, La migration des LT vers la tumeur mêle le roulement, l'adhésion, l'extravasation et le chimiotactisme. La structure tumorale et son micro-environnement entravent l'infiltration des LT 146,147,148. Cette étape est essentielle pour le succès de la thérapie de CAR-T cell

, Ainsi, l'absence des récepteurs de chimiokines à la surface des CAR-T cells limite l'accès aux tumeurs 149. Nous avons précédemment décrit la manière dont certaines conceptions de CART cells sont réalisées avec des récepteurs aux chimiokines pour augmenter leur migration

, Le choix du site d'injection des LT en fonction du cancer est une approche envisageable pour optimiser la thérapie. Par exemple, l'injection intracérébrale des cellules CAR-T a permis d'éviter le phénomène de barrière hémato

, De même, une étude menée sur les cancers pleuraux a mise en évidence la différence d'efficacité entre deux voies d'administration

, L'administration intra-pleurale de CAR

, Plusieurs méthodes visent une meilleure précision : la sélection fine de TAA, la densité d'expression des TAA 6 , des CAR ciblant deux antigènes, des CAR avec des commutateurs (gène suicide), L'un des principaux freins à l'utilisation de CAR-T cells est la toxicité hors-cibles

, Lymphocyte T à récepteur d'antigène chimérique Tableau 7 : Essais cliniques en cours de CAR-T cell ciblant des cancers solides, vol.2, p.130

. Bibliographie-bibliographie-1, J. Owen, J. Punt, and S. Stranford, Immunologie-7e édition: Le cours de Janis Kuby avec questions de révision, 2014.

M. Sharpe and N. Mount, Genetically modified T cells in cancer therapy: opportunities and challenges, Dis. Model. Mech, vol.8, pp.337-350, 2015.

A. J. Smith, J. Oertle, D. Warren, and D. Prato, Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: Summary and perspective, J. Cell. Immunother

A. K. Abbas, Les bases de l'immunologie fondamentale et clinique

. Masson, , 2016.

L. Robb, Cytokine receptors and hematopoietic differentiation, Oncogene, vol.26, pp.6715-6723, 2007.

A. D. Fesnak, C. H. June, and B. L. Levine, Engineered T cells: the promise and challenges of cancer immunotherapy, Nat. Rev. Cancer, vol.16, pp.566-581, 2016.

R. H. Schwartz, Annu. Rev. Immunol, vol.21, pp.305-334, 2003.

L. Zitvogel, A. Tesniere, and G. Kroemer, Cancer despite immunosurveillance: immunoselection and immunosubversion, Nat. Rev. Immunol, vol.6, pp.715-727, 2006.

R. Gu&#xe9, L. Hugues, and S. Phanie, Th17 Cell Plasticity and Functions in Cancer Immunity, BioMed Res. Int, p.314620, 2015.

A. B. Frey, Suppression of T cell responses in the tumor microenvironment, Vaccine, vol.33, pp.7393-7400, 2015.

, Multiple-Purpose Immunotherapy for Cancer

(. Immunologie, . Lucienne, and . Bach-jean-françois, Librairie Lavoisier Available

, /descriptif-9782257205292, 2017.

A. Jemal, Global cancer statistics. CA. Cancer J. Clin, vol.61, pp.69-90, 2011.

M. Wang, B. Yin, H. Y. Wang, and R. Wang, Current advances in T-cell-based cancer immunotherapy, Immunotherapy, vol.6, pp.1265-1278, 2014.

L. 'essentiel-de-la-biologie-cellulaire, (. Bruce, . Bray-dennis, J. Hopkin-karen, L. Alexander et al., , p.115

W. L. Peter and . Lavoisier,

P. A. Oliveira, Chemical carcinogenesis, An. Acad. Bras. Cienc, vol.79, pp.593-616, 2007.

S. I. Grivennikov, F. R. Greten, and M. Karin, Immunity, inflammation, and cancer

, Cell, vol.140, pp.883-899, 2010.

M. D. Vesely, M. H. Kershaw, R. D. Schreiber, and M. J. Smyth, Natural innate and adaptive immunity to cancer, Annu. Rev. Immunol, vol.29, pp.235-271, 2011.

M. Burnet, Cancer; a biological approach. I. The processes of control, Br. Med. J, vol.1, pp.779-786, 1957.

G. P. Dunn, A. T. Bruce, H. Ikeda, L. J. Old, and R. D. Schreiber, Cancer immunoediting: from immunosurveillance to tumor escape, Nat. Immunol, vol.3, pp.991-998, 2002.

G. P. Dunn, L. J. Old, and R. D. Schreiber, The three Es of cancer immunoediting

, Annu. Rev. Immunol, vol.22, pp.329-360, 2004.

M. J. Smyth, G. P. Dunn, and R. D. Schreiber, Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity, Adv. Immunol, vol.90, pp.1-50, 2006.

M. D. Vesely and R. D. Schreiber, Cancer immunoediting: antigens, mechanisms, and implications to cancer immunotherapy, Ann. N. Y. Acad. Sci, vol.1284, pp.1-5, 2013.

