Y. Yamazaki, Apolipoprotein E as a Therapeutic Target in Alzheimer's Disease: A Review of Basic Research and Clinical Evidence, CNS Drugs, vol.30, issue.9, pp.773-89, 2016.

H. M. Schipper, MicroRNA expression in Alzheimer blood mononuclear cells, Gene Regul Syst Bio, vol.1, pp.263-74, 2007.

T. Lashley, Molecular biomarkers of Alzheimer's disease: progress and prospects, Dis Model Mech, issue.5, p.11, 2018.

O. Shaham, A plasma signature of human mitochondrial disease revealed through metabolic profiling of spent media from cultured muscle cells, Proc Natl Acad Sci U S A, vol.107, issue.4, pp.1571-1576, 2010.

J. L. Griffin and J. P. Shockcor, Metabolic profiles of cancer cells, Nat Rev Cancer, vol.4, issue.7, pp.551-61, 2004.

M. Kori, Metabolic Biomarkers and Neurodegeneration: A Pathway Enrichment Analysis of Alzheimer's Disease, Parkinson's Disease, and Amyotrophic Lateral Sclerosis, OMICS, vol.20, issue.11, pp.645-661, 2016.

L. M. Smith and N. L. Kelleher, Proteoform: a single term describing protein complexity, Nat Methods, vol.10, issue.3, pp.186-193, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02086222

E. A. Ponomarenko, The Size of the Human Proteome: The Width and Depth, Int J Anal Chem, p.7436849, 2016.

X. Dang, The first pilot project of the consortium for top-down proteomics: a status report, Proteomics, vol.14, issue.10, pp.1130-1170, 2014.

Y. I. Li, RNA splicing is a primary link between genetic variation and disease, Science, vol.352, issue.6285, pp.600-604, 2016.

E. T. Wang, Alternative isoform regulation in human tissue transcriptomes, Nature, vol.456, issue.7221, pp.470-476, 2008.

E. Picardi, REDIportal: a comprehensive database of A-to-I RNA editing events in humans, Nucleic Acids Res, vol.45, issue.D1, pp.750-757, 2017.

A. A. Su and L. Randau, A-to-I and C-to-U editing within transfer RNAs, Biochemistry (Mosc), vol.76, issue.8, pp.932-939, 2011.

B. Vanderperre, Direct detection of alternative open reading frames translation products in human significantly expands the proteome, PLoS One, vol.8, issue.8, p.70698, 2013.

R. B. Loftfield and D. Vanderjagt, The frequency of errors in protein biosynthesis, Biochem J, vol.128, issue.5, pp.1353-1359, 1972.

R. Aebersold, How many human proteoforms are there?, Nat Chem Biol, vol.14, issue.3, pp.206-214, 2018.

O. Pagel, Current strategies and findings in clinically relevant post-translational modification-specific proteomics, Expert Rev Proteomics, vol.12, issue.3, pp.235-53, 2015.

R. Ree, S. Varland, and T. Arnesen, Spotlight on protein N-terminal acetylation, Exp Mol Med, vol.50, issue.7, p.90, 2018.

J. Dai, Identification of degradation products formed during performic oxidation of peptides and proteins by high-performance liquid chromatography with matrixassisted laser desorption/ionization and tandem mass spectrometry, Rapid Commun Mass Spectrom, vol.19, issue.9, pp.1130-1138, 2005.

Z. Zhang, Identification of lysine succinylation as a new post-translational modification, Nat Chem Biol, vol.7, issue.1, pp.58-63, 2011.

Z. Xie, Lysine succinylation and lysine malonylation in histones, Mol Cell Proteomics, vol.11, issue.5, pp.100-107, 2012.

K. Zhang, Identification and verification of lysine propionylation and butyrylation in yeast core histones using PTMap software, J Proteome Res, vol.8, issue.2, pp.900-906, 2009.

M. Tan, Lysine glutarylation is a protein posttranslational modification regulated by SIRT5, Cell Metab, vol.19, issue.4, pp.605-622, 2014.

M. Tan, Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification, Cell, vol.146, issue.6, pp.1016-1044, 2011.

C. Choudhary, The growing landscape of lysine acetylation links metabolism and cell signalling, Nat Rev Mol Cell Biol, vol.15, issue.8, pp.536-50, 2014.

J. Murn and Y. Shi, The winding path of protein methylation research: milestones and new frontiers, Nat Rev Mol Cell Biol, vol.18, issue.8, pp.517-527, 2017.

L. Afjehi-sadat and B. A. Garcia, Comprehending dynamic protein methylation with mass spectrometry, Curr Opin Chem Biol, vol.17, issue.1, pp.12-21, 2013.

H. Lodish, B. A. Zipursky, and S. L. , Molecular Cell Biology, 2000.

C. Xu and D. T. Ng, Glycosylation-directed quality control of protein folding, Nat Rev Mol Cell Biol, vol.16, issue.12, pp.742-52, 2015.

M. Rape, Ubiquitylation at the crossroads of development and disease, Nat Rev Mol Cell Biol, vol.19, issue.1, pp.59-70, 2018.

O. Kerscher, R. Felberbaum, and M. Hochstrasser, Modification of proteins by ubiquitin and ubiquitin-like proteins, Annu Rev Cell Dev Biol, vol.22, pp.159-80, 2006.

C. M. Hickey, N. R. Wilson, and M. Hochstrasser, Function and regulation of SUMO proteases, Nat Rev Mol Cell Biol, vol.13, issue.12, pp.755-66, 2012.

I. A. Hendriks and A. C. Vertegaal, A comprehensive compilation of SUMO proteomics, Nat Rev Mol Cell Biol, vol.17, issue.9, pp.581-95, 2016.

J. G. Perez-silva, The Degradome database: expanding roles of mammalian proteases in life and disease, Nucleic Acids Res, vol.44, issue.D1, pp.351-356, 2016.

J. Griffiths, A brief history of mass spectrometry, Anal Chem, vol.80, issue.15, pp.5678-83, 2008.

A. Hamdi, Fast and facile preparation of nanostructured silicon surfaces for laser desorption/ionization mass spectrometry of small compounds, Rapid Commun Mass Spectrom, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02133855

C. Junot, High resolution mass spectrometry based techniques at the crossroads of metabolic pathways, Mass Spectrom Rev, vol.33, issue.6, pp.471-500, 2014.

T. Shi, Targeted quantification of low ng/mL level proteins in human serum without immunoaffinity depletion, J Proteome Res, vol.12, issue.7, pp.3353-61, 2013.

M. A. Kuzyk, Multiple reaction monitoring-based, multiplexed, absolute quantitation of 45 proteins in human plasma, Mol Cell Proteomics, vol.8, issue.8, pp.1860-77, 2009.

V. Lange, Selected reaction monitoring for quantitative proteomics: a tutorial, Mol Syst Biol, vol.4, p.222, 2008.

P. Picotti and R. Aebersold, Selected reaction monitoring-based proteomics: workflows, potential, pitfalls and future directions, Nat Methods, vol.9, issue.6, pp.555-66, 2012.

S. Gallien, S. Y. Kim, and B. Domon, Large-Scale Targeted Proteomics Using Internal Standard Triggered-Parallel Reaction Monitoring (IS-PRM), Mol Cell Proteomics, vol.14, issue.6, pp.1630-1674, 2015.

B. Rougemont, Scout-MRM: Multiplexed Targeted Mass Spectrometry-Based Assay without Retention Time Scheduling Exemplified by Dickeya dadantii Proteomic Analysis during Plant Infection, Anal Chem, vol.89, issue.3, pp.1421-1426, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01518912

B. T. Chait and . Chemistry, Mass spectrometry: bottom-up or top-down? Science, vol.314, pp.65-71, 2006.

J. V. Olsen, A dual pressure linear ion trap Orbitrap instrument with very high sequencing speed, Mol Cell Proteomics, vol.8, issue.12, pp.2759-69, 2009.

C. Wu, A protease for 'middle-down' proteomics, Nat Methods, vol.9, issue.8, pp.822-826, 2012.

E. I. Chen, Optimization of mass spectrometry-compatible surfactants for shotgun proteomics, J Proteome Res, vol.6, issue.7, pp.2529-2567, 2007.

