al., Prognostic significance of CEBPA mutations in a large cohort of younger adult patients with acute myeloid leukemia: Impact of double CEBPA mutations and the interaction with FLT3 and NPM1 mutations, J Clin Oncol, 2010.

R. F. Schlenk, K. Döhner, and J. Krauter, Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia, N Engl J Med, 2008.
DOI : 10.1056/nejmoa074306

R. E. Gale, K. Lamb, and C. Allen, Simpson's paradox and the impact of different DNMT3A mutations on outcome in younger adults with acute myeloid leukemia, J Clin Oncol, 2015.

N. Shayegi, M. Kramer, and M. Bornhäuser, The level of residual disease based on mutant NPM1 is an independent prognostic factor for relapse and survival in AML, Blood, 2013.

M. Hubmann, T. Köhnke, and E. Hoster, Molecular response assessment by quantitative real-time polymerase chain reaction after induction therapy in NPM1mutated patients identifies those at high risk of relapse, Haematologica, 2014.

J. Krönke, L. Bullinger, and V. Teleanu, Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia, Blood, 2013.

A. Ivey, R. K. Hills, and M. A. Simpson, Assessment of Minimal Residual Disease in Standard-Risk AML, N Engl J Med, 2016.
DOI : 10.1056/nejmoa1507471

URL : http://discovery.ucl.ac.uk/1475365/1/nejmoa1507471.pdf

P. Gorello, G. Cazzaniga, and F. Alberti, Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations

, Leukemia, vol.20, issue.6, pp.1103-1108, 2006.

M. A. Quail, M. Smith, and P. Coupland, A tale of three next generation sequencing platforms: comparison of Ion Torrent, Pacific Biosciences and Illumina MiSeq sequencers, BMC Genomics, 2012.

A. Kohlmann, N. Nadarajah, and T. Alpermann, Monitoring of residual disease by nextgeneration deep-sequencing of RUNX1 mutations can identify acute myeloid leukemia patients with resistant disease, Leukemia, 2014.

P. J. Sykes, S. H. Neoh, M. J. Brisco, E. Hughes, J. Condon et al., Quantitation of targets for PCR by use of limiting dilution, Biotechniques, 1992.

B. Vogelstein and K. W. Kinzler, Digital PCR. Genetics, 1999.

S. Olmedillas-lópez, M. García-arranz, and D. García-olmo, Current and Emerging Applications of Droplet Digital PCR in Oncology, Mol Diagn Ther, 2017.

G. Roloff, C. Lai, C. Hourigan, and L. Dillon, Technical Advances in the Measurement of Residual Disease in Acute Myeloid Leukemia, J Clin Med, 2017.

D. Digital and . Table-of-contents,

J. F. Huggett, C. A. Foy, and V. Benes, The digital MIQE guidelines: Minimum information for publication of quantitative digital PCR experiments, Clin Chem, 2013.

K. A. Heyries, C. Tropini, and M. Vaninsberghe, Megapixel digital PCR, Nat Methods, 2011.
DOI : 10.1038/nmeth.1640

B. J. Hindson, K. D. Ness, and D. A. Masquelier, High-throughput droplet digital PCR system for absolute quantitation of DNA copy number, AnalChem, vol.83, pp.8604-8610, 2011.
DOI : 10.1021/ac202028g

URL : https://doi.org/10.1021/ac202028g

D. Pekin, Y. Skhiri, and J. C. Baret, Quantitative and sensitive detection of rare mutations using droplet-based microfluidics, Lab Chip, 2011.
DOI : 10.1039/c1lc20128j

Y. Lo, F. Lun, and K. Chan, Digital PCR for the molecular detection of fetal chromosomal aneuploidy, Proc Natl Acad Sci, 2007.

R. Sanders, J. F. Huggett, C. A. Bushell, S. Cowen, D. J. Scott et al., Evaluation of digital PCR for absolute DNA quantification, Anal Chem, 2011.
DOI : 10.1021/ac103230c

S. Dube, J. Qin, and R. Ramakrishnan, Mathematical analysis of copy number variation in a DNA sample using digital PCR on a nanofluidic device, PLoS One, 2008.

S. Bhat, J. Herrmann, P. Armishaw, P. Corbisier, and K. R. Emslie, Single molecule detection in nanofluidic digital array enables accurate measurement of DNA copy number, Anal Bioanal Chem, 2009.

I. I. Apport, O. Caen, and P. Nizard, PCR digitale en micro-compartiments pour la détection quantitative d 'ADN tumoral circulant, p.31, 2015.

R. H. Sedlak and K. R. Jerome, Viral diagnostics in the era of digital polymerase chain reaction, Diagn Microbiol Infect Dis, 2013.

H. Quentmeier, M. P. Martelli, and W. G. Dirks, Cell line OCI/AML3 bears exon-12 NPM gene mutation-A and cytoplasmic expression of nucleophosmin, Leukemia, vol.19, issue.10, pp.1760-1767, 2005.

Y. Uchiyama, M. Nakashima, and S. Watanabe, Ultra-sensitive droplet digital PCR for detecting a low-prevalence somatic GNAQ mutation in Sturge-Weber syndrome, Sci Rep, vol.6, 2016.
DOI : 10.1038/srep22985

URL : https://www.nature.com/articles/srep22985.pdf

H. H. Zhu, X. H. Zhang, and Y. Z. Qin, MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: Results from the AML05 multicenter trial, Blood, vol.121, issue.20, pp.4056-4062, 2013.

M. Balsat, A. Renneville, and X. Thomas, Postinduction minimal residual disease predicts outcome and benefit from allogeneic stem cell transplantation in acute myeloid leukemia with NPM1 mutation: A study by the acute leukemia French association group, J Clin Oncol, vol.35, issue.2, pp.185-193, 2017.

C. Brunetti, L. Anelli, and A. Zagaria, Droplet Digital PCR Is a Reliable Tool for Monitoring Minimal Residual Disease in Acute Promyelocytic Leukemia, J Mol Diagnostics, vol.19, issue.3, 2017.
DOI : 10.1016/j.jmoldx.2017.01.004

E. Onecha, M. Linares, and I. Rapado, Novel deep targeted sequencing method for minimal residual disease monitoring in acute myeloid leukemia. Haematologica. 2018:haematol, 2018.
DOI : 10.3324/haematol.2018.194712

URL : http://www.haematologica.org/content/early/2018/08/07/haematol.2018.194712.full.pdf

D. Drandi, E. Genuardi, and I. Dogliotti, Highly sensitive MYD88 l265p mutation detection by droplet digital polymerase chain reaction in waldenström macroglobulinemia, Haematologica, 2018.
DOI : 10.3324/haematol.2017.186528

URL : http://www.haematologica.org/content/early/2018/03/16/haematol.2017.186528.full.pdf

A. Minervini, F. Minervini, C. Anelli, and L. , Droplet digital PCR analysis of NOTCH1 gene mutations in chronic lymphocytic leukemia, Oncotarget, 2016.

M. Bill, J. Grimm, and M. Jentzsch, Digital droplet PCR-based absolute quantification of pre-transplant NPM1 mutation burden predicts relapse in acute myeloid leukemia patients, Ann Hematol, 2018.

D. Drandi, L. Kubiczkova-besse, and S. Ferrero, Minimal Residual Disease Detection by Droplet Digital PCR in Multiple Myeloma, Mantle Cell Lymphoma, and Follicular Lymphoma: A Comparison with Real-Time PCR, J Mol Diagn, 2015.

V. Der-velden, G. Cazzaniga, and A. Schrauder, Analysis of minimal residual