M. Yáñez-mó, Biological properties of extracellular vesicles and their physiological functions, Journal of Extracellular Vesicles, vol.4, issue.0, p.29, 2015.

W. Yiran, Global scientific trends on exosome research during 2007-2016: A bibliometric analysis, Oncotarget, vol.5, issue.0, p.24, 2017.

E. Chargaff, The Biological Significance of the Thromboplastic Protein of Blood, Journal of Biological Chemistry, vol.166, issue.1, pp.189-197, 1924.

P. Wolf, The Nature and Significance of Platelet Products in Human Plasma, British Journal of Haematology, vol.13, issue.3, pp.269-288, 1924.

. Bin-tao-pan, Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor, Cell, vol.33, issue.3, pp.967-978, 1924.

, B lymphocytes secrete antigen-presenting vesicles, The Journal of Experimental Medicine, vol.183, issue.3, pp.1161-1172, 1924.

C. Théry, Exosomes: Composition, biogenesis and function, Nature Reviews Immunology, vol.2, issue.8, p.27, 2002.

. Basant-kumar-thakur, Double-stranded DNA in exosomes: A novel biomarker in cancer detection, Cell Research, vol.24, issue.6, pp.766-769, 1924.

J. Ratajczak, Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: Evidence for horizontal transfer of mRNA and protein delivery, Leukemia, vol.20, issue.5, p.35, 1924.

H. Valadi, Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nature Cell Biology, vol.9, issue.6, p.148, 2007.

J. Skog, Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers, Nature Cell Biology, vol.10, issue.12, p.32, 2008.

C. P. Lai, Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters, Nature Communications, vol.6, p.7029, 1924.

M. Guescini, Astrocytes and Glioblastoma cells release exosomes carrying mtDNA, Journal of Neural Transmission, vol.117, issue.1, p.24, 2010.
DOI : 10.1007/s00702-009-0288-8

L. Balaj, Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences, Nature Communications, vol.2, p.180, 1924.
DOI : 10.1038/ncomms1180

URL : https://www.nature.com/articles/ncomms1180.pdf

C. Kahlert, Identification of Double Stranded Genomic DNA Spanning all Chromosomes with Mutated KRAS and p53 DNA in the Serum Exosomes of Patients with Pancreatic Cancer, Journal of Biological Chemistry, pp.113-532267, 1924.

M. Colombo, Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles, Annual Review of Cell and Developmental Biology, vol.30, p.28, 2014.

A. Llorente, Cholesterol regulates prostasome release from secretory lysosomes in PC-3 human prostate cancer cells, European Journal of Cell Biology, vol.86, issue.7, pp.405-415, 1925.

K. Laulagnier, Mast cell-and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization, Biochemical Journal, vol.380, issue.1, pp.161-171, 1925.

I. Parolini, Microenvironmental pH Is a Key Factor for Exosome Traffic in Tumor Cells, Journal of Biological Chemistry, vol.284, issue.49, p.28, 1925.

A. A. Sander and . Kooijmans, Exosome mimetics: A novel class of drug delivery systems, International Journal of Nanomedicine, vol.7, p.25, 2012.

J. Tyson and . Smyth, Examination of the specificity of tumor cell derived exosomes with tumor cells in vitro, Biochimica Et Biophysica Acta, vol.1838, issue.11, pp.2954-2965, 1925.

D. Fitzner, Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis, J Cell Sci, vol.124, issue.3, p.50, 2011.

J. R. Edgar, Q&A: What are exosomes, exactly?, BMC Biology, vol.14, p.27, 2016.

H. W. King, Hypoxic enhancement of exosome release by breast cancer cells, BMC Cancer, vol.12, p.26, 2012.

A. Savina, Exosome Release Is Regulated by a Calcium-dependent Mechanism in K562 Cells, Journal of Biological Chemistry, vol.278, issue.22, p.148, 2003.

E. Segura, ICAM-1 on exosomes from mature dendritic cells is critical for efficient naive T-cell priming, Blood, vol.106, issue.1, pp.216-223, 1926.

L. Kilpinen, Extracellular membrane vesicles from umbilical cord blood-derived MSC protect against ischemic acute kidney injury, a feature that is lost after inflammatory conditioning, Journal of Extracellular Vesicles, vol.2, 1926.

M. D. Beckler, Proteomic analysis of exosomes from mutant KRAS colon cancer cells identifies intercellular transfer of mutant KRAS, Molecular & cellular proteomics: MCP, vol.12, issue.2, pp.343-355, 1926.

J. Bow and . Tauro, Oncogenic H-ras reprograms Madin-Darby canine kidney (MDCK) cell-derived exosomal proteins following epithelial-mesenchymal transition, Molecular & cellular proteomics: MCP, vol.12, issue.8, p.27, 2013.

G. Olivier and . De-jong, Cellular stress conditions are reflected in the protein and RNA content of endothelial cell-derived exosomes, Journal of Extracellular Vesicles, vol.1, 1926.

P. Kucharzewska, Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development, Proceedings of the National Academy of Sciences, vol.110, issue.18, pp.7312-7317, 1926.

H. Kalra, Focus on Extracellular Vesicles: Introducing the Next Small Big Thing, International Journal of Molecular Sciences, vol.17, issue.2, p.53, 2016.

C. Théry, Membrane vesicles as conveyors of immune responses, Nature Reviews Immunology, vol.9, issue.8, p.35, 1927.

E. Van-der-pol, Classification, functions, and clinical relevance of extracellular vesicles, Pharmacological Reviews, vol.64, issue.3, p.45, 2012.

. Samir-el-andaloussi, Extracellular vesicles: Biology and emerging therapeutic opportunities, Nature Reviews. Drug Discovery, vol.12, issue.5, p.29, 2013.

C. Théry, Isolation and characterization of exosomes from cell culture supernatants and biological fluids, Current Protocols in Cell Biology, vol.37, p.50, 1927.

F. G. Harry and . Heijnen, Activated Platelets Release Two Types of Membrane Vesicles: Microvesicles by Surface Shedding and Exosomes Derived From Exocytosis of Multivesicular Bodies and alphaGranules, Blood, vol.94, issue.11, p.27, 1999.

C. Théry, Proteomic Analysis of Dendritic Cell-Derived Exosomes: A Secreted Subcellular Compartment Distinct from Apoptotic Vesicles, The Journal of Immunology, vol.166, issue.12, p.27, 2001.

, Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes, The Journal of Cell Biology, vol.101, issue.3, p.27, 1985.

S. Keerthikumar, Proteogenomic analysis reveals exosomes are more oncogenic than ectosomes, Oncotarget, vol.6, issue.17, p.27, 2015.

. Suresh-mathivanan, Exosomes: Extracellular organelles important in intercellular communication, Journal of Proteomics, vol.73, issue.10, p.29, 2010.

R. Crescitelli, Distinct RNA profiles in subpopulations of extracellular vesicles: Apoptotic bodies, microvesicles and exosomes, Journal of Extracellular Vesicles, vol.2, p.27, 2013.

J. Kowal, Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes, Proceedings of the National Academy of Sciences, vol.113, issue.8, pp.968-977, 1928.

C. Théry, Isolation and Characterization of Exosomes from Cell Culture Supernatants and Biological Fluids, Current Protocols in Cell Biology, p.28, 2001.

M. Paulaitis, Dynamic Scaling of Exosome Sizes, Langmuir, 1928.

E. Van-der-pol, Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles, Journal of Thrombosis and Haemostasis, vol.14, issue.1, p.46, 2016.

A. Montecalvo, Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes, Blood, vol.119, issue.3, pp.756-766, 1928.

J. Katrin and . Svensson, Exosome uptake depends on ERK1/2-heat shock protein 27 signalling and lipid raft-mediated endocytosis negatively regulated by caveolin-1, Journal of Biological Chemistry, pp.112-445403, 1928.

S. Gatti, Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury, Nephrology Dialysis Transplantation, vol.26, issue.5, pp.1474-1483, 1929.

I. Del-conde, Tissue-factor-bearing microvesicles arise from lipid rafts and fuse with activated platelets to initiate coagulation, Blood, vol.106, issue.5, pp.1604-1611, 1929.

G. Raposo, B lymphocytes secrete antigen-presenting vesicles, The Journal of Experimental Medicine, vol.183, issue.3, pp.1161-1172, 1929.
DOI : 10.1084/jem.183.3.1161

URL : http://jem.rupress.org/content/183/3/1161.full.pdf

J. Wolfers, Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming, Nature Medicine, vol.7, issue.3, p.30, 2001.

K. Pramod and . Giri, Exosomes Derived from M. Bovis BCG Infected Macrophages Activate Antigen-Specific CD4+ and CD8+ T Cells In Vitro and In Vivo, PLOS ONE, vol.3, issue.6, p.30, 2008.

J. D. Walker, Cytomegalovirus-infected human endothelial cells can stimulate allogeneic CD4+ memory T cells by releasing antigenic exosomes, Journal of immunology, vol.182, issue.3, p.30, 1950.

L. Luketic, Antigen Presentation by Exosomes Released from Peptide-Pulsed Dendritic Cells Is not Suppressed by the Presence of Active CTL, The Journal of Immunology, vol.179, issue.8, p.31, 2007.

S. Utsugi-kobukai, MHC class I-mediated exogenous antigen presentation by exosomes secreted from immature and mature bone marrow derived dendritic cells, Immunology Letters, vol.89, issue.2, p.31, 2003.

C. Admyre, Direct exosome stimulation of peripheral humanT cells detected by ELISPOT, European Journal of Immunology, vol.36, issue.7, p.31, 2006.

N. Chaput, Exosomes as Potent Cell-Free Peptide-Based Vaccine. II. Exosomes in CpG Adjuvants Efficiently Prime Naive Tc1 Lymphocytes Leading to Tumor Rejection, The Journal of Immunology, vol.172, issue.4, p.31, 2004.

