, , p.32

C. Bonnans, J. Chou, and Z. Werb, Remodelling the extracellular matrix in development and disease, Nature reviews. Molecular cell biology 15, vol.12, p.40, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01952416

B. A. Bonsing, Allelotype analysis of flow-sorted breast cancer cells demonstrates genetically related diploid and aneuploid subpopulations in primary tumors and lymph node metastases". en, Genes, Chromosomes and Cancer 28, vol.2, pp.1098-2264, 2000.

,

/. %28sici-%,

, , p.40

V. Bronte, Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards". eng, Nature Communications, vol.7, p.71, 2016.

B. Brücher, I. S. Ldm, and . Jamall, Epistemology of the origin of cancer: a new paradigm, BMC cancer, vol.14, p.35, 2014.

M. Cella, Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon, 1999.

, Nature Medicine 5, vol.8, p.50

D. S. Chen and I. Mellman, Oncology meets immunology: the cancerimmunity cycle". eng, Immunity 39.1, p.43, 2013.

L. Chen, Inflammatory responses and inflammation-associated diseases in organs, Oncotarget 9, vol.6, pp.1949-2553, 2017.

A. Ciaccio, T. Laura, and . Bormann, The Sapir-Whorf hypothesis in the domain of colors: evidence from healthy and aphasic speakers". en, Rivista Italiana di Filosofia del Linguaggio, vol.3, pp.1-14, 2013.

L. Cohn, Antigen delivery to early endosomes eliminates the superiority of human blood BDCA3+ dendritic cells at cross presentation". eng, The Journal of Experimental Medicine, vol.210, p.50, 2013.

M. Collin and V. Bigley, Human dendritic cell subsets: an update, Immunology 154, vol.1, pp.19-2805, 2018.

M. Collin, N. Mcgovern, and M. Haniffa, Human dendritic cell subsets, Immunology 140.1, vol.167, pp.19-2805, 2013.

M. Colonna, G. Trinchieri, and Y. Liu, Plasmacytoid dendritic cells in immunity, Nature Immunology 5, vol.12, p.50, 2004.

A. Conesa, A survey of best practices for RNA-seq data analysis". eng, Genome Biology, vol.17, p.62, 2016.

J. Cortés, Pertuzumab Monotherapy After Trastuzumab-Based Treatment and Subsequent Reintroduction of Trastuzumab: Activity and Tolerability in Patients With Advanced Human Epidermal Growth Factor Receptor 2Positive Breast Cancer". en, Journal of Clinical Oncology, vol.30, p.39, 2012.

D. Croft, Reactome: a database of reactions, pathways and biological processes". eng, Nucleic Acids Research, vol.39, p.62, 2011.

C. Curtis, The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups, Nature 486.7403, pp.346-352, 2012.

M. Dalod, Dendritic cell maturation: functional specialization through signaling specificity and transcriptional programming, The EMBO Journal, vol.33, pp.261-4189, 2014.

, , p.53, 201488027.

G. Danaei, Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors". eng, Lancet, p.36, 2005.

D. G. Denardo, P. Andreu, and L. M. Coussens, Interactions between lymphocytes and myeloid cells regulate proversus anti-tumor immunity, eng. In: Cancer Metastasis Reviews, vol.29, p.35, 2010.

D. G. Denardo, B. Jairo, and . Barreto, CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages". eng, Cancer Cell, vol.16, issue.2, pp.1878-3686, 2009.

D. G. Denardo, J. Donal, and . Brennan, Leukocyte Complexity Predicts Breast Cancer Survival and Functionally Regulates Response to Chemotherapy". en, Cancer Discovery. issn, p.43, 2011.

G. Dennis, DAVID: Database for Annotation, Visualization, and Integrated Discovery, Genome Biology 4.9, p.62, 2003.

M. S. Diamond, Type I interferon is selectively required by dendritic cells for immune rejection of tumors, The Journal of experimental medicine 208, vol.10, p.170, 2011.

M. Dillies, A comprehensive evaluation of normalization methods for Illumina high-throughput RNA sequencing data analysis, Briefings in Bioinformatics, vol.14, p.60, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01521274

A. Dobin, STAR: ultrafast universal RNA-seq aligner". en, Bioinformatics 29.1, p.60, 2013.

M. R. Doherty, Interferon, p.170, 2017.

A. M. Dudek, Immature, Semi-Mature, and Fully Mature Dendritic Cells: Toward a DC-Cancer Cells Interface That Augments Anticancer Immunity". eng, Frontiers in Immunology, vol.4, p.173, 2013.

G. P. Dunn, T. Allen, and . Bruce, Cancer immunoediting: from immunosurveillance to tumor escape, Nature Immunology, vol.3, p.42, 2002.

G. P. Dunn, J. Lloyd, R. D. Old, and . Schreiber, The immunobiology of cancer immunosurveillance and immunoediting". eng, Immunity 21, vol.2, p.35, 2004.

G. P. Dunn, A critical function for type I interferons in cancer immunoediting, Nature immunology 6.7, p.170, 2005.

S. Dushyanthen, Relevance of tumor-infiltrating lymphocytes in breast cancer". en, BMC Medicine 13.1. issn, p.39, 2015.

J. Faget, ICOS-ligand expression on plasmacytoid dendritic cells supports breast cancer progression by promoting the accumulation of immunosuppressive CD4+ T cells". eng, Cancer Research 72, vol.23, pp.6130-6141, 2012.

G. Ferlazzo and B. Morandi, Cross-Talks between Natural Killer Cells and Distinct Subsets of Dendritic Cells, 2014.

, Frontiers in Immunology, vol.5, p.32

M. Fuertes, Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells, J Exp Med, vol.208, p.170, 2011.

F. Fuhrmann, Adequate immune response ensured by binary IL-2 and graded CD25 expression in a murine transfer model, p.32, 2018.

D. I. Gabrilovich and S. Nagaraj, Myeloid-derived suppressor cells as regulators of the immune system, Nature Reviews. Immunology, vol.9, p.43, 2009.

