J. F. Huggett, C. A. Foy, V. Benes, K. Emslie, J. A. Garson et al., The Digital MIQE Guidelines: Minimum Information for Publication of Quantitative Digital PCR Experiments, Clinical Chemistry, vol.59, issue.6, pp.892-902, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02992959

, Global surfactants market expected to reach $64 bn by 2025, says Allied Market Research, Focus on Surfactants, vol.2018, issue.10, pp.3-4, 2018.

R. K. Saiki, D. H. Gelfand, S. Stoffel, S. J. Scharf, R. Higuchi et al., Primer-directed enzymatic amplification of DNA with a thermostable DNA polymerase, Science, vol.239, issue.4839, pp.487-491, 1988.

P. Simmonds, P. Balfe, J. F. Peutherer, C. A. Ludlam, J. O. Bishop et al., Human immunodeficiency virus-infected individuals contain provirus in small numbers of peripheral mononuclear cells and at low copy numbers., Journal of Virology, vol.64, issue.2, pp.864-872, 1990.

A. A. Morley, Digital PCR: A brief history, Biomolecular Detection and Quantification, vol.1, issue.1, pp.1-2, 2014.

R. Higuchi, G. Dollinger, P. S. Walsh, and R. Griffith, Simultaneous Amplification and Detection of Specific DNA Sequences, Bio/Technology, vol.10, issue.4, pp.413-417, 1992.

B. Vogelstein and K. W. Kinzler, Digital PCR, Proceedings of the National Academy of Sciences, vol.96, issue.16, pp.9236-9241, 1999.

F. Diehl, M. Li, Y. He, K. W. Kinzler, B. Vogelstein et al., BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions, Nature Methods, vol.3, issue.7, pp.551-559, 2006.

F. Diehl, K. Schmidt, M. A. Choti, K. Romans, S. Goodman et al., Circulating mutant DNA to assess tumor dynamics, Nature Medicine, vol.14, issue.9, pp.985-990, 2008.

D. Pekin, Y. Skhiri, J. C. Baret, D. Le-corre, L. Mazutis et al., Quantitative and sensitive detection of rare mutations using droplet-based microfluidics, Lab on a Chip, vol.11, issue.13, p.2156, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02148770

M. Y. Chang, S. Ahn, M. Y. Kim, J. H. Han, H. R. Park et al., One-step noninvasive prenatal testing (NIPT) for autosomal recessive homozygous point mutations using digital PCR, Scientific Reports, vol.8, issue.1, p.2877, 2018.

J. Beck, M. Oellerich, and E. Schütz, A Universal Droplet Digital PCR Approach for Monitoring of Graft Health After Transplantation Using a Preselected SNP Set, Methods in Molecular Biology, vol.1768, pp.335-348, 2018.

H. C. Macher, N. García-fernández, A. Adsuar-gómez, M. Porras-lópez, A. González-calle et al., Donor-specific circulating cell free DNA as a noninvasive biomarker of graft injury in heart transplantation, Clinica Chimica Acta, vol.495, pp.590-597, 2019.

S. Pholwat, S. Stroup, S. Foongladda, and E. Houpt, Digital PCR to Detect and Quantify Heteroresistance in Drug Resistant Mycobacterium tuberculosis, PLoS ONE, vol.8, issue.2, p.e57238, 2013.

A. S. Whale, C. A. Bushell, P. R. Grant, S. Cowen, I. Gutierrez-aguirre et al., Detection of Rare Drug Resistance Mutations by Digital PCR in a Human Influenza A Virus Model System and Clinical Samples, Journal of Clinical Microbiology, vol.54, issue.2, pp.392-400, 2015.

L. P. Zheng, L. Y. Chen, X. Y. Liao, Z. H. Xu, Z. Y. Chen et al., Case report: primary resistance to osimertinib in erlotinib-pretreated lung adenocarcinoma with EGFR T790 M mutation, BMC Cancer, vol.18, issue.1, p.1070, 2018.