C. Théry, Indirect activation of naïve CD4 + T cells by dendritic cell-derived exosomes, Nature Immunology, vol.3, issue.12, p.31, 2002.

D. Skokos, Mast Cell-Derived Exosomes Induce Phenotypic and Functional Maturation of Dendritic Cells and Elicit Specific Immune Responses In Vivo, The Journal of Immunology, vol.170, issue.6, p.31, 2003.

J. Klibi, Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells, Blood, vol.113, issue.9, p.31, 2009.

A. Clayton, Human Tumor-Derived Exosomes Selectively Impair Lymphocyte Responses to Interleukin-2, Cancer Research, vol.67, issue.15, p.31, 2007.
DOI : 10.1158/0008-5472.can-06-3456

URL : http://cancerres.aacrjournals.org/content/canres/67/15/7458.full.pdf

C. Liu, Murine Mammary Carcinoma Exosomes Promote Tumor Growth by Suppression of NK Cell Function, The Journal of Immunology, vol.176, issue.3, p.31, 2006.
DOI : 10.4049/jimmunol.176.3.1375

URL : http://www.jimmunol.org/content/176/3/1375.full.pdf

A. Clayton, Human Tumor-Derived Exosomes Down-Modulate NKG2D Expression, The Journal of Immunology, vol.180, issue.11, p.31, 2008.
DOI : 10.4049/jimmunol.180.11.7249

URL : http://www.jimmunol.org/content/180/11/7249.full.pdf

S. Yu, Tumor Exosomes Inhibit Differentiation of Bone Marrow Dendritic Cells, The Journal of Immunology, vol.178, issue.11, p.31, 2007.
DOI : 10.4049/jimmunol.178.11.6867

X. Xiang, Induction of myeloid-derived suppressor cells by tumor exosomes, International journal of cancer. Journal international du cancer, vol.124, issue.11, p.31, 2009.
DOI : 10.1002/ijc.24249

URL : http://europepmc.org/articles/pmc2757307?pdf=render

H. Pêche, Presentation of donor major histocompatibility complex antigens by bone marrow dendritic cell-derived exosomes modulates allograft rejection1, Transplantation, vol.76, issue.10, p.31, 2003.

S. Kim, Effective Treatment of Inflammatory Disease Models with Exosomes Derived from Dendritic Cells Genetically Modified to Express IL-4, The Journal of Immunology, vol.179, issue.4, p.31, 2007.
DOI : 10.4049/jimmunol.179.4.2242

URL : http://www.jimmunol.org/content/179/4/2242.full.pdf

N. Prado, Exosomes from Bronchoalveolar Fluid of Tolerized Mice Prevent Allergic Reaction, The Journal of Immunology, vol.181, issue.2, p.31, 2008.
DOI : 10.4049/jimmunol.181.2.1519

URL : http://www.jimmunol.org/content/181/2/1519.full.pdf

A. G. Thompson, Extracellular vesicles in neurodegenerative disease-pathogenesis to biomarkers, Nature Reviews Neurology, vol.12, issue.6, p.31, 2016.
DOI : 10.1038/nrneurol.2016.68

J. Fauré, Exosomes are released by cultured cortical neurones, Molecular and Cellular Neuroscience, vol.31, issue.4, p.31, 2006.

M. Chivet, Emerging role of neuronal exosomes in the central nervous system, Frontiers in Physiology, vol.3, p.31, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00733770

A. M. Janas, Exosomes and other extracellular vesicles in neural cells and neurodegenerative diseases, Biochimica et Biophysica Acta-Biomembranes, vol.1858, issue.6, p.31, 2016.
DOI : 10.1016/j.bbamem.2016.02.011

URL : https://doi.org/10.1016/j.bbamem.2016.02.011

B. Zhang, Synaptic Vesicle Size and Number Are Regulated by a Clathrin Adaptor Protein Required for Endocytosis, Neuron, vol.21, issue.6, p.31, 1998.
DOI : 10.1016/s0896-6273(00)80664-9

URL : https://doi.org/10.1016/s0896-6273(00)80664-9

R. Jahn, Molecular machines governing exocytosis of synaptic vesicles, Nature, vol.490, issue.7419, p.31, 2012.
DOI : 10.1038/nature11320

URL : http://europepmc.org/articles/pmc4461657?pdf=render

V. Budnik, Extracellular vesicles round off communication in the nervous system, Nature reviews. Neuroscience, vol.17, issue.3, p.31, 2016.

E. Krämer-albers, Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons?, PROTEOMICS-Clinical Applications, vol.1, issue.11, p.31, 2007.

F. Antonucci, Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism, The EMBO Journal, vol.31, issue.5, p.31, 2012.
DOI : 10.1038/emboj.2011.489

URL : http://emboj.embopress.org/content/31/5/1231.full.pdf

L. Morel, Neuronal Exosomal miRNA-dependent Translational Regulation of Astroglial Glutamate Transporter GLT1, Journal of Biological Chemistry, vol.288, issue.10, p.31, 2013.
DOI : 10.1074/jbc.m112.410944

URL : http://www.jbc.org/content/288/10/7105.full.pdf

C. A. Escudero, The p75 neurotrophin receptor evades the endolysosomal route in neuronal cells, favouring multivesicular bodies specialised for exosomal release, Journal of Cell Science, vol.127, issue.9, p.31, 2014.

B. Kumar, Acute myeloid leukemia transforms the bone marrow niche into a leukemiapermissive microenvironment through exosome secretion, Leukemia, p.31, 2017.
DOI : 10.1038/leu.2017.259

URL : https://www.nature.com/articles/leu2017259.pdf

Y. Lee, Oligodendroglia metabolically support axons and contribute to neurodegeneration, Nature, vol.487, issue.7408, p.31, 2012.
DOI : 10.1038/nature11314

URL : http://europepmc.org/articles/pmc3408792?pdf=render

H. K. Lee, Mesenchymal Stem Cells Deliver Exogenous miRNAs to Neural Cells and Induce Their Differentiation and Glutamate Transporter Expression, Stem Cells and Development, vol.23, issue.23, p.31, 2014.
DOI : 10.1089/scd.2014.0146

S. A. Bellingham, Exosomes: Vehicles for the transfer of toxic proteins associated with neurodegenerative diseases?, Membrane Physiology and Membrane Biophysics, vol.3, p.31, 2012.

X. Cao, MicroRNA biomarkers of Parkinson's disease in serum exosome-like microvesicles, Neuroscience Letters, vol.644, p.31, 2017.

A. M. Miranda, Neuronal lysosomal dysfunction releases exosomes harboring APP C-terminal fragments and unique lipid signatures, Nature Communications, vol.9, issue.1, p.31, 2018.
DOI : 10.1038/s41467-017-02533-w

URL : https://www.nature.com/articles/s41467-017-02533-w.pdf

F. Fenizia, EGFR mutations in lung cancer: From tissue testing to liquid biopsy, Future Oncology, vol.11, issue.11, p.32, 2015.
DOI : 10.2217/fon.15.23

A. Lewis, Pancreatic cancer: Are "liquid biopsies" ready for prime-time?, World Journal of Gastroenterology, vol.22, issue.32, p.32, 2016.
DOI : 10.3748/wjg.v22.i32.7175

URL : https://doi.org/10.3748/wjg.v22.i32.7175

M. Gerlinger, Intratumor heterogeneity and branched evolution revealed by multiregion sequencing, The New England Journal of Medicine, vol.366, issue.10, p.32, 2012.

A. Panagiotara, Exosomes: A Cancer Theranostics Road Map, Public Health Genomics, vol.20, issue.2, p.32, 2017.
DOI : 10.1159/000478253

URL : https://www.karger.com/Article/Pdf/478253

T. Steuber, Serum markers for prostate cancer: A rational approach to the literature, European Urology, vol.54, issue.1, pp.31-40, 1932.
DOI : 10.1016/j.eururo.2008.01.034

M. R. Fernando, New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes, PLoS ONE, vol.12, issue.8, p.32, 2017.

H. W. Hou, Isolation and retrieval of circulating tumor cells using centrifugal forces, Scientific Reports, vol.3, p.32, 2013.
DOI : 10.1038/srep01259

URL : https://www.nature.com/articles/srep01259.pdf

L. Guo, Extracellular vesicles and their diagnostic and prognostic potential in cancer, Translational Cancer Research, vol.6, issue.3, p.32, 2017.
DOI : 10.21037/tcr.2017.06.32

M. Tkach, Communication by Extracellular Vesicles: Where We Are and Where We Need to Go, Cell, vol.164, issue.6, p.32, 2016.
DOI : 10.1016/j.cell.2016.01.043

URL : https://doi.org/10.1016/j.cell.2016.01.043

M. Kanada, Signaling by Extracellular Vesicles Advances Cancer Hallmarks, Trends in Cancer, vol.2, issue.2, p.32, 2016.
DOI : 10.1016/j.trecan.2015.12.005

O. Lorraine and . Driscoll, Expanding on Exosomes and Ectosomes in Cancer, New England Journal of Medicine, vol.372, issue.24, p.32, 2015.

M. Wiktoria and . Suchorska, The role of exosomes in tumor progression and metastasis (Review), Oncology Reports, vol.35, issue.3, p.32, 2016.

C. Honorary, Research Director, Founder of RevInterCell, a et al. Cell-derived Extracellular Vesicles Open New Perspectives for Cancer Research, ) and UPMC, vol.1, p.32, 2015.