J. Rodriguez, Does trypsin cut before proline?, J Proteome Res, vol.7, issue.1, pp.300-305, 2008.

E. Vandermarliere, M. Mueller, and L. Martens, Getting intimate with trypsin, the leading protease in proteomics, Mass Spectrom Rev, vol.32, issue.6, pp.453-65, 2013.

M. S. Kim, A draft map of the human proteome, Nature, vol.509, issue.7502, pp.575-81, 2014.

P. Roepstorff and J. Fohlman, Proposal for a common nomenclature for sequence ions in mass spectra of peptides, Biomed Mass Spectrom, vol.11, issue.11, p.601, 1984.

R. S. Johnson, Novel fragmentation process of peptides by collision-induced decomposition in a tandem mass spectrometer: differentiation of leucine and isoleucine, Anal Chem, vol.59, issue.21, pp.2621-2626, 1987.

P. Giansanti, An Augmented Multiple-Protease-Based Human Phosphopeptide Atlas, Cell Rep, vol.11, issue.11, pp.1834-1877, 2015.

S. Gauci, Lys-N and trypsin cover complementary parts of the phosphoproteome in a refined SCX-based approach, Anal Chem, vol.81, issue.11, pp.4493-501, 2009.

D. L. Swaney, C. D. Wenger, and J. J. Coon, Value of using multiple proteases for largescale mass spectrometry-based proteomics, J Proteome Res, vol.9, issue.3, pp.1323-1332, 2010.

X. Guo, Confetti: a multiprotease map of the HeLa proteome for comprehensive proteomics, Mol Cell Proteomics, vol.13, issue.6, pp.1573-84, 2014.

N. Nagaraj, Deep proteome and transcriptome mapping of a human cancer cell line, Mol Syst Biol, vol.7, p.548, 2011.

J. C. Tran, Mapping intact protein isoforms in discovery mode using top-down proteomics, Nature, vol.480, issue.7376, pp.254-262, 2011.

A. L. Capriotti, Intact protein separation by chromatographic and/or electrophoretic techniques for top-down proteomics, J Chromatogr A, pp.8760-76, 1218.

P. O. Schmit, Towards a routine application of Top-Down approaches for labelfree discovery workflows, J Proteomics, vol.175, pp.12-26, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01757595

I. Ntai, Precise characterization of KRAS4b proteoforms in human colorectal cells and tumors reveals mutation/modification cross-talk, Proc Natl Acad Sci U S A, vol.115, issue.16, pp.4140-4145, 2018.

J. F. Kellie, Quantitative measurement of intact alpha-synuclein proteoforms from post-mortem control and Parkinson's disease brain tissue by intact protein mass spectrometry, Sci Rep, vol.4, p.5797, 2014.

C. Chanthamontri, J. Liu, and S. A. Mcluckey, Charge State Dependent Fragmentation of Gaseous alpha-Synuclein Cations via Ion Trap and Beam-Type Collisional Activation, Int J Mass Spectrom, vol.283, issue.1-3, pp.9-16, 2009.

Y. Xia, X. Liang, and S. A. Mcluckey, Ion trap versus low-energy beam-type collisioninduced dissociation of protonated ubiquitin ions, Anal Chem, vol.78, issue.4, pp.1218-1245, 2006.

J. R. Cannon, Characterization of green fluorescent proteins by 193 nm ultraviolet photodissociation mass spectrometry, Proteomics, vol.14, issue.10, pp.1165-73, 2014.

T. P. Cleland, High-Throughput Analysis of Intact Human Proteins Using UVPD and HCD on an Orbitrap Mass Spectrometer, J Proteome Res, vol.16, issue.5, pp.2072-2079, 2017.

S. Barelli, Plasma/serum proteomics: pre-analytical issues, Expert Rev Proteomics, vol.4, issue.3, pp.363-70, 2007.

A. J. Rai and F. Vitzthum, Effects of preanalytical variables on peptide and protein measurements in human serum and plasma: implications for clinical proteomics, Expert Rev Proteomics, vol.3, issue.4, pp.409-435, 2006.

M. K. Tuck, Standard operating procedures for serum and plasma collection: early detection research network consensus statement standard operating procedure integration working group, J Proteome Res, vol.8, issue.1, pp.113-120, 2009.

T. Shi, IgY14 and SuperMix immunoaffinity separations coupled with liquid chromatography-mass spectrometry for human plasma proteomics biomarker discovery, Methods, vol.56, issue.2, pp.246-53, 2012.

M. Dupin, Impact of Serum and Plasma Matrices on the Titration of Human Inflammatory Biomarkers Using Analytically Validated SRM Assays, J Proteome Res, vol.15, issue.8, pp.2366-78, 2016.

A. Incamps, Impact of biological matrix on inflammatory protein biomarker quantification based on targeted mass spectrometry, Bioanalysis, vol.10, issue.17, pp.1383-1399, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02316826

G. S. Omenn, Overview of the HUPO Plasma Proteome Project: results from the pilot phase with 35 collaborating laboratories and multiple analytical groups, generating a core dataset of 3020 proteins and a publicly-available database, Proteomics, vol.5, issue.13, pp.3226-3271, 2005.

N. L. Anderson and N. G. Anderson, The human plasma proteome: history, character, and diagnostic prospects, Mol Cell Proteomics, vol.1, issue.11, pp.845-67, 2002.

T. Farrah, A high-confidence human plasma proteome reference set with estimated concentrations in PeptideAtlas, Mol Cell Proteomics, vol.10, issue.9, pp.110-006353, 2011.

L. Thadikkaran, Recent advances in blood-related proteomics, Proteomics, vol.5, issue.12, pp.3019-3053, 2005.

Z. Ouyang, Pellet digestion: a simple and efficient sample preparation technique for LC-MS/MS quantification of large therapeutic proteins in plasma, Bioanalysis, vol.4, issue.1, pp.17-28, 2012.

C. Hirtz, Absolute quantification of 35 plasma biomarkers in human saliva using targeted MS, Bioanalysis, vol.8, issue.1, pp.43-53, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01842381

B. Mertens, Assessing a multiplex-targeted proteomics approach for the clinical diagnosis of periodontitis using saliva samples, Bioanalysis, vol.10, issue.1, pp.35-45, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01842210

N. R. Barthelemy, Differential Mass Spectrometry Profiles of Tau Protein in the Cerebrospinal Fluid of Patients with Alzheimer's Disease, Progressive Supranuclear Palsy, and Dementia with Lewy Bodies, J Alzheimers Dis, vol.51, issue.4, pp.1033-1076, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01842422

N. R. Barthelemy, Tau Protein Quantification in Human Cerebrospinal Fluid by Targeted Mass Spectrometry at High Sequence Coverage Provides Insights into Its Primary Structure Heterogeneity, J Proteome Res, vol.15, issue.2, pp.667-76, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01842426

P. Bros, Antibody-free quantification of seven tau peptides in human CSF using targeted mass spectrometry, Front Neurosci, vol.9, p.302, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01842470

F. Becher, A simple and rapid LC-MS/MS method for therapeutic drug monitoring of cetuximab: a GPCO-UNICANCER proof of concept study in head-and-neck cancer patients, vol.7, p.2714, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01606760

C. Hirtz, From radioimmunoassay to mass spectrometry: a new method to quantify orexin-A (hypocretin-1) in cerebrospinal fluid, p. 25162. 85. Lame, M.E., E.E. Chambers, and M. Blatnik, Quantitation of amyloid beta peptides, vol.6, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01842392

. Abeta, Abeta(1-40), and Abeta(1-42) in human cerebrospinal fluid by ultraperformance liquid chromatography-tandem mass spectrometry, Anal Biochem, vol.419, issue.2, pp.133-142, 2011.

J. Pannee, A selected reaction monitoring (SRM)-based method for absolute quantification of Abeta38, Abeta40, and Abeta42 in cerebrospinal fluid of Alzheimer's disease patients and healthy controls, J Alzheimers Dis, vol.33, issue.4, pp.1021-1053, 2013.

E. Portelius, Determination of beta-amyloid peptide signatures in cerebrospinal fluid using immunoprecipitation-mass spectrometry, J Proteome Res, vol.5, issue.4, pp.1010-1016, 2006.

