Y. Shi and J. O. Thomas, The transport of proteins into the nucleus requires the 70-kilodalton heat shock protein or its cytosolic cognate, Mol Cell Biol, vol.12, pp.2186-92, 1992.

H. Murakami, D. Pain, and G. Blobel, 70-kD heat shock-related protein is one of at least two distinct cytosolic factors stimulating protein import into mitochondria, J Cell Biol, vol.107, pp.2051-57, 1988.

R. P. Beckmann, L. E. Mizzen, and W. J. Welch, Interaction of Hsp 70 with newly synthesized proteins: implications for protein folding and assembly, Science, vol.248, pp.850-854, 1990.

J. S. Seo, Y. M. Park, J. I. Kim, E. H. Shim, C. W. Kim et al., T cell lymphoma in transgenic mice expressing the human Hsp70 gene
DOI : 10.1006/bbrc.1996.0103

, Biochem Biophys Res Commun, vol.218, pp.582-589, 1996.

V. Z. Volloch and M. Y. Sherman, Oncogenic potential of Hsp72, Oncogene, vol.18, pp.3648-51, 1999.
DOI : 10.1038/sj.onc.1202525

URL : https://www.nature.com/articles/1202525.pdf

M. Jaattela, Over-expression of hsp70 confers tumorigenicity to mouse fibrosarcoma cells, Int J Cancer, vol.60, pp.689-93, 1995.

L. M. Vargas-roig, F. E. Gago, O. Tello, J. C. Aznar, and D. R. Ciocca, Heat shock protein expression and drug resistance in breast cancer patients treated with induction chemotherapy, Int J Cancer, vol.79, pp.468-75, 1998.

J. Nylandsted, M. Rohde, K. Brand, L. Bastholm, F. Elling et al., Selective depletion of heat shock protein 70 (Hsp70) activates a tumor-specific death program that is independent of caspases and bypasses Bcl-2, Proc Natl Acad Sci U S A, vol.97, pp.7871-7877, 2000.

S. Gurbuxani, J. M. Bruey, A. Fromentin, N. Larmonier, A. Parcellier et al., Selective depletion of inducible HSP70 enhances immunogenicity of rat colon cancer cells, Oncogene, vol.20, pp.7478-85, 2001.

H. M. Beere, B. B. Wolf, K. Cain, D. D. Mosser, A. Mahboubi et al., Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome, Nat Cell Biol, vol.2, pp.469-75, 2000.

L. Ravagnan, S. Gurbuxani, S. A. Susin, C. Maisse, E. Daugas et al., Heat-shock protein 70 antagonizes apoptosis-inducing factor, Nat Cell Biol, vol.3, pp.839-882, 2001.
DOI : 10.1038/ncb0901-839

URL : https://hal.archives-ouvertes.fr/hal-01606079

G. Chiosis, Discovery and development of purine-scaffold Hsp90 inhibitors, Curr Top Med Chem, vol.6, pp.1183-91, 2006.
DOI : 10.1517/17460441.3.1.99

G. Chiosis, M. N. Timaul, B. Lucas, P. N. Munster, F. F. Zheng et al., A small molecule designed to bind to the adenine nucleotide pocket of Hsp90 causes Her2 degradation and the growth arrest and differentiation of breast cancer cells, Chem Biol, vol.8, pp.289-99, 2001.

L. Neckers and K. Neckers, Heat-shock protein 90 inhibitors as novel cancer chemotherapeutic agents, Expert Opin Emerg Drugs, vol.7, pp.277-88, 2002.
DOI : 10.1517/14728214.7.2.277

S. Z. Usmani, R. Bona, and Z. Li, 17 AAG for HSP90 inhibition in cancer-from bench to bedside, Curr Mol Med, vol.9, pp.654-64, 2009.

S. Gurbuxani, E. Schmitt, C. Cande, A. Parcellier, A. Hammann et al., Heat shock protein 70 binding inhibits the nuclear import of apoptosis-inducing factor, Oncogene, vol.22, pp.6669-78, 2003.

E. Schmitt, A. Parcellier, S. Gurbuxani, C. Cande, A. Hammann et al., Chemosensitization by a non-apoptogenic heat shock protein 70-binding apoptosis-inducing factor mutant, Cancer Res, vol.63, pp.8233-8273, 2003.

A. J. Steele, A. G. Prentice, A. V. Hoffbrand, B. C. Yogashangary, S. M. Hart et al., 2-Phenylacetylenesulfonamide (PAS) induces p53independent apoptotic killing of B-chronic lymphocytic leukemia (CLL) cells, Blood, vol.114, pp.1217-1242, 2009.

J. I. Leu, J. Pimkina, A. Frank, M. E. Murphy, and D. L. George, A small molecule inhibitor of inducible heat shock protein 70, Mol Cell, vol.36, pp.15-27, 2009.

S. N. Thibodeau, G. Bren, and D. Schaid, Microsatellite instability in cancer of the proximal colon, Science, vol.260, pp.816-819, 1993.

L. A. Aaltonen, Clues to the pathogenesis of familial colorectal cancer, Science, vol.260, pp.812-816, 1993.

Y. Ionov, M. A. Peinado, S. Malkhosyan, D. Shibata, and M. Perucho, Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis, Nature, vol.363, pp.558-561, 1993.

A. Duval and R. Hamelin, Mutations at coding repeat sequences in mismatch repairdeficient human cancers: toward a new concept of target genes for instability, Cancer Res, vol.62, pp.2447-2454, 2002.

A. Walther, R. Houlston, and I. Tomlinson, Association between chromosomal instability and prognosis in colorectal cancer: a meta-analysis, Gut, vol.57, pp.941-950, 2008.

G. Giannini, Mutations of an intronic repeat induce impaired MRE11 expression in primary human cancer with microsatellite instability, Oncogene, vol.23, pp.2640-2647, 2004.

D. J. Sargent, Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer, J. Clin. Oncol, vol.28, pp.3219-3226, 2010.

A. Zaanan, Impact of p53 expression and microsatellite instability on stage III colon cancer disease-free survival in patients treated by 5-fluorouracil and leucovorin with or without oxaliplatin, Ann. Oncol, vol.21, pp.772-780, 2010.

N. Yamagishi, Y. Saito, and T. Hatayama, Mammalian 105 kDa heat shock family proteins suppress hydrogen peroxide-induced apoptosis through a p38 MAPKdependent mitochondrial pathway in HeLa cells, FEBS J, vol.275, pp.4558-4570, 2008.

S. Hosaka, Synthetic small interfering RNA targeting heat shock protein 105 induces apoptosis of various cancer cells both in vitro and in vivo, Cancer Sci, vol.97, pp.623-632, 2006.

N. Yamagishi, K. Ishihara, Y. Saito, and T. Hatayama, Hsp105 family proteins suppress staurosporine-induced apoptosis by inhibiting the translocation of Bax to mitochondria in HeLa cells, Exp. Cell Res, vol.312, pp.3215-3223, 2006.

C. Siatskas, J. Underwood, A. Ramezani, R. G. Hawley, and J. A. Medin, Specific pharmacological dimerization of KDR in lentivirally transduced human hematopoietic cells activates anti-apoptotic and proliferative mechanisms, FASEB J, vol.19, pp.1752-1754, 2005.

M. Kai, Heat shock protein 105 is overexpressed in a variety of human tumors, Oncol. Rep, vol.10, pp.1777-1782, 2003.

O. Slaby, Significant overexpression of Hsp110 gene during colorectal cancer progression, Oncol. Rep, vol.21, pp.1235-1241, 2009.

A. Trott, L. Shaner, and K. A. Morano, The molecular chaperone Sse1 and the growth control protein kinase Sch9 collaborate to regulate protein kinase A activity in Saccharomyces cerevisiae, Genetics, vol.170, pp.1009-1021, 2005.

C. Andréasson, J. Fiaux, H. Rampelt, S. Druffel-augustin, and B. Bukau, Insights into the structural dynamics of the Hsp110-Hsp70 interaction reveal the mechanism for nucleotide exchange activity, Proc. Natl. Acad. Sci. USA 105, pp.16519-16524, 2008.

X. Y. Wang, Targeted immunotherapy using reconstituted chaperone complexes of heat shock protein 110 and melanoma-associated antigen gp100, Cancer Res, vol.63, pp.2553-2560, 2003.

M. H. Manjili, Development of a recombinant HSP110-HER-2/neu vaccine using the chaperoning properties of HSP110, Cancer Res, vol.62, pp.1737-1742, 2002.

M. H. Manjili, HSP110-HER2/neu chaperone complex vaccine induces protective immunity against spontaneous mammary tumors in HER-2/neu transgenic mice, J. Immunol, vol.171, pp.4054-4061, 2003.

E. Ceballos, Inhibitory effect of c-Myc on p53-induced apoptosis in leukemia cells. Microarray analysis reveals defective induction of p53 target genes and upregulation of chaperone genes, Oncogene, vol.24, pp.4559-4571, 2005.

K. Gotoh, Apg-2 has a chaperone-like activity similar to Hsp110 and is overexpressed in hepatocellular carcinomas, FEBS Lett, vol.560, pp.19-24, 2004.

N. Suraweera, Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR, Gastroenterology, vol.123, pp.1804-1811, 2002.

O. Buhard, Multipopulation analysis of polymorphisms in five mononucleotide repeats used to determine the microsatellite instability status of human tumors, J. Clin. Oncol, vol.24, pp.241-251, 2006.

W. M. Grady, Mutation of the type II transforming growth factor-beta receptor is coincident with the transformation of human colon adenomas to malignant carcinomas, Cancer Res, vol.58, pp.3101-3104, 1998.

A. Duval, The mutator pathway is a feature of immunodeficiency-related lymphomas, Proc. Natl. Acad. Sci. USA, vol.101, pp.5002-5007, 2004.
URL : https://hal.archives-ouvertes.fr/hal-00023370

A. L. Rérole, Peptides and aptamers targeting HSP70: a novel approach for anticancer chemotherapy, Cancer Res, vol.71, pp.484-495, 2011.

G. Jego, A. Hazoume, R. Seigneuric, and C. Garrido, Targeting heat shock proteins in cancer, Cancer Lett. published online, issue.13, 2010.