R. D. Schreiber, L. J. Old, and M. J. Smyth, Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion, Science, vol.331, pp.1565-1570, 2011.

V. Shankaran, IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity, Nature, vol.410, pp.1107-1111, 2001.

Y. Meirow, J. Kanterman, and M. Baniyash, Paving the Road to Tumor Development and Spreading: Myeloid-Derived Suppressor Cells are Ruling the Fate, Front. Immunol, vol.6, p.523, 2015.

B. Engels, Relapse or eradication of cancer is predicted by peptide-MHC affinity, Cancer Cell, vol.23, pp.516-526, 2013.

J. B. Swann and M. J. Smyth, Immune surveillance of tumors, J. Clin. Invest, vol.117, pp.1137-1146, 2007.

J. D. Bui and R. D. Schreiber, Cancer immunosurveillance, immunoediting and inflammation: independent or interdependent processes?, Curr. Opin. Immunol, vol.19, pp.203-208

. Bibliographie, , 2007.

E. Vivier, E. Tomasello, M. Baratin, T. Walzer, and S. Ugolini, Functions of natural killer cells, Nat. Immunol, vol.9, pp.503-510, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00294184

S. Sivori, CpG and double-stranded RNA trigger human NK cells by Toll-like receptors: Induction of cytokine release and cytotoxicity against tumors and dendritic cells

, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.10116-10121, 2004.

M. J. Smyth, Differential Tumor Surveillance by Natural Killer (Nk) and Nkt Cells, J. Exp. Med, vol.191, pp.661-668, 2000.

A. Mantovani and A. Sica, Macrophages, innate immunity and cancer: balance, tolerance, and diversity, Curr. Opin. Immunol, vol.22, pp.231-237, 2010.

K. Kersten, C. Salvagno, and K. E. De-visser, Exploiting the Immunomodulatory Properties of Chemotherapeutic Drugs to Improve the Success of Cancer Immunotherapy

, Front. Immunol, vol.6, p.516, 2015.

C. E. Lewis and J. W. Pollard, Distinct role of macrophages in different tumor microenvironments, Cancer Res, vol.66, pp.605-612, 2006.

Z. G. Fridlender, Polarization of tumor-associated neutrophil phenotype by TGFbeta: 'N1' versus 'N2' TAN, Cancer Cell, vol.16, pp.183-194, 2009.

Y. Nefedova, Regulation of dendritic cell differentiation and antitumor immune response in cancer by pharmacologic-selective inhibition of the janus-activated kinase

, /signal transducers and activators of transcription 3 pathway, Cancer Res, vol.65, pp.9525-9535, 2005.

J. Galaine, C. Borg, Y. Godet, and O. Adotévi, Interest of Tumor-Specific CD4 T Helper 1 Cells for Therapeutic Anticancer Vaccine, vol.3, pp.490-502, 2015.

W. Zou, Immunosuppressive networks in the tumour environment and their therapeutic relevance, Nat. Rev. Cancer, vol.5, pp.263-274, 2005.

L. M. Coussens and Z. Werb, Inflammation and cancer, Nature, vol.420, pp.860-867, 2002.

S. Farkona, E. P. Diamandis, and I. M. Blasutig, Cancer immunotherapy: the beginning of the end of cancer, BMC Med, vol.14, 2016.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, pp.646-674, 2011.

P. Sharma, K. Wagner, J. D. Wolchok, and J. P. Allison, Novel cancer immunotherapy agents with survival benefit: recent successes and next steps, Nat. Rev. Cancer, vol.11, pp.805-812

S. A. Grupp, Chimeric antigen receptor-modified T cells for acute lymphoid leukemia, N. Engl. J. Med, vol.368, pp.1509-1518, 2013.

S. A. Rosenberg, Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report, N. Engl. J. Med, vol.319, pp.1676-1680, 1988.

D. E. Gilham, Adoptive T-cell therapy for cancer in the United kingdom: a review of activity for the British Society of Gene and Cell Therapy annual meeting, 2015.

G. Hum and . Ther, , vol.26, pp.276-285, 2015.

T. S. Park, S. A. Rosenberg, and R. A. Morgan, Treating Cancer with Genetically

T. Engineered and . Cells, Trends Biotechnol, vol.29, pp.550-557, 2011.

M. H. Kershaw, J. A. Westwood, and P. K. Darcy, Gene-engineered T cells for cancer therapy, Nat. Rev. Cancer, vol.13, pp.525-541, 2013.

C. Yee, The use of endogenous T cells for adoptive transfer, Immunol. Rev, vol.257, pp.250-263, 2014.

H. Shi, L. Liu, and Z. Wang, Improving the efficacy and safety of engineered T cell therapy for cancer, Cancer Lett, vol.328, pp.191-197, 2013.

D. N. Khalil, E. L. Smith, R. J. Brentjens, and J. D. Wolchok, The future of cancer treatment: immunomodulation, CARs and combination immunotherapy, Nat. Rev. Clin

S. J. Hamieh, M. Sadelain, and M. , The pharmacology of secondgeneration chimeric antigen receptors, Nat. Rev. Drug Discov, vol.13, pp.499-509, 2015.