M. Gilar, Orthogonality of separation in two-dimensional liquid chromatography, Anal Chem, vol.77, pp.6426-6460, 2005.

H. Keshishian, Quantification of cardiovascular biomarkers in patient plasma by targeted mass spectrometry and stable isotope dilution, Mol Cell Proteomics, vol.8, issue.10, pp.2339-2388, 2009.

H. Keshishian, Quantitative, multiplexed assays for low abundance proteins in plasma by targeted mass spectrometry and stable isotope dilution, Mol Cell Proteomics, vol.6, issue.12, pp.2212-2241, 2007.

T. Shi, Antibody-free, targeted mass-spectrometric approach for quantification of proteins at low picogram per milliliter levels in human plasma/serum, Proc Natl Acad Sci, vol.109, issue.38, pp.15395-400, 2012.

S. Roche, Depletion of one, six, twelve or twenty major blood proteins before proteomic analysis: the more the better?, J Proteomics, vol.72, issue.6, pp.945-51, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00410072

E. Bellei, High-abundance proteins depletion for serum proteomic analysis: concomitant removal of non-targeted proteins, Amino Acids, vol.40, issue.1, pp.145-56, 2011.

B. B. Patel, Assessment of two immunodepletion methods: off-target effects and variations in immunodepletion efficiency may confound plasma proteomics, J Proteome Res, vol.11, issue.12, pp.5947-58, 2012.

S. Li, Digging More Missing Proteins Using an Enrichment Approach with ProteoMiner, J Proteome Res, vol.16, issue.12, pp.4330-4339, 2017.

A. Altomare, An in depth proteomic analysis based on ProteoMiner, affinity chromatography and nano-HPLC-MS/MS to explain the potential health benefits of bovine colostrum, J Pharm Biomed Anal, vol.121, pp.297-306, 2016.

B. R. Fonslow, Improvements in proteomic metrics of low abundance proteins through proteome equalization using ProteoMiner prior to MudPIT, J Proteome Res, vol.10, issue.8, pp.3690-700, 2011.

S. Hartwig, Combinatorial hexapeptide ligand libraries (ProteoMiner): an innovative fractionation tool for differential quantitative clinical proteomics, Arch Physiol Biochem, vol.115, issue.3, pp.155-60, 2009.

E. Boschetti and P. G. Righetti, The ProteoMiner in the proteomic arena: a nondepleting tool for discovering low-abundance species, J Proteomics, vol.71, issue.3, pp.255-64, 2008.

P. G. Righetti and E. Boschetti, The ProteoMiner and the FortyNiners: searching for gold nuggets in the proteomic arena, Mass Spectrom Rev, vol.27, issue.6, pp.596-608, 2008.

J. Travis, Isolation of albumin from whole human plasma and fractionation of albumin-depleted plasma, Biochem J, vol.157, issue.2, pp.301-307, 1976.

J. Travis and R. Pannell, Selective removal of albumin from plasma by affinity chromatography, Clin Chim Acta, vol.49, issue.1, pp.49-52, 1973.

J. Vialaret, Albumin depletion of human serum to improve quantitative clinical proteomics, Current Topics in Peptide & Protein Research, vol.19, pp.53-62, 2018.

B. Krastins, Rapid development of sensitive, high-throughput, quantitative and highly selective mass spectrometric targeted immunoassays for clinically important proteins in human plasma and serum, Clin Biochem, vol.46, issue.6, pp.399-410, 2013.

M. Dupre, Multiplex quantification of protein toxins in human biofluids and food matrices using immunoextraction and high-resolution targeted mass spectrometry, Anal Chem, vol.87, issue.16, pp.8473-80, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02083903

J. Gao, Multiplex Immuno-MALDI-TOF MS for Targeted Quantification of Protein Biomarkers and Their Proteoforms Related to Inflammation and Renal Dysfunction, Anal Chem, vol.90, issue.5, pp.3366-3373, 2018.

N. L. Anderson, Mass spectrometric quantitation of peptides and proteins using Stable Isotope Standards and Capture by Anti-Peptide Antibodies (SISCAPA), J Proteome Res, vol.3, issue.2, pp.235-279, 2004.

Y. C. Hsiao, Development of a Multiplexed Assay for Oral Cancer Candidate Biomarkers Using Peptide Immunoaffinity Enrichment and Targeted Mass Spectrometry, Mol Cell Proteomics, vol.16, issue.10, pp.1829-1849, 2017.

P. J. Ippoliti, Automated Microchromatography Enables Multiplexing of Immunoaffinity Enrichment of Peptides to Greater than 150 for Targeted MS-Based Assays, Anal Chem, vol.88, issue.15, pp.7548-55, 2016.

M. Razavi, High precision quantification of human plasma proteins using the automated SISCAPA Immuno-MS workflow, N Biotechnol, vol.33, issue.5, pp.494-502, 2016.

E. Kuhn, Interlaboratory evaluation of automated, multiplexed peptide immunoaffinity enrichment coupled to multiple reaction monitoring mass spectrometry for quantifying proteins in plasma, Mol Cell Proteomics, vol.11, issue.6, pp.111-013854, 2012.

X. Yang, Mass Spectrometric Quantitation of Tubulin Acetylation from Pepsin-Digested Rat Brain Tissue Using a Novel Stable-Isotope Standard and Capture by Anti-Peptide Antibody (SISCAPA) Method, Anal Chem, vol.90, issue.3, pp.2155-2163, 2018.

J. Jiang, Development of an immuno tandem mass spectrometry (iMALDI) assay for EGFR diagnosis, Proteomics Clin Appl, vol.1, issue.12, pp.1651-1660, 2007.

J. Jiang, An immunoaffinity tandem mass spectrometry (iMALDI) assay for detection of Francisella tularensis, Anal Chim Acta, vol.605, issue.1, pp.70-79, 2007.

N. L. Anderson, Precision of heavy-light peptide ratios measured by maldi-tof mass spectrometry, J Proteome Res, vol.11, issue.3, pp.1868-78, 2012.

H. Liu, Quantitation of a recombinant monoclonal antibody in monkey serum by liquid chromatography-mass spectrometry, Anal Biochem, vol.414, issue.1, pp.147-53, 2011.

C. W. Damen, The bioanalysis of the monoclonal antibody trastuzumab by highperformance liquid chromatography with fluorescence detection after immuno-affinity purification from human serum, J Pharm Biomed Anal, vol.50, issue.5, pp.861-867, 2009.

J. R. Whiteaker, Antibody-based enrichment of peptides on magnetic beads for mass-spectrometry-based quantification of serum biomarkers, Anal Biochem, vol.362, issue.1, pp.44-54, 2007.

M. Fernandez-ocana, Clinical pharmacokinetic assessment of an anti-MAdCAM monoclonal antibody therapeutic by LC-MS/MS, Anal Chem, vol.84, issue.14, pp.5959-67, 2012.

I. Onami, A versatile method for protein-based antigen bioanalysis in nonclinical pharmacokinetics studies of a human monoclonal antibody drug by an immunoaffinity liquid chromatography-tandem mass spectrometry, J Chromatogr A, vol.1334, pp.64-71, 2014.

M. A. Willrich, Quantitation of infliximab using clonotypic peptides and selective reaction monitoring by LC-MS/MS, Int Immunopharmacol, vol.28, issue.1, pp.513-533, 2015.

G. Liu, A novel and cost effective method of removing excess albumin from plasma/serum samples and its impacts on LC-MS/MS bioanalysis of therapeutic proteins, Anal Chem, vol.86, issue.16, pp.8336-8379, 2014.

C. Gong, Post-pellet-digestion precipitation and solid phase extraction: A practical and efficient workflow to extract surrogate peptides for ultra-high performance liquid chromatography--tandem mass spectrometry bioanalysis of a therapeutic antibody in the low ng/mL range, J Chromatogr A, vol.1424, pp.27-36, 2015.

N. Iwamoto, Fully validated LCMS bioanalysis of Bevacizumab in human plasma using nano-surface and molecular-orientation limited (nSMOL) proteolysis, Drug Metab Pharmacokinet, vol.31, issue.1, pp.46-50, 2016.

C. Fuente-garcia, Search for proteomic biomarkers related to bovine preslaughter stress using liquid isoelectric focusing (OFFGEL) and mass spectrometry, J Proteomics, 2018.