C. J. Gobbo, S. Gaucher-di-stasio, J. Weidmann, C. Guzzo, and . Garrido, Annexe 2 : Quantification of HSP27 and HSP70 molecular chaperone activities, Methods Mol Biol

, Fetal bovine serum (FBS) (Lonza)

, DMEM w/FBS: DMEM supplemented with 10% FBS

, Cell culture grade trypsin solution (Lonza)

, Ethylenediaminetetraacetic acid (EDTA): 1% (Sigma)

, Sterile washing buffer: HBSS (Lonza)

, Transfection reagent: DreamFectTM Gold ® (Oz biosciences)

D. and /. Fbs-10%,

, Sterile washing buffer: HBSS (Lonza)

-. Opti and . Medium,

. Human, HA hHSP27 or hHSP70

, Sterile washing buffer: HBSS (Lonza)

D. and /. Fbs-10%,

, Phosphate-buffered saline (PBS) 1× (Lonza)

, Lysis buffer: 50 mM HEPES, 0.25 M EDTA, 4 M NaCl, 0.1% NP40

, Antiprotease: Prepare in advance in sterile water. Store at ?20°C

, Lowry protein assay

, Sonicator. 1. 96-well plate (flat bottom)

, BSA: Prepare in advance at 100 g/L, store at ?20°C

, DTT: Prepare in advance at 1 M, store at ?20°C

, Sodium phosphate buffer at 50 mM

, Hsp27 recombinant protein: Store separate aliquots at ?80°C (see Note 2)

, For protease inhibition, add 300 mL of antiprotease to 10 mL lysis buffer. Keep the buffer on ice (see Note 11)

, Add 300 mL of lysis buffer (see Note 12)

, Transfer in microtube and vortex for 10 s. Keep on ice during 20 min for cell lysis

, Sonicate for 10 s, power 10%

, Adjust protein concentration of all samples at 2 mg/mL with sodium phosphate buffer or Tris-HCl buffer

, Samples can be stored at ?80°C supplemented with 3% of glycerol before aggregation test (see Note 14)

R. A. Laskey, B. M. Honda, A. D. Mills, N. R. Morris, A. H. Wyllie et al., , 1978.

, Chromatin assembly and transcription in eggs and oocytes of Xenopus laevis, Cold Spring Harb Symp Quant Biol, vol.42, pp.171-178

R. P. Beckmann, L. E. Mizzen, and W. J. Welch, Interaction of Hsp 70 with newly synthesized proteins: implications for protein folding and assembly, Science, vol.248, pp.850-854, 1990.

P. De-los-rios, A. Ben-zvi, O. Slutsky, A. Azem, and P. Goloubinoff, Hsp70 chaperones accelerate protein translocation and the unfolding of stable protein aggregates by entropic pulling, Proc Natl Acad Sci. USA, vol.103, pp.6166-6171, 2006.

Y. Shi and J. O. Thomas, The transport of proteins into the nucleus requires the 70-kilodalton heat shock protein or its cytosolic cognate, Mol Cell Biol, vol.12, pp.2186-2192, 1992.

E. A. Nollen, J. F. Brunsting, H. Roelofsen, L. A. Weber, and H. H. Kampinga, In vivo chaperone activity of heat shock protein 70 and thermotolerance, Mol Cell Biol, vol.19, pp.2069-2079, 1999.

M. Ehrnsperger, S. Gräber, M. Gaestel, and J. Buchner, Binding of non-native protein to Hsp25 during heat shock creates a reservoir of folding intermediates for reactivation, EMBO J, vol.16, pp.221-229, 1997.

R. Shashidharamurthy, H. A. Koteiche, J. Dong, and H. S. Mchaourab, Mechanism of chaperone function in small heat shock proteins: dissociation of the HSP27 oligomer is required for recognition and binding of destabilized T4 lysozyme, J Biol Chem, vol.280, pp.5281-5289, 2005.

J. M. Bruey, C. Paul, A. Fromentin, S. Hilpert, A. P. Arrigo et al., Differential regulation of HSP27 oligomerization in tumor cells grown in vitro and in vivo, Oncogene, vol.19, pp.4855-4863, 2000.

B. Lelj-garolla and A. G. Mauk, Selfassociation and chaperone activity of Hsp27 are thermally activated, J Biol Chem, vol.281, pp.8169-8174, 2006.

V. T. Devita and E. Chu, A history of cancer chemotherapy, Cancer Res, vol.68, pp.8643-8653, 2008.

C. A. Lipinski, F. Lombardo, B. W. Dominy, and P. J. Feeney, Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings, Adv Drug Deliv Rev, vol.46, pp.3-26, 2001.

K. Imai and A. Takaoka, Comparing antibody and smallmolecule therapies for cancer, Nat Rev Cancer, vol.6, pp.714-727, 2006.

J. A. Papin, T. Hunter, B. O. Palsson, and S. Subramaniam, Reconstruction of cellular signalling networks and analysis of their properties, Nat Rev Mol Cell Biol, vol.6, pp.99-111, 2005.

R. C. Ladner, Antibodies cut down to size, Nat Biotechnol, vol.25, pp.875-877, 2007.

E. Lacana, S. Amur, P. Mummanneni, H. Zhao, and F. W. Frueh, The emerging role of pharmacogenomics in biologics, Clin Pharmacol Ther, vol.82, pp.466-471, 2007.

A. L. Nelson, E. Dhimolea, and J. M. Reichert, Development trends for human monoclonal antibody therapeutics, Nat Rev Drug Discov, vol.9, pp.767-774

, Oncotarget, vol.2, pp.557-561, 2011.

I. Kola and J. Landis, Can the pharmaceutical industry reduce attrition rates?, Nat Rev Drug Discov, vol.3, pp.711-715, 2004.

R. Seigneuric, L. Markey, D. S. Nuyten, C. Dubernet, C. T. Evelo et al., From Nanotechnology to Nanomedicine: applications to cancer research, Curr Molecular Medicine, vol.10, issue.7, pp.640-52, 2010.

P. Colas, B. Cohen, T. Jessen, I. Grishina, J. Mccoy et al., Genetic selection of peptide aptamers that recognize and inhibit cyclin-dependent kinase 2, Nature, vol.380, pp.548-550, 1996.

A. P. Mann, R. C. Bhavane, A. Somasunderam, L. Montalvoortiz, B. Ghaghada et al., Thioaptamer conjugated liposomes for tumor vasculature targeting, Oncotarget, vol.2, pp.298-304

C. Borghouts, C. Kunz, and B. Groner, Peptide aptamers: recent developments for cancer therapy, Expert Opin Biol Ther, vol.5, pp.783-797, 2005.

I. C. Baines and P. Colas, Peptide aptamers as guides for smallmolecule drug discovery, Drug Discov Today, vol.11, pp.334-341, 2006.

R. C. Ladner, Constrained peptides as binding entities, Trends Biotechnol, vol.13, pp.426-430, 1995.

O. C. Farokhzad, J. Cheng, B. A. Teply, I. Sherifi, J. S. Kantoff et al., Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo, Proc Natl Acad Sci, vol.103, pp.6315-6320, 2006.

A. F. Rhee, J. W. Richie, J. P. Radovic-moreno, A. F. Langer, R. Farokhzad et al., New frontiers in nanotechnology for cancer treatment, Urol Oncol, vol.26, pp.74-85, 2008.

E. W. Ng, D. T. Shima, P. Calias, E. T. Cunningham, J. Guyer et al., Adamis AP. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease, Nat Rev Drug Discov, vol.5, pp.123-132, 2006.

S. Soundararajan, L. Wang, V. Sridharan, W. Chen, C. et al., Plasma membrane nucleolin is a receptor for the anticancer aptamer AS1411 in MV4-11 leukemia cells, Mol Pharmacol, vol.76, pp.984-991, 2009.

K. Butz, C. Denk, A. Ullmann, M. Scheffner, and F. Hoppe-seyler, Induction of apoptosis in human papillomaviruspositive cancer cells by peptide aptamers targeting the viral E6 oncoprotein, Proc Natl Acad Sci, vol.97, pp.6693-6697, 2000.

C. Kunz, C. Borghouts, C. Buerger, and B. Groner, Peptide aptamers with binding specificity for the intracellular domain of the ErbB2 receptor interfere with AKT signaling and sensitize breast cancer cells to Taxol, Mol Cancer Res, vol.4, pp.983-998, 2006.

E. Guida, A. Bisso, C. Fenollar-ferrer, M. Napoli, C. Anselmi et al., Peptide aptamers targeting mutant p53 induce apoptosis in tumor cells, Cancer Res, vol.68, pp.6550-6558, 2008.

A. L. Rerole, J. Gobbo, D. Thonel, A. Schmitt, E. et al., Peptides and Aptamers Targeting HSP70: A Novel Approach for Anticancer Chemotherapy, Cancer Res, 2011.

B. Gibert, E. Hadchity, A. Czekalla, M. T. Aloy, P. Colas et al., Inhibition of heat shock protein 27 (HspB1) tumorigenic functions by peptide aptamers, Oncogene, 2011.

L. Tourneau, C. Lee, J. J. Siu, and L. L. , Dose escalation methods in phase I cancer clinical trials, J Natl Cancer Inst, vol.101, pp.708-720, 2009.

L. Tourneau, C. Dieras, V. Tresca, P. Cacheux, W. Paoletti et al., Current challenges for the early clinical development of anticancer drugs in the era of molecularly targeted agents, Target Oncol, vol.5, pp.65-72

T. R. Fleming and D. L. Demets, Surrogate End Points in Clinical Trials: Are We Being Misled?, Ann Intern Med, pp.605-613, 1996.

S. Johnson, D. Evans, S. Laurenson, D. Paul, A. G. Davies et al., Surface-immobilized peptide aptamers as probe molecules for protein detection, Anal Chem, vol.80, pp.978-983, 2008.

, Getting personal, Nature, vol.473, p.253, 2011.

R. A. Weinberg, The Biology of Cancer, 2006.

G. Weissmann, C. Bernard, and J. Folkman, nothing makes sense in medicine except in the light of biology, Faseb J, vol.22, pp.943-946, 2008.

R. G. Seigneuric, J. L. Chasse, P. Auger, and A. Bardou, Simulated interactions between a Class III antiarrhythmic drug and a figure 8 reentry, Acta Biotheor, vol.53, pp.265-275, 2005.

W. A. Catterall, Ion channel voltage sensors: structure, function, and pathophysiology, Neuron, vol.67, pp.915-928, 2010.