K. Al-nedawi, Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells, Nature Cell Biology, vol.10, issue.5, pp.619-624, 1932.
DOI : 10.1038/ncb1725

H. Peinado, Melanoma exosomes educate bone marrow progenitor cells toward a prometastatic phenotype through MET, Nature Medicine, vol.18, issue.6, p.32, 2012.
DOI : 10.1038/nm.2753

URL : http://europepmc.org/articles/pmc3645291?pdf=render

B. S. Hong, Colorectal cancer cell-derived microvesicles are enriched in cell cycle-related mRNAs that promote proliferation of endothelial cells, BMC Genomics, vol.10, issue.1, p.32, 2009.

T. Kogure, Intercellular nanovesicle-mediated microRNA transfer: A mechanism of environmental modulation of hepatocellular cancer cell growth, Hepatology, vol.54, issue.4, p.32, 2011.

W. Zhu, Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth in vivo, Cancer Letters, vol.315, issue.1, p.32, 2012.

H. Xiao, Mast cell exosomes promote lung adenocarcinoma cell proliferation-role of KIT-stem cell factor signaling, Cell communication and signaling: CCS, vol.12, p.32, 2014.

C. Yeung, Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1, Nature Communications, vol.7, p.32, 2016.

Z. Zhang, Loss of exosomal miR-320a from cancer-associated fibroblasts contributes to HCC proliferation and metastasis, Cancer Letters, vol.397, pp.33-42, 1932.

S. Josson, Stromal fibroblast-derived miR-409 promotes epithelial-to-mesenchymal transition and prostate tumorigenesis, Oncogene, vol.34, issue.21, p.2690, 1932.

L. Lugini, Exosomes from human colorectal cancer induce a tumor-like behavior in colonic mesenchymal stromal cells, Oncotarget, vol.7, issue.31, pp.50086-50098, 1932.

D. Kumar, Biomolecular characterization of exosomes released from cancer stem cells: Possible implications for biomarker and treatment of cancer, Oncotarget, vol.6, issue.5, p.32, 2015.

J. Wu, Role of stem cell-derived exosomes in cancer, Oncology Letters, vol.13, issue.5, pp.2855-2866, 1932.

A. Sharma, Role of stem cell derived exosomes in tumor biology, International Journal of Cancer, vol.142, issue.6, p.32, 2018.

T. N. Minh and . Le, miR-200-containing extracellular vesicles promote breast cancer cell metastasis, The Journal of Clinical Investigation, vol.124, issue.12, p.33, 2014.

A. Zomer, In Vivo Imaging Reveals Extracellular Vesicle-Mediated Phenocopying of Metastatic Behavior. Cell, vol.161, issue.5, pp.1046-1057, 1932.

J. P. Webber, Differentiation of tumour-promoting stromal myofibroblasts by cancer exosomes, Oncogene, vol.34, issue.3, p.32, 2015.

D. Hoshino, Exosome secretion is enhanced by invadopodia and drives invasive behavior, Cell Reports, vol.5, issue.5, p.32, 2013.

I. Lazar, Adipocyte Exosomes Promote Melanoma Aggressiveness through Fatty Acid Oxidation: A Novel Mechanism Linking Obesity and Cancer, Cancer Research, vol.76, issue.14, p.32, 2016.

S. Taverna, Role of exosomes released by chronic myelogenous leukemia cells in angiogenesis, International Journal of Cancer, vol.130, issue.9, pp.2033-2043, 1932.

J. Katrin and . Svensson, Hypoxia triggers a proangiogenic pathway involving cancer cell microvesicles and PAR-2-mediated heparin-binding EGF signaling in endothelial cells, Proceedings of the National Academy of Sciences, vol.108, issue.32, p.32, 2011.

J. L. Hood, Paracrine induction of endothelium by tumor exosomes, Laboratory Investigation; a Journal of Technical Methods and Pathology, vol.89, issue.11, p.32, 2009.

D. Millimaggi, Tumor vesicle-associated CD147 modulates the angiogenic capability of endothelial cells, Neoplasia, vol.9, issue.4, p.32, 2007.

J. Wang, Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression, The Journal of Pathology, vol.239, issue.2, p.32, 2016.
DOI : 10.1002/path.4712

N. Kosaka, Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis, The Journal of Biological Chemistry, vol.288, issue.15, p.32, 2013.
DOI : 10.1074/jbc.m112.446831

URL : http://www.jbc.org/content/288/15/10849.full.pdf

G. Fan, Hypoxic exosomes promote angiogenesis, Blood, vol.124, issue.25, pp.3669-3670, 1932.
DOI : 10.1182/blood-2014-10-607846

URL : http://www.bloodjournal.org/content/124/25/3669.full.pdf

H. Tadokoro, Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells, The Journal of Biological Chemistry, vol.288, issue.48, p.32, 2013.

X. Liang, Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a, Journal of Cell Science, vol.129, issue.11, p.32, 2016.
DOI : 10.1242/jcs.170373

URL : http://jcs.biologists.org/content/129/11/2182.full.pdf

N. Kosaka, Decoding the Secret of Cancer by Means of Extracellular Vesicles, Journal of Clinical Medicine, vol.5, issue.2, p.32, 2016.

. Wei-xian-chen, Exosomes from Drug-Resistant Breast Cancer Cells Transmit Chemoresistance by a Horizontal Transfer of MicroRNAs, PLOS ONE, vol.9, issue.4, p.32, 2014.

Y. Wei, Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells, Breast Cancer Research and Treatment, vol.147, issue.2, p.32, 2014.
DOI : 10.1007/s10549-014-3037-0

L. Qu, Exosome-Transmitted lncARSR Promotes Sunitinib Resistance in Renal Cancer by Acting as a Competing Endogenous RNA, Cancer Cell, vol.29, issue.5, p.32, 2016.

Y. Chen, Breast cancer resistance protein (BCRP)-containing circulating microvesicles contribute to chemoresistance in breast cancer, Oncology Letters, vol.10, issue.6, p.32, 2015.

T. Aung, Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3, Proceedings of the National Academy of Sciences of the United States of America, vol.108, p.32, 2011.

K. E. Richards, Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells, Oncogene, vol.36, issue.13, p.32, 2017.
DOI : 10.1038/onc.2016.353

URL : http://europepmc.org/articles/pmc5366272?pdf=render

C. Mirjam and . Boelens, Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways, Cell, vol.159, issue.3, p.32, 2014.

K. Shedden, Expulsion of small molecules in vesicles shed by cancer cells: Association with gene expression and chemosensitivity profiles, Cancer Research, vol.63, issue.15, pp.4331-4337, 2003.

V. Ciravolo, Potential role of HER2-overexpressing exosomes in countering trastuzumabbased therapy, Journal of Cellular Physiology, vol.227, issue.2, p.32, 2012.

T. L. Whiteside, Tumor-Derived Exosomes and Their Role in Cancer Progression, Advances in Clinical Chemistry, vol.74, p.32, 2016.

. Morad-rémy-muhsin-sharafaldine, Procoagulant and immunogenic properties of melanoma exosomes, microvesicles and apoptotic vesicles, Oncotarget, vol.7, issue.35, p.32, 2016.

M. Iero, Tumour-released exosomes and their implications in cancer immunity, Cell Death and Differentiation, vol.15, issue.1, p.32, 2008.

M. Szajnik, Tumor-Derived Microvesicles Induce, Expand and Up-Regulate Biological Activities of Human Regulatory T Cells (Treg), PLOS ONE, vol.5, issue.7

U. Eva and . Wieckowski, Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes, Journal of Immunology, vol.183, issue.6, p.32, 1950.

G. Cai, A Regulatory Role for IL-10 Receptor Signaling in Development and B Cell Help of T Follicular Helper Cells in Mice, The Journal of Immunology, vol.189, issue.3, p.32, 2012.

T. Condamine, Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function, Trends in Immunology, vol.32, issue.1, p.32, 2011.

R. Valenti, Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-beta-mediated suppressive activity on T lymphocytes, Cancer Research, vol.66, issue.18, pp.9290-9298, 1932.

. Muller-fabbri, MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response, Proceedings of the National Academy of Sciences of the United States of America, vol.109, pp.2110-2116, 1932.

X. Ying, Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages, Oncotarget, vol.7, issue.28, pp.43076-43087, 1932.

S. Sukhvinder and . Sidhu, The microvesicle as a vehicle for EMMPRIN in tumor-stromal interactions, Oncogene, vol.23, issue.4, p.32, 2004.

M. Aga, Exosomal HIF1? supports invasive potential of nasopharyngeal carcinoma-associated LMP1-positive exosomes, Oncogene, vol.33, issue.37, p.32, 2014.

A. Zomer, In Vivo Imaging Reveals Extracellular Vesicle-Mediated Phenocopying of Metastatic Behavior. Cell, vol.161, issue.5, pp.1046-1057, 1932.

B. Costa-silva, Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver, Nature Cell Biology, vol.17, issue.6, p.32, 2015.

Y. Miranda and . Fong, Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis, Nature Cell Biology, vol.17, issue.2, p.32, 2015.

Y. Liu, Tumor Exosomal RNAs Promote Lung Pre-metastatic Niche Formation by Activating Alveolar Epithelial TLR3 to Recruit Neutrophils, Cancer Cell, vol.30, issue.2, p.32, 2016.

A. Hoshino, Tumour exosome integrins determine organotropic metastasis, Nature, vol.527, issue.7578, pp.329-335, 1932.

D. Antonopoulos, Cancer's smart bombs: Tumor-derived exosomes target lung epithelial cells triggering pre-metastatic niche formation, Journal of Thoracic Disease, vol.9, issue.4, p.32, 2017.

N. Tominaga, Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier, Nature Communications, vol.6, p.32, 2015.

L. Zhang, Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth, Nature, vol.527, issue.7576, p.32, 2015.

S. A. Bliss, Mesenchymal Stem Cell-Derived Exosomes Stimulate Cycling Quiescence and Early Breast Cancer Dormancy in Bone Marrow, Cancer Research, vol.76, issue.19, p.32, 2016.