L. R. Beldarrain, Use of liquid isoelectric focusing (OFFGEL) on the discovery of meat tenderness biomarkers, J Proteomics, vol.183, pp.25-33, 2018.

H. Kraiem, Two-Dimensional Isoelectric Focusing OFFGEL, Micro-Fluidic Labon-Chip Electrophoresis and FTIR for Assessment of Long-Term Stability of rhG-CSF Formulation, IEEE Trans Nanobioscience, vol.16, issue.8, pp.694-702, 2017.

B. M. Naveena, In-gel and OFFGEL-based proteomic approach for authentication of meat species from minced meat and meat products, J Sci Food Agric, vol.98, issue.3, pp.1188-1196, 2018.

S. Michelland, Low-molecular-weight color pI markers to monitor on-line the peptide focusing process in OFFGEL fractionation, Electrophoresis, vol.38, issue.16, pp.2034-2041, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01596279

B. M. Naveena, OFFGEL electrophoresis and tandem mass spectrometry approach compared with DNA-based PCR method for authentication of meat species from raw and cooked ground meat mixtures containing cattle meat, water buffalo meat and sheep meat, Food Chem, vol.233, pp.311-320, 2017.

A. E. Aziz and T. M. , Fractionation and proteomic analysis of the Walterinnesia aegyptia snake venom using OFFGEL and MALDI-TOF-MS techniques, Electrophoresis, vol.36, issue.20, pp.2594-605, 2015.

S. Magdeldin, Complementary protein and peptide OFFGEL fractionation for high-throughput proteomic analysis, Anal Chem, vol.87, issue.16, pp.8481-8489, 2015.

A. Neill, Characterization of Recombinant Monoclonal Antibody Charge Variants Using OFFGEL Fractionation, Weak Anion Exchange Chromatography, and Mass Spectrometry, Anal Chem, vol.87, issue.12, pp.6204-6215, 2015.

A. Moreda-pineiro, N. Garcia-otero, and P. Bermejo-barrera, A review on preparative and semi-preparative offgel electrophoresis for multidimensional protein/peptide assessment, Anal Chim Acta, vol.836, pp.1-17, 2014.

C. Franchin, OFFGEL fractionation of peptides: where really is your sample?, J Chromatogr A, vol.1355, pp.278-83, 2014.

S. Magdeldin, Deep proteome mapping of mouse kidney based on OFFGel prefractionation reveals remarkable protein post-translational modifications, J Proteome Res, vol.13, issue.3, pp.1636-1682, 2014.

S. M. Zargar, Unraveling the iron deficiency responsive proteome in Arabidopsis shoot by iTRAQ-OFFGEL approach, Plant Signal Behav, issue.8, pp.10-4161, 2013.

A. Rideau, Two-step OFFGEL approach for effective peptide separation compatible with iTRAQ labeling, Proteomics, vol.13, issue.22, pp.3261-3267, 2013.

N. Garcia-otero, Study of extraction procedures for protein analysis in plankton samples by OFFGEL electrophoresis hyphenated with Lab-on-a-chip technology, Talanta, vol.115, pp.631-672, 2013.

N. Garcia-otero, Two-dimensional isoelectric focusing OFFGEL and microfluidic lab-on-chip electrophoresis for assessing dissolved proteins in seawater, Anal Chem, vol.85, issue.12, pp.5909-5925, 2013.

E. Tobolkina, Segmented field OFFGEL(R) electrophoresis, Electrophoresis, vol.33, issue.22, pp.3331-3339, 2012.

C. Abdallah, Optimization of iTRAQ labelling coupled to OFFGEL fractionation as a proteomic workflow to the analysis of microsomal proteins of Medicago truncatula roots, Proteome Sci, vol.10, issue.1, p.37, 2012.

H. Konecna, Exploration of beer proteome using OFFGEL prefractionation in combination with two-dimensional gel electrophoresis with narrow pH range gradients, J Agric Food Chem, vol.60, issue.10, pp.2418-2444, 2012.

J. Gannon and K. Ohlendieck, Subproteomic analysis of basic proteins in aged skeletal muscle following offgel pre-fractionation, Mol Med Rep, vol.5, issue.4, pp.993-1000, 2012.

E. M. Keidel, Evaluation of protein loading techniques and improved separation in OFFGEL isoelectric focusing, Electrophoresis, vol.32, issue.13, pp.1659-66, 2011.

S. Krishnan, OFFgel-based multidimensional LC-MS/MS approach to the cataloguing of the human platelet proteome for an interactomic profile, Electrophoresis, pp.686-95, 2011.

M. L. Mena, OFFGEL isoelectric focusing and polyacrylamide gel electrophoresis separation of platinum-binding proteins, J Chromatogr A, vol.1218, issue.9, pp.1281-90, 2011.

Y. Zhang, Comprehensive analysis of low-abundance proteins in human urinary exosomes using peptide ligand library technology, peptide OFFGEL fractionation and nanoHPLC-chip-MS/MS, Electrophoresis, pp.3797-807, 2010.

M. Balasubramani, Characterization and detection of cellular and proteomic alterations in stable stathmin-overexpressing, taxol-resistant BT549 breast cancer cells using offgel IEF/PAGE difference gel electrophoresis, Mutat Res, vol.722, issue.2, pp.154-64, 2011.

D. O. Azulay, H. Neubert, and M. F. Ocana, Visualisation tool for peptide fractionation data in proteomics: application to OFFGEL isoelectric focussing, BMC Bioinformatics, vol.11, p.371, 2010.

C. M. Warren, Sub-proteomic fractionation, iTRAQ, and OFFGEL-LC-MS/MS approaches to cardiac proteomics, J Proteomics, vol.73, issue.8, pp.1551-61, 2010.

E. Ernoult, A proteomic approach for plasma biomarker discovery with iTRAQ labelling and OFFGEL fractionation, J Biomed Biotechnol, p.927917, 2010.

B. Manadas, Comparative analysis of OFFGel, strong cation exchange with pH gradient, and RP at high pH for first-dimensional separation of peptides from a membrane-enriched protein fraction, Proteomics, vol.9, issue.22, pp.5194-5202, 2009.

E. Ernoult, E. Gamelin, and C. Guette, Improved proteome coverage by using iTRAQ labelling and peptide OFFGEL fractionation, Proteome Sci, vol.6, p.27, 2008.

J. Chenau, Peptides OFFGEL electrophoresis: a suitable pre-analytical step for complex eukaryotic samples fractionation compatible with quantitative iTRAQ labeling, Proteome Sci, vol.6, p.9, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00313760

S. Fraterman, Combination of peptide OFFGEL fractionation and label-free quantitation facilitated proteomics profiling of extraocular muscle, Proteomics, vol.7, issue.18, pp.3404-3420, 2007.

H. T. Lam, Modeling the isoelectric focusing of peptides in an OFFGEL multicompartment cell, J Proteome Res, vol.6, issue.5, pp.1666-76, 2007.

P. Horth, Efficient fractionation and improved protein identification by peptide OFFGEL electrophoresis, Mol Cell Proteomics, vol.5, issue.10, pp.1968-74, 2006.

S. Lehmann, Stable Isotope Labeling by Amino acid in Vivo (SILAV): a new method to explore protein metabolism, Rapid Commun Mass Spectrom, vol.29, issue.20, pp.1917-1942, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01842452

S. E. Ong, Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics, Mol Cell Proteomics, vol.1, issue.5, pp.376-86, 2002.

R. Zhang, Fractionation of isotopically labeled peptides in quantitative proteomics, Anal Chem, vol.73, issue.21, pp.5142-5151, 2001.

R. J. Beynon and J. M. Pratt, Metabolic labeling of proteins for proteomics, Mol Cell Proteomics, vol.4, issue.7, pp.857-72, 2005.

X. Yao, Proteolytic 18O labeling for comparative proteomics: model studies with two serotypes of adenovirus, Anal Chem, vol.73, issue.13, pp.2836-2878, 2001.

S. P. Gygi, Quantitative analysis of complex protein mixtures using isotope-coded affinity tags, Nat Biotechnol, vol.17, issue.10, pp.994-1003, 1999.

A. Thompson, Tandem mass tags: a novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS, Anal Chem, vol.75, issue.8, pp.1895-904, 2003.