H. Akamatsu, Y. Koh, I. Okamoto, D. Fujimoto, A. Bessho et al., Clinical significance of monitoring EGFR mutation in plasma using multiplexed digital PCR in EGFR mutated patients treated with afatinib (West Japan Oncology Group 8114LTR study), Lung Cancer, vol.131, pp.128-133, 2019.

A. S. Whale, J. F. Huggett, S. Cowen, V. Speirs, J. Shaw et al., Comparison of microfluidic digital PCR and conventional quantitative PCR for measuring copy number variation, Nucleic Acids Research, vol.40, issue.11, pp.e82-e82, 2012.

J. Qin, R. C. Jones, and R. Ramakrishnan, Studying copy number variations using a nanofluidic platform, Nucleic Acids Research, vol.36, issue.18, pp.e116-e116, 2008.

K. Cassinari, O. Quenez, G. Joly-hélas, L. Beaussire, N. Le-meur et al., A Simple, Universal, and Cost-Efficient Digital PCR Method for the Targeted Analysis of Copy Number Variations, Clinical Chemistry, vol.65, issue.9, pp.1153-1160, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02339190

M. Lodrini, A. Sprüssel, K. Astrahantseff, D. Tiburtius, R. Konschak et al., Using droplet digital PCR to analyze MYCN and ALK copy number in plasma from patients with neuroblastoma, Oncotarget, vol.8, issue.49, pp.85234-85251, 2017.

L. A. El-khattabi, C. Rouillac-le-sciellour, D. Le-tessier, A. Luscan, A. Coustier et al., Could Digital PCR Be an Alternative as a Non-Invasive Prenatal Test for Trisomy 21: A Proof of Concept Study, PLOS ONE, vol.11, issue.5, p.e0155009, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01322786

M. Alikian, P. Ellery, M. Forbes, G. Gerrard, D. Kasperaviciute et al., Next-Generation Sequencing-Assisted DNA-Based Digital PCR for a Personalized Approach to the Detection and Quantification of Residual Disease in Chronic Myeloid Leukemia Patients, The Journal of Molecular Diagnostics, vol.18, issue.2, pp.176-189, 2016.

I. Dogliotti, D. Drandi, E. Genuardi, and S. Ferrero, New Molecular Technologies for Minimal Residual Disease Evaluation in B-Cell Lymphoid Malignancies, Journal of Clinical Medicine, vol.7, issue.9, p.288, 2018.

T. J. Henrich, S. Gallien, J. Z. Li, F. Pereyra, and D. R. Kuritzkes, Low-level detection and quantitation of cellular HIV-1 DNA and 2-LTR circles using droplet digital PCR, Journal of Virological Methods, vol.186, issue.1-2, pp.68-72, 2012.

S. Rutsaert, K. Bosman, W. Trypsteen, M. Nijhuis, and L. Vandekerckhove, Digital PCR as a tool to measure HIV persistence, Retrovirology, vol.15, issue.1, p.16, 2018.

W. Trypsteen, M. Kiselinova, L. Vandekerckhove, and W. De-spiegelaere, Diagnostic utility of droplet digital PCR for HIV reservoir quantification, Journal of Virus Eradication, vol.2, issue.3, pp.162-169, 2016.

M. Fitarelli-kiehl, F. Yu, R. Ashtaputre, K. W. Leong, I. Ladas et al., Denaturation-Enhanced Droplet Digital PCR for Liquid Biopsies, Clinical Chemistry, vol.64, issue.12, pp.1762-1771, 2018.

J. Regan and G. Karlin-neumann, Phasing DNA Markers Using Digital PCR, Methods in Molecular Biology, vol.1768, pp.489-512, 2018.

N. Chen and I. Schrijver, Allelic discrimination of cis-trans relationships by digital polymerase chain reaction: GJB2 (p.V27I/p.E114G) and CFTR (p.R117H/5T), Genetics in Medicine, vol.13, issue.12, pp.1025-1031, 2011.