D. D. Vizio, Large oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease, The American Journal of Pathology, vol.181, issue.5, p.32, 2012.

A. C. Leal, Tumor-Derived Exosomes Induce the Formation of Neutrophil Extracellular Traps: Implications For The Establishment of Cancer-Associated Thrombosis, Scientific Reports, vol.7, issue.1, p.6438, 1932.

I. Edit and . Buzas, Emerging role of extracellular vesicles in inflammatory diseases, Nature Reviews. Rheumatology, vol.10, issue.6, p.32, 2014.

H. Peinado, Melanoma exosomes educate bone marrow progenitor cells toward a prometastatic phenotype through MET, Nature Medicine, vol.18, issue.6, p.35, 2012.

J. Skog, Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers, Nature Cell Biology, vol.10, issue.12, p.35, 2008.
DOI : 10.1038/ncb1800

URL : http://europepmc.org/articles/pmc3423894?pdf=render

R. Bhome, Exosomal microRNAs (exomiRs): Small molecules with a big role in cancer, Cancer Letters, vol.420, p.33, 2018.

F. Cappello, Exosome levels in human body fluids: A tumor marker by themselves?, European Journal of Pharmaceutical Sciences, vol.96, p.33, 2017.

M. Logozzi, High Levels of Exosomes Expressing CD63 and Caveolin-1 in Plasma of Melanoma Patients, PLOS ONE, vol.4, issue.4, p.45, 2009.

C. Federici, Exosome release and low pH belong to a framework of resistance of human melanoma cells to cisplatin, PloS One, vol.9, issue.2, p.33, 2014.

A. Caivano, High serum levels of extracellular vesicles expressing malignancy-related markers are released in patients with various types of hematological neoplastic disorders, Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine, vol.36, issue.12, p.33, 2015.

D. Duijvesz, Immuno-based detection of extracellular vesicles in urine as diagnostic marker for prostate cancer, International Journal of Cancer, vol.137, issue.12, p.33, 2015.

J. Silva, Analysis of exosome release and its prognostic value in human colorectal cancer, Genes, Chromosomes & Cancer, vol.51, issue.4, p.33, 2012.

. Estibaliz-alegre, Circulating melanoma exosomes as diagnostic and prognosis biomarkers, International Journal of Clinical Chemistry, vol.454, p.33, 2016.

Y. Matsumoto, Quantification of plasma exosome is a potential prognostic marker for esophageal squamous cell carcinoma, Oncology Reports, vol.36, issue.5, p.33, 2016.

H. Shao, Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma, Nature Communications, vol.6, p.6999, 1934.

D. Zocco, Extracellular Vesicles as Shuttles of Tumor Biomarkers and Anti-Tumor Drugs, Frontiers in Oncology, vol.4, p.34, 2014.

A. Schwab, Extracellular vesicles from infected cells: Potential for direct pathogenesis, Frontiers in Microbiology, vol.6, p.34, 2015.
DOI : 10.3389/fmicb.2015.01132

URL : https://www.frontiersin.org/articles/10.3389/fmicb.2015.01132/pdf

A. Marcilla, Extracellular vesicles in parasitic diseases, Journal of Extracellular Vesicles, vol.3, p.34, 2014.
DOI : 10.3402/jev.v3.25040

URL : https://doi.org/10.3402/jev.v3.25040

X. Delabranche, Microparticles and infectious diseases, Medecine Et Maladies Infectieuses, vol.42, issue.8, p.34, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00789006

J. H. Ellwanger, Exosomes in HIV infection: A review and critical look, Infection, Genetics and Evolution, vol.53, p.34, 2017.

M. N. Madison, Implications in HIV-1 Pathogenesis. Viruses, vol.7, pp.4093-4118, 1934.

S. Chettimada, Exosome markers associated with immune activation and oxidative stress in HIV patients on antiretroviral therapy, Scientific Reports, vol.8, issue.1, p.7227, 1934.

C. Demarino, Antiretroviral Drugs Alter the Content of Extracellular Vesicles from HIV-1-Infected Cells, Scientific Reports, vol.8, issue.1, p.7653, 1934.

H. Lei, Protein kinase A-dependent translocation of Hsp90 alpha impairs endothelial nitric-oxide synthase activity in high glucose and diabetes, The Journal of Biological Chemistry, vol.282, issue.13, p.34, 2007.

H. Sheng, Insulinoma-released exosomes or microparticles are immunostimulatory and can activate autoreactive T cells spontaneously developed in nonobese diabetic mice, Journal of Immunology, vol.187, issue.4, p.34, 1950.

R. Bashratyan, Insulinoma-released exosomes activate autoreactive marginal zone-like B cells that expand endogenously in prediabetic NOD mice, European Journal of Immunology, vol.43, issue.10, p.34, 2013.
DOI : 10.1002/eji.201343376

URL : http://europepmc.org/articles/pmc3832688?pdf=render

C. P. Hasilo, Presence of diabetes autoantigens in extracellular vesicles derived from human islets, Scientific Reports, vol.7, issue.1

M. Garcia-contreras, Exosomes as biomarkers and therapeutic tools for type 1 diabetes mellitus, European Review for Medical and Pharmacological Sciences, vol.21, issue.12, p.34, 2017.

. Zhong-bin-deng, Adipose tissue exosome-like vesicles mediate activation of macrophageinduced insulin resistance, Diabetes, vol.58, issue.11, p.34, 2009.

M. Lee, Exosomes as the source of biomarkers of metabolic diseases, Annals of Pediatric Endocrinology & Metabolism, vol.21, issue.3, p.34, 2016.

J. Withrow, Extracellular vesicles in the pathogenesis of rheumatoid arthritis and osteoarthritis, Arthritis Research & Therapy, vol.18, p.35, 2016.

S. Cosenza, Pathogenic or Therapeutic Extracellular Vesicles in Rheumatic Diseases: Role of Mesenchymal Stem Cell-Derived Vesicles, International Journal of Molecular Sciences, vol.18, issue.4, p.35, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01623928

K. Skriner, Association of citrullinated proteins with synovial exosomes, Arthritis and Rheumatism, vol.54, issue.12, p.35, 2006.

E. Boilard, Platelets amplify inflammation in arthritis via collagen-dependent microparticle production, Science, vol.327, issue.5965, p.35, 2010.
DOI : 10.1126/science.1181928

URL : http://europepmc.org/articles/pmc2927861?pdf=render

M. Kurowska-stolarska, MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis, Proceedings of the National Academy of Sciences of the United States of America, vol.108, p.35, 2011.

M. Chiara-deregibus, Endothelial progenitor cell derived microvesicles activate an angiogenic program in endothelial cells by a horizontal transfer of mRNA, Blood, vol.110, issue.7, p.35, 2007.

M. Herrera, Human liver stem cell-derived microvesicles accelerate hepatic regeneration in hepatectomized rats, Journal of Cellular and Molecular Medicine, vol.14, issue.6b, p.35, 2010.

G. Raposo, B lymphocytes secrete antigen-presenting vesicles, The Journal of Experimental Medicine, vol.183, issue.3, p.35, 1996.
DOI : 10.1084/jem.183.3.1161

URL : http://jem.rupress.org/content/183/3/1161.full.pdf

R. Yeo, Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery, Advanced Drug Delivery Reviews, vol.65, issue.3, p.35, 2013.

G. Henry and . Lamparski, Production and characterization of clinical grade exosomes derived from dendritic cells, Journal of Immunological Methods, vol.270, issue.2, p.35, 2002.

. Samir-el-andaloussi, Extracellular vesicles: Biology and emerging therapeutic opportunities, Nature Reviews Drug Discovery, vol.12, issue.5, p.35, 2013.

K. Trajkovic, Ceramide triggers budding of exosome vesicles into multivesicular endosomes, Science, issue.5867, p.35, 2008.

A. Bobrie, Exosome Secretion: Molecular Mechanisms and Roles in Immune Responses, Traffic, vol.12, issue.12, p.35, 2011.
DOI : 10.1111/j.1600-0854.2011.01225.x

K. Al-nedawi, Endothelial expression of autocrine VEGF upon the uptake of tumor-derived microvesicles containing oncogenic EGFR, Proceedings of the National Academy of Sciences, vol.106, issue.10, p.35, 2009.

G. Luize and . Lima, Tumor-derived microvesicles modulate the establishment of metastatic melanoma in a phosphatidylserine-dependent manner, Cancer Letters, vol.283, issue.2, p.35, 2009.

G. Cai, A Regulatory Role for IL-10 Receptor Signaling in Development and B Cell Help of T Follicular Helper Cells in Mice, The Journal of Immunology, vol.189, issue.3, p.35, 2012.

M. Baj-krzyworzeka, Tumour-derived microvesicles carry several surface determinants and mRNA of tumour cells and transfer some of these determinants to monocytes, Cancer immunology, immunotherapy: CII, vol.55, issue.7, p.35, 2006.

Y. Zhang, Secreted monocytic miR-150 enhances targeted endothelial cell migration, Molecular Cell, vol.39, issue.1, p.35, 2010.

L. Alvarez-erviti, Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes, Nature Biotechnology, vol.29, issue.4, p.35, 2011.

R. Kojima, Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson's disease treatment, Nature Communications, vol.9, issue.1, p.35, 2018.

F. Collino, Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs, PloS One, vol.5, issue.7, p.35, 2010.

J. Derrick and . Gibbings, Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity, Nature Cell Biology, vol.11, issue.9, p.35, 2009.

J. Szebeni, Complement activation-related pseudoallergy caused by liposomes, micellar carriers of intravenous drugs, and radiocontrast agents, Critical Reviews in Therapeutic Drug Carrier Systems, vol.18, issue.6, p.35, 2001.