P. L. Ross, Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents, Mol Cell Proteomics, vol.3, issue.12, pp.1154-69, 2004.

S. P. Gygi, Proteome analysis of low-abundance proteins using multidimensional chromatography and isotope-coded affinity tags, J Proteome Res, vol.1, issue.1, pp.47-54, 2002.

E. C. Yi, Increased quantitative proteome coverage with (13)C/(12)C-based, acidcleavable isotope-coded affinity tag reagent and modified data acquisition scheme, Proteomics, vol.5, issue.2, pp.380-387, 2005.

Y. H. Ahn, Quantitative analysis of aberrant protein glycosylation in liver cancer plasma by AAL-enrichment and MRM mass spectrometry, Analyst, vol.138, issue.21, pp.6454-62, 2013.

T. Werner, High-resolution enabled TMT 8-plexing, Anal Chem, vol.84, issue.16, pp.7188-94, 2012.

G. C. Mcalister, Increasing the multiplexing capacity of TMTs using reporter ion isotopologues with isobaric masses, Anal Chem, vol.84, issue.17, pp.7469-78, 2012.

P. V. Bondarenko, D. Chelius, and T. A. Shaler, Identification and relative quantitation of protein mixtures by enzymatic digestion followed by capillary reversed-phase liquid chromatography-tandem mass spectrometry, Anal Chem, vol.74, issue.18, pp.4741-4750, 2002.

S. Purvine, Shotgun collision-induced dissociation of peptides using a time of flight mass analyzer, Proteomics, vol.3, issue.6, pp.847-50, 2003.

L. C. Gillet, Targeted data extraction of the MS/MS spectra generated by dataindependent acquisition: a new concept for consistent and accurate proteome analysis, Mol Cell Proteomics, vol.11, issue.6, pp.111-016717, 2012.

C. Ludwig, Data-independent acquisition-based SWATH-MS for quantitative proteomics: a tutorial, Mol Syst Biol, vol.14, issue.8, p.8126, 2018.

F. Meier, A. B. , M. Frank, A. Ha, E. Voytik et al., Parallel accumulation -serial fragmentation combined with data-independent acquisition (diaPASEF): Bottom-up proteomics with near optimal ion usage, 2019.

V. Brun, Isotope dilution strategies for absolute quantitative proteomics, J Proteomics, vol.72, issue.5, pp.740-749, 2009.

V. Brun, Isotope-labeled protein standards: toward absolute quantitative proteomics, Mol Cell Proteomics, vol.6, issue.12, pp.2139-2188, 2007.

M. Zeiler, A Protein Epitope Signature Tag (PrEST) library allows SILAC-based absolute quantification and multiplexed determination of protein copy numbers in cell lines, Mol Cell Proteomics, vol.11, issue.3, pp.111-009613, 2012.

S. E. Kaiser, Protein standard absolute quantification (PSAQ) method for the measurement of cellular ubiquitin pools, Nat Methods, vol.8, issue.8, pp.691-697, 2011.

S. Lai, A combined tryptic peptide and winged peptide internal standard approach for the determination of alpha-lactalbumin in dairy products by ultra high performance liquid chromatography with tandem mass spectrometry, J Sep Sci, vol.38, issue.10, pp.1800-1806, 2015.

M. Faria, Comparison of a stable isotope labeled (SIL) peptide and an extended SIL peptide as internal standards to track digestion variability of an unstable signature peptide during quantification of a cancer biomarker, human osteopontin, from plasma using capillary microflow LC-MS/MS, J Chromatogr B Analyt Technol Biomed Life Sci, vol.1001, pp.156-68, 2015.

C. M. Shuford, Absolute Protein Quantification by Mass Spectrometry: Not as Simple as Advertised, Anal Chem, vol.89, issue.14, pp.7406-7415, 2017.

K. J. Bronsema, R. Bischoff, and N. C. Van-de-merbel, High-sensitivity LC-MS/MS quantification of peptides and proteins in complex biological samples: the impact of enzymatic digestion and internal standard selection on method performance, Anal Chem, vol.85, issue.20, pp.9528-9563, 2013.

K. J. Bronsema, R. Bischoff, and N. C. Van-de-merbel, Internal standards in the quantitative determination of protein biopharmaceuticals using liquid chromatography coupled to mass spectrometry, J Chromatogr B Analyt Technol Biomed Life Sci, pp.1-14, 2012.

J. Shi, Determining Allele-Specific Protein Expression (ASPE) Using a Novel QconCAT-Based Proteomics Method, J Proteome Res, 2019.

S. K. Stelder, Correction to "Stoichiometry, Absolute Abundance, and Localization of Proteins in the Bacillus cereus Spore Coat Insoluble Fraction Determined Using a QconCAT Approach, J Proteome Res, vol.17, issue.7, p.2562, 2018.

S. K. Stelder, Stoichiometry, Absolute Abundance, and Localization of Proteins in the Bacillus cereus Spore Coat Insoluble Fraction Determined Using a QconCAT Approach, J Proteome Res, vol.17, issue.2, pp.903-917, 2018.

D. G. Smith, Design and expression of a QconCAT protein to validate Hi3 protein quantification of influenza vaccine antigens, J Proteomics, vol.146, pp.133-173, 2016.

H. Pertl-obermeyer, Quantitation of Vacuolar Sugar Transporter Abundance Changes Using QconCAT Synthtetic Peptides, Front Plant Sci, vol.7, p.411, 2016.

K. B. Scott, I. V. Turko, and K. W. Phinney, QconCAT: Internal Standard for Protein Quantification, Methods Enzymol, vol.566, pp.289-303, 2016.

S. W. Holman, L. Mclean, and C. E. Eyers, RePLiCal: A QconCAT Protein for Retention Time Standardization in Proteomics Studies, J Proteome Res, vol.15, issue.3, pp.1090-102, 2016.

M. D. Harwood, Application of an LC-MS/MS method for the simultaneous quantification of human intestinal transporter proteins absolute abundance using a QconCAT technique, J Pharm Biomed Anal, vol.110, pp.27-33, 2015.

R. Voges, Absolute quantification of Corynebacterium glutamicum glycolytic and anaplerotic enzymes by QconCAT, J Proteomics, vol.113, pp.366-77, 2015.

J. Wei, High-throughput absolute quantification of proteins using an improved two-dimensional reversed-phase separation and quantification concatemer (QconCAT) approach, Anal Bioanal Chem, vol.406, issue.17, pp.4183-93, 2014.

Z. M. Al-majdoub, Quantification of the proteins of the bacterial ribosome using QconCAT technology, J Proteome Res, vol.13, issue.3, pp.1211-1233, 2014.

M. R. Russell, Alternative fusion protein strategies to express recalcitrant QconCAT proteins for quantitative proteomics of human drug metabolizing enzymes and transporters, J Proteome Res, vol.12, issue.12, pp.5934-5976, 2013.

T. A. Zimmerman, Quantification of transferrin in human serum using both QconCAT and synthetic internal standards, Anal Chem, vol.85, issue.21, pp.10362-10370, 2013.

R. Chawner, QconCAT standard for calibration of ion mobility-mass spectrometry systems, J Proteome Res, vol.11, issue.11, pp.5564-72, 2012.

P. J. Brownridge, Absolute multiplexed protein quantification using QconCAT technology, Methods Mol Biol, vol.893, pp.267-93, 2012.

R. J. Austin, IQcat: multiplexed protein quantification by isoelectric QconCAT, Proteomics, vol.12, issue.13, pp.2078-83, 2012.

S. L. Bislev, Quantotypic properties of QconCAT peptides targeting bovine host response to Streptococcus uberis, J Proteome Res, vol.11, issue.3, pp.1832-1875, 2012.

K. M. Carroll, F. Lanucara, and C. E. Eyers, Quantification of proteins and their modifications using QconCAT technology, Methods Enzymol, vol.500, pp.113-144, 2011.

K. M. Carroll, Absolute quantification of the glycolytic pathway in yeast: deployment of a complete QconCAT approach, Mol Cell Proteomics, vol.10, issue.12, pp.111-007633, 2011.

P. Brownridge, Global absolute quantification of a proteome: Challenges in the deployment of a QconCAT strategy, Proteomics, vol.11, issue.15, pp.2957-70, 2011.