S. Menzel, J. Qin, N. Vasavda, S. L. Thein, and R. Ramakrishnan, Experimental Generation of SNP Haplotype Signatures in Patients with Sickle Cell Anaemia, PLoS ONE, vol.5, issue.9, p.e13004, 2010.

G. R. Oxnard, C. P. Paweletz, Y. Kuang, S. L. Mach, A. O'connell et al., Noninvasive Detection of Response and Resistance in EGFR-Mutant Lung Cancer Using Quantitative Next-Generation Genotyping of Cell-Free Plasma DNA, Clinical Cancer Research, vol.20, issue.6, pp.1698-1705, 2014.

M. Falabella, L. Sun, J. Barr, A. Z. Pena, E. E. Kershaw et al., Single-Step qPCR and dPCR Detection of Diverse CRISPR-Cas9 Gene Editing Events in Vivo, G3: Genes|Genomes|Genetics, vol.7, issue.10, pp.3533-3542, 2017.

L. B. Pinheiro, H. O?brien, J. Druce, H. Do, P. Kay et al., Interlaboratory Reproducibility of Droplet Digital Polymerase Chain Reaction Using a New DNA Reference Material Format, Analytical Chemistry, vol.89, issue.21, pp.11243-11251, 2017.

A. S. Devonshire, I. Honeyborne, A. Gutteridge, A. S. Whale, G. Nixon et al., Highly Reproducible Absolute Quantification ofMycobacterium tuberculosisComplex by Digital PCR, Analytical Chemistry, vol.87, issue.7, pp.3706-3713, 2015.

L. Dong, Y. Meng, Z. Sui, J. Wang, L. Wu et al., Comparison of four digital PCR platforms for accurate quantification of DNA copy number of a certified plasmid DNA reference material, Scientific Reports, vol.5, issue.1, p.13174, 2015.

A. S. Whale, G. M. Jones, J. Pav?i?, T. Dreo, N. Redshaw et al., Assessment of Digital PCR as a Primary Reference Measurement Procedure to Support Advances in Precision Medicine, Clinical Chemistry, vol.64, issue.9, pp.1296-1307, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02299498

J. Pavsic, A. Devonshire, A. Blejec, C. A. Foy, F. Van-heuverswyn et al., Inter-laboratory assessment of different digital PCR platforms for quantification of human cytomegalovirus DNA, Anal Bioanal Chem, vol.409, pp.2601-2615, 2017.

P. Corbisier, L. Pinheiro, S. Mazoua, A. M. Kortekaas, P. Y. Chung et al., DNA copy number concentration measured by digital and droplet digital quantitative PCR using certified reference materials, Analytical and Bioanalytical Chemistry, vol.407, issue.7, pp.1831-1840, 2015.

S. Bhat and K. R. Emslie, Digital polymerase chain reaction for characterisation of DNA reference materials, Biomolecular Detection and Quantification, vol.10, pp.47-49, 2016.

R. J. Haynes, M. C. Kline, B. Toman, C. Scott, P. Wallace et al., Standard Reference Material 2366 for Measurement of Human Cytomegalovirus DNA, The Journal of Molecular Diagnostics, vol.15, issue.2, pp.177-185, 2013.

H. J. He, B. Das, M. H. Cleveland, L. Chen, C. E. Camalier et al., Development and interlaboratory evaluation of a NIST Reference Material RM 8366 for EGFR and MET gene copy number measurements, Clinical Chemistry and Laboratory Medicine (CCLM), vol.57, issue.8, pp.1142-1152, 2019.

H. White, L. Deprez, P. Corbisier, V. Hall, F. Lin et al., A certified plasmid reference material for the standardisation of bcr-abl1 mRNA quantification by real-time quantitative PCR, Leukemia, vol.29, pp.369-76, 2015.