J. Szebeni, Complement activation-related pseudoallergy: A new class of drug-induced acute immune toxicity, Toxicology, vol.216, issue.2-3, p.35, 2005.

E. Consortium, EV-TRACK: Transparent reporting and centralizing knowledge in extracellular vesicle research, Nature Methods, vol.14, issue.3, p.155, 2017.

K. W. Witwer, Standardization of sample collection, isolation and analysis methods in extracellular vesicle research, Journal of Extracellular Vesicles, vol.2, p.155, 2013.

J. Lötvall, Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles, Journal of Extracellular Vesicles, vol.3, p.36, 2014.

K. W. Witwer, Updating the MISEV minimal requirements for extracellular vesicle studies: Building bridges to reproducibility, Journal of Extracellular Vesicles, vol.6, issue.1, p.36, 2017.

S. Gholizadeh, Microfluidic approaches for isolation, detection, and characterization of extracellular vesicles: Current status and future directions, Biosensors and Bioelectronics, vol.91, pp.588-605, 1937.

R. J. Lobb, Optimized exosome isolation protocol for cell culture supernatant and human plasma, Journal of Extracellular Vesicles, vol.4, p.52, 2015.

B. György, Detection and isolation of cell-derived microparticles are compromised by protein complexes resulting from shared biophysical parameters, Blood, vol.117, issue.4, p.52, 2011.

. Liron-issman, Cryogenic Transmission Electron Microscopy Nanostructural Study of Shed Microparticles, PLOS ONE, vol.8, issue.12, p.149, 2013.

S. A. Melo, Cancer Exosomes Perform Cell-Independent MicroRNA Biogenesis and Promote Tumorigenesis, Cancer cell, vol.26, issue.5, p.37, 2014.

Y. Yuana, Atomic force microscopy: A novel approach to the detection of nanosized blood microparticles, Journal of Thrombosis and Haemostasis, vol.8, issue.2, p.38, 2010.

J. Hardij, Characterisation of tissue factor-bearing extracellular vesicles with AFM: Comparison of air-tapping-mode AFM and liquid Peak Force AFM, Journal of Extracellular Vesicles, vol.2, p.38, 2013.

B. Whitehead, Tumour exosomes display differential mechanical and complement activation properties dependent on malignant state: Implications in endothelial leakiness, Journal of Extracellular Vesicles, vol.4, issue.0, p.38, 2015.

C. M. Hoo, A comparison of atomic force microscopy (AFM) and dynamic light scattering (DLS) methods to characterize nanoparticle size distributions, Journal of Nanoparticle Research, vol.10, issue.1, p.39, 2008.

E. Van-der-pol, Optical and non-optical methods for detection and characterization of microparticles and exosomes, Journal of Thrombosis and Haemostasis, vol.8, issue.12, p.39, 2010.

L. N. Sybren and . Maas, Possibilities and limitations of current technologies for quantification of biological extracellular vesicles and synthetic mimics, Journal of Controlled Release, vol.200, p.40, 2015.

T. Ito, A Carbon Nanotube-Based Coulter Nanoparticle Counter, Accounts of Chemical Research, vol.37, issue.12, p.40, 2004.

R. Vogel, Quantitative Sizing of Nano/Microparticles with a Tunable Elastomeric Pore Sensor, Analytical Chemistry, vol.83, issue.9, pp.3499-3506, 1940.

W. Anderson, Observations of Tunable Resistive Pulse Sensing for Exosome Analysis: Improving System Sensitivity and Stability, Langmuir, vol.31, issue.23, p.40, 2015.

D. Kozak, Simultaneous Size and ?-Potential Measurements of Individual Nanoparticles in Dispersion Using Size-Tunable Pore Sensors, ACS Nano, vol.6, issue.8, p.40, 2012.

J. C. Akers, Comparative Analysis of Technologies for Quantifying Extracellular Vesicles (EVs) in Clinical Cerebrospinal Fluids (CSF), PLOS ONE, vol.11, issue.2, p.43, 2016.

T. P. Burg, Vacuum-Packaged Suspended Microchannel Resonant Mass Sensor for Biomolecular Detection, Journal of Microelectromechanical Systems, vol.15, issue.6, p.40, 2006.

E. Arif and . Cetin, Determining therapeutic susceptibility in multiple myeloma by single-cell mass accumulation, Nature Communications, vol.8, issue.1, p.40, 2017.

M. Mark and . Stevens, Drug sensitivity of single cancer cells is predicted by changes in mass accumulation rate, Nature Biotechnology, p.40, 2016.

N. Cermak, High-throughput measurement of single-cell growth rates using serial microfluidic mass sensor arrays, Nature Biotechnology, p.40, 2016.

R. J. Kimmerling, A microfluidic platform enabling single-cell RNA-seq of multigenerational lineages, Nature Communications, vol.7, p.40, 2016.

J. Shaw-bagnall, Deformability-based cell selection with downstream immunofluorescence analysis, Integrative Biology, p.41, 2016.

A. K. Bryan, Measurement of mass, density, and volume during the cell cycle of yeast, Proceedings of the National Academy of Sciences, vol.107, issue.3, p.41, 2010.

S. Olcum, Weighing nanoparticles in solution at the attogram scale, Proceedings of the National Academy of Sciences, vol.111, issue.4, p.174, 1941.

V. Agache, An embedded microchannel in a MEMS plate resonator for ultrasensitive mass sensing in liquid, Lab on a Chip, vol.11, issue.15, p.41, 2011.

C. Hadji, Hollow MEMS mass sensors for real-time particles weighing and sizing from a few 10 nm to the #x03BC;m scale, 28th IEEE International Conference on Micro Electro Mechanical Systems (MEMS), p.41, 2015.

P. Thomas and . Burg, Weighing of biomolecules, single cells and single nanoparticles in fluid, Nature, vol.446, issue.7139, p.41, 2007.

I. Tatischeff, Fast characterisation of cell-derived extracellular vesicles by nanoparticles tracking analysis, cryo-electron microscopy, and Raman tweezers microspectroscopy, Journal of Extracellular Vesicles, vol.1, p.41, 2012.

A. Gualerzi, Raman spectroscopy uncovers biochemical tissue-related features of extracellular vesicles from mesenchymal stromal cells, Scientific Reports, vol.7, issue.1, p.45, 2017.

S. Stremersch, Identification of Individual Exosome-Like Vesicles by Surface Enhanced Raman Spectroscopy, Small, vol.12, issue.24, p.41

J. Park, Exosome Classification by Pattern Analysis of Surface-Enhanced Raman Spectroscopy Data for Lung Cancer Diagnosis, Analytical Chemistry, vol.89, issue.12, p.41, 2017.

C. Krafft, A specific spectral signature of serum and plasma-derived extracellular vesicles for cancer screening, Nanomedicine: Nanotechnology, Biology and Medicine, vol.13, issue.3, p.41, 2017.

L. Grasso, Molecular screening of cancer-derived exosomes by surface plasmon resonance spectroscopy, Analytical and Bioanalytical Chemistry, vol.407, issue.18, p.41, 2015.

H. Im, Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor, Nature Biotechnology, vol.32, issue.5, p.45, 2014.

L. M. Déborah and . Rupert, Determination of exosome concentration in solution using surface plasmon resonance spectroscopy, Analytical Chemistry, vol.86, issue.12, p.41, 2014.

S. Castorph, Structure Parameters of Synaptic Vesicles Quantified by Small-Angle X-Ray Scattering, Biophysical Journal, vol.98, issue.7, p.45, 2010.

H. Christian and D. Aass, Fluorescent particles in the antibody solution result in false TFand CD14-positive microparticles in flow cytometric analysis, Cytometry. Part A: The Journal of the International Society for Analytical Cytology, vol.79, issue.12, p.43, 2011.

A. Samuel and . Stoner, High sensitivity flow cytometry of membrane vesicles, Cytometry Part A, vol.89, issue.2, p.43, 2016.

E. Van-der-pol, Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing, Journal of Thrombosis and Haemostasis, vol.12, issue.7, p.43, 2014.

J. Els and . Van-der-vlist, Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry, Nature Protocols, vol.7, issue.7, p.43, 2012.

E. Van-der and P. , Single vs. swarm detection of microparticles and exosomes by flow cytometry, Journal of Thrombosis and Haemostasis, vol.10, issue.5, p.43, 2012.

A. Suetsugu, Imaging exosome transfer from breast cancer cells to stroma at metastatic sites in orthotopic nude-mouse models, Advanced Drug Delivery Reviews, vol.65, issue.3, p.43, 2013.

A. Zomer, In Vivo Imaging Reveals Extracellular Vesicle-Mediated Phenocopying of Metastatic Behavior. Cell, vol.161, issue.5, p.45, 2015.

Y. Yoshioka, Ultra-sensitive liquid biopsy of circulating extracellular vesicles using ExoScreen, Nature Communications, vol.5, p.45, 2014.

M. He, Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology, Lab on a Chip, vol.14, issue.19, p.54, 1944.

Y. Kitai, DNA-Containing Exosomes Derived from Cancer Cells Treated with Topotecan Activate a STING-Dependent Pathway and Reinforce Antitumor Immunity, The Journal of Immunology, vol.198, issue.4, p.44, 2017.

K. Ueda, Antibody-coupled monolithic silica microtips for highthroughput molecular profiling of circulating exosomes, Scientific Reports, vol.4, p.44, 2014.

E. Zeringer, Methods for the extraction and RNA profiling of exosomes, World Journal of Methodology, vol.3, issue.1, p.44, 2013.

K. Rekker, Comparison of serum exosome isolation methods for microRNA profiling, Clinical Biochemistry, vol.47, issue.1-2, p.44, 2014.