W. Castro-borges, Abundance of tegument surface proteins in the human blood fluke Schistosoma mansoni determined by QconCAT proteomics, J Proteomics, vol.74, issue.9, pp.1519-1552, 2011.

N. Swainston, D. Jameson, and K. Carroll, A QconCAT informatics pipeline for the analysis, visualization and sharing of absolute quantitative proteomics data, Proteomics, vol.11, issue.2, pp.329-362, 2011.

J. Rivers, Absolute multiplexed quantitative analysis of protein expression during muscle development using QconCAT, Mol Cell Proteomics, vol.6, issue.8, pp.1416-1443, 2007.

J. M. Pratt, Multiplexed absolute quantification for proteomics using concatenated signature peptides encoded by QconCAT genes, Nat Protoc, vol.1, issue.2, pp.1029-1072, 2006.

D. C. Liebler and L. J. Zimmerman, Targeted quantitation of proteins by mass spectrometry, Biochemistry, vol.52, issue.22, pp.3797-806, 2013.

P. Picotti, R. Aebersold, and B. Domon, The implications of proteolytic background for shotgun proteomics, Mol Cell Proteomics, vol.6, issue.9, pp.1589-98, 2007.

A. N. Hoofnagle, Recommendations for the Generation, Quantification, Storage, and Handling of Peptides Used for Mass Spectrometry-Based Assays, Clin Chem, vol.62, issue.1, pp.48-69, 2016.

P. D. Compton, N. L. Kelleher, and J. Gunawardena, Estimating the Distribution of Protein Post-Translational Modification States by Mass Spectrometry, J Proteome Res, vol.17, issue.8, pp.2727-2734, 2018.

D. Domanski, L. C. Murphy, and C. H. Borchers, Assay development for the determination of phosphorylation stoichiometry using multiple reaction monitoring methods with and without phosphatase treatment: application to breast cancer signaling pathways, Anal Chem, vol.82, issue.13, pp.5610-5630, 2010.

A. Wolf-yadlin, Multiple reaction monitoring for robust quantitative proteomic analysis of cellular signaling networks, Proc Natl Acad Sci, vol.104, issue.14, pp.5860-5865, 2007.

Z. Sheng, Ser1292 autophosphorylation is an indicator of LRRK2 kinase activity and contributes to the cellular effects of PD mutations, Sci Transl Med, vol.4, issue.164, pp.164-161, 2012.

J. Navarrete-perea, Streamlined Tandem Mass Tag (SL-TMT) Protocol: An Efficient Strategy for Quantitative (Phospho)proteome Profiling Using Tandem Mass Tag-Synchronous Precursor Selection-MS3, J Proteome Res, vol.17, issue.6, pp.2226-2236, 2018.

L. Xu, An MRM-based workflow for absolute quantitation of lysine-acetylated metabolic enzymes in mouse liver, Analyst, vol.140, issue.23, pp.7868-75, 2015.

H. Mirzaei, Characterizing the connectivity of poly-ubiquitin chains by selected reaction monitoring mass spectrometry, Mol Biosyst, vol.6, issue.10, pp.2004-2018, 2010.

J. M. Held, Targeted quantitation of site-specific cysteine oxidation in endogenous proteins using a differential alkylation and multiple reaction monitoring mass spectrometry approach, Mol Cell Proteomics, vol.9, issue.7, pp.1400-1410, 2010.

T. Yoneyama, Quantitative targeted absolute proteomics-based large-scale quantification of proline-hydroxylated alpha-fibrinogen in plasma for pancreatic cancer diagnosis, J Proteome Res, vol.12, issue.2, pp.753-62, 2013.

S. R. Danielson, Preferentially increased nitration of alpha-synuclein at tyrosine-39 in a cellular oxidative model of Parkinson's disease, Anal Chem, vol.81, issue.18, pp.7823-7831, 2009.

Y. H. Ahn, A lectin-coupled, targeted proteomic mass spectrometry (MRM MS) platform for identification of multiple liver cancer biomarkers in human plasma, J Proteomics, vol.75, issue.17, pp.5507-5522, 2012.

H. J. Sung, Large-scale isotype-specific quantification of Serum amyloid A 1/2 by multiple reaction monitoring in crude sera, J Proteomics, vol.75, issue.7, pp.2170-80, 2012.

P. Oeckl, Alpha-, Beta-, and Gamma-synuclein Quantification in Cerebrospinal Fluid by Multiple Reaction Monitoring Reveals Increased Concentrations in Alzheimer's and Creutzfeldt-Jakob Disease but No Alteration in Synucleinopathies, Mol Cell Proteomics, vol.15, issue.10, pp.3126-3138, 2016.

H. Steen, Phosphorylation analysis by mass spectrometry: myths, facts, and the consequences for qualitative and quantitative measurements, Mol Cell Proteomics, vol.5, issue.1, pp.172-81, 2006.

Z. Wu, Impact of Phosphorylation on the Mass Spectrometry Quantification of Intact Phosphoproteins, Anal Chem, vol.90, issue.8, pp.4935-4939, 2018.

A. W. Schmid, Alpha-synuclein post-translational modifications as potential biomarkers for Parkinson disease and other synucleinopathies, Mol Cell Proteomics, vol.12, issue.12, pp.3543-58, 2013.

H. D. Seckler, A Targeted, Differential Top-Down Proteomic Methodology for Comparison of ApoA-I Proteoforms in Individuals with High and Low HDL Efflux Capacity, J Proteome Res, vol.17, issue.6, pp.2156-2164, 2018.

T. A. Addona, Multi-site assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma, Nat Biotechnol, vol.27, issue.7, pp.633-674, 2009.

S. E. Abbatiello, Large-Scale Interlaboratory Study to Develop, Analytically Validate and Apply Highly Multiplexed, Quantitative Peptide Assays to Measure Cancer-Relevant Proteins in Plasma, Mol Cell Proteomics, vol.14, issue.9, pp.2357-74, 2015.

P. Oeckl, P. Steinacker, and M. Otto, Comparison of Internal Standard Approaches for SRM Analysis of Alpha-Synuclein in Cerebrospinal Fluid, J Proteome Res, vol.17, issue.1, pp.516-523, 2018.

S. L. Arnold, F. Stevison, and N. Isoherranen, Impact of Sample Matrix on Accuracy of Peptide Quantification: Assessment of Calibrator and Internal Standard Selection and Method Validation, Anal Chem, vol.88, issue.1, pp.746-53, 2016.

E. Nouri-nigjeh, Effects of calibration approaches on the accuracy for LC-MS targeted quantification of therapeutic protein, Anal Chem, vol.86, issue.7, pp.3575-84, 2014.

K. B. Scott, I. V. Turko, and K. W. Phinney, Quantitative performance of internal standard platforms for absolute protein quantification using multiple reaction monitoring-mass spectrometry, Anal Chem, vol.87, issue.8, pp.4429-4464, 2015.

C. M. Shuford, Peptide production and decay rates affect the quantitative accuracy of protein cleavage isotope dilution mass spectrometry (PC-IDMS), Mol Cell Proteomics, vol.11, issue.9, pp.814-837, 2012.

D. R. Barnidge, Evaluation of a cleavable stable isotope labeled synthetic peptide for absolute protein quantification using LC-MS/MS, J Proteome Res, vol.3, issue.3, pp.658-61, 2004.

J. Cao, A rapid, reproducible, on-the-fly orthogonal array optimization method for targeted protein quantification by LC/MS and its application for accurate and sensitive quantification of carbonyl reductases in human liver, Anal Chem, vol.82, issue.7, pp.2680-2689, 2010.

D. Lebert, DIGESTIF: a universal quality standard for the control of bottom-up proteomics experiments, J Proteome Res, vol.14, issue.2, pp.787-803, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02083909

X. Duan, High-throughput method development for sensitive, accurate, and reproducible quantification of therapeutic monoclonal antibodies in tissues using orthogonal array optimization and nano liquid chromatography/selected reaction monitoring mass spectrometry, Anal Chem, vol.84, issue.10, pp.4373-82, 2012.

P. Brownridge and R. J. Beynon, The importance of the digest: proteolysis and absolute quantification in proteomics, Methods, vol.54, issue.4, pp.351-60, 2011.