T. F. Gajewski, H. Schreiber, and Y. Fu, Innate and adaptive immune cells in the tumor microenvironment". eng, Nature Immunology, vol.14, p.169, 2013.

P. García-teijido, Tumor-Infiltrating Lymphocytes in Triple Negative Breast Cancer: The Future of Immune Targeting". eng, Clinical Medicine Insights. Oncology, vol.10, p.39, 2016.

A. Gardner and B. Ruffell, Dendritic Cells and Cancer Immunity, Trends in Immunology, vol.37, pp.1471-4906, 2016.

,

C. Ghirelli, Breast Cancer Cell-Derived GM-CSF Licenses Regulatory Th2 Induction by Plasmacytoid Predendritic Cells in Aggressive Disease Subtypes, 2015.

, Cancer Research 75, vol.14, p.71

F. Ginhoux and M. Guilliams, Tissue-Resident Macrophage Ontogeny and Homeostasis". eng, pp.439-449, 2016.

, , vol.56, p.34

F. Ginhoux and S. Jung, Monocytes and macrophages: developmental pathways and tissue homeostasis, 2014.

, Nature Reviews. Immunology, vol.14, p.45

S. Gordon, Alternative activation of macrophages". eng, Nature Reviews, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00474829

, , p.46

D. Gosselin, Environment drives selection and function of enhancers controlling tissue-specific macrophage identities". eng, Cell 159, vol.6, p.34, 2014.

D. J. Gough, Constitutive type I interferon modulates homeostatic balance through tonic signaling, p.170, 2012.

M. Grandclaudon, Th17 pathway and prognosis in Triple Negative breast cancer, p.31

T. Granot, Dendritic Cells Display Subset and Tissue-Specific Maturation Dynamics over, Human Life". eng. In: Immunity, vol.46, p.50, 2017.

M. Greaves and C. C. Maley, , 2012.

, Clonal evolution in cancer, Nature 481, vol.7381, p.39

M. Griffith, Alternative expression analysis by RNA sequencing". eng, Nature Methods 7.10, p.171, 2010.

M. Kanehisa and S. Goto, KEGG: Kyoto Encyclopedia of Genes and Genomes, Nucleic Acids Research, vol.28, p.62, 2000.

M. King, Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2". eng, Science, vol.302, p.36, 2003.

D. C. Koboldt, Comprehensive molecular portraits of human breast tumours". en. In: Nature 490, vol.7418, p.37, 2012.

S. Koren and M. Bentires-alj, Breast Tumor Heterogeneity: Source of Fitness, Hurdle for Therapy, Molecular Cell, vol.60, p.38, 2015.

L. Mcnally, S. P. Brown, and A. L. Jackson, Cooperation and the evolution of intelligence". en, Proc, 2012.

R. Soc, , p.27

C. E. Meacham and S. J. Morrison, Tumour heterogeneity and cancer cell plasticity". eng. In: Nature 501, vol.7467, p.39, 2013.

R. Medzhitov, Origin and physiological roles of inflammation". eng, p.34, 2008.

A. Mildner and S. Jung, Development and function of dendritic cell subsets". eng, Immunity 40.5, p.167, 2014.

J. C. Miller, Deciphering the transcriptional network of the DC lineage, Nature immunology, vol.13, p.168, 2012.

J. C. Mills, DNA microarrays and beyond: completing the journey from tissue to cell, Nature Cell Biology, vol.3, p.57, 2001.

M. Mojic, K. Takeda, and Y. Hayakawa, The Dark Side of IFN-gamma: Its Role in Promoting Cancer Immunoevasion, International journal of molecular sciences 19.1. issn, p.171, 2017.

J. Mora, U. H. Rodrigo, and . Von-andrian, T-cell homing specificity and plasticity: new concepts and future challenges, 2006.

. English, Trends in Immunology, vol.27, issue.06, p.33

J. Mora, M. R. Rodrigo, and . Bono, Selective imprinting of gut-homing T cells by Peyer's patch dendritic cells". eng. In: Nature 424, vol.6944, p.168, 2003.

A. Mortazavi, Mapping and quantifying mammalian transcriptomes by RNA-Seq". eng, Nature Methods 5, vol.7, p.60, 2008.

H. Mukai, Treatment strategy for HER2-positive breast cancer". eng, International Journal of Clinical Oncology, vol.15, p.39, 2010.

H. Nakano, Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses". eng, Nature Immunology, vol.10, p.52, 2009.

A. M. Newman, Robust enumeration of cell subsets from tissue expression profiles, In: Nature methods, vol.12, pp.1-10, 2014.

,

%. 7b-%-5c-%-%7d5cnhttp, , p.57

M. E. Newman, The structure and function of complex networks, SIAM Review 45, vol.2, p.31, 2003.

M. J. Newport, A mutation in the interferon-gamma-receptor gene and susceptibility to mycobacterial infection". eng, The New England Journal of Medicine, vol.335, p.29, 1996.

G. V. Niel, D. Gisela, G. Angelo, and . Raposo, Shedding light on the cell biology of extracellular vesicles, 2018.

, Nature Reviews. Molecular Cell Biology, vol.19, p.26

K. J. Niklas, A. Stuart, and . Newman, , 2013.

, The origins of multicellular organisms, Evolution & Development, vol.15, p.28

G. Nizzoli, Human CD1c+ dendritic cells secrete high levels of IL-12 and potently prime cytotoxic T-cell responses". eng, Blood 122, vol.6, p.50, 2013.

U. O'doherty, Human blood contains two subsets of dendritic cells, one immunologically mature and the other immature". eng, pp.19-2805, 1994.

L. S. Ojalvo and W. King, High-density gene expression analysis of tumor-associated macrophages from mouse mammary tumors". eng, The American Journal of Pathology, vol.174, p.62, 2009.

L. S. Ojalvo, A. Charles, and . Whittaker, Gene Expression Analysis of Macrophages That Facilitate Tumor Invasion Supports a Role for Wnt-Signaling in Mediating Their Activity in Primary Mammary Tumors". en, The Journal of Immunology, vol.184, pp.1550-6606, 2010.