H. Wulff, N. A. Castle, and L. A. Pardo, Voltage-gated potassium channels as therapeutic targets, Nat Rev Drug Discov, vol.8, pp.982-1001, 2009.
DOI : 10.1038/nrd2983

URL : http://europepmc.org/articles/pmc2790170?pdf=render

G. Manning, D. B. Whyte, R. Martinez, T. Hunter, and S. Sudarsanam, The protein kinase complement of the human genome, Science, vol.298, pp.1912-1934, 2002.

M. Vieth, J. J. Sutherland, D. H. Robertson, and R. M. Campbell, Kinomics: characterizing the therapeutically validated kinase space, Drug Discov Today, vol.10, pp.839-846, 2005.
DOI : 10.1016/s1359-6446(05)03477-x

P. Colas, The eleven-year switch of peptide aptamers, J Biol, vol.7, issue.2, 2008.
DOI : 10.1186/jbiol64

URL : https://hal.archives-ouvertes.fr/hal-00416052

M. B. Sporn, Perspective: The big C-for Chemoprevention, Nature, vol.471, pp.10-11, 2011.

E. D. Pleasance, P. J. Stephens, S. O'meara, D. J. Mcbride, A. Meynert et al., A small-cell lung cancer genome with complex signatures of tobacco exposure, Oncotarget, vol.463, pp.557-561, 2011.

, Frantz S. Drug discovery: playing dirty, Nature, vol.437, pp.942-943, 2005.

G. V. Paolini, R. H. Shapland, W. P. Van-hoorn, J. S. Mason, and A. L. Hopkins, Global mapping of pharmacological space, Nat Biotechnol, vol.24, pp.805-815, 2006.
DOI : 10.1038/nbt1228

M. A. Yildirim, K. I. Goh, M. E. Cusick, A. L. Barabasi, and M. Vidal, Drug-target network, Nat Biotechnol, vol.25, pp.1119-1126, 2007.

N. Neznanov, A. P. Komarov, L. Neznanova, P. Stanhope-baker, A. Gudkov et al., primary tumor), (ii) entering the circulation (intravasation), (iii) exiting the circulation (extravasation), (iv) the colonization of distant organs (Fidler, 2003). Current clinical investigations into the detection of CTCs are based on the cell receptor EpCAM, Proteotoxic stress targeted therapy (PSTT): induction of protein misfolding enhances the antitumor effect of the proteasome inhibitor bortezomib, vol.2, p.70, 2002.

(. Modi, The problem of this intrinsic variability in HSP expression may be circumvented by using time series, assessing HSPs with longitudinal data by following-up every single patient as a function of time. In order to compare HSP levels between patients, adjusting for age, gender, and ethnicity may be necessary. The role of circulating HSP70 and/or HSP70 antibodies in the peripheral circulation in the context of aging is interesting, 1998.

N. Arispe, M. Doh, O. Simakova, B. Kurganov, D. Maio et al., Hsc70 and Hsp70 interact with phosphatidylserine on the surface of PC12 cells resulting in a decrease of viability, FASEB J, vol.18, pp.1636-1645, 2004.

S. Banerjee, C. F. Lin, K. A. Skinner, L. M. Schiffhauer, J. Peacock et al., Heat shock protein 27 differentiates tolerogenic macrophages that may support human breast cancer progression, Cancer Res, vol.71, pp.318-327, 2011.
DOI : 10.1158/0008-5472.can-10-1778

URL : http://cancerres.aacrjournals.org/content/71/2/318.full.pdf

S. Basu, R. J. Binder, R. Suto, K. M. Anderson, and P. K. Srivastava, Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NFkappa B pathway, Int. Immunol, vol.12, pp.1539-1546, 2000.

J. M. Bruey, C. Ducasse, P. Bonniaud, L. Ravagnan, S. A. Susin et al., Hsp27 negatively regulates cell death by interacting with cytochrome c, Nat. Cell Biol, vol.2, pp.645-652, 2000.
DOI : 10.1038/35023595

S. K. Calderwood, Heat shock proteins in breast cancer progression-a suitable case for treatment?, Int. J. Hyperthermia, vol.26, pp.681-685, 2010.

C. Castilla, B. Congregado, J. M. Conde, R. Medina, F. J. Torrubia et al., Immunohistochemical expression of Hsp60 correlates with tumor progression and hormone resistance in prostate cancer, Urology, vol.76, pp.1-6, 2010.

F. Chalmin, S. Ladoire, G. Mignot, J. Vincent, M. Bruchard et al., , 2010.

, Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells, J. Clin. Invest, vol.120, pp.457-471

A. K. Chaturvedi, N. E. Caporaso, H. A. Katki, H. L. Wong, N. Chatterjee et al., C-reactive protein and risk of lung cancer, J. Clin. Oncol, vol.28, pp.2719-2726, 2010.

D. R. Ciocca, C. , and S. K. , Heat shock proteins in cancer: diagnostic, prognostic, predictive, and treatment implications, Cell Stress Chaperones, vol.10, pp.86-103, 2005.

P. A. Cornford, A. R. Dodson, K. F. Parsons, A. D. Desmond, A. Woolfenden et al., Heat shock protein expression independently predicts clinical outcome in prostate cancer, Cancer Res, vol.60, pp.7099-7105, 2000.

A. De-maio, Extracellular heat shock proteins, cellular export vesicles, and the stress observation system: a form of communication during injury, infection, and cell damage. It is never known how far a controversial finding will go! Dedicated to Ferruccio Ritossa, Cell Stress Chaperones, vol.16, pp.235-249, 2011.

C. Desmetz, F. Bibeau, F. Boissiere, V. Bellet, P. Rouanet et al., , 2008.

, Proteomics-based identification of HSP60 as a tumor-associated antigen in early stage breast cancer and ductal carcinoma in situ, J. Proteome Res, vol.7, pp.3830-3837

D. Tommaso, L. Destro, A. Seok, J. Y. Balladore, E. Terracciano et al., The application of markers (HSP70 GPC3 and GS) in liver biopsies is useful for detection of hepatocellular carcinoma, J. Hepatol, vol.50, pp.746-754, 2009.

C. Didelot, D. Lanneau, M. Brunet, A. L. Joly, A. De-thonel et al., Anti-cancer therapeutic approaches based on intracellular and extracellular heat shock proteins, Curr. Med. Chem, vol.14, pp.2839-2847, 2007.

C. Dorard, A. De-thonel, A. Collura, L. Marisa, M. Svrcek et al., Expression of a mutant HSP110 sensitizes colorectal cancer cells to chemotherapy and improves disease prognosis, Nat. Med, 2011.

M. J. Duffy, Serum tumor markers in breast cancer: are they of clinical value?, Clin. Chem, vol.52, pp.345-351, 2006.

G. O. Elpek, S. Karaveli, T. Simsek, N. Keles, and N. H. Aksoy, Expression of heat-shock proteins hsp27, hsp70 and hsp90 in malignant epithelial tumour of the ovaries, APMIS, vol.111, pp.523-530, 2003.

A. L. Evdonin, M. G. Martynova, O. A. Bystrova, I. V. Guzhova, B. A. Margulis et al., The release of Hsp70 from A431 carcinoma cells is mediated by secretory-like granules, Eur. J. Cell Biol, vol.85, pp.443-455, 2006.

I. J. Fidler, The pathogenesis of cancer metastasis: the "seed and soil" hypothesis revisited, Nat. Rev. Cancer, vol.3, pp.453-458, 2003.

Y. Fujita, T. Nakanishi, Y. Miyamoto, M. Hiramatsu, H. Mabuchi et al., Lett, vol.263, pp.280-290, 2008.

G. Fung, R. Seigneuric, S. Krishnan, R. B. Rao, B. G. Wouters et al., Reducing a biomarkers list via mathematical programming: application to gene signatures to detect time-dependent hypoxia in cancer, Icmla 2007: Sixth International Conference On Machine Learning And Applications, Proceedings, pp.482-487, 2007.

S. Gallucci and P. Matzinger, Danger signals: SOS to the immune system, Curr. Opin. Immunol, vol.13, pp.114-119, 2001.

C. Garrido, M. Brunet, C. Didelot, Y. Zermati, E. Schmitt et al., Heat shock proteins 27 and 70: anti-apoptotic proteins with tumorigenic properties, Cell Cycle, vol.5, pp.2592-2601, 2006.

M. Ghayour-mobarhan, H. Saber, and G. A. Ferns, The potential role of heat shock protein 27 in cardiovascular disease, Clin. Chim. Acta, 2011.

A. Glaessgen, S. Jonmarker, A. Lindberg, B. Nilsson, R. Lewensohn et al., Heat shock proteins 27, 60 and 70 as prognostic markers of prostate cancer, APMIS, vol.116, pp.888-895, 2008.

B. Hamrita, K. Chahed, M. Kabbage, C. L. Guillier, M. Trimeche et al., Identification of tumor antigens that elicit a humoral immune response in breast cancer patients' sera by sero, 2008.

, Clin. Chim. Acta, vol.393, pp.95-102

L. E. Hightower and P. T. Guidon, Selective release from cultured mammalian cells of heatshock (stress) proteins that resemble glia-axon transfer proteins, J. Cell. Physiol, vol.138, pp.257-266, 1989.

C. Hunter-lavin, E. L. Davies, M. M. Bacelar, M. J. Marshall, S. M. Andrew et al., Hsp70 release from peripheral blood mononuclear cells, Biochem. Biophys. Res. Commun, vol.324, pp.511-517, 2004.

Y. J. Hwang, S. P. Lee, S. Y. Kim, Y. H. Choi, M. J. Kim et al., Expression of heat shock protein 60 kDa is upregulated in cervical cancer, 2009.

, J, vol.50, pp.399-406

J. P. Ioannidis, Why most published research findings are false, PLoS. Med, vol.2, p.124, 2005.

A. Isidoro, E. Casado, A. Redondo, P. Acebo, E. Espinosa et al., Breast carcinomas fulfill the Warburg hypothesis and provide metabolic markers of cancer prognosis, Carcinogenesis, vol.26, pp.2095-2104, 2005.

G. Jego, A. Hazoumé, R. Seigneuric, and C. Garrido, Targeting heat shock proteins in cancer, Cancer Lett, 2010.

A. L. Joly, G. Wettstein, G. Mignot, F. Ghiringhelli, and C. Garrido, Dual role of heat shock proteins as regulators of apoptosis and innate immunity, J. Innate Immun, vol.2, pp.238-247, 2010.

A. Kamal, L. Thao, J. Sensintaffar, L. Zhang, M. F. Boehm et al., A highaffinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors, Nature, vol.425, pp.407-410, 2003.