C. Pritchard, Quantification of human growth hormone in serum with a labeled protein as an internal standard: essential considerations, Anal Chem, vol.86, issue.13, pp.6525-6557, 2014.

J. R. Clamp and L. Hough, The Periodate Oxidation of Amino Acids with Reference to Studies on Glycoproteins, Biochem J, vol.94, pp.17-24, 1965.

A. Meister, Transamination and associated deamidation of asparagine and glutamine, J Biol Chem, vol.197, issue.1, pp.319-349, 1952.

A. Kraut, Peptide storage: are you getting the best return on your investment? Defining optimal storage conditions for proteomics samples, J Proteome Res, vol.8, issue.7, pp.3778-85, 2009.

S. Dolman, Investigation of carryover of peptides in nano-liquid chromatography/mass spectrometry using packed and monolithic capillary columns, J Chromatogr B Analyt Technol Biomed Life Sci, vol.912, pp.56-63, 2013.

H. John, Analytical procedures for quantification of peptides in pharmaceutical research by liquid chromatography-mass spectrometry, Anal Bioanal Chem, vol.378, issue.4, pp.883-97, 2004.

S. J. Bark and V. Hook, Differential recovery of peptides from sample tubes and the reproducibility of quantitative proteomic data, J Proteome Res, vol.6, issue.11, pp.4511-4517, 2007.

I. Van-den-broek, Quantitative bioanalysis of peptides by liquid chromatography coupled to (tandem) mass spectrometry, J Chromatogr B Analyt Technol Biomed Life Sci, vol.872, issue.1-2, pp.1-22, 2008.

R. W. Mahley and S. C. Rall, Apolipoprotein E: far more than a lipid transport protein, Annu Rev Genomics Hum Genet, vol.1, pp.507-544, 2000.

Y. Huang and R. W. Mahley, Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer's diseases, Neurobiol Dis, pp.3-12, 2014.

M. H. Dominiczak and M. J. Caslake, Apolipoproteins: metabolic role and clinical biochemistry applications, Ann Clin Biochem, vol.48, pp.498-515, 2011.

R. W. Mahley, Apolipoprotein E: cholesterol transport protein with expanding role in cell biology, Science, vol.240, issue.4852, pp.622-652, 1988.

P. B. Verghese, J. M. Castellano, and D. M. Holtzman, Apolipoprotein E in Alzheimer's disease and other neurological disorders, Lancet Neurol, vol.10, issue.3, pp.241-52, 2011.

A. R. Roda, L. Montoliu-gaya, and S. Villegas, The Role of Apolipoprotein E Isoforms in Alzheimer's Disease, J Alzheimers Dis, vol.68, issue.2, pp.459-471, 2019.

C. Koopal, A. D. Marais, and F. L. Visseren, Familial dysbetalipoproteinemia: an underdiagnosed lipid disorder, Curr Opin Endocrinol Diabetes Obes, vol.24, issue.2, pp.133-139, 2017.

S. Soderlund, Increased apolipoprotein E level and reduced high-density lipoprotein mean particle size associate with low high-density lipoprotein cholesterol and features of metabolic syndrome, Metabolism, vol.59, issue.10, pp.1502-1511, 2010.

E. H. Corder, Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease, Nat Genet, vol.7, issue.2, pp.180-184, 1994.

A. M. Saunders, Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer's disease, Neurology, vol.43, issue.8, pp.1467-72, 1993.

H. Bickeboller, Apolipoprotein E and Alzheimer disease: genotype-specific risks by age and sex, Am J Hum Genet, vol.60, issue.2, pp.439-485, 1997.

E. Genin, APOE and Alzheimer disease: a major gene with semi-dominant inheritance, Mol Psychiatry, vol.16, issue.9, pp.903-910, 2011.

D. A. Brouwer, J. J. Van-doormaal, and F. A. Muskiet, Clinical chemistry of common apolipoprotein E isoforms, J Chromatogr B Biomed Appl, vol.678, issue.1, pp.23-41, 1996.

B. Rochat, Validation of hepcidin quantification in plasma using LC-HRMS and discovery of a new hepcidin isoform, Bioanalysis, issue.5, pp.2509-2529, 2013.

E. H. Kemna, Mass spectrometry-based hepcidin measurements in serum and urine: analytical aspects and clinical implications, Clin Chem, vol.53, issue.4, pp.620-628, 2007.

H. Kulaksiz, The iron-regulatory peptide hormone hepcidin: expression and cellular localization in the mammalian kidney, J Endocrinol, vol.184, issue.2, pp.361-70, 2005.

C. H. Park, Hepcidin, a urinary antimicrobial peptide synthesized in the liver, J Biol Chem, vol.276, issue.11, pp.7806-7816, 2001.

O. Itkonen, Binding of hepcidin to plasma proteins, Clin Chem, vol.58, issue.7, pp.1158-60, 2012.

E. Ramos, Minihepcidins prevent iron overload in a hepcidin-deficient mouse model of severe hemochromatosis, Blood, vol.120, issue.18, pp.3829-3865, 2012.

J. J. Kroot, Hepcidin in human iron disorders: diagnostic implications, Clin Chem, vol.57, issue.12, pp.1650-69, 2011.

C. Delaby, V. J. Bros, P. Gabelle, A. Lefebvre, T. Puy et al., Clinical measurement of Hepcidin-25 in human serum: Is quantitative mass spectrometry up to the job?, EuPA Open Proteomics, vol.3, pp.60-67, 2014.

U. Kobold, Quantification of hepcidin-25 in human serum by isotope dilution micro-HPLC-tandem mass spectrometry, Clin Chem, vol.54, issue.9, pp.1584-1590, 2008.

S. S. Bansal, Quantitation of hepcidin in human urine by liquid chromatographymass spectrometry, Anal Biochem, vol.384, issue.2, pp.245-53, 2009.

F. Wolff, Hepcidin-25: Measurement by LC-MS/MS in serum and urine, reference ranges and urinary fractional excretion, Clin Chim Acta, vol.423, pp.99-104, 2013.

S. S. Bansal, Quantification of hepcidin using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, Rapid Commun Mass Spectrom, vol.23, issue.11, pp.1531-1573, 2009.

P. Bros, Impurity determination for hepcidin by liquid chromatography-high resolution and ion mobility mass spectrometry for the value assignment of candidate primary calibrators, Anal Bioanal Chem, vol.409, issue.10, pp.2559-2567, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01785651

Y. Dauvilliers, Association between serum hepcidin level and restless legs syndrome, Mov Disord, vol.33, issue.4, pp.618-627, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01842236

C. Delaby, Quantification of hepcidin-25 in human cerebrospinal fluid using LC-MS/MS, Bioanalysis, vol.9, issue.4, pp.337-347, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01792350

O. Itkonen, Preanalytical factors and reference intervals for serum hepcidin LC-MS/MS method, Clin Chim Acta, vol.413, issue.7-8, pp.696-701, 2012.

J. J. Kroot, Pre)analytical imprecision, between-subject variability, and daily variations in serum and urine hepcidin: implications for clinical studies, Anal Biochem, vol.389, issue.2, pp.124-133, 2009.

J. J. Kroot, Results of the first international round robin for the quantification of urinary and plasma hepcidin assays: need for standardization, Haematologica, vol.94, issue.12, pp.1748-52, 2009.

A. C. Swensen, Whole blood and urine bioactive Hepcidin-25 determination using liquid chromatography mass spectrometry, Anal Biochem, vol.517, pp.23-30, 2017.

V. Houbart, Hepcidin determination in dried blood by microfluidic LC-MS/MS: comparison of DBS and volumetric absorptive microsampling for matrix effect and recovery, Bioanalysis, vol.7, issue.21, pp.2789-99, 2015.

T. Ganz, Immunoassay for human serum hepcidin, Blood, vol.112, issue.10, pp.4292-4299, 2008.

S. Lasocki, Impact of iron deficiency diagnosis using hepcidin mass spectrometry dosage methods on hospital stay and costs after a prolonged ICU stay: Study protocol for a multicentre, randomised, single-blinded medico-economic trial, Anaesth Crit Care Pain Med, vol.36, issue.6, pp.391-396, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01745569

D. M. Ecker, S. D. Jones, and H. L. Levine, The therapeutic monoclonal antibody market, MAbs, vol.7, issue.1, pp.9-14, 2015.