Y. Okabe and R. Medzhitov, Tissue biology perspective on macrophages, Nature Immunology, vol.17, p.34, 2016.

J. Olweus, Dendritic cell ontogeny: a human dendritic cell lineage of myeloid origin". eng, Proceedings of the National Academy of Sciences of the United States of America 94, vol.23, p.50, 1997.

D. Ortuño, Embryonic Neural Stem Cell Differentiation to Aldynoglia Induced by Olfactory Bulb Ensheathing Cell-Conditioned Medium, p.58, 2011.

P. A. Ott, F. S. Hodi, and C. Robert, CTLA-4 and PD, 2013.

/. Blockade, New Immunotherapeutic Modalities with Durable Clinical Benefit in Melanoma Patients". en. In: Clinical Cancer Research, vol.19, pp.5300-5309

N. Pandis, Cytogenetic comparison of primary tumors and lymph node metastases in breast cancer patients, Genes, Chromosomes & Cancer, vol.22, p.40, 1998.

B. S. Parker, J. Rautela, and P. Hertzog, Antitumour actions of interferons: implications for cancer therapy, Nature reviews. Cancer, vol.16, p.171, 2016.

J. Parkin and B. Cohen, An overview of the immune system, eng. In: Lancet (London, p.45, 2001.

P. Paszek, Population robustness arising from cellular heterogeneity". en, Proceedings of the National Academy of Sciences 107.25, p.32, 2010.

D. Peng, Myeloid-Derived Suppressor Cells Endow Stem-like Qualities to Breast Cancer Cells through IL6/STAT3 and NO/NOTCH Cross-talk Signaling, 2016.

, Cancer Research 76, vol.11, p.43

E. K. Persson, , 2013.

, IRF4 transcription-factor-dependent

, Immunity 38.5, p.50

E. Picardi, Large-scale detection and analysis of RNA editing in grape mtDNA by RNA deep-sequencing". eng, Nucleic Acids Research, vol.38, p.60, 2010.

M. Plantinga, Conventional and monocyte-derived CD11b(+) dendritic cells initiate and maintain T helper 2 cellmediated immunity to house dust mite allergen". eng, Immunity 38, vol.2, p.52, 2013.

V. Poli, L. Fagnocchi, and A. Zippo, Tumorigenic Cell Reprogramming and Cancer Plasticity: Interplay between Signaling, Microenvironment, and Epigenetics. en, 2018.

L. Poulin and . Franz, Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells". eng, The Journal of Experimental Medicine, vol.207, p.50, 2010.

A. Prat and J. S. Parker, Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer". eng, Breast cancer research: BCR 12.5, R68. issn: 1465-542X, p.38, 2010.

A. Prat and E. Pineda, Clinical implications of the intrinsic molecular subtypes of breast cancer". eng, Breast (Edinburgh, Scotland) 24 Suppl, vol.2, p.37, 2015.

B. Pulendran, K. Palucka, and J. Banchereau, Sensing Pathogens and Tuning Immune Responses". en. In: Science 293, vol.5528, pp.36-8075, 2001.

L. Pyfferoen, The transcriptome of lung tumor-infiltrating dendritic cells reveals a tumor-supporting phenotype and a microRNA signature with negative impact on clinical outcome, eng. In: Oncoimmunology, vol.6, p.62, 2017.

. Quail and . Joyce, Microenvironmental regulation of tumor progression and metastasis". In: Nature medicine 19, vol.11, p.40, 2013.

I. Raphael, T cell subsets and their signature cytokines in autoimmune and inflammatory diseases, p.31, 2015.

J. C. Rieckmann, Social network architecture of human immune cells unveiled by quantitative proteomics". en, Nature Immunology, vol.18, pp.1529-2916, 2017.

D. Risso, Normalization of RNA-seq data using factor analysis of control genes or samples". eng, Nature Biotechnology, vol.32, p.60, 2014.

M. C. Rissoan, Reciprocal control of T helper cell and dendritic cell differentiation". eng, ) 283.5405, p.31, 1999.

M. E. Ritchie, limma powers differential expression analyses for RNAsequencing and microarray studies". eng, Nucleic Acids Research, vol.43, 2015.

A. G. Rivenbark, M. Siobhan, W. B. Oconnor, and . Coleman, Molecular and Cellular Heterogeneity in Breast Cancer, The American Journal of Pathology, vol.183, p.38, 2013.

G. Robertson, De novo assembly and analysis of RNA-seq data". eng, Nature Methods 7.11, p.60, 2010.

M. D. Robinson, J. Davis, G. K. Mccarthy, and . Smyth, edgeR: a Bioconductor package for differential expression analysis of digital gene expression data, Bioinformatics 26.1, pp.1367-4803, 2010.

L. B. Rodda, Single-Cell RNA Sequencing of Lymph Node Stromal Cells Reveals Niche-Associated Heterogeneity, English. In: Immunity 48, vol.5, pp.30143-30145, 2018.

C. Sánchez-torres, CD16+ and CD16 human blood monocyte subsets differentiate in vitro to dendritic cells with different abilities to stimulate CD4+, 2001.

, International Immunology 13, vol.12, p.45

S. M. Santini, Type I Interferon as a Powerful Adjuvant for Monocyte-Derived Dendritic Cell Development and Activity in Vitro and in Hu-Pbl-Scid Mice, The Journal of Experimental Medicine, vol.191, p.171, 2000.

C. Scheel, Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast, 2011.

, Cell 145, vol.6, p.41

A. Schlitzer, IRF4 transcription factor-dependent CD11b+ dendritic cells in human and mouse control mucosal IL-17 cytokine responses, eng. In: Immunity 38, vol.5, p.50, 2013.

H. A. Schreiber, Intestinal monocytes and macrophages are required for T cell polarization in response to Citrobacter rodentium, The Journal of Experimental Medicine, vol.210, p.52, 2013.