. Walter-w-chen, BEAMing and Droplet Digital PCR Analysis of Mutant IDH1 mRNA in Glioma Patient Serum and Cerebrospinal Fluid Extracellular Vesicles, Molecular Therapy. Nucleic Acids, vol.2, issue.7, p.45, 2013.

S. Sharma, Structural-mechanical characterization of nanoparticles-Exosomes in human saliva, using correlative AFM, FESEM and force spectroscopy, ACS nano, vol.4, issue.4, p.45, 2010.

V. Sokolova, Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy, Colloids and Surfaces B: Biointerfaces, vol.87, issue.1, p.45, 2011.

R. A. Dragovic, Sizing and phenotyping of cellular vesicles using Nanoparticle Tracking Analysis, Nanomedicine, vol.7, issue.6, p.45, 2011.

L. N. Sybren and . Maas, Quantification and Size-profiling of Extracellular Vesicles Using Tunable Resistive Pulse Sensing, Journal of Visualized Experiments : JoVE, issue.92, p.45, 2014.

S. Robert, Standardization of platelet-derived microparticle counting using calibrated beads and a Cytomics FC500 routine flow cytometer: A first step towards multicenter studies?, Journal of Thrombosis and Haemostasis, vol.7, issue.1, p.45, 2009.

M. Zhang, Methods and Technologies for Exosome Isolation and Characterization, Small Methods, vol.0, issue.0

F. Yang, Exosome separation using microfluidic systems: Size-based, immunoaffinity-based and dynamic methodologies, Biotechnology Journal, vol.46, p.53, 2017.

L. M. Déborah and . Rupert, Methods for the physical characterization and quantification of extracellular vesicles in biological samples, Biochimica et Biophysica Acta (BBA)-General Subjects, vol.46, issue.1, pp.3164-3179, 2017.

S. Gholizadeh, Microfluidic Approaches for Isolation, Detection, and Characterization of Extracellular Vesicles: Current Status and Future Directions, Biosensors and Bioelectronics, vol.46, 2017.

. Zamila-khatun, Elucidating diversity of exosomes: Biophysical and molecular characterization methods, Nanomedicine, vol.11, issue.17, pp.2359-2377, 2016.

J. Ko, Detection and isolation of circulating exosomes and microvesicles for cancer monitoring and diagnostics using micro-/nano-based devices, The Analyst, vol.141, issue.2, pp.450-460, 2016.

V. Sunkara, Emerging techniques in the isolation and characterization of extracellular vesicles and their roles in cancer diagnostics and prognostics, The Analyst, vol.141, issue.2, p.46, 2016.

R. Szatanek, Isolation of extracellular vesicles: Determining the correct approach (Review), International Journal of Molecular Medicine, vol.36, issue.1, pp.11-17, 2015.

P. Miko?aj and . Zaborowski, Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study, BioScience, vol.65, issue.8, p.48, 2015.

. Fatemeh-momen-heravi, Current methods for the isolation of extracellular vesicles, Biological Chemistry, vol.394, issue.10, p.46, 2013.

M. Maeki, Advances in microfluidics for lipid nanoparticles and extracellular vesicles and applications in drug delivery systems, Advanced Drug Delivery Reviews, vol.46, 2018.

E. Pariset, Vesicles: Isolation Methods. Advanced Biosystems, vol.1, issue.5, p.47, 2017.

. Fatemeh-momen-heravi, Impact of Biofluid Viscosity on Size and Sedimentation Efficiency of the Isolated Microvesicles, Frontiers in Physiology, vol.3, p.48, 2012.

A. Mikhail and . Livshts, Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol, Scientific Reports, vol.5, p.48, 2015.

J. Caradec, Reproducibility and efficiency of serum-derived exosome extraction methods, Clinical Biochemistry, vol.47, p.49, 2014.

R. Cantin, Discrimination between exosomes and HIV-1: Purification of both vesicles from cell-free supernatants, Journal of Immunological Methods, vol.338, issue.1-2, p.49, 2008.

Y. Yuana, Co-isolation of extracellular vesicles and high-density lipoproteins using density gradient ultracentrifugation, Journal of Extracellular Vesicles, vol.3, issue.0, p.49, 2014.

I. Vanni, Exosomes: A new horizon in lung cancer, Drug Discovery Today, vol.22, issue.6, p.49, 2017.

. Douglasd and . Taylor, Exosome Isolation for Proteomic Analyses and RNA Profiling, Serum/Plasma Proteomics, p.50, 2011.

J. Van-deun, The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling, Journal of Extracellular Vesicles, vol.3, issue.0, p.50, 2014.

T. Yamada, Comparison of Methods for Isolating Exosomes from Bovine Milk, Journal of Veterinary Medical Science, vol.74, issue.11, p.50, 2012.

I. Helwa, A Comparative Study of Serum Exosome Isolation Using Differential Ultracentrifugation and Three Commercial Reagents, PLOS ONE, vol.12, issue.1, p.50, 2017.

X. Gallart-palau, Extracellular vesicles are rapidly purified from human plasma by PRotein Organic Solvent PRecipitation (PROSPR), Scientific Reports, vol.5, p.50, 2015.
DOI : 10.1038/srep14664

URL : https://www.nature.com/articles/srep14664.pdf

J. Bow and . Tauro, Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes, Methods, vol.56, issue.2, p.50, 2012.

J. Stephen and . Gould, As we wait: Coping with an imperfect nomenclature for extracellular vesicles, Journal of Extracellular Vesicles, vol.2, issue.0, p.50, 2013.

C. Chen, Microfluidic isolation and transcriptome analysis of serum microvesicles, Lab on a chip, vol.10, issue.4, p.53, 2010.
DOI : 10.1039/b916199f

URL : http://europepmc.org/articles/pmc3136803?pdf=render

C. Chen, Paper-based immunoaffinity devices for accessible isolation and characterization of extracellular vesicles, Microfluidics and Nanofluidics, vol.16, issue.5, p.51, 2014.

L. Balaj, Heparin affinity purification of extracellular vesicles, Scientific Reports, vol.5, p.10266, 1951.

A. Ghosh, Rapid Isolation of Extracellular Vesicles from Cell Culture and Biological Fluids Using a Synthetic Peptide with Specific Affinity for Heat Shock Proteins, PLOS ONE, vol.9, issue.10, p.51, 2014.

C. Jaco and . Knol, Peptide-mediated 'miniprep' isolation of extracellular vesicles is suitable for high-throughput proteomics, EuPA Open Proteomics, vol.11, p.51, 2016.

S. Cho, Isolation of extracellular vesicle from blood plasma using electrophoretic migration through porous membrane, Sensors and Actuators, B: Chemical, vol.233, p.52, 2016.

R. T. Davies, Microfluidic filtration system to isolate extracellular vesicles from blood, Lab on a Chip, vol.12, issue.24, p.51, 2012.

N. Heath, Rapid isolation and enrichment of extracellular vesicle preparations using anion exchange chromatography, Scientific Reports, vol.8, issue.1, p.52, 2018.

L. Muller, Isolation of Biologically-Active Exosomes from Human Plasma, Journal of immunological methods, vol.411, p.52, 2014.

M. L. Alvarez, Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers, Kidney International, vol.82, issue.9, p.52, 2012.

R. Lai, Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury, Stem Cell Research, vol.4, issue.3, p.52, 2010.
DOI : 10.1016/j.scr.2009.12.003

URL : https://doi.org/10.1016/j.scr.2009.12.003

L. Michael and . Merchant, Microfiltration isolation of human urinary exosomes for characterization by MS, PROTEOMICS-Clinical Applications, vol.4, issue.1, p.52, 2010.

A. Gámez-valero, Size-Exclusion Chromatography-based isolation minimally alters Extracellular Vesicles' characteristics compared to precipitating agents, Scientific Reports, vol.6, p.52, 2016.

A. Hashemi-gheinani, Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery, Scientific Reports, vol.8, issue.1, p.52, 2018.

M. Sáenz-cuesta, Methods for Extracellular Vesicles Isolation in a Hospital Setting, Frontiers in Immunology, vol.6, p.53, 2015.

D. Issadore, Miniature magnetic resonance system for point-of-care diagnostics, Lab on a Chip, vol.11, issue.13, p.54, 2011.

J. Rho, Magnetic Nanosensor for Detection and Profiling of Erythrocyte-Derived Microvesicles, ACS nano, vol.7, issue.12, p.54, 2013.

Z. Zhao, A microfluidic ExoSearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis, Lab on a Chip, vol.16, issue.3, p.54, 2016.

S. Jeong, Integrated Magneto-Electrochemical Sensor for Exosome Analysis, ACS nano, vol.10, issue.2, pp.1802-1809, 1954.

S. Jaideep and . Dudani, Rapid inertial solution exchange for enrichment and flow cytometric detection of microvesicles, Biomicrofluidics, vol.9, issue.1, p.54, 2015.

H. Bruus, Acoustofluidics 7: The acoustic radiation force on small particles, Lab on a Chip, vol.12, issue.6, p.55, 2012.

K. Lee, Acoustic Purification of Extracellular Microvesicles, ACS Nano, vol.9, issue.3, p.55, 2015.

M. Wu, Isolation of exosomes from whole blood by integrating acoustics and microfluidics, Proceedings of the National Academy of Sciences, p.56, 2017.

S. Sitar, Size Characterization and Quantification of Exosomes by Asymmetrical-Flow Field-Flow Fractionation, Analytical Chemistry, vol.87, issue.18, p.56, 2015.

D. Müller, Nanoparticle separation with a miniaturized asymmetrical flow field-flow fractionation cartridge, Frontiers in Chemistry, vol.3, p.57, 2015.