H. H. Kampinga, J. Hageman, M. J. Vos, H. Kubota, R. M. Tanguay et al., Guidelines for the nomenclature of the human heat shock proteins, Cell Stress Chaperones, vol.14, pp.105-111, 2009.

A. A. Khalil, N. F. Kabapy, S. F. Deraz, and C. Smith, Heat shock proteins in oncology: diagnostic biomarkers or therapeutic targets?, Biochim. Biophys. Acta, vol.1816, pp.89-104, 2011.

J. Kocsis, B. Madaras, E. K. Toth, G. Fust, and Z. Prohaszka, Serum level of soluble 70-kD heat shock protein is associated with high mortality in patients with colorectal cancer without distant metastasis, Cell Stress Chaperones, vol.15, pp.143-151, 2010.

K. E. Krueger, The potential of serum proteomics for detection of cancer: promise or only hope?, Onkologie, vol.29, pp.498-499, 2006.

S. P. Langdon, G. J. Rabiasz, G. L. Hirst, R. J. King, R. A. Hawkins et al., Expression of the heat shock protein HSP27 in human ovarian cancer, Clin. Cancer Res, vol.1, pp.1603-1609, 1995.

, Frontiers in Oncology | Molecular and Cellular Oncology, vol.1, issue.8, 2011.

. Seigneuric, HSPs as cancer danger signals

D. Lanneau, G. Wettstein, P. Bonniaud, and C. Garrido, Heat shock proteins: cell protection through protein triage, ScientificWorldJournal, vol.10, pp.1543-1552, 2010.

T. Lebret, R. W. Watson, V. Molinie, A. O'neill, C. Gabriel et al., Heat shock proteins HSP27, HSP60, HSP70, and HSP90: expression in bladder carcinoma, Cancer, vol.98, pp.970-977, 2003.

H. Ledford, Cancer theory faces doubts, Nature, vol.472, p.273, 2011.

I. N. Lee, C. H. Chen, J. C. Sheu, H. S. Lee, G. T. Huang et al., Identification of human hepatocellular carcinoma-related biomarkers by two-dimensional difference gel electrophoresis and mass spectrometry, J. Proteome Res, vol.4, pp.2062-2069, 2005.

S. O. Lim, S. G. Park, J. H. Yoo, Y. M. Park, H. J. Kim et al., Expression of heat shock proteins (HSP27, HSP60, HSP70, HSP90, GRP78, GRP94) in hepatitis B virus-related hepatocellular carcinomas and dysplastic nodules, World J. Gastroenterol, vol.11, pp.2072-2079, 2005.

K. S. Looi, E. S. Nakayasu, R. A. Diaz, E. M. Tan, I. C. Almeida et al., Using proteomic approach to identify tumorassociated antigens as markers in hepatocellular carcinoma, J. Proteome Res, vol.7, pp.4004-4012, 2008.

A. J. Macario, F. Cappello, G. Zummo, C. De-macario, and E. , Chaperonopathies of senescence and the scrambling of interactions between the chaperoning and the immune systems, Ann. N. Y. Acad. Sci, vol.1197, pp.85-93, 2011.

A. J. Macario, C. De-macario, and E. , The pathology of cellular anti-stress mechanisms: a new frontier, Stress, vol.7, pp.243-249, 2004.

A. J. Macario, C. De-macario, and E. , Sick chaperones, cellular stress, and disease, N. Engl. J. Med, vol.353, pp.1489-1501, 2005.

S. S. Mambula, C. , and S. K. , Heat shock protein 70 is secreted from tumor cells by a nonclassical pathway involving lysosomal endosomes, J. Immunol, vol.177, pp.7849-7857, 2006.

H. Mischak, G. Allmaier, R. Apweiler, T. Attwood, M. Baumann et al., Recommendations for biomarker identification and qualification in clinical proteomics, vol.2, pp.46-88, 2010.

H. Miyake, M. Muramaki, T. Kurahashi, A. Takenaka, and M. Fujisawa, Expression of potential molecular markers in prostate cancer: correlation with clinicopathological outcomes in patients undergoing radical prostatectomy, Urol. Oncol, vol.28, pp.145-151, 2010.

H. Miyake, M. Muramaki, T. Kurahashi, K. Yamanaka, I. Hara et al., Enhanced expression of heat shock protein 27 following neoadjuvant hormonal therapy is associated with poor clinical outcome in patients undergoing radical prostatectomy for prostate cancer, Anticancer Res, vol.26, pp.1583-1587, 2006.

S. Modi, A. T. Stopeck, M. S. Gordon, D. Mendelson, D. B. Solit et al., Combination of trastuzumab and tanespimycin (17AAG, KOS-953) is safe and active in trastuzumab-refractory HER-2 overexpressing breast cancer: a phase I dose-escalation study, J. Clin. Oncol, vol.25, pp.5410-5417, 2007.

D. Mori, Y. Nakafusa, K. Miyazaki, and O. Tokunaga, Differential expression of Janus kinase 3 (JAK3), matrix metalloproteinase 13 (MMP13), heat shock protein 60 (HSP60), and mouse double minute 2 (MDM2) in human colorectal cancer progression using human cancer cDNA microarrays, Pathol. Res. Pract, vol.201, pp.777-789, 2005.

W. Nickel and M. Seedorf, Unconventional mechanisms of protein transport to the cell surface of eukaryotic cells, Annu. Rev. Cell Dev. Biol, vol.24, pp.287-308, 2008.

R. Njemini, I. Bautmans, O. O. Onyema, K. Van-puyvelde, C. Demanet et al., Circulating heat shock protein 70 in health, aging and disease, BMC Immunol, vol.12, p.24, 2011.

R. Njemini, C. Demanet, and T. Mets, Comparison of two ELISAs for the determination of Hsp70 in serum, J. Immunol. Methods, vol.306, pp.176-182, 2005.

A. Olejek, A. Damasiewicz-bodzek, P. Bodzek, T. Wielkoszynski, J. Zamlynski et al., Concentrations of antibodies against heat shock protein 27 in the sera of women with ovarian carcinoma, Int. J. Gynecol. Cancer, vol.19, pp.1516-1520, 2009.

E. F. Petricoin, A. M. Ardekani, B. A. Hitt, P. J. Levine, V. A. Fusaro et al., Use of proteomic patterns in serum to identify ovarian cancer, Lancet, vol.359, pp.572-577, 2002.

E. Pick, Y. Kluger, J. M. Giltnane, C. Moeder, R. L. Camp et al., , 2007.

, High HSP90 expression is associated with decreased survival in breast cancer, Cancer Res, vol.67, pp.2932-2937

A. G. Pockley, J. Shepherd, C. , and J. M. , Detection of heat shock protein 70 (Hsp70) and antiHsp70 antibodies in the serum of normal individuals, Immunol. Invest, vol.27, pp.367-377, 1998.

R. K. Ramanathan, M. J. Egorin, C. Erlichman, S. C. Remick, S. S. Ramalingam et al., Phase I pharmacokinetic and pharmacodynamic study of 17-dimethylaminoethylamino17-demethoxygeldanamycin, an inhibitor of heat-shock protein 90, in patients with advanced solid tumors, J. Clin. Oncol, vol.28, pp.1520-1526, 2010.

C. G. Rao, D. Chianese, G. V. Doyle, M. C. Miller, T. Russell et al., Expression of epithelial cell adhesion molecule in carcinoma cells present in blood and primary and metastatic tumors, Int. J. Oncol, vol.27, pp.49-57, 2005.

I. M. Rea, S. Mcnerlan, and A. G. Pockley, Serum heat shock protein and anti-heat shock protein antibody levels in aging, Exp. Gerontol, vol.36, pp.341-352, 2001.

Z. Rui, J. Jian-guo, T. Yuan-peng, P. Hai, B. et al., Use of serological proteomic methods to find biomarkers associated with breast cancer, Proteomics, vol.3, pp.433-439, 2003.
DOI : 10.1002/pmic.200390058

I. Sakai, H. Miyake, A. Takenaka, and M. Fujisawa, Expression of potential molecular markers in renal cell carcinoma: impact on clinicopathological outcomes in patients undergoing radical nephrectomy, BJU Int, vol.104, pp.942-946, 2009.

D. Schilling, M. Gehrmann, C. Steinem, A. De-maio, A. G. Pockley et al., Binding of heat shock protein 70 to extracellular phosphatidylserine promotes killing of normoxic and hypoxic tumor cells, FASEB J, vol.23, pp.2467-2477, 2009.

E. Schmitt, M. Gehrmann, M. Brunet, G. Multhoff, and C. Garrido, Intracellular and extracellular functions of heat shock proteins: repercussions in cancer therapy, J. Leukoc. Biol, vol.81, pp.15-27, 2007.

V. Sebastiani, C. Botti, U. Di-tondo, P. Visca, L. Pizzuti et al., Tissue microarray analysis of FAS, Bcl-2, Bcl-x, ER, PgR, Hsp60, p53 and Her2-neu in breast carcinoma, Anticancer Res, vol.26, pp.2983-2987, 2006.

R. Seigneuric, J. Gobbo, P. Colas, and C. Garrido, Targeting cancer with peptide aptamers, Oncotarget, vol.2, pp.557-561, 2011.
DOI : 10.18632/oncotarget.297

URL : http://www.oncotarget.com/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=297&path%5B%5D=470

R. Seigneuric, L. Markey, D. S. Nuyten, C. Dubernet, C. T. Evelo et al., From nanotechnology to nanomedicine: applications to cancer research, Curr. Mol. Med, vol.10, pp.640-652, 2010.
DOI : 10.2174/156652410792630634

R. Seigneuric, N. A. Van-riel, M. H. Starmans, A. Van-erk, C. T. Evelo et al., Systems Biology Applied to Cancer Research. Hershey, NY: Medical Information Science Reference, 2009.

A. Shamaei-tousi, A. Steptoe, K. O'donnell, J. Palmen, J. W. Stephens et al., Plasma heat shock protein 60 and cardiovascular disease risk: the role of psychosocial, genetic, and biological factors, Cell Stress Chaperones, vol.12, pp.384-392, 2007.