P. See, Mapping the human DC lineage through the integration of high-dimensional techniques, eng. In: Science, 2017.

C. Seebode, J. Lehmann, and S. Emmert, Photocarcinogenesis and Skin Cancer Prevention Strategies". eng, Anticancer Research, vol.36, issue.3, p.30, 2016.

E. Segura and S. Amigorena, Inflammatory dendritic cells in mice and humans, Trends in Immunology, vol.34, pp.1471-4981, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00846095

, , vol.167, p.52

E. Segura and M. Touzot, Human inflammatory dendritic cells induce Th17 cell differentiation". eng, Immunity 38, vol.2, p.52, 2013.

S. Selvaraj and J. Natarajan, Microarray Data Analysis and Mining Tools, pp.95-99, 2011.

A. N. Seo, Tumour-infiltrating CD8+ lymphocytes as an independent predictive factor for pathological complete response to primary systemic therapy in breast cancer, British Journal of Cancer, vol.109, p.43, 2013.

F. Shi, Organ-specific features of natural killer cells". eng, Nature Reviews. Immunology, vol.11, p.33, 2011.

A. Sica and A. Mantovani, Macrophage plasticity and polarization: in vivo veritas". eng, The Journal of Clinical Investigation, vol.122, p.46, 2012.

F. P. Siegal, The nature of the principal type 1 interferon-producing cells in human blood". eng, Science, vol.284, pp.36-8075, 1999.

H. Sinn and H. Kreipe, A Brief Overview of the WHO Classification of Breast Tumors, 2013.

, Breast Care, p.37

T. Sørlie, Repeated observation of breast tumor subtypes in independent gene expression data sets". en. In: Proceedings of the National Academy of Sciences 100, vol.14, p.38, 2003.

V. Soumelis, Systems approaches to unravel innate immune cell diversity, environmental plasticity and functional specialization, Current opinion in immunology, vol.32, pp.952-7915, 2015.

S. Spranger, Mechanisms of tumor escape in the context of the T-cellinflamed and the non-T-cell-inflamed tumor microenvironment". eng, International Immunology, vol.28, p.43, 2016.

J. Stagg and B. Allard, Immunotherapeutic approaches in triplenegative breast cancer: latest research and clinical prospects". en, Therapeutic Advances in Medical Oncology 5.3, p.53, 2013.

S. E. Stanton, S. Adams, and M. L. ,

. Disis, Variation in the Incidence and Magnitude of Tumor-Infiltrating Lymphocytes in Breast Cancer Subtypes: A Systematic Review". eng, JAMA oncology 2.10, p.169, 2016.

R. M. Steinman and Z. A. Cohn, Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution". eng, The Journal of Experimental Medicine, vol.137, p.46, 1973.

A. Stiff and W. Carson, Investigations of interferon-lambda for the treatment of cancer, Journal of innate immunity 7.3, p.170, 2015.

C. Stockmann, The Impact of the Immune System on Tumor: Angiogenesis and Vascular Remodeling, Frontiers in Oncology 4. issn: 2234-943X, 2014.

Y. Sun, Risk Factors and Preventions of Breast Cancer". eng. In: International Journal of Biological Sciences, vol.13, issue.11, p.36, 2017.

H. Takaba and H. Takayanagi, The Mechanisms of T Cell Selection in the, Thymus". English. In: Trends in Immunology, vol.38, issue.17, p.29, 2017.

A. R. Tan, Checkpoint Inhibitors in Breast Cancer: Changing the Therapeutic Landscape. en. url: https : / / am . asco . org / checkpoint -inhibitorsbreast-cancer-changing-therapeuticlandscape, p.44, 2018.

S. Tay, Single-cell NF-kappaB dynamics reveal digital activation and analogue information processing". eng, Nabreast cancer". eng. In: Breast Cancer Research and Treatment, vol.163, p.43, 2010.

J. Yin, Comprehensive analysis of immune evasion in breast cancer by single-cell RNA-seq". en. In: bioRxiv, p.368605, 2018.

L. A. Yurkovetskiy, M. Joseph, A. V. Pickard, and . Chervonsky, Microbiota and autoimmunity: exploring new avenues". eng, Cell Host & Microbe, vol.17, pp.1934-6069, 2015.

, , p.35

L. C. Zaba, G. James, M. A. Krueger, and . Lowes, Resident and "inflammatory" dendritic cells in human skin, The Journal of Investigative Dermatology 129, vol.2, p.52, 2009.

L. A. Zenewicz, Unraveling the genetics of autoimmunity". eng, Cell 140, vol.6, p.35, 2010.

Y. Zhou, Prognostic value of tumor-infiltrating Foxp3+ regulatory T cells in patients with breast cancer: a meta-analysis, Journal of Cancer, vol.8, p.43, 2017.

L. Ziegler-heitbrock, Nomenclature of monocytes and dendritic cells in blood, Blood 116, vol.16, p.50, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00611173

B. Zygmunt and M. Veldhoen, T helper cell differentiation more than just cytokines". eng, Advances in Immunology, vol.109, pp.1557-8445, 2011.

,

N. Lucas, , vol.1

V. Saada,

. Martin-e-fernadez-zapico, , vol.8, p.9

M. T. Howard, , vol.8, p.9

R. L. King, Sandrine Niyongere, 10 M'boyba Khadija Diop, vol.8, p.4, 2009.

P. Fenaux, Eric Padron, 10 Vassili Soumelis, 5 Nathalie Droin, 1,4 Mrinal M Patnaik, 12* Eric Solary, 1,2,3* Running head: Clonal PDC in chronic myelomonocytic leukemia 1 -INSERM U1170, Gustave Roussy Cancer Center, Gustave Roussy Cancer Center, issue.7

C. Inserm-us23 and . Center,

-. and I. Curie,

U. -inserm, C. Umr8104, and I. Cochin, 12 -Division of Hematology, Service d'hématologie biologique

C. Waskow, K. Liu, and G. Darrasse-jèze, FMS-like tyrosine kinase 3 is required for dendritic cell development in peripheral lymphoid tissues, Nat Immunol, vol.9, issue.6, pp.676-683, 2008.