P. Corbisier, S. Bhat, L. Partis, V. Rui-dan-xie, and K. R. Emslie, Absolute quantification of genetically modified MON810 maize (Zea mays L.) by digital polymerase chain reaction, Analytical and Bioanalytical Chemistry, vol.396, issue.6, pp.2143-2150, 2009.

P. Guertler, L. Grohmann, H. Naumann, M. Pavlovic, and U. Busch, Development of event-specific qPCR detection methods for genetically modified alfalfa events J101, J163 and KK179, Biomolecular Detection and Quantification, vol.17, p.100076, 2019.

Y. Jiang, H. Yang, S. Quan, Y. Liu, P. Shen et al., Development of certified matrix-based reference material of genetically modified rice event TT51-1 for real-time PCR quantification, Analytical and Bioanalytical Chemistry, vol.407, issue.22, pp.6731-6739, 2015.

, Updates to the International System of Units (SI) brochure (8th edition), Chemistry International, vol.36, issue.6, 2014.

M. C. Kline, E. L. Romsos, and D. L. Duewer, Evaluating Digital PCR for the Quantification of Human Genomic DNA: Accessible Amplifiable Targets, Analytical Chemistry, vol.88, issue.4, pp.2132-2139, 2016.

, Biotechnology - Requirements for evaluating the performance of quantification methods for nucleic acid target sequences - qPCR and dPCR, p.2019

H. B. Yoo, S. R. Park, L. Dong, J. Wang, Z. Sui et al., International Comparison of Enumeration-Based Quantification of DNA Copy-Concentration Using Flow Cytometric Counting and Digital Polymerase Chain Reaction, Anal Chem, vol.88, pp.12169-76

, In vitro diagnostic medical devices ? Requirements for international harmonisation protocols establishing metrological traceability of values assigned to calibrators and human samples, ISO Guide, vol.17511, p.2020

S. Bustin and J. Huggett, qPCR primer design revisited, Biomolecular Detection and Quantification, vol.14, pp.19-28, 2017.

S. A. Bustin, V. Benes, J. A. Garson, J. Hellemans, J. Huggett et al., The MIQE Guidelines: Minimum Information for Publication of Quantitative Real-Time PCR Experiments, Clinical Chemistry, vol.55, issue.4, pp.611-622, 2009.

M. Krumbholz, K. Goerlitz, C. Albert, J. Lawlor, M. Suttorp et al., Large amplicon droplet digital PCR for DNAbased monitoring of pediatric chronic myeloid leukaemia, J Cell Mol Med, vol.23, pp.4955-61, 2019.

A. Lievens, S. Jacchia, D. Kagkli, C. Savini, and M. Querci, Measuring Digital PCR Quality: Performance Parameters and Their Optimization, PLOS ONE, vol.11, issue.5, p.e0153317, 2016.

A. S. Whale, S. Cowen, C. A. Foy, and J. F. Huggett, Methods for Applying Accurate Digital PCR Analysis on Low Copy DNA Samples, PLoS ONE, vol.8, issue.3, p.e58177, 2013.

M. Vynck, J. Vandesompele, N. Nijs, B. Menten, A. De-ganck et al., Flexible analysis of digital PCR experiments using generalized linear mixed models, Biomolecular Detection and Quantification, vol.9, pp.1-13, 2016.

A. S. Whale, J. F. Huggett, and S. Tzonev, Fundamentals of multiplexing with digital PCR, Biomolecular Detection and Quantification, vol.10, pp.15-23, 2016.

J. F. Huggett, S. Cowen, and C. A. Foy, Considerations for Digital PCR as an Accurate Molecular Diagnostic Tool, Clinical Chemistry, vol.61, issue.1, pp.79-88, 2015.