D. B. Solit, C. , and G. , Development and application of Hsp90 inhibitors, Drug Discov. Today, vol.13, pp.38-43, 2008.

C. H. Song, S. Y. Park, K. Y. Eom, J. H. Kim, S. W. Kim et al., Potential prognostic value of heat-shock protein 90 in the presence of phosphatidylinositol3-kinase overexpression or loss of PTEN, in invasive breast cancers, Proc. Natl. Acad. Sci. U.S.A, vol.12, pp.733-738, 2010.

K. Suzuki, Y. Ito, K. Wakai, M. Kawado, S. Hashimoto et al., Serum heat shock protein 70 levels and lung cancer risk: a case-control study nested in a large cohort study. Cancer Epidemiol, Biomarkers Prev, vol.15, pp.1733-1737, 2006.

M. Taipale, D. F. Jarosz, and S. Lindquist, HSP90 at the hub of protein homeostasis: emerging mechanistic insights, Nat. Rev. Mol. Cell Biol, vol.11, pp.515-528, 2010.

D. F. Terry, D. F. Wyszynski, V. G. Nolan, G. Atzmon, E. A. Schoenhofen et al., Serum heat shock protein 70 level as a biomarker of exceptional longevity, Mech. Ageing Dev, vol.127, pp.862-868, 2006.

J. P. Thiery, Epithelialmesenchymal transitions in tumour progression, Nat. Rev. Cancer, vol.2, pp.442-454, 2002.

J. P. Thiery, H. Acloque, R. Y. Huang, and M. A. Nieto, , 2009.

, Epithelial-mesenchymal transitions in development and disease, Cell, vol.139, pp.871-890

V. L. Vega, M. Rodriguez-silva, T. Frey, M. Gehrmann, J. C. Diaz et al., Hsp70 translocates into the plasma membrane after stress and is released into the extracellular environment in a membrane-associated form that activates macrophages, J. Immunol, vol.180, pp.4299-4307, 2008.

L. Whitesell and S. L. Lindquist, HSP90 and the chaperoning of cancer, Nat. Rev. Cancer, vol.5, pp.761-772, 2005.

M. S. Willis and C. Patterson, Hold me tight: role of the heat shock protein family of chaperones in cardiac disease, Circulation, vol.122, pp.1740-1751, 2010.

L. Xu, S. Chen, and R. C. Bergan, MAPKAPK2 and HSP27 are downstream effectors of p38 MAP kinase-mediated matrix metalloproteinase type 2 activation and cell invasion in human prostate cancer, Oncogene, vol.25, pp.2987-2998, 2006.

. Bibliographie,

C. Admyre, S. M. Johansson, K. R. Qazi, J. Filén, R. Lahesmaa et al., Exosomes with immune modulatory features are present in human breast milk, Journal of immunology, vol.179, issue.3, pp.1969-78, 1950.

J. Amin, J. Ananthan, and R. Voellmy, Key Features of Heat Shock Regulatory Elements, vol.8, 1988.

F. Andre, B. Escudier, E. Angevin, T. Tursz, and L. Zitvogel, Annals of oncology : official journal of the European Society for, Medical Oncology / ESMO, vol.15, issue.4, 2004.

A. Appert, C. Nam, N. Lobato, E. Priego, R. N. Miguel et al., , 2009.

, Targeting LMO2 with a peptide aptamer establishes a necessary function in overt T-cell neoplasia, Cancer research, vol.69, issue.11

A. Asea, E. Kabingu, M. A. Stevenson, and S. K. Calderwood, HSP70 peptidembearing and peptide-negative preparations act as chaperokines, Cell stress & chaperones, vol.5, issue.5, pp.425-456, 2000.

A. Asea, C. Jean-pierre, P. Kaur, P. Rao, I. M. Linhares et al., Heat shock protein-containing exosomes in mid-trimester amniotic fluids, Journal of reproductive immunology, vol.79, issue.1, pp.12-19, 2008.

R. Bagatell and L. Whitesell, Altered Hsp90 function in cancer : A unique therapeutic opportunity Minireview Altered Hsp90 function in cancer : A unique therapeutic opportunity, pp.1021-1030, 2004.

I. C. Baines and P. Colas, Peptide aptamers as guides for small-molecule drug discovery. Drug discovery today, vol.11, pp.334-375, 2006.

G. M. Balaburski, J. I. Leu, N. Beeharry, S. Hayik, M. D. Andrake et al., A modified HSP70 inhibitor shows broad activity as an anticancer agent, 2013.

L. Balaj, R. Lessard, L. Dai, Y. Cho, S. L. Pomeroy et al., Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences, Nature communications, vol.2, p.180, 2011.

J. J. Barrott, T. Haystead, and J. , Hsp90, an unlikely ally in the war on cancer, The FEBS journal, vol.280, issue.6, pp.1381-96, 2013.

S. Basu, R. J. Binder, R. Suto, K. M. Anderson, and P. K. Srivastava, Necrotic but not apoptotic cell death releases heat shock proteins , which deliver a partial maturation signal to dendritic cells and activate the NF-? B pathway, pp.1539-1546, 2000.

H. Bausinger, D. Lipsker, U. Ziylan, S. Manié, J. Briand et al., , 2002.

, Endotoxin-free heat-shock protein 70 fails to induce APC activation, European journal of immunology, vol.32, issue.12, pp.3708-3721

V. Baylot, C. Andrieu, M. Katsogiannou, D. Taieb, S. Garcia et al., , 2011.

, OGX-427 inhibits tumor progression and enhances gemcitabine chemotherapy in pancreatic cancer. Cell death & disease, 2, e221

H. M. Beere, B. B. Wolf, K. Cain, D. D. Mosser, A. Mahboubi et al., Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome, Nature cell biology, vol.2, issue.8, pp.469-75, 2000.

H. Bendz, S. C. Ruhland, J. Maya, O. Hainzl, S. Riegelsberger et al., Human Heat Shock Protein 70 Enhances Tumor Antigen Presentation through Complex Formation and Intracellular Antigen Delivery without Innate Immune Signaling * ?, 2007.

R. J. Binder, N. E. Blachere, and P. K. Srivastava, Heat shock protein-chaperoned peptides but not free peptides introduced into the cytosol are presented efficiently by major histocompatibility complex I molecules, The Journal of biological chemistry, vol.276, issue.20, pp.17163-71, 2001.

A. Bobrie, M. Colombo, G. Raposo, and C. Théry, Exosome secretion: molecular mechanisms and roles in immune responses, Traffic, issue.12, pp.1659-68, 2011.

T. J. Borges, L. Wieten, M. J. Van-herwijnen, F. Broere, R. Van-der-zee et al., The anti-inflammatory mechanisms of Hsp70, Frontiers in immunology, vol.3, p.95, 2012.

P. Bottoni, B. Giardina, and R. Scatena, Proteomic profiling of heat shock proteins: An emerging molecular approach with direct pathophysiological and clinical implications, Proteomics. Clinical applications, vol.3, issue.6, pp.636-53, 2009.

M. J. Braunstein, S. S. Scott, C. M. Scott, S. Behrman, P. Walter et al., , 2011.

, Antimyeloma Effects of the Heat Shock Protein 70 Molecular Chaperone Inhibitor MAL3-101, Journal of oncology, p.232037, 2011.

J. M. Bruey, C. Ducasse, P. Bonniaud, L. Ravagnan, S. A. Susin et al., Hsp27 negatively regulates cell death by interacting with cytochrome c, Nature cell biology, vol.2, issue.9, pp.645-52, 2000.

M. Brunet-simioni, L. De-thonel, G. Bossis, E. Fourmaux, and C. Garrido, Heat shock protein 27 is involved in SUMO-2/3 modification of heat shock factor 1 and thereby modulates the transcription factor activity, Oncogene, vol.28, issue.37, pp.3332-3376, 2009.

S. I. Buschow, E. N. Nolte-'t-hoen, G. Van-niel, M. S. Pols, T. Ten-broeke et al., MHC II in dendritic cells is targeted to lysosomes or T cell-induced exosomes via distinct multivesicular body pathways, Traffic, issue.10, pp.1528-1570, 2009.

F. Chalmin, S. Ladoire, G. Mignot, J. Vincent, M. Bruchard et al., Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells, vol.120, pp.457-471, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00451697

N. Chaput and C. Théry, Exosomes: immune properties and potential clinical implementations, Seminars in immunopathology, vol.33, issue.5, pp.419-459, 2011.

T. Chen and X. Cao, Stress for maintaining memory: HSP70 as a mobile messenger for innate and adaptive immunity, European journal of immunology, vol.40, issue.6, pp.1541-1545, 2010.

P. Colas, B. Cohen, T. Jessen, I. Grishina, J. Mccoy et al., Genetic selection of peptide aptamers that recognize and inhibit cyclin-dependent kinase 2, Nature, vol.380, issue.6574, 1996.

M. Crawford, R. Woodman, and P. Ko-ferrigno, Peptide aptamers: tools for biology and drug discovery, Briefings in functional genomics & proteomics, vol.2, issue.1, pp.72-81, 2003.

A. G. Cuenca, M. J. Delano, K. M. Kelly-scumpia, C. Moreno, P. O. Scumpia et al., A paradoxical role for myeloid-derived suppressor cells in sepsis and trauma. Molecular medicine, vol.17, pp.281-92, 2011.

C. Dai, L. Whitesell, A. B. Rogers, and S. Lindquist, Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis, Cell, vol.130, issue.6, pp.1005-1023, 2007.

A. De-maio, Extracellular heat shock proteins, cellular export vesicles, and the Stress Observation System: a form of communication during injury, infection, and cell damage. It is never known how far a controversial finding will go! Dedicated to Ferruccio Ritossa, Cell stress & chaperones, vol.16, issue.3, pp.235-284, 2011.

A. De-maio and D. Vazquez, Extracellular Heat Shock Proteins: A New Location, a New Function, Shock, 2013.

A. De-thonel, V. Mezger, and C. Garrido, Implication of Heat Shock Factors in Tumorigenesis: Therapeutical Potential, Cancers, vol.3, issue.4, pp.1158-1181, 2011.

A. De-thonel, J. Vandekerckhove, D. Lanneau, S. Selvakumar, G. Courtois et al., HSP27 controls GATA-1 protein level during erythroid cell differentiation, Blood, vol.116, issue.1, pp.85-96, 2010.

C. Didelot, E. Schmitt, M. Brunet, L. Maingret, A. Parcellier et al., Heat shock proteins: endogenous modulators of apoptotic cell death, Handbook of experimental pharmacology, issue.172, pp.171-98, 2006.