B. Cisse, M. L. Caton, and M. Lehner, Transcription Factor E2-2 Is an Essential and Specific Regulator of Plasmacytoid Dendritic Cell Development, Cell, vol.135, issue.1, p.48, 2008.

M. Guilliams, F. Ginhoux, and C. Jakubzick, Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny, Nat Rev Immunol, vol.14, issue.8, pp.571-579, 2014.

F. P. Siegal, N. Kadowaki, and M. Shodell, The nature of the principal type 1 interferonproducing cells in human blood, Science, vol.284, issue.5421, pp.1835-1842, 1999.

J. A. Villadangos and L. Young, Antigen-Presentation Properties of Plasmacytoid Dendritic Cells, Immunity, vol.29, issue.3, p.61, 2008.

N. Kadowaki, S. Antonenko, J. Lau, and Y. Liu, Natural Interferon / -Producing Cells Link Innate and Adaptive Immunity, J Exp Med, vol.192, issue.2, pp.219-245, 2000.

W. Vermi, M. Soncini, L. Melocchi, S. Sozzani, and F. Facchetti, Plasmacytoid dendritic cells and cancer, J Leukoc Biol, vol.90, issue.4, p.90, 2011.

W. Zou, V. Machelon, and A. Coulomb-l'hermin, Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells, Nat Med, vol.7, issue.12, pp.1339-1385, 2001.

S. Wei, I. Kryczek, and L. Zou, Plasmacytoid Dendritic Cells Induce CD8+ Regulatory T Cells In Human Ovarian Carcinoma, Cancer Res, vol.65, issue.12, pp.5020-5026, 2005.

C. Conrad, J. Gregorio, and Y. Wang, Plasmacytoid Dendritic Cells Promote Immunosuppression in Ovarian Cancer via ICOS Costimulation of Foxp3+ T-Regulatory Cells, Cancer Res, vol.72, issue.20, pp.5240-5249, 2012.

S. Demoulin, M. Herfs, P. Delvenne, and P. Hubert, Tumor microenvironment converts plasmacytoid dendritic cells into immunosuppressive/tolerogenic cells: insight into the molecular mechanisms, J Leukoc Biol, vol.93, issue.3, pp.343-52, 2013.

M. Hishizawa, K. Imada, T. Kitawaki, M. Ueda, N. Kadowaki et al., Depletion and impaired interferon--producing capacity of blood plasmacytoid dendritic cells in human Tcell leukaemia virus type I-infected individuals, Br J Haematol, vol.125, issue.5, p.75, 2004.

N. B. Mami, M. Mohty, T. Aurran-schleinitz, D. Olive, and B. Gaugler, Blood dendritic cells in patients with chronic lymphocytic leukaemia, Immunobiology, vol.213, issue.6, pp.493-501, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00483520

M. Deininger, J. W. Tyner, and E. Solary, Turning the tide in myelodysplastic / myeloproliferative neoplasms, Nat Rev Cancer, vol.17, issue.7, p.40, 2017.

D. A. Arber, A. Orazi, and R. Hasserjian, The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia, Blood, vol.127, issue.20, pp.2391-405, 2016.

R. Itzykson, O. Kosmider, and A. Renneville, Prognostic score including gene mutations in chronic myelomonocytic leukemia, J Clin Oncol, vol.31, pp.2428-2464, 2013.

J. Merlevede, N. Droin, and T. Qin, Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents, Nat Commun, vol.7, p.10767, 2016.

R. Itzykson, O. Kosmider, and A. Renneville, Clonal architecture of chronic myelomonocytic leukemias, Blood, vol.121, issue.12, pp.2186-98, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00923316

E. Padron, J. S. Painter, and S. Kunigal, GM-CSF-dependent pSTAT5 sensitivity is a feature with therapeutic potential in chronic myelomonocytic leukemia, Blood, vol.121, issue.25, p.5068, 2013.

M. M. Patnaik and A. Tefferi, Chronic myelomonocytic leukemia: 2016 update on diagnosis, risk stratification, and management, Am J Hematol, vol.91, issue.6, p.42, 2016.

T. De-witte, D. Bowen, and M. Robin, Allogeneic hematopoietic stem cell transplantation for MDS and CMML: recommendations from an international expert panel, Blood, vol.129, issue.13, pp.1753-62, 2017.

E. Solary and R. Itzykson, How I treat chronic myelomonocytic leukemia, Blood, vol.130, issue.2, p.36, 2017.

Y. Chen, J. Chou, R. P. Ketterling, L. Letendre, and C. Li, Histologic and Immunohistochemical Study of Bone Marrow Monocytic Nodules in 21 Cases With Myelodysplasia, Am J Clin Pathol, vol.120, issue.6, p.81, 2003.

W. Vermi, F. Facchetti, and S. Rosati, Nodal and extranodal tumor-forming accumulation of plasmacytoid monocytes/interferon-producing cells associated with myeloid disorders, Am J Surg Pathol, vol.28, issue.5, p.95, 2004.

A. Orazi, R. Chiu, O. Malley, and D. P. , Chronic myelomonocytic leukemia: the role of bone marrow biopsy immunohistology, Mod Pathol, vol.19, issue.12, pp.1536-1581, 2006.

N. L. Harris and Z. Demirjian, Plasmacytoid T-zone cell proliferation in a patient with chronic myelomonocytic leukemia. Histologic and immunohistologic characterization, Am J Surg Pathol, vol.15, issue.1, p.95, 1991.

H. K. Müller-hermelink, H. Stein, G. Steinmann, and K. Lennert, Malignant lymphoma of plasmacytoid T-cells. Morphologic and immunologic studies characterizing a special type of T-cell, Am J Surg Pathol, vol.7, issue.8, pp.849-62

F. Facchetti, C. De-wolf-peeters, D. Y. Mason, K. Pulford, J. J. Van-den-oord et al.,

T. Plasmacytoid and . Cells, Immunohistochemical evidence for their monocyte/macrophage origin, Am J Pathol, vol.133, issue.1, p.15, 1988.