V. Taly, D. Pekin, L. Benhaim, S. K. Kotsopoulos, D. Le-corre et al., Multiplex Picodroplet Digital PCR to Detect KRAS Mutations in Circulating DNA from the Plasma of Colorectal Cancer Patients, Clinical Chemistry, vol.59, issue.12, pp.1722-1731, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02299581

L. Miotke, B. T. Lau, R. T. Rumma, and H. P. Ji, High sensitivity detection and quantitation of DNA copy number and single nucleotide variants with single color droplet digital PCR, Anal Chem, vol.86, pp.2618-2642, 2014.

Q. Zhong, S. Bhattacharya, S. Kotsopoulos, J. Olson, V. Taly et al., Multiplex digital PCR: breaking the one target per color barrier of quantitative PCR, Lab on a Chip, vol.11, issue.13, p.2167, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02299588

B. J. Hindson, K. D. Ness, D. A. Masquelier, P. Belgrader, N. J. Heredia et al., High-throughput droplet digital PCR system for absolute quantitation of DNA copy number, Anal Chem, vol.83, pp.8604-8614, 2011.

R. Sanders, D. J. Mason, C. A. Foy, and J. F. Huggett, Considerations for accurate gene expression measurement by reverse transcription quantitative PCR when analysing clinical samples, Analytical and Bioanalytical Chemistry, vol.406, issue.26, pp.6471-6483, 2014.

, International vocabulary of metrology?basic and general concepts and associated terms, Bureau International des Poids et Measures (BIPM), 2012.

L. Aigrain, Y. Gu, and M. A. Quail, Quantitation of next generation sequencing library preparation protocol efficiencies using droplet digital PCR assays - a systematic comparison of DNA library preparation kits for Illumina sequencing, BMC Genomics, vol.17, issue.1, p.458, 2016.

J. D. Robin, A. T. Ludlow, R. Laranger, W. E. Wright, and J. W. Shay, Comparison of DNA Quantification Methods for Next Generation Sequencing, Scientific Reports, vol.6, issue.1, p.24067, 2016.

R. Sanders, D. J. Mason, C. A. Foy, and J. F. Huggett, Evaluation of Digital PCR for Absolute RNA Quantification, PLoS ONE, vol.8, issue.9, p.e75296, 2013.

U. Mock, I. Hauber, and B. Fehse, Digital PCR to assess gene-editing frequencies (GEF-dPCR) mediated by designer nucleases, Nature Protocols, vol.11, issue.3, pp.598-615, 2016.

G. Nixon, J. A. Garson, P. Grant, E. Nastouli, C. A. Foy et al., Comparative Study of Sensitivity, Linearity, and Resistance to Inhibition of Digital and Nondigital Polymerase Chain Reaction and Loop Mediated Isothermal Amplification Assays for Quantification of Human Cytomegalovirus, Analytical Chemistry, vol.86, issue.9, pp.4387-4394, 2014.

B. K. Jacobs, E. Goetghebeur, and L. Clement, Impact of variance components on reliability of absolute quantification using digital PCR, BMC Bioinformatics, vol.15, issue.1, p.283, 2014.

M. C. Strain, S. M. Lada, T. Luong, S. E. Rought, S. Gianella et al., Highly Precise Measurement of HIV DNA by Droplet Digital PCR, PLoS ONE, vol.8, issue.4, p.e55943, 2013.

M. Jones, J. Williams, K. Gärtner, R. Phillips, J. Hurst et al., Low copy target detection by Droplet Digital PCR through application of a novel open access bioinformatic pipeline, ?definetherain?, Journal of Virological Methods, vol.202, pp.46-53, 2014.

B. G. Brink, J. Meskas, and R. R. Brinkman, ddPCRclust: an R package and Shiny app for automated analysis of multiplexed ddPCR data, Bioinformatics, vol.34, issue.15, pp.2687-2689, 2018.

D. Attali, R. Bidshahri, C. Haynes, and J. Bryan, ddpcr: an R package and web application for analysis of droplet digital PCR data, F1000Research, vol.5, p.1411, 2016.