C. M. Dobson, Protein folding and misfolding, Nature, vol.426, issue.6968, pp.884-90, 2003.

C. Dorard, A. De-thonel, A. Collura, L. Marisa, M. Svrcek et al., Expression of a mutant HSP110 sensitizes colorectal cancer cells to chemotherapy and improves disease prognosis, Nature medicine, vol.17, issue.10, pp.1283-1292, 2011.

D. Duijvesz, T. Luider, C. H. Bangma, and G. Jenster, Exosomes as biomarker treasure chests for prostate cancer, European urology, vol.59, issue.5, pp.823-854, 2011.

D. P. Easton, Y. Kaneko, and J. R. Subjeck, The Hsp110 and Grp170 stress proteins : newly recognized relatives of the Hsp70s INTRODUCTION, vol.5, pp.276-290, 2000.

C. G. Evans, L. Chang, and J. E. Gestwicki, Heat shock protein 70 (hsp70) as an emerging drug target, Journal of medicinal chemistry, vol.53, issue.12, pp.4585-602, 2010.

A. L. Evdonin, M. G. Martynova, O. A. Bystrova, I. V. Guzhova, B. A. Margulis et al., The release of Hsp70 from A431 carcinoma cells is mediated by secretory-like granules, European journal of cell biology, vol.85, issue.6, pp.443-55, 2006.

J. Fauré, G. Lachenal, M. Court, J. Hirrlinger, C. Chatellard-causse et al., Exosomes are released by cultured cortical neurones, Molecular and cellular neurosciences, vol.31, issue.4, pp.642-650, 2006.

M. Ferrarini, S. Heltai, M. R. Zocchi, and C. Rugarli, Unusual expression and localization of heat-shock proteins in human tumor cells, International journal of cancer. Journal international du cancer, vol.51, issue.4, pp.613-622, 1992.

B. Fevrier, D. Vilette, F. Archer, D. Loew, W. Faigle et al., Cells release prions in association with exosomes, Proceedings of the National Academy of Sciences of the United States of America, vol.101, issue.26, pp.9683-9691, 2004.

S. W. Fewell, B. W. Day, and J. L. Brodsky, Identification of an inhibitor of hsc70-mediated protein translocation and ATP hydrolysis, The Journal of biological chemistry, vol.276, issue.2, pp.910-914, 2001.

R. A. Floto, P. A. Macary, J. M. Boname, T. S. Mien, B. Kampmann et al., Dendritic cell stimulation by mycobacterial Hsp70 is mediated through CCR5, Science, issue.5798, pp.454-462, 2006.

M. Fujimoto, H. Izu, K. Seki, K. Fukuda, T. Nishida et al., HSF4 is required for normal cell growth and differentiation during mouse lens development, The EMBO journal, vol.23, issue.21, pp.4297-306, 2004.

A. Gallo and I. Alevizos, Isolation of circulating microRNA in saliva, Methods in molecular biology, vol.1024, pp.183-90, 2013.

B. Gao and M. Tsan, Endotoxin contamination in recombinant human heat shock protein 70 (Hsp70) preparation is responsible for the induction of tumor necrosis factor alpha release by murine macrophages, The Journal of biological chemistry, vol.278, issue.1, pp.174-183, 2003.

C. Garrido, L. Galluzzi, M. Brunet, P. E. Puig, C. Didelot et al., Mechanisms of cytochrome c release from mitochondria, Cell death and differentiation, vol.13, issue.9, pp.1423-1456, 2006.

C. Garrido and E. Solary, A role of HSPs in apoptosis through "protein triage, Cell death and differentiation, vol.10, issue.6, pp.619-639, 2003.

C. Garrido, A. Collura, K. Berthenet, A. Lagrange, and A. Duval, Mutation d ' HSP110 dans les cancers colorectaux, pp.9-46, 2012.

M. Gehrmann, G. Liebisch, G. Schmitz, R. Anderson, C. Steinem et al., Tumor-specific Hsp70 plasma membrane localization is enabled by the glycosphingolipid Gb3, PloS one, vol.3, issue.4, 2008.

A. R. Goloudina, O. N. Demidov, and C. Garrido, Inhibition of HSP70: a challenging anticancer strategy, Cancer letters, vol.325, issue.2, pp.117-141, 2012.

F. Guo, C. Sigua, P. Bali, P. George, W. Fiskus et al., Mechanistic role of heat shock protein 70 in Bcr-Abl-mediated resistance to apoptosis in human acute leukemia cells, Blood, vol.105, issue.3, pp.1246-55, 2005.

S. Gurbuxani, E. Schmitt, C. Cande, A. Parcellier, A. Hammann et al., Heat shock protein 70 binding inhibits the nuclear import of apoptosis-inducing factor, Oncogene, vol.22, issue.43, pp.6669-78, 2003.

B. N. Hannafon and W. Ding, Intercellular Communication by Exosome-Derived microRNAs in Cancer, International journal of molecular sciences, vol.14, issue.7, pp.14240-69, 2013.

M. Hantschel, K. Pfister, . Jordan, R. Scholz, R. Andreesen et al., Hsp70 plasma membrane expression on primary tumor biopsy material and bone marrow of leukemic patients, Cell stress & chaperones, vol.5, issue.5, pp.438-480, 2000.

S. Hao, Z. Ye, F. Li, Q. Meng, M. Qureshi et al., Epigenetic transfer of metastatic activity by uptake of highly metastatic B16 melanoma cell-released exosomes, Experimental oncology, vol.28, issue.2, pp.126-157, 2006.

M. Haug, C. P. Schepp, H. Kalbacher, G. E. Dannecker, and U. Holzer, 70-kDa heat shock proteins: specific interactions with HLA-DR molecules and their peptide fragments, European journal of immunology, vol.37, issue.4, pp.1053-63, 2007.

B. Henderson, S. K. Calderwood, A. R. Coates, I. Cohen, W. Van-eden et al., Caught with their PAMPs down? The extracellular signalling actions of molecular chaperones are not due to microbial contaminants, Cell stress & chaperones, vol.15, issue.2, pp.123-164, 2010.

L. E. Hightower and P. T. Guidon, Selective release from cultured mammalian cells of heatshock (stress) proteins that resemble glia-axon transfer proteins, Journal of cellular physiology, vol.138, issue.2, pp.257-66, 1989.

C. R. Ireson and L. R. Kelland, Discovery and development of anticancer aptamers, Molecular cancer therapeutics, vol.5, issue.12, pp.2957-62, 2006.

N. Isambert, A. Hollebecque, Y. Berge, H. , V. I. Brienza et al., A Phase I study of Debio 0932 , an oral HSP90 inhibitor, 2012.

M. Jäättelä, Over-expression of hsp70 confers tumorigenicity to mouse fibrosarcoma cells, International journal of cancer. Journal international du cancer, vol.60, issue.5, pp.689-93, 1995.

S. E. Jackson, Hsp90: structure and function, Topics in current chemistry, vol.328, pp.155-240, 2013.

G. Jego, A. Hazoumé, R. Seigneuric, and C. Garrido, Targeting heat shock proteins in cancer, Cancer letters, vol.332, issue.2, pp.275-85, 2013.

K. Jhaveri and S. Modi, HSP90 inhibitors for cancer therapy and overcoming drug resistance, Advances in pharmacology, vol.65, pp.471-517, 2012.

U. K. Jinwal, Y. Miyata, J. Koren, J. R. Jones, J. H. Trotter et al., Chemical manipulation of hsp70 ATPase activity regulates tau stability, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.29, issue.39, pp.12079-88, 2009.

R. M. Johnstone, M. Adam, J. R. Hammond, L. Orr, and C. Turbide, Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes), The Journal of biological chemistry, vol.262, issue.19, pp.9412-9432, 1987.

M. Kai, T. Nakatsura, H. Egami, S. Senju, Y. Nishimura et al., Heat shock protein 105 is overexpressed in a variety of human tumors, Oncology reports, vol.10, issue.6, pp.1777-82

M. Kallio, Y. Chang, M. Manuel, T. Alastalo, M. Rallu et al., Brain abnormalities, defective meiotic chromosome synapsis and female subfertility in HSF2 null mice, The EMBO journal, vol.21, issue.11, pp.2591-601, 2002.

A. Kamal, L. Thao, J. Sensintaffar, L. Zhang, M. F. Boehm et al., A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors, Nature, vol.425, issue.6956, pp.407-417, 2003.

H. H. Kampinga, J. Hageman, M. J. Vos, H. Kubota, R. M. Tanguay et al., Guidelines for the nomenclature of the human heat shock proteins, Cell stress & chaperones, vol.14, issue.1, pp.105-116, 2009.

J. Kao, E. C. Ko, S. Eisenstein, A. G. Sikora, S. Fu et al., Targeting immune suppressing myeloid-derived suppressor cells in oncology, Critical reviews in oncology/hematology, vol.77, issue.1, pp.12-21, 2011.

M. Katakowski, B. Buller, X. Zheng, Y. Lu, T. Rogers et al., Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth, Cancer letters, vol.335, issue.1, pp.201-205, 2013.

S. Kostenko and U. Moens, Heat shock protein 27 phosphorylation: kinases, phosphatases, functions and pathology. Cellular and molecular life sciences : CMLS, vol.66, pp.3289-307, 2009.

M. C. Kuppner, R. Gastpar, S. Gelwer, E. Nössner, O. Ochmann et al., The role of heat shock protein (hsp70) in dendritic cell maturation: hsp70 induces the maturation of immature dendritic cells but reduces DC differentiation from monocyte precursors, European journal of immunology, vol.31, issue.5, pp.1602-1611, 2001.

S. Kusmartsev, F. Cheng, B. Yu, Y. Nefedova, E. Sotomayor et al., , 2003.

, All-trans-retinoic acid eliminates immature myeloid cells from tumor-bearing mice and improves the effect of vaccination, Cancer research, vol.63, issue.15, pp.4441-4450

R. C. Lai, R. W. Yeo, K. H. Tan, and S. K. Lim, Exosomes for drug delivery-a novel application for the mesenchymal stem cell, Biotechnology advances, vol.31, issue.5, pp.543-51

J. E. Lancet, I. Gojo, M. Burton, M. Quinn, S. M. Tighe et al., Phase I study of the heat shock protein 90 inhibitor alvespimycin (KOS-1022, 17-DMAG) administered intravenously twice weekly to patients with acute myeloid leukemia, Leukemia, vol.24, issue.4, pp.699-705, 2010.