A. Villani, R. Satija, and G. Reynolds, Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors, Science, vol.356, issue.6335, p.4573, 2017.

P. See, C. Dutertre, and J. Chen, Mapping the human DC lineage through the integration of high-dimensional techniques, Science, vol.356, issue.6342, p.3009, 2017.

E. Such, U. Germing, and L. Malcovati, Development and validation of a prognostic scoring system for patients with chronic myelomonocytic leukemia, Blood, vol.121, issue.15, pp.3005-3020, 2013.

M. M. Patnaik, T. L. Lasho, and P. Vijayvargiya, Prognostic interaction between ASXL1 and TET2 mutations in chronic myelomonocytic leukemia, Blood Cancer J, vol.6, issue.1, pp.385-416, 2016.

Y. Liu and . Ipc, Professional Type 1 Interferon-Producing Cells and Plasmacytoid Dendritic Cell Precursors, Annu Rev Immunol, vol.23, issue.1, pp.275-306, 2005.

E. I. Boyle, S. Weng, and J. Gollub, TermFinder-open source software for accessing Gene Ontology information and finding significantly enriched Gene Ontology terms associated with a list of genes, Bioinformatics, vol.20, issue.18, pp.3710-3715, 2004.

G. Yu, L. Wang, Y. Han, and Q. He, clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters, OMICS : a Journal of Integrative Biology, vol.16, issue.5, pp.284-287, 2012.

J. Lee, G. Breton, and T. Oliveira, Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow, J Exp Med, vol.212, issue.3, pp.385-99, 2015.

J. Lee, G. Breton, and A. Aljoufi, Clonal analysis of human dendritic cell progenitor using a stromal cell culture, J Immunol Methods, vol.425, p.21, 2015.

W. Chen, S. Antonenko, and J. M. Sederstrom, Thrombopoietin cooperates with FLT3-ligand in the generation of plasmacytoid dendritic cell precursors from human hematopoietic progenitors, Blood, vol.103, issue.7, pp.2547-53, 2004.

S. Demoulin, P. Roncarati, P. Delvenne, and P. Hubert, Production of large numbers of plasmacytoid dendritic cells with functional activities from CD34+ hematopoietic progenitor cells: Use of interleukin-3, Exp Hematol, vol.40, issue.4, pp.268-78, 2012.

A. Pardanani, K. K. Reichard, and D. Zblewski, CD123 immunostaining patterns in systemic mastocytosis: differential expression in disease subgroups and potential prognostic value, Leukemia, vol.30, issue.4, pp.914-922, 2016.

A. Tzankov, K. Hebeda, and M. Kremer, Plasmacytoid dendritic cell proliferations and neoplasms involving the bone marrow: Summary of the workshop cases submitted to the 18th Meeting of the European Association for Haematopathology (EAHP) organized by the European Bone Marrow Working Group, Ann Hematol, vol.96, issue.5, pp.765-77, 2016.

T. Matsui, J. E. Connolly, and M. Michnevitz, CD2 Distinguishes Two Subsets of Human Plasmacytoid Dendritic Cells with Distinct Phenotype and Functions, J Immunol, vol.182, issue.11, pp.6815-6838, 2009.

L. Zitvogel, L. Galluzzi, O. Kepp, M. J. Smyth, and G. Kroemer, Type I interferons in anticancer immunity, Nat Rev Immunol, vol.15, pp.405-414, 2015.

M. Touzot, M. Grandclaudon, and A. Cappuccio, Combinatorial flexibility of cytokine function during human T helper cell differentiation, Nat Commun, vol.5, p.3987, 2014.

J. Kiladjian, S. Giraudier, and B. Cassinat, Interferon-alpha for the therapy of myeloproliferative neoplasms: targeting the malignant clone, Leukemia, vol.30, issue.4, p.81, 2016.

D. Chauhan, A. V. Singh, and M. Brahmandam, Functional Interaction of Plasmacytoid Dendritic Cells with Multiple Myeloma Cells: A Therapeutic Target, Cancer Cell, vol.16, issue.4, p.23, 2009.

S. G. Alculumbre, V. Saint-andré, D. Domizio, and J. , Diversification of human plasmacytoid predendritic cells in response to a single stimulus, Nat Immunol, vol.19, issue.1, pp.63-75, 2018.

D. Reynaud, E. Pietras, and K. Barry-holson, IL-6 controls leukemic multipotent progenitor cell fate and contributes to chronic myelogenous leukemia development, Cancer Cell, vol.20, issue.5, p.661, 2011.

R. S. Welner, G. Amabile, and D. Bararia, Treatment of chronic myelogenous leukemia by blocking cytokine alterations found in normal stem and progenitor cells, Cancer Cell, vol.27, issue.5, p.671, 2015.

C. Alfaro, M. F. Sanmamed, and M. E. Rodríguez-ruiz, Interleukin-8 in cancer pathogenesis, treatment and follow-up, Cancer Treat Rev, vol.60, pp.24-31, 2017.

A. Sinclair, L. Park, and M. Shah, CXCR2 and CXCL4 regulate survival and self-renewal of hematopoietic stem/progenitor cells, Blood, vol.128, issue.3, p.371, 2016.

A. Tobler, B. Moser, and B. Dewald, Constitutive expression of interleukin-8 and its receptor in human myeloid and lymphoid leukemia, Blood, vol.82, issue.8, pp.2517-2542, 1993.

A. Shafat, M. S. Mehta, and T. K. , MIF-Induced Stromal PKC /IL8 Is Essential in Human Acute Myeloid Leukemia, Cancer Res, vol.77, issue.2, pp.303-314, 2017.

C. Schinke, O. Giricz, and W. Li, IL8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells, Blood, vol.125, issue.20, p.3144, 2015.

D. Kingston, M. A. Schmid, N. Onai, A. Obata-onai, D. Baumjohann et al., The concerted action of GM-CSF and Flt3-ligand on in vivo dendritic cell homeostasis, Blood, vol.114, issue.4, p.43, 2009.