A. Chiu, M. Ayub, C. Dive, G. Brady, and C. J. Miller, twoddpcr: an R/Bioconductor package and Shiny app for Droplet Digital PCR analysis, Bioinformatics, vol.33, issue.17, pp.2743-2745, 2017.

B. K. Jacobs, E. Goetghebeur, J. Vandesompele, A. De-ganck, N. Nijs et al., Model-Based Classification for Digital PCR: Your Umbrella for Rain, Analytical Chemistry, vol.89, issue.8, pp.4461-4467, 2017.

D. Dobnik, B. Spilsberg, A. Bogo?alec-ko?ir, D. ?tebih, D. Morisset et al., Multiplex Droplet Digital PCR Protocols for Quantification of GM Maize Events, Methods in Molecular Biology, vol.1768, pp.69-98, 2018.

W. Trypsteen, M. Vynck, J. De-neve, P. Bonczkowski, M. Kiselinova et al., ddpcRquant: threshold determination for single channel droplet digital PCR experiments, Analytical and Bioanalytical Chemistry, vol.407, issue.19, pp.5827-5834, 2015.

C. Liu, W. Zhou, T. Zhang, K. Jiang, H. Li et al., An automated approach to classification of duplex assay for digital droplet PCR, Journal of Bioinformatics and Computational Biology, vol.16, issue.03, p.1850003, 2018.

T. Dreo, M. Pirc, ?. Ram?ak, J. Pav?i?, M. Milavec et al., Optimising droplet digital PCR analysis approaches for detection and quantification of bacteria: a case study of fire blight and potato brown rot, Analytical and Bioanalytical Chemistry, vol.406, issue.26, pp.6513-6528, 2014.

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, Analytical and Bioanalytical Chemistry, vol.394, issue.2, pp.457-467, 2009.

S. C. Taylor, J. Carbonneau, D. N. Shelton, and G. Boivin, Optimization of Droplet Digital PCR from RNA and DNA extracts with direct comparison to RT-qPCR: Clinical implications for quantification of Oseltamivir-resistant subpopulations, Journal of Virological Methods, vol.224, pp.58-66, 2015.

M. Vynck, J. Vandesompele, and O. Thas, Quality control of digital PCR assays and platforms, Analytical and Bioanalytical Chemistry, vol.409, issue.25, pp.5919-5931, 2017.

A. K. Witte, P. Mester, S. Fister, M. K. Witte, D. Schoder et al., A Systematic Investigation of Parameters Influencing Droplet Rain in the Listeria monocytogenes prfA Assay - Reduction of Ambiguous Results in ddPCR, PLOS ONE, vol.11, issue.12, p.e0168179, 2016.

K. J. Bosman, M. Nijhuis, P. M. Van-ham, A. M. Wensing, K. Vervisch et al., Comparison of digital PCR platforms and semi-nested qPCR as a tool to determine the size of the HIV reservoir, Scientific Reports, vol.5, issue.1, p.13811, 2015.

M. Kiselinova, A. O. Pasternak, W. De-spiegelaere, D. Vogelaers, B. Berkhout et al., Comparison of Droplet Digital PCR and Seminested Real-Time PCR for Quantification of Cell-Associated HIV-1 RNA, PLoS ONE, vol.9, issue.1, p.e85999, 2014.

J. Ruelle, V. Yfantis, A. Duquenne, and P. Goubau, Validation of an ultrasensitive digital droplet PCR assay for HIV-2 plasma RNA quantification, Journal of the International AIDS Society, vol.17, p.19675, 2014.

S. F. Destgroth, The evaluation of limiting dilution assays, Journal of Immunological Methods, vol.49, issue.2, pp.R11-R23, 1982.

A. B. Ko?ir, C. Divieto, J. Pav?i?, S. Pavarelli, D. Dobnik et al., Droplet volume variability as a critical factor for accuracy of absolute quantification using droplet digital PCR, Analytical and Bioanalytical Chemistry, vol.409, issue.28, pp.6689-6697, 2017.