T. H. Oude-munnink, Therapeutic drug monitoring of monoclonal antibodies in inflammatory and malignant disease: Translating TNF-alpha experience to oncology, Clin Pharmacol Ther, vol.99, issue.4, pp.419-450, 2016.

D. Mulleman, Infliximab concentration monitoring improves the control of disease activity in rheumatoid arthritis, Arthritis Res Ther, vol.11, issue.6, p.178, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00375858

B. Gao, Evidence for therapeutic drug monitoring of targeted anticancer therapies, J Clin Oncol, vol.30, issue.32, pp.4017-4042, 2012.

L. Minasian, Optimizing dosing of oncology drugs, Clin Pharmacol Ther, vol.96, issue.5, pp.572-581, 2014.

N. Ferrara, H. P. Gerber, and J. Lecouter, The biology of VEGF and its receptors, Nat Med, vol.9, issue.6, pp.669-76, 2003.

N. Ferrara, Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer, Nat Rev Drug Discov, vol.3, issue.5, pp.391-400, 2004.

J. Folkman, Tumor angiogenesis: therapeutic implications, N Engl J Med, vol.285, issue.21, pp.1182-1188, 1971.

H. Hurwitz, Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer, N Engl J Med, vol.350, issue.23, pp.2335-2377, 2004.

J. F. Lu, Clinical pharmacokinetics of bevacizumab in patients with solid tumors, Cancer Chemother Pharmacol, vol.62, issue.5, pp.779-86, 2008.

M. Caulet, Bevacizumab Pharmacokinetics Influence Overall and Progression-Free Survival in Metastatic Colorectal Cancer Patients, Clin Pharmacokinet, vol.55, issue.11, pp.1381-1394, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01769811

H. Neubert, White Paper on Recent Issues in Bioanalysis: focus on immunogenicity assays by hybrid LBA/LCMS and regulatory feedback (Part 2 -PK, PD & ADA assays by hybrid LBA/LCMS & regulatory agencies' inputs on bioanalysis, 2018.

B. Desilva, Recommendations for the bioanalytical method validation of ligandbinding assays to support pharmacokinetic assessments of macromolecules, Pharm Res, vol.20, issue.11, pp.1885-900, 2003.

C. W. Damen, Development and validation of an enzyme-linked immunosorbent assay for the quantification of trastuzumab in human serum and plasma, Anal Biochem, vol.391, issue.2, pp.114-134, 2009.

A. N. Hoofnagle and M. H. Wener, The fundamental flaws of immunoassays and potential solutions using tandem mass spectrometry, J Immunol Methods, vol.347, issue.1-2, pp.3-11, 2009.

E. Ezan and F. Bitsch, Critical comparison of MS and immunoassays for the bioanalysis of therapeutic antibodies, Bioanalysis, vol.1, issue.8, pp.1375-88, 2009.

M. El-amrani, Quantification of active infliximab in human serum with liquid chromatography-tandem mass spectrometry using a tumor necrosis factor alpha -based pre-analytical sample purification and a stable isotopic labeled infliximab bio-similar as internal standard: A target-based, sensitive and cost-effective method, J Chromatogr A, vol.1454, pp.42-50, 2016.

H. H. Chiu, Development of a general method for quantifying IgG-based therapeutic monoclonal antibodies in human plasma using protein G purification coupled with a two internal standard calibration strategy using LC-MS/MS, Anal Chim Acta, vol.1019, pp.93-102, 2018.

H. H. Chiu, Development of an LC-MS/MS method with protein G purification strategy for quantifying bevacizumab in human plasma, Anal Bioanal Chem, vol.409, issue.28, pp.6583-6593, 2017.

G. Nugue, Monitoring monoclonal antibody delivery in oncology: the example of bevacizumab, PLoS One, vol.8, issue.8, p.72021, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00854640

A. Nakamura, High performance plasma amyloid-beta biomarkers for Alzheimer's disease, Nature, vol.554, issue.7691, pp.249-254, 2018.

S. E. Schindler, High-precision plasma beta-amyloid 42/40 predicts current and future brain amyloidosis, 2019.

M. Wang, J. Chen, and I. V. Turko, 15N-labeled full-length apolipoprotein E4 as an internal standard for mass spectrometry quantification of apolipoprotein E isoforms, Anal Chem, vol.84, pp.8340-8344, 2012.

J. L. Proc, A quantitative study of the effects of chaotropic agents, surfactants, and solvents on the digestion efficiency of human plasma proteins by trypsin, J Proteome Res, vol.9, issue.10, pp.5422-5459, 2010.

R. Simon, Total ApoE and ApoE4 isoform assays in an Alzheimer's disease casecontrol study by targeted mass spectrometry (n=669): a pilot assay for methioninecontaining proteotypic peptides, Mol Cell Proteomics, vol.11, issue.11, pp.1389-403, 2012.

E. Martinez-morillo, Assessment of peptide chemical modifications on the development of an accurate and precise multiplex selected reaction monitoring assay for apolipoprotein e isoforms, J Proteome Res, vol.13, issue.2, pp.1077-87, 2014.

O. V. Krokhin, W. Ens, and K. G. Standing, Characterizing degradation products of peptides containing N-terminal Cys residues by (off-line high-performance liquid chromatography)/matrix-assisted laser desorption/ionization quadrupole time-of-flight measurements, Rapid Commun Mass Spectrom, vol.17, issue.22, pp.2528-2562, 2003.

E. Martinez-morillo, Total apolipoprotein E levels and specific isoform composition in cerebrospinal fluid and plasma from Alzheimer's disease patients and controls, Acta Neuropathol, vol.127, issue.5, pp.633-676, 2014.

M. Olkowicz, Development and analytical comparison of microflow and nanoflow liquid chromatography/mass spectrometry procedures for quantification of cardiac troponin T in mouse hearts, Talanta, vol.131, pp.510-530, 2015.

G. Peslova, Hepcidin, the hormone of iron metabolism, is bound specifically to alpha-2-macroglobulin in blood, Blood, vol.113, issue.24, pp.6225-6261, 2009.

D. Girelli, E. Nemeth, and D. W. Swinkels, Hepcidin in the diagnosis of iron disorders, Blood, vol.127, issue.23, pp.2809-2822, 2016.

J. B. Jordan, Hepcidin revisited, disulfide connectivity, dynamics, and structure, J Biol Chem, vol.284, issue.36, pp.24155-67, 2009.

D. W. Swinkels, Advances in quantitative hepcidin measurements by time-of-flight mass spectrometry, PLoS One, vol.3, issue.7, p.2706, 2008.

H. N. Hunter, The solution structure of human hepcidin, a peptide hormone with antimicrobial activity that is involved in iron uptake and hereditary hemochromatosis, J Biol Chem, vol.277, issue.40, pp.37597-603, 2002.

C. M. Laarakkers, Improved mass spectrometry assay for plasma hepcidin: detection and characterization of a novel hepcidin isoform, PLoS One, vol.8, issue.10, p.75518, 2013.

I. C. Macdougall, Current status of the measurement of blood hepcidin levels in chronic kidney disease, Clin J Am Soc Nephrol, vol.5, issue.9, pp.1681-1690, 2010.

L. N. Van-der-vorm, Toward Worldwide Hepcidin Assay Harmonization: Identification of a Commutable Secondary Reference Material, Clin Chem, vol.62, issue.7, pp.993-1001, 2016.

S. Rogstad, A Retrospective Evaluation of the Use of Mass Spectrometry in FDA Biologics License Applications, J Am Soc Mass Spectrom, vol.28, issue.5, pp.786-794, 2017.

Q. Zhang, Generic automated method for liquid chromatography-multiple reaction monitoring mass spectrometry based monoclonal antibody quantitation for preclinical pharmacokinetic studies, Anal Chem, vol.86, issue.17, pp.8776-84, 2014.

J. F. Jourdil, Infliximab quantitation in human plasma by liquid chromatographytandem mass spectrometry: towards a standardization of the methods?, Anal Bioanal Chem, vol.409, issue.5, pp.1195-1205, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02454688

F. A. Administration, Bioanalytical method validation guidance for industry, 327. Agency, E.M., Bioanalytical method validation, 2011.

D. H. Chace, Mass Spectrometry in the Clinical Laboratory: General Principles and Guidance, 2007.