D. Lanneau, M. Brunet, E. Frisan, E. Solary, M. Fontenay et al., Heat shock proteins: essential proteins for apoptosis regulation, Journal of cellular and molecular medicine, vol.12, issue.3, pp.743-61, 2008.

L. Lattuada, A. Barge, G. Cravotto, G. B. Giovenzana, and L. Tei, The synthesis and application of polyamino polycarboxylic bifunctional chelating agents, Chemical Society reviews, vol.40, issue.5, pp.3019-3068, 2011.

J. I. Leu, J. Pimkina, A. Frank, M. E. Murphy, and D. L. George, A small molecule inhibitor of inducible heat shock protein 70, Molecular cell, vol.36, issue.1, pp.15-27, 2009.

J. Lewis, A. Devin, A. Miller, Y. Lin, Y. Rodriguez et al., Disruption of hsp90 function results in degradation of the death domain kinase, receptor-interacting protein (RIP), and blockage of tumor necrosis factor-induced nuclear factor-kappaB activation, The Journal of biological chemistry, vol.275, issue.14, pp.10519-10545, 2000.

C. Y. Li, J. S. Lee, Y. G. Ko, J. I. Kim, and J. S. Seo, Heat shock protein 70 inhibits apoptosis downstream of cytochrome c release and upstream of caspase-3 activation, The Journal of biological chemistry, vol.275, issue.33, pp.25665-71, 2000.

W. Liao, M. Wu, H. Wang, Y. Tien, and J. Lin, Serum heat shock protein 27 is increased in chronic pancreatitis and pancreatic carcinoma, Pancreas, vol.38, issue.4, pp.422-428, 2009.

D. Lindau, P. Gielen, M. Kroesen, P. Wesseling, and G. J. Adema, The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells, Immunology, vol.138, issue.2, pp.105-120, 2013.

S. Lindquist, The heat-shock response. Annual review of biochemistry, vol.55, pp.1151-91, 1986.

S. Liu, Bifunctional coupling agents for radiolabeling of biomolecules and target-speci fi c delivery of metallic radionuclides ?, vol.60, pp.1347-1370, 2008.

Y. Loriot, A. Zoubeidi, and M. E. Gleave, Targeted therapies in metastatic castration-resistant prostate cancer: beyond the androgen receptor, The Urologic clinics of North America, vol.39, issue.4, pp.517-548, 2012.

L. Lv, Y. Wan, Y. Lin, W. Zhang, M. Yang et al., Anticancer drugs cause release of exosomes with heat shock proteins from human hepatocellular carcinoma cells that elicit effective natural killer cell antitumor responses in vitro, The Journal of biological chemistry, vol.287, issue.19, pp.15874-85, 2012.

A. J. Macario and E. Conway-de-macario, Sick chaperones, cellular stress, and disease. The New England journal of medicine, vol.353, pp.1489-501, 2005.

S. S. Mambula, S. K. Calderwood, and E. Alerts, Nonclassical Pathway Involving Lysosomal Endosomes, vol.1, 2013.

D. H. Margineantu, C. B. Emerson, D. Diaz, and D. M. Hockenbery, Hsp90 inhibition decreases mitochondrial protein turnover, PloS one, vol.2, issue.10, p.1066, 2007.

R. U. Mattoo, S. K. Sharma, S. Priya, A. Finka, and P. Goloubinoff, Hsp110 is a bona fide chaperone using ATP to unfold stable misfolded polypeptides and reciprocally collaborate with hsp70 to solubilize protein aggregates, The Journal of biological chemistry, vol.288, issue.29, pp.21399-411, 2013.

J. R. Mcconnell and S. R. Mcalpine, Heat shock proteins 27, 40, and 70 as combinational and dual therapeutic cancer targets, Bioorganic & medicinal chemistry letters, vol.23, issue.7, pp.1923-1931, 2013.

D. R. Mcmillan, X. Xiao, L. Shao, K. Graves, and I. J. Benjamin, Targeted disruption of heat shock transcription factor 1 abolishes thermotolerance and protection against heat-inducible apoptosis, The Journal of biological chemistry, vol.273, issue.13, pp.7523-7531, 1998.

K. W. Moore, R. De-waal-malefyt, R. L. Coffman, and A. Garra, Interleukin-10 and the interleukin-10 receptor, Annual review of immunology, vol.19, pp.683-765, 2001.

G. Multhoff, Heat shock protein 70 ( Hsp70 ): Membrane location , export and immunological relevance, vol.43, pp.229-237, 2007.

G. Multhoffx, C. Botzler, M. Wiesnet, E. Muller, T. Meier et al., A STRESS-INDUCIBLE 72-kDa HEAT-SHOCK PROTEIN ( HSP72 ) IS EXPRESSED ON THE SURFACE OF HUMAN TUMOR CELLS, vol.279, pp.272-279, 1995.

M. E. Murphy, The HSP70 family and cancer, Carcinogenesis, vol.34, issue.6, pp.1181-1189, 2013.

S. Nagaraj, M. Collazo, C. A. Corzo, J. Youn, M. Ortiz et al., Regulatory myeloid suppressor cells in health and disease, Cancer research, vol.69, issue.19, pp.7503-7509, 2009.

A. C. Newton, Regulation of the ABC kinases by phosphorylation: protein kinase C as a paradigm, The Biochemical journal, vol.370, pp.361-71, 2003.

Y. Onishi, Z. Fehervari, T. Yamaguchi, and S. Sakaguchi, Foxp3+ natural regulatory T cells preferentially form aggregates on dendritic cells in vitro and actively inhibit their maturation, Proceedings of the National Academy of Sciences of the United States of America, vol.105, pp.10113-10121, 2008.

P. Ostling, J. K. Björk, P. Roos-mattjus, V. Mezger, and L. Sistonen, Heat shock factor 2 (HSF2) contributes to inducible expression of hsp genes through interplay with HSF1, The Journal of biological chemistry, vol.282, issue.10, pp.7077-86, 2007.

M. Ostrowski, N. B. Carmo, S. Krumeich, I. Fanget, G. Raposo et al., Rab27a and Rab27b control different steps of the exosome secretion pathway, Nature cell biology, vol.12, issue.1, pp.19-30, 2010.

M. Ottenhausen, I. Bodhinayake, M. Banu, K. Kesavabhotla, A. Ray et al., Industry progress report on neuro-oncology: Biotech update, Journal of neuro-oncology, 2013.

C. Paul, F. Manero, S. Gonin, C. Kretz-remy, S. Virot et al., Hsp27 as a negative regulator of cytochrome C release, Molecular and cellular biology, vol.22, issue.3, pp.816-850, 2002.
URL : https://hal.archives-ouvertes.fr/hal-00121161

H. Peinado, M. Ale?kovi?, S. Lavotshkin, I. Matei, B. Costa-silva et al., Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET, Nature medicine, vol.18, issue.6, pp.883-91, 2012.

K. Pfister, J. Radons, R. Busch, J. G. Tidball, M. Pfeifer et al., Patient survival by Hsp70 membrane phenotype: association with different routes of metastasis, Cancer, vol.110, issue.4, pp.926-961, 2007.

A. Poliakov, M. Spilman, T. Dokland, C. L. Amling, and J. A. Mobley, Structural heterogeneity and protein composition of exosome-like vesicles (prostasomes) in human semen, The Prostate, vol.69, issue.2, pp.159-67, 2009.

I. Poschke and R. Kiessling, On the armament and appearances of human myeloid-derived suppressor cells, Clinical immunology, vol.144, issue.3, pp.250-68, 2012.

P. Raber, A. C. Ochoa, and P. C. Rodríguez, Metabolism of L-arginine by myeloid-derived suppressor cells in cancer: mechanisms of T cell suppression and therapeutic perspectives, Immunological investigations, pp.614-648, 2012.

F. Raimondo, L. Morosi, C. Chinello, F. Magni, and M. Pitto, Advances in membranous vesicle and exosome proteomics improving biological understanding and biomarker discovery, Proteomics, vol.11, issue.4, 2011.

A. Rajan, R. J. Kelly, J. B. Trepel, Y. S. Kim, S. V. Alarcon et al., A phase I study of PF-04929113 (SNX-5422), an orally bioavailable heat shock protein 90 inhibitor, in patients with refractory solid tumor malignancies and lymphomas, Clinical cancer research : an official journal of the American Association for Cancer Research, vol.17, issue.21, pp.6831-6840, 2011.

S. Rana, S. Yue, D. Stadel, and M. Zöller, Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. The international journal of biochemistry & cell biology, vol.44, pp.1574-84, 2012.

F. Rappa, F. Farina, G. Zummo, S. David, C. Campanella et al., , 2012.

, HSP-molecular chaperones in cancer biogenesis and tumor therapy: an overview, Anticancer research, vol.32, issue.12, pp.5139-50

L. Ravagnan, S. Gurbuxani, S. A. Susin, C. Maisse, E. Daugas et al., Heat-shock protein 70 antagonizes apoptosis-inducing factor, vol.3, pp.839-843, 2001.
URL : https://hal.archives-ouvertes.fr/hal-01606079

A. Rebillard, X. Tekpli, O. Meurette, O. Sergent, G. Lemoigne-muller et al., Cisplatin-induced apoptosis involves membrane fluidification via inhibition of NHE1 in human colon cancer cells, Cancer research, vol.67, issue.16, pp.7865-74, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00690301

M. Retzlaff, F. Hagn, L. Mitschke, M. Hessling, F. Gugel et al., Asymmetric activation of the hsp90 dimer by its cochaperone aha1, Molecular cell, vol.37, issue.3, pp.344-54, 2010.

J. Ribeil, Y. Zermati, J. Vandekerckhove, S. Cathelin, J. Kersual et al., Hsp70 regulates erythropoiesis by preventing caspase-3-mediated cleavage of GATA-1, Nature, vol.445, issue.7123, pp.102-107, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00092601

M. C. Rosenhagen, C. S?ti, U. Schmidt, G. M. Wochnik, F. U. Hartl et al., The heat shock protein 90-targeting drug cisplatin selectively inhibits steroid receptor activation, Molecular endocrinology, issue.10, p.17, 1991.

K. Ruchalski, H. Mao, Z. Li, Z. Wang, S. Gillers et al., Distinct hsp70 domains mediate apoptosis-inducing factor release and nuclear accumulation, The Journal of biological chemistry, vol.281, issue.12, pp.7873-80, 2006.