L. Huang, Continuous particle separation through deterministic lateral displacement, Science, vol.304, issue.5673, p.62, 2004.
DOI : 10.1126/science.1094567

H. Benjamin and . Wunsch, Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm, Nature Nanotechnology, vol.57, p.178, 2016.

J. Mcgrath, Deterministic lateral displacement for particle separation: A review, vol.14, p.57, 2014.

D. W. Inglis, Critical particle size for fractionation by deterministic lateral displacement, Lab on a Chip, vol.6, issue.5, p.187, 2006.

D. W. Inglis, Critical particle size for fractionation by deterministic lateral displacement, vol.6, p.60, 2006.

D. John-alan, Microfluidic Separation of Blood Components through Deterministic Lateral Displacement. Dissertation presented to the faculty of Princeton University in Candidacy for the Degree of Doctor of Philosophy, vol.63, p.195, 2008.

T. Kulrattanarak, Analysis of mixed motion in deterministic ratchets via experiment and particle simulation, Microfluidics and Nanofluidics, vol.10, issue.4, p.121, 1960.

B. R. Long, Multidirectional sorting modes in deterministic lateral displacement devices, Physical Review E (Statistical, Nonlinear, and Soft Matter Physics), vol.78, issue.4, p.61, 2008.

B. M. Dincau, Deterministic lateral displacement (DLD) in the high Reynolds number regime: High-throughput and dynamic separation characteristics, Microfluidics and Nanofluidics, vol.22, issue.6, p.62, 2018.

Y. S. Lubbersen, Suspension separation with deterministic ratchets at moderate Reynolds numbers, Chemical Engineering Science, vol.73, p.108, 2012.

. Gaetano-d'avino, Non-Newtonian deterministic lateral displacement separator: Theory and simulations, Rheologica Acta, vol.52, issue.3, p.62, 2013.

M. Heller, A theoretical analysis of the resolution due to diffusion and size dispersion of particles in deterministic lateral displacement devices, Journal of Micromechanics and Microengineering, vol.18, issue.7, p.62, 2008.

M. Heller, A theoretical analysis of the resolution due to diffusion and size-dispersion of particles in deterministic lateral displacement devices, Journal of Micromechanics and Microengineering, vol.18, issue.7, p.62, 2008.

S. Cerbelli, Separation of polydisperse particle mixtures by deterministic lateral displacement. The impact of particle diffusivity on separation efficiency, Asia-Pacific Journal of Chemical Engineering, vol.7, p.63, 2012.

P. Christopher and . Cheng, Quantification of Wall Shear Stress in Large Blood Vessels Using Lagrangian Interpolation Functions with Cine Phase-Contrast Magnetic Resonance Imaging, Annals of Biomedical Engineering, vol.30, issue.8, p.63, 2002.

K. Kaushik and . Rangharajan, Effect of Microstructure Geometric Form on Surface Shear Stress, Journal of Fluids Engineering, vol.139, issue.1, p.63, 2016.

A. Dmitry and . Fedosov, Deformation and dynamics of red blood cells in flow through cylindrical microchannels, vol.10, p.63, 2014.

J. Dupire, Full dynamics of a red blood cell in shear flow, Proceedings of the National Academy of Sciences, vol.109, issue.51, p.63, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01957799

R. Quek, Separation of deformable particles in deterministic lateral displacement devices, Physical Review E, vol.83, issue.5, 1964.

C. S. Peskin, The immersed boundary method, Acta Numerica, vol.11, pp.479-517, 1964.

D. Krueger, Deformability-based red blood cell separation in deterministic lateral displacement devices-A simulation study, Biomicrofluidics, vol.8, issue.5, p.65, 2014.

Z. Zhang, Behavior of rigid and deformable particles in deterministic lateral displacement devices with different post shapes, The Journal of Chemical Physics, vol.143, issue.24, p.68, 2015.

G. Kabacaoglu, Optimal design of deterministic lateral displacement device for viscosity contrast based cell sorting, p.66, 2018.

X. Fu, Numerical investigation of polygonal particle separation in microfluidic channels, Microfluidics and Nanofluidics, vol.20, issue.7, p.66, 2016.

. Shashi-ranjan, DLD pillar shape design for efficient separation of spherical and non-spherical bioparticles, Lab on a Chip, vol.14, issue.21, p.91, 1966.

H. Sam and . Au, Microfluidic Isolation of Circulating Tumor Cell Clusters by Size and Asymmetry, Scientific Reports, vol.7, issue.1, p.178, 1966.

J. Wei, Numerical Study of Pillar Shapes in Deterministic Lateral Displacement Microfluidic Arrays for Spherical Particle Separation, IEEE transactions on nanobioscience, vol.14, issue.6, p.68, 2015.

K. Loutherback, Deterministic Microfluidic Ratchet, Physical Review Letters, vol.102, issue.4, p.68, 2009.

K. Loutherback, Improved performance of deterministic lateral displacement arrays with triangular posts, Microfluidics and Nanofluidics, vol.9, issue.6, p.88, 1968.

J. Hyun, Improved pillar shape for deterministic lateral displacement separation method to maintain separation efficiency over a long period of time, Separation and Purification Technology, vol.172, p.70, 2017.

M. Al-fandi, New design for the separation of microorganisms using microfluidic deterministic lateral displacement. Robotics and Computer-Integrated Manufacturing, vol.27, p.70, 2011.

J. P. Dijkshoorn, Reducing the critical particle diameter in (highly) asymmetric sieve-based lateral displacement devices, Scientific Reports, vol.7, issue.1, p.70, 2017.

S. Du, Liquid-based stationary phase for deterministic lateral displacement separation in microfluidics, Soft Matter, p.70, 2017.

S. Feng, Maximizing particle concentration in deterministic lateral displacement arrays, Biomicrofluidics, vol.11, issue.2, p.97, 2017.

J. P. Beech, Tipping the balance of deterministic lateral displacement devices using dielectrophoresis, Lab on a Chip, vol.9, issue.18, p.70, 2009.

. Kerwin-kwek-zeming, Real-time modulated nanoparticle separation with an ultra-large dynamic range, vol.16, p.178, 2015.

T. J. Bowman, Inertia and scaling in deterministic lateral displacement, Biomicrofluidics, vol.7, issue.6, 1970.

S. Du, Gravity driven deterministic lateral displacement for suspended particles in a 3D obstacle array, Scientific Reports, vol.6, p.70, 2016.

D. J. Collins, Particle separation using virtual deterministic lateral displacement (vDLD), Lab on a Chip, vol.14, issue.9, p.70, 2014.

J. A. Davis, Deterministic hydrodynamics: Taking blood apart, Proceedings of the National Academy of Sciences of the United States of America, vol.103, pp.14779-14784, 1971.

D. W. Inglis, Microfluidic device for label-free measurement of platelet activation, vol.8, p.178, 2008.

D. W. Inglis, Scaling deterministic lateral displacement arrays for high throughput and dilution-free enrichment of leukocytes, Journal of Micromechanics and Microengineering, vol.21, issue.5, p.71, 2011.

J. P. Beech, Sorting cells by size, shape and deformability, vol.12, pp.1048-1051, 1971.
DOI : 10.1039/c2lc21083e

URL : http://lup.lub.lu.se/search/ws/files/1650408/2369057.pdf

. Kerwin-kwek-zeming, Rotational separation of non-spherical bioparticles using I-shaped pillar arrays in a microfluidic device, Nature Communications, vol.4, p.1625, 1971.

D. Holmes, Separation of blood cells with differing deformability using deterministic lateral displacement, Interface Focus, vol.4, issue.6, p.178, 2014.

J. Choi, On-chip Extraction of Intracellular Molecules in White, Blood Cells from Whole Blood. Scientific Reports, vol.5, p.15167, 1971.

C. I. Civin, Automated leukocyte processing by microfluidic deterministic lateral displacement, Cytometry Part A, vol.89, issue.12, pp.1073-1083, 1971.
DOI : 10.1002/cyto.a.23019

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/cyto.a.23019

E. Henry, Sorting cells by their dynamical properties, Scientific Reports, vol.6, p.34375, 1971.
DOI : 10.1038/srep34375

URL : https://www.nature.com/articles/srep34375.pdf

. Kerwin-kwek-zeming, Asymmetrical Deterministic Lateral Displacement Gaps for Dual Functions of Enhanced Separation and Throughput of Red Blood Cells, Scientific Reports, vol.6, p.91, 1971.

B. Kim, Deterministic Migration-Based Separation of White Blood Cells, Small, vol.12, issue.37, pp.5159-5168, 1971.

M. Yamada, Slanted, asymmetric microfluidic lattices as size-selective sieves for continuous particle/cell sorting, Lab on a Chip, vol.17, issue.2, pp.304-314, 1971.

T. Jing, Single Cell Analysis of Leukocyte Protease Activity Using Integrated ContinuousFlow Microfluidics, Analytical Chemistry, vol.88, issue.23, pp.11750-11757, 1971.

R. Campos-gonzález, Deterministic Lateral Displacement: The Next-Generation CAR T-Cell Processing?, p.71, 2018.

J. V. Green, Deterministic Lateral Displacement as a Means to Enrich Large Cells for Tissue Engineering, Analytical Chemistry, vol.81, issue.21, pp.9178-9182, 1972.

K. Loutherback, Deterministic separation of cancer cells from blood at 10 mL/min, AIP Advances, vol.2, issue.4, p.42107, 1972.

Z. Liu, Rapid isolation of cancer cells using microfluidic deterministic lateral displacement structure, Biomicrofluidics, vol.7, issue.1, p.11801, 1972.

H. Okano, Enrichment of circulating tumor cells in tumor-bearing mouse blood by a deterministic lateral displacement microfluidic device, Biomedical Microdevices, vol.17, issue.3, p.59, 1972.