K. R. Emslie, J. L. H.-mclaughlin, K. Griffiths, M. Forbes-smith, L. B. Pinheiro et al., Droplet Volume Variability and Impact on Digital PCR Copy Number Concentration Measurements, Analytical Chemistry, vol.91, issue.6, pp.4124-4131, 2019.

N. Majumdar, S. Banerjee, M. Pallas, T. Wessel, and P. Hegerich, Poisson Plus Quantification for Digital PCR Systems, Scientific Reports, vol.7, issue.1, p.9617, 2017.

M. Vynck and O. Thas, Reducing Bias in Digital PCR Quantification Experiments: The Importance of Appropriately Modeling Volume Variability, Analytical Chemistry, vol.90, issue.11, pp.6540-6547, 2018.

J. Tellinghuisen, Partition Volume Variability in Digital Polymerase Chain Reaction Methods: Polydispersity Causes Bias but Can Improve Precision, Analytical Chemistry, vol.88, issue.24, pp.12183-12187, 2016.

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, vol.3, issue.8, p.e2876, 2008.

L. D. Brown, T. T. Cai, A. Dasgupta, A. Agresti, B. A. Coull et al., Interval estimation for a binomial proportion-comment-rejoinder, Stat Sci, vol.16, pp.101-134, 2001.

S. Lefever, J. Hellemans, F. Pattyn, D. R. Przybylski, C. Taylor et al., RDML: structured language and reporting guidelines for real-time quantitative PCR data, Nucleic Acids Research, vol.37, issue.7, pp.2065-2069, 2009.

J. M. Ruijter, S. Lefever, J. Anckaert, J. Hellemans, M. W. Pfaffl et al., RDML-Ninja and RDMLdb for standardized exchange of qPCR data, BMC Bioinformatics, vol.16, issue.1, p.197, 2015.

J. Björkman, D. ?vec, E. Lott, M. Kubista, and R. Sjöback, Differential amplicons (?Amp)?a new molecular method to assess RNA integrity, Biomolecular Detection and Quantification, vol.6, pp.4-12, 2016.

A. Sta?hlberg, J. Ha?kansson, X. Xian, H. Semb, and M. Kubista, Properties of the Reverse Transcription Reaction in mRNA Quantification, Clinical Chemistry, vol.50, issue.3, pp.509-515, 2004.

S. A. Bustin, V. Benes, J. A. Garson, J. Hellemans, J. Huggett et al., Primer Sequence Disclosure: A Clarification of the MIQE Guidelines, Clinical Chemistry, vol.57, issue.6, pp.919-921, 2011.

A. Nour, A. M. Azhar, E. Damanhouri, G. Bustin, and S. A. , Five years MIQE guidelines: the case of the Arabian countries, PLoS One, vol.9, p.88266, 2014.

S. A. Bustin, Why the need for qPCR publication guidelines??The case for MIQE, Methods, vol.50, issue.4, pp.217-226, 2010.

S. A. Bustin, J. F. Beaulieu, J. Huggett, R. Jaggi, F. S. Kibenge et al., MIQE précis: Practical implementation of minimum standard guidelines for fluorescence-based quantitative real-time PCR experiments, BMC Molecular Biology, vol.11, issue.1, p.74, 2010.

S. A. Bustin and C. T. Wittwer, MIQE: A Step Toward More Robust and Reproducible Quantitative PCR, Clinical Chemistry, vol.63, issue.9, pp.1537-1538, 2017.

M. Dooms, A. Chango, E. Barbour, P. Pouillart, and A. M. Abdel-nour, Improving biological relevancy of transcriptional biomarkers experiments by applying the MIQE guidelines to pre-clinical and clinical trials, Methods, vol.59, issue.1, pp.147-153, 2013.

G. Johnson, T. Nolan, and S. A. Bustin, Real-Time Quantitative PCR, Pathogen Detection and MIQE, PCR Detection of Microbial Pathogens, vol.943, pp.1-16, 2012.