C. Saegusa, T. Tanaka, S. Tani, S. Itohara, K. Mikoshiba et al., Decreased basal mucus secretion by Slp2-a-deficient gastric surface mucous cells. Genes to cells : devoted to molecular & cellular mechanisms, vol.11, pp.623-654, 2006.

T. Sakurai, M. Kudo, A. Umemura, G. He, A. M. Elsharkawy et al., P38? Inhibits Liver Fibrogenesis and Consequent Hepatocarcinogenesis By Curtailing Accumulation of Reactive Oxygen Species, Cancer research, vol.73, issue.1, pp.215-239, 2013.

A. Savina, M. Furlán, M. Vidal, and M. I. Colombo, Exosome release is regulated by a calcium-dependent mechanism in K562 cells, The Journal of biological chemistry, vol.278, issue.22, 2003.

D. Schilling, M. Gehrmann, C. Steinem, A. De-maio, A. G. Pockley et al., Binding of heat shock protein 70 to extracellular phosphatidylserine promotes killing of normoxic and hypoxic tumor cells, FASEB journal : official publication of the Federation of American Societies for Experimental Biology, vol.23, issue.8, pp.2467-77, 2009.

E. Schmitt, M. Gehrmann, M. Brunet, G. Multhoff, and C. Garrido, Intracellular and extracellular functions of heat shock proteins: repercussions in cancer therapy, Journal of leukocyte biology, vol.81, issue.1, pp.15-27, 2007.

E. Schmitt, L. Maingret, P. Puig, A. Rerole, F. Ghiringhelli et al., Heat shock protein 70 neutralization exerts potent antitumor effects in animal models of colon cancer and melanoma, Cancer research, vol.66, issue.8, pp.4191-4198, 2006.

E. Schmitt, A. Parcellier, S. Gurbuxani, C. Cande, A. Hammann et al., Chemosensitization by a Non-apoptogenic Heat Shock Protein 70-Binding ApoptosisInducing Factor Mutant Chemosensitization by a Non-apoptogenic Heat Shock Protein 70Binding Apoptosis-Inducing Factor Mutant, pp.8233-8240, 2003.

A. Schneider and M. Simons, Exosomes: vesicular carriers for intercellular communication in neurodegenerative disorders, Cell and tissue research, vol.352, issue.1, pp.33-47, 2013.

R. Seigneuric, H. Mjahed, J. Gobbo, A. Joly, K. Berthenet et al., Heat shock proteins as danger signals for cancer detection, Frontiers in oncology, vol.1, p.37, 2011.

K. Sidera and E. Patsavoudi, ND ES SC RIB ND ES SC RIB, vol.7, issue.10, pp.1564-1568, 2008.

R. Singh, S. Kolvraa, and S. I. Rattan, Genetics of human longevity with emphasis on the relevance of HSP70 as candidate genes, Frontiers in bioscience : a journal and virtual library, vol.12, pp.4504-4517, 2007.

O. Slaby, K. Sobkova, M. Svoboda, I. Garajova, P. Fabian et al., Significant overexpression of Hsp110 gene during colorectal cancer progression, pp.1235-1241, 2009.

S. Soga, S. Akinaga, and Y. Shiotsu, Hsp90 inhibitors as anti-cancer agents, from basic discoveries to clinical development, Current pharmaceutical design, vol.19, issue.3, pp.366-76, 2013.

R. Somasundaram and M. Herlyn, Melanoma exosomes: messengers of metastasis, Nature medicine, vol.18, issue.6, pp.853-857, 2012.

A. S. Sreedhar, E. Kalmár, P. Csermely, and Y. Shen, Hsp90 isoforms: functions, expression and clinical importance, FEBS letters, vol.562, issue.1-3, pp.11-16, 2004.

S. Stangl, M. Gehrmann, J. Riegger, K. Kuhs, I. Riederer et al., Targeting membrane heat-shock protein 70 (Hsp70) on tumors by cmHsp70.1 antibody, Proceedings of the National Academy of Sciences of the United States of America, vol.108, pp.733-741, 2011.

E. Strom, S. Sathe, P. G. Komarov, O. B. Chernova, I. Pavlovska et al., Small-molecule inhibitor of p53 binding to mitochondria protects mice from gamma radiation, Nature chemical biology, vol.2, issue.9, pp.474-483, 2006.

B. J. Tauro, D. W. Greening, R. A. Mathias, H. Ji, S. Mathivanan et al., Comparison of ultracentrifugation , density gradient separation , and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes, METHODS, 2012.

D. D. Taylor, S. Akyol, and C. Taylor, Pregnancy-associated exosomes and their modulation of T cell signaling, Journal of immunology, vol.176, issue.3, pp.1534-1576, 1950.

C. Théry, L. Duban, E. Segura, P. Véron, O. Lantz et al., Indirect activation of naïve CD4+ T cells by dendritic cell-derived exosomes, Nature immunology, vol.3, issue.12, pp.1156-62, 2002.

C. Théry, M. Ostrowski, and E. Segura, Membrane vesicles as conveyors of immune responses, Nature reviews. Immunology, vol.9, issue.8, pp.581-93, 2009.

C. Théry, L. Zitvogel, and S. Amigorena, Exosomes: composition, biogenesis and function, Nature reviews. Immunology, vol.2, issue.8, pp.569-79, 2002.

T. Paredes, P. Esser, J. Admyre, C. Nord, M. Rahman et al., Bronchoalveolar lavage fluid exosomes contribute to cytokine and leukotriene production in allergic asthma, Allergy, vol.67, issue.7, pp.911-920, 2012.

K. Trajkovic, C. Hsu, S. Chiantia, L. Rajendran, D. Wenzel et al., Ceramide triggers budding of exosome vesicles into multivesicular endosomes, Science, issue.5867, pp.1244-1251, 2008.

E. G. Trams, C. J. Lauter, N. Salem, and U. Heine, Exfoliation of membrane ecto-enzymes in the form of micro-vesicles, Biochimica et biophysica acta, vol.645, issue.1, pp.63-70, 1981.

M. Triantafilou and K. Triantafilou, Heat-shock protein 70 and heat-shock protein 90 associate with Toll-like receptor 4 in response to bacterial lipopolysaccharide, Biochemical Society transactions, vol.32, pp.636-645, 2004.

M. Tytell, S. G. Greenberg, and R. J. Lasek, Heat shock-like protein is transferred from glia to axon, Brain research, vol.363, issue.1, pp.161-165, 1986.

R. M. Vabulas, P. Ahmad-nejad, C. Da-costa, T. Miethke, C. J. Kirschning et al., Endocytosed HSP60s use toll-like receptor 2 (TLR2) and TLR4 to activate the toll/interleukin-1 receptor signaling pathway in innate immune cells, The Journal of biological chemistry, vol.276, issue.33, pp.31332-31341, 2001.

H. Valadi, K. Ekström, A. Bossios, M. Sjöstrand, J. J. Lee et al., Exosomemediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nature cell biology, vol.9, issue.6, pp.654-663, 2007.

B. W. Van-balkom, T. Pisitkun, M. C. Verhaar, and M. A. Knepper, Exosomes and the kidney: prospects for diagnosis and therapy of renal diseases, Kidney international, vol.80, issue.11, 2011.

J. G. Van-den-boorn, J. Dassler, C. Coch, M. Schlee, and G. Hartmann, Exosomes as nucleic acid nanocarriers. Advanced drug delivery reviews, vol.65, pp.331-336, 2013.

A. V. Vlassov, S. Magdaleno, R. Setterquist, and R. Conrad, Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials, Biochimica et biophysica acta, issue.7, pp.940-948, 2012.

B. Vogelstein, D. Lane, and A. J. Levine, Surfing the p53 network, Nature, vol.408, issue.6810, pp.307-317, 2000.

T. J. Wadas, E. H. Wong, G. R. Weisman, and C. J. Anderson, Coordinating radiometals of copper, gallium, indium, yttrium, and zirconium for PET and SPECT imaging of disease, Chemical reviews, vol.110, issue.5, pp.2858-902, 2010.

H. Whetstone and C. Lingwood, 3'Sulfogalactolipid binding specifically inhibits Hsp70 ATPase activity in vitro, Biochemistry, vol.42, issue.6, pp.1611-1618, 2003.

C. Whibley, P. D. Pharoah, and M. Hollstein, p53 polymorphisms: cancer implications, Nature reviews. Cancer, vol.9, issue.2, pp.95-107, 2009.

R. L. Williams and S. Urbé, The emerging shape of the ESCRT machinery, Nature reviews. Molecular cell biology, vol.8, issue.5, pp.355-68, 2007.

D. S. Williamson, J. Borgognoni, A. Clay, Z. Daniels, P. Dokurno et al., Novel adenosine-derived inhibitors of 70 kDa heat shock protein, discovered through structure-based design, Journal of medicinal chemistry, vol.52, issue.6, pp.1510-1513, 2009.

B. Wu, C. Hunt, and R. Morimoto, Structure and expression of the human gene encoding major heat shock protein HSP70, Molecular and cellular biology, vol.5, issue.2, pp.330-371, 1985.

X. Xiang, Y. Liu, X. Zhuang, S. Zhang, S. Michalek et al., TLR2-mediated expansion of MDSCs is dependent on the source of tumor exosomes, The American journal of pathology, vol.177, issue.4, pp.1606-1616, 2010.

L. Xiao, X. Lu, and D. M. Ruden, Effectiveness of hsp90 inhibitors as anti-cancer drugs. Mini reviews in medicinal chemistry, vol.6, pp.1137-1180, 2006.

X. Xiao, X. Zuo, A. A. Davis, D. R. Mcmillan, B. B. Curry et al., HSF1 is required for extra-embryonic development, postnatal growth and protection during inflammatory responses in mice, The EMBO journal, vol.18, issue.21, pp.5943-52, 1999.

N. Yamagishi, K. Ishihara, Y. Saito, and T. Hatayama, Hsp105 family proteins suppress staurosporine-induced apoptosis by inhibiting the translocation of Bax to mitochondria in HeLa cells, Experimental cell research, issue.17, pp.3215-3238, 2006.

R. R. Zielinski, B. J. Eigl, and K. N. Chi, Targeting the apoptosis pathway in prostate cancer, Cancer journal, vol.19, issue.1, pp.79-89, 2013.

K. C. Zimmermann, C. Bonzon, and D. R. Green, The machinery of programmed cell death, Pharmacology & therapeutics, vol.92, issue.1, pp.57-70, 2001.