E. Esashi, Y. Wang, O. Perng, X. Qin, Y. Liu et al., The Signal Transducer STAT5 Inhibits Plasmacytoid Dendritic Cell Development by Suppressing Transcription Factor IRF8, Immunity, vol.28, issue.4, p.20, 2008.

S. Niyongere, N. Lucas, and J. M. Zhou, Heterogeneous expression of cytokines accounts for clinical diversity and refines prognostication in CMML, Leukemia

C. Desterke, C. Bilhou-nabéra, and B. Guerton, FLT3-Mediated p38-MAPK Activation Participates in the Control of Megakaryopoiesis in Primary Myelofibrosis, Cancer Res, vol.71, issue.8, pp.2901-2916, 2011.

S. Y. Kordasti, W. Ingram, and J. Hayden, CD4+CD25high Foxp3+ regulatory T cells in myelodysplastic syndrome (MDS), Blood, vol.110, issue.3, p.50, 2007.

I. Kotsianidis, I. Bouchliou, and E. Nakou, Kinetics, function and bone marrow trafficking of CD4+CD25+FOXP3+ regulatory T cells in myelodysplastic syndromes (MDS), Leukemia, vol.23, issue.3, pp.510-518, 2008.

A. O. Kittang, S. Kordasti, and K. E. Sand, Expansion of myeloid derived suppressor cells correlates with number of T regulatory cells and disease progression in myelodysplastic syndrome, Oncoimmunology, vol.5, issue.2, p.1062208, 2016.

V. Sisirak, J. Faget, and M. Gobert, Impaired IFN-Production by Plasmacytoid Dendritic Cells Favors Regulatory T-cell Expansion That May Contribute to Breast Cancer Progression, Cancer Res, vol.72, issue.20, pp.5188-97, 2012.

A. Ray, D. S. Das, and Y. Song, A novel agent SL-401 induces anti-myeloma activity by targeting plasmacytoid dendritic cells, osteoclastogenesis and cancer stem-like cells, Leukemia, vol.31, issue.12, pp.2652-2660, 2017.

, Alongside PD-1 + T cells within tumors, CD8 + PD-1 -ILT2 + T cells are a major intratumor cytotoxic population selectively inhibited by the immune checkpoint

, Alix Jacquier, vol.1, 2005.

H. Department, S. Cea-drf, and . Hospital, 3 Department of Medical Oncology

&. Taipei, /. Hospital, Y. University, and . Shiang, Republic of China the interest of therapeutic targeting of the HLA-G:ILT2 checkpoint in HLA-G-positive tumors, either concomitantly to anti-PD1/PD-L1

R. J. Motzer, B. Escudier, D. F. Mcdermott, S. George, H. J. Hammers et al., Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma, New Engl J Med, vol.373, pp.1803-1816, 2015.

R. J. Motzer, N. M. Tannir, D. F. Mcdermott, O. A. Frontera, B. Melichar et al., Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma, New Engl J Med, vol.378, pp.1277-90, 2018.

N. Rouas-freiss, J. Lemaoult, J. Verine, T. Roux, D. Culine et al., Intratumor heterogeneity of immune checkpoints in primary renal cell cancer: Focus on HLA-G/ILT2/ILT4, Oncoimmunology, vol.6, 2017.

E. D. Carosella, B. Favier, N. Rouas-freiss, P. Moreau, and J. Lemaoult, Beyond the increasing complexity of the immunomodulatory HLA-G molecule, Blood, vol.111, pp.4862-70, 2008.

E. D. Carosella, N. Rouas-freiss, D. T. Roux, P. Moreau, and J. Lemaoult, HLA-G: An Immune Checkpoint Molecule, Advances in immunology, vol.127, pp.33-144, 2015.

A. Naji, L. Rond, S. Durrbach, A. Krawice-radanne, I. Creput et al., CD3+CD4low and CD3+CD8low are induced by HLA-G: novel human peripheral blood suppressor T-cell subsets involved in transplant acceptance, Blood, vol.110, pp.3936-3984, 2007.

S. Agaugue, E. D. Carosella, and N. Rouas-freiss, Role of HLA-G in tumor escape through expansion of myeloid-derived suppressor cells and cytokinic balance in favor of Th2 versus Th1/Th17, Blood, vol.117, pp.7021-7052, 2011.

E. C. Ibrahim, N. Guerra, M. J. Lacombe, E. Angevin, S. Chouaib et al., Tumor-specific up-regulation of the nonclassical class I HLA-G antigen expression in renal carcinoma, Cancer research, vol.61, pp.6838-6883, 2001.
URL : https://hal.archives-ouvertes.fr/hal-00092851

E. C. Ibrahim, Y. Allory, F. Commo, B. Gattegno, P. Callard et al., Altered pattern of major histocompatibility complex expression in renal carcinoma: tumor-specific expression of the nonclassical human leukocyte antigen-G molecule is restricted to clear cell carcinoma while up-regulation of other major histocompatibility complex antigens is primarily distributed in all subtypes of renal carcinoma, The American journal of pathology, vol.162, pp.501-509, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00092847

B. L. Li, A. Lin, X. J. Zhang, X. Zhang, J. G. Zhang et al., Characterization of HLA-G expression in renal cell carcinoma, Tissue antigens, vol.74, pp.213-234, 2009.

C. C. Chang, S. P. Murphy, and S. Ferrone, Differential in vivo and in vitro HLA-G expression in melanoma cells: potential mechanisms, Human immunology, vol.64, pp.1057-63, 2003.

V. Rebmann, S. Wagner, and H. Grosse-wilde, HLA-G expression in malignant melanoma, Seminars in cancer biology, vol.17, pp.422-431, 2007.

W. N. Melsted, L. L. Johansen, J. Lock-andersen, N. Behrendt, J. O. Eriksen et al., HLA class Ia and Ib molecules and FOXP3+TILs in relation to the prognosis of malignant melanoma patients, Clinical Immunology, vol.183, pp.191-198, 2017.