N. Murat-karabacak, marker-free isolation of circulating tumor cells from blood samples, Nature Protocols, vol.9, issue.3, pp.694-710, 1972.

F. Fachin, Monolithic Chip for High-throughput Blood Cell Depletion to Sort Rare Circulating Tumor Cells, Scientific Reports, vol.7, 1972.

X. Jiang, Microfluidic isolation of platelet-covered circulating tumor cells, 1972.

H. Stefan and . Holm, Separation of parasites from human blood using deterministic lateral displacement, Lab on a Chip, vol.11, issue.7, pp.1326-1332, 1972.

D. W. Inglis, Highly accurate deterministic lateral displacement device and its application to purification of fungal spores, Biomicrofluidics, vol.4, issue.2, p.72, 2010.

J. András and . Laki, Separation of Microvesicles from Serological Samples Using Deterministic Lateral Displacement Effect. BioNanoScience, vol.5, issue.1, pp.48-54, 1972.

J. P. Beech, Separation of pathogenic bacteria by chain length, Analytica Chimica Acta, vol.1000, pp.223-231, 1972.

T. Bowman, Force driven separation of drops by deterministic lateral displacement, Lab on a Chip, vol.12, issue.16, p.2903

N. Tottori, Separation of main and satellite droplets in a deterministic lateral displacement microfluidic device, vol.7, p.72, 2017.

. Kerwin-kwek-zeming, Fluorescent label-free quantitative detection of nano-sized bioparticles using a pillar array, Nature Communications, vol.9, issue.1, p.119, 2018.

H. Benjamin and . Wunsch, Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm, Nature Nanotechnology, vol.11, issue.11, pp.936-940, 1972.

P. Schwartz, A second-order accurate method for solving the signed distance function equation, Communications in Applied Mathematics and Computational Science, vol.5, issue.1, p.76, 2010.

Z. Zhang, Behavior of rigid and deformable particles in deterministic lateral displacement devices with different post shapes, The Journal of Chemical Physics, vol.143, issue.24, p.79, 2015.

R. Vernekar, Anisotropic permeability in deterministic lateral displacement arrays, vol.17, p.133, 1979.
DOI : 10.1039/c7lc00785j

URL : https://pubs.rsc.org/en/content/articlepdf/2017/lc/c7lc00785j

E. Pariset, Anticipating Cutoff Diameters in Deterministic Lateral Displacement (DLD) Microfluidic Devices for an Optimized Particle Separation, Small, vol.13, issue.37, p.121, 2017.

D. W. Inglis, Efficient microfluidic particle separation arrays, Applied Physics Letters, vol.94, issue.1, p.117, 2009.
DOI : 10.1063/1.3068750

T. Kulrattanarak, Mixed motion in deterministic ratchets due to anisotropic permeability, Journal of Colloid and Interface Science, vol.354, issue.1, pp.7-14, 0121.

J. Cohen, Sepsis: A roadmap for future research. The Lancet Infectious Diseases, vol.15, pp.581-614, 0138.
DOI : 10.1016/s1473-3099(15)70112-x

P. Zhang, Characterization of Spherulites as a Lipidic Carrier for Low and High Molecular Weight Agents, Pharmaceutical research, vol.30, issue.6, p.152, 2013.

T. Baranyai, Isolation of Exosomes from Blood Plasma: Qualitative and Quantitative Comparison of Ultracentrifugation and Size Exclusion Chromatography Methods, PLoS ONE, vol.10, issue.12, p.159, 2015.

A. N. Böing, Single-step isolation of extracellular vesicles by size-exclusion chromatography, Journal of Extracellular Vesicles, vol.3, p.159, 2014.

J. Lötvall, Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles, Journal of Extracellular Vesicles, vol.3, p.160, 2014.

M. Kotmakci, Exosome Isolation: Is There an Optimal Method with Regard to Diagnosis or Treatment?, p.162, 2017.

N. Murat-karabacak, marker-free isolation of circulating tumor cells from blood samples, Nature Protocols, vol.9, issue.3, p.182, 0167.

H. Stefan and . Holm, Separation of parasites from human blood using deterministic lateral displacement, Lab on a Chip, vol.11, issue.7, p.178, 2011.

J. A. Davis, Deterministic hydrodynamics: Taking blood apart, Proceedings of the National Academy of Sciences, vol.103, issue.40, pp.14779-14784, 0178.
DOI : 10.1073/pnas.0605967103

URL : http://www.pnas.org/content/103/40/14779.full.pdf

J. András and . Laki, Separation of Microvesicles from Serological Samples Using Deterministic Lateral Displacement Effect. BioNanoScience, vol.5, issue.1, p.178, 2014.

X. Li, Study on the mechanism of droplet formation in T-junction microchannel, Chemical Engineering Science, vol.69, issue.1, p.182, 2012.

Y. Leslie and . Yeo, Microfluidic Devices for Bioapplications, Small, vol.7, issue.1, p.182, 2011.

D. Velasco, Microfluidic Encapsulation of Cells in Polymer Microgels, Small, vol.8, issue.11, p.182, 2012.

D. Kang, Droplet microfluidics for single-molecule and single-cell analysis in cancer research, diagnosis and therapy, TrAC Trends in Analytical Chemistry, vol.58, p.182, 2014.

S. Mashaghi, Droplet microfluidics: A tool for biology, chemistry and nanotechnology, TrAC Trends in Analytical Chemistry, vol.82, p.182, 2016.
DOI : 10.1016/j.trac.2016.05.019

URL : http://ro.uow.edu.au/cgi/viewcontent.cgi?article=5004&context=smhpapers

M. Zagnoni, Chapter 2-Droplet Microfluidics for High-throughput Analysis of Cells and Particles, Cell Biology, vol.102, p.182, 2011.

Y. Evgenia and . Basova, Droplet microfluidics in (bio)chemical analysis, Analyst, vol.140, issue.1, p.182, 2014.

M. Godin, Measuring the mass, density, and size of particles and cells using a suspended microchannel resonator, Applied Physics Letters, vol.91, issue.12, p.182, 2007.

B. Moon, Water-in-Water Droplets by Passive Microfluidic Flow Focusing, Analytical Chemistry, vol.88, issue.7, p.182, 2016.
DOI : 10.1021/acs.analchem.6b00225

Y. Yan, Numerical simulation of junction point pressure during droplet formation in a microfluidic T-junction, Chemical Engineering Science, vol.84, p.183, 2012.

P. Garstecki, Formation of droplets and bubbles in a microfluidic T-junction-scaling and mechanism of break-up, Lab on a Chip, vol.6, issue.3, p.189, 2006.

S. Zhang, Prediction of sizes and frequencies of nanoliter-sized droplets in cylindrical T-junction microfluidics, Chemical Engineering Science, vol.138, p.189, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01278233

A. Gupta, Droplet formation and stability of flows in a microfluidic T-junction, Applied Physics Letters, vol.94, issue.16, p.164107, 0192.

F. Pineda, Using Electrofluidic Devices As Hyper-elastic Strain Sensors, Microelectron. Eng, vol.144, issue.C, pp.27-31, 0193.
DOI : 10.1016/j.mee.2015.02.013

C. Parent, Quantitative biological assays with on-chip calibration using versatile architecture and collapsible chambers, Sensors and Actuators B: Chemical, vol.261, pp.106-114, 0195.
DOI : 10.1016/j.snb.2018.01.111

URL : https://hal.archives-ouvertes.fr/hal-01756825

, ? Polyethyleneglycol-trimethylsulfoxide

E. Pariset, C. Parent, Y. Fouillet, F. Boizot, N. Verplanck et al., Separation of Biological Particles in a Modular Platform of Cascaded Deterministic Lateral Displacement Modules, Scientific Reports, vol.8, p.17762, 2018.

E. Pariset, C. Pudda, F. Boizot, N. Verplanck, F. Revol-cavalier et al., Purification of complex samples: implementation of a modular and reconfigurable droplet-based microfluidic platform with cascaded deterministic lateral displacement separation modules, PLOS ONE, vol.13, issue.5, pp.1-18, 2018.

E. Pariset, C. Pudda, F. Boizot, N. Verplanck, J. Berthier et al., Anticipating Cutoff Diameters in Deterministic Lateral Displacement (DLD) Microfluidic Devices for an Optimized Particle Separation, vol.13, p.37, 2017.

E. Pariset, V. Agache, and A. Millet, Extracellular Vesicles: Isolation Methods, Advanced Biosystems, vol.1, pp.1-12, 2017.

. Patents-n-o-e.n-;-e, J. Pariset, A. Berthier, V. Thuaire, and . Agache, Système de tri de particules par gamme de tailles, pp.18-52121, 2018.

N. , E. N-;-e, C. Pariset, F. Parent, Y. Boizot et al., Dispositif d'injection d'un échantillon fluidique, p.61263, 2017.

N. , E. N-;-e, F. Pariset, V. Revol-cavalier, and . Agache, Equipement de tri de particules présentes dans un échantillon fluidique, pp.18-52121, 2016.

E. Pariset, F. Revol-cavalier, F. Boizot, C. Pudda, A. Thuaire et al., Deterministic Lateral Displacement: Finite element modeling and experimental validation for particle trajectory and separation, Paris. Oral Presentation E. Pariset, J. Berthier, F. Revol-Cavalier, vol.17, 2017.

E. Pariset, D. Gosselin, V. Agache, J. Berthier, ;. Gdr-micro-et-nanofluidique et al., Modeling the steric effect in COMSOL for Deterministic Lateral Displacement based separation, Poster Presentation Awards L'Oréal-UNESCO French Fellowship for Women in Science, 2015.

, Travel Grant for the 20 th International Conference on Miniaturized Systems for Chemistry and Life Sciences, 2017.