A. Lin, C. C. Zhu, H. X. Chen, B. F. Chen, X. Zhang et al., Clinical relevance and functional implications for human leucocyte antigen-g expression in non-small-cell lung cancer, Journal of cellular and molecular medicine, vol.14, pp.2318-2347, 2010.

S. M. Yie, H. Yang, S. R. Ye, K. Li, D. D. Dong et al., Expression of human leucocyte antigen G (HLA-G) is associated with prognosis in non-small cell lung cancer, Lung Cancer, vol.58, pp.267-74, 2007.

L. Loumagne, J. Baudhuin, B. Favier, F. Montespan, and E. D. Carosella, Rouas-Freiss N. In vivo evidence that secretion of HLA-G by immunogenic tumor cells allows their evasion from immunosurveillance, International journal of cancer Journal international du cancer, vol.135, pp.2107-2124, 2014.

M. Colonna, F. Navarro, T. Bellon, M. Llano, P. Garcia et al., A common inhibitory receptor for major histocompatibility complex class I molecules on human lymphoid and myelomonocytic cells, The Journal of experimental medicine, vol.186, pp.1809-1827, 1997.

D. Saverino, M. Fabbi, F. Ghiotto, A. Merlo, S. Bruno et al., The CD85/LIR-1/ILT2 inhibitory receptor is expressed by all human T lymphocytes and down-regulates their functions, Journal of Immunology, vol.165, pp.3742-55, 2000.

D. E. Speiser, D. Valmori, D. Rimoldi, M. J. Pittet, D. Lienard et al., CD28-negative cytolytic effector T cells frequently express NK receptors and are present at variable proportions in circulating lymphocytes from healthy donors and melanoma patients, European journal of immunology, vol.29, pp.1990-1999, 1999.

N. T. Young, M. Uhrberg, J. H. Phillips, L. L. Lanier, and P. Parham, Differential expression of leukocyte receptor complex-encoded Ig-like receptors correlates with the transition from effector to memory CTL, Journal of Immunology, vol.166, pp.3933-3974, 2001.

M. N. Ince, B. Harnisch, Z. Xu, S. K. Lee, C. Lange et al., Increased expression of the natural killer cell inhibitory receptor CD85j/ILT2 on antigen-specific effector CD8 T cells and its impact on CD8 T-cell function, Immunology, vol.112, pp.531-573, 2004.

J. Northfield, M. Lucas, H. Jones, N. T. Young, and P. Klenerman, Does memory improve with age? CD85j (ILT-2/LIR-1) expression on CD81 T cells correlates with 'memory inflation' in human cytomegalovirus infection, Immunology and cell biology, vol.83, pp.182-190, 2005.

C. E. Gustafson, Q. Qi, J. Hutter-saunders, S. Gupta, R. Jadhav et al., Immune Checkpoint Function of CD85j in CD8 T Cell Differentiation and Aging, Frontiers in immunology, vol.8, 2017.

K. Rothe, D. Quandt, K. Schubert, M. Rossol, M. Klingner et al., Latent Cytomegalovirus Infection in Rheumatoid Arthritis and Increased Frequencies of Cytolytic LIR-1+CD8+T Cells, Arthritis & Rheumatology, vol.68, pp.337-383, 2016.

N. Anfossi, J. M. Doisne, M. A. Peyrat, S. Ugolini, O. Bonnaud et al., Coordinated expression of ig-like inhibitory MHC class I receptors and acquisition of cytotoxic function in human CD8(+) T cells, Journal of Immunology, vol.173, pp.7223-7232, 2004.

P. Costa, S. Rusconi, D. Mavilio, M. Fogli, G. Murdaca et al., Differential disappearance of inhibitory natural killer cell receptors during HAART and possible impairment of HIV-1-specific CD8 cytotoxic T lymphocytes, AIDS, vol.15, pp.965-74, 2001.

N. Servant, E. Gravier, P. Gestraud, C. Laurent, C. Paccard et al., EMA -A R package for Easy Microarray data analysis, BMC research notes, vol.3, p.277, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00984710

F. Sallusto, D. Lenig, R. Forster, M. Lipp, and A. Lanzavecchia, Two subsets of memory T lymphocytes with distinct homing potentials and effector functions, Nature, vol.401, pp.708-720, 1999.

S. Chevrier, J. H. Levine, V. Zanotelli, K. Silina, D. Schulz et al., An Immune Atlas of Clear Cell Renal Cell Carcinoma, Cell, vol.169, pp.736-785, 2017.

Y. D. Mahnke, T. M. Brodie, F. Sallusto, M. Roederer, and E. Lugli, The who's who of T-cell differentiation: human memory T-cell subsets, European journal of immunology, vol.43, pp.2797-809, 2013.

M. Strioga, V. Pasukoniene, and D. Characiejus, CD8+ CD28-and CD8+ CD57+ T cells and their role in health and disease, Immunology, vol.134, pp.17-32, 2011.

S. Kuttruff, S. Koch, A. Kelp, G. Pawelec, H. Rammensee et al., NKp80 defines and stimulates a reactive subset of CD8 T cells, Blood, vol.113, pp.358-69, 2009.

Y. Simoni, E. Becht, M. Fehlings, C. Y. Loh, S. Koo et al., Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates, Nature, vol.557, pp.575-584, 2018.

L. Gal, F. A. Riteau, B. Sedlik, C. Khalil-daher, I. Menier et al., HLA-G-mediated inhibition of antigen-specific cytotoxic T lymphocytes, International immunology, vol.11, pp.1351-1357, 1999.

G. Curigliano, C. Criscitiello, L. Gelao, and A. Goldhirsch, Clinical cancer research : an official journal of the American Association for, Cancer Research, vol.19, pp.5564-71, 2013.

T. K. Choueiri, M. N. Fishman, B. Escudier, D. F. Mcdermott, C. G. Drake et al.,

, Immunomodulatory Activity of Nivolumab in Metastatic Renal Cell Carcinoma. Clinical Cancer Research, vol.22, pp.5461-71, 2016.