M. Sadelain, R. Brentjens, and I. Rivière, The basic principles of chimeric antigen receptor design, Cancer Discov, vol.3, pp.388-398, 2013.

M. Haji-fatahaliha, CAR-modified T-cell therapy for cancer: an updated review

. Artif, Cells Nanomedicine Biotechnol, vol.44, pp.1339-1349, 2016.

H. J. Jackson, S. Rafiq, and R. J. Brentjens, Driving CAR T-cells forward, Nat. Rev

, Clin. Oncol, vol.13, pp.370-383, 2016.

D. Abate-daga and M. L. Davila, CAR models: next-generation CAR modifications for enhanced T-cell function, Mol. Ther.-Oncolytics, vol.3, p.16014, 2016.

M. Schubert, Chimeric Antigen Receptor T Cell Therapy Targeting CD19

, Positive Leukemia and Lymphoma in the Context of Stem Cell Transplantation. Hum. Gene Ther, vol.27, pp.758-771, 2016.

J. Maher, R. J. Brentjens, G. Gunset, I. Rivière, M. Sadelain et al.,

, Nat. Biotechnol, vol.20, pp.70-75, 2002.

C. Carpenito, Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains, Proc. Natl. Acad. Sci

, A, vol.106, pp.3360-3365, 2009.

X. Zhong, M. Matsushita, J. Plotkin, I. Riviere, and M. Sadelain, Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication, Mol. Ther. J. Am. Soc. Gene Ther, vol.18, pp.413-420, 2010.

M. C. Jensen and S. R. Riddell, Designing chimeric antigen receptors to effectively and safely target tumors, Curr. Opin. Immunol, vol.33, pp.9-15, 2015.

C. Berger, Safety of Targeting ROR1 in Primates with Chimeric Antigen Receptor-Modified T Cells, Cancer Immunol. Res, vol.3, pp.206-216, 2015.

A. Hombach, A. A. Hombach, and H. Abken, Adoptive immunotherapy with genetically engineered T cells: modification of the IgG1 Fc 'spacer' domain in the extracellular moiety of chimeric antigen receptors avoids 'off-target' activation and unintended initiation of an innate immune response, Gene Ther, vol.17, pp.1206-1213, 2010.

M. Norelli, M. Casucci, C. Bonini, and A. Bondanza, Clinical pharmacology of CART cells: Linking cellular pharmacodynamics to pharmacokinetics and antitumor effects

, Biochim. Biophys. Acta BBA-Rev. Cancer, vol.1865, pp.90-100, 2016.

K. Töpfer, DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy, J. Immunol. Baltim. Md, pp.3201-3212, 2015.

X. Wang and I. Rivière, Clinical manufacturing of CAR T cells: foundation of a promising therapy, Mol. Ther. Oncolytics, vol.3, p.16015, 2016.

A. Fesnak, C. Lin, D. L. Siegel, M. V. Maus, and . Car-t, Cell Therapies From

, Transfusion Medicine Perspective. Transfus. Med. Rev, vol.30, pp.139-145, 2016.

S. Gill, M. V. Maus, and D. L. Porter, Chimeric antigen receptor T cell therapy: 25 years in the making, Blood Rev, vol.30, pp.157-167, 2016.

M. A. Kay, State-of-the-art gene-based therapies: the road ahead, Nat. Rev. Genet, vol.12, pp.316-328, 2011.

J. E. Vargas, Retroviral vectors and transposons for stable gene therapy

, Advances, current challenges and perspectives, ResearchGate, vol.14, 2016.

X. Wang and I. Rivière, Manufacture of tumor-and virus-specific T lymphocytes for Bibliographie 119 adoptive cell therapies, Cancer Gene Ther, vol.22, pp.85-94, 2015.

A. Deichmann and M. Schmidt, Biosafety considerations using gamma-retroviral vectors in gene therapy, Curr. Gene Ther, vol.13, pp.469-477, 2013.

S. Hacein-bey-abina, A Serious Adverse Event after Successful Gene Therapy for X-Linked Severe Combined Immunodeficiency, N. Engl. J. Med, vol.348, pp.255-256, 2003.

J. Mátrai, Hepatocyte-targeted expression by integrase-defective lentiviral vectors induces antigen-specific tolerance in mice with low genotoxic risk, Hepatol. Baltim. Md, vol.53, pp.1696-1707, 2011.

R. Monjezi, Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors, Leukemia, 2016.

Y. Xu, Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15, Blood, vol.123, pp.3750-3759, 2014.

, Adoptive Therapy With Chimeric Antigen Receptor-Modified T C... : The Cancer Journal. LWW Available at

. Antigen, , 2017.

D. Sommermeyer, Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo, Leukemia, vol.30, pp.492-500, 2016.

C. J. Turtle, CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients, J. Clin. Invest, vol.126, pp.2123-2138

C. Stemberger, Novel serial positive enrichment technology enables clinical multiparameter cell sorting, PloS One, vol.7, p.35798, 2012.

C. L. Bonifant, H. J. Jackson, R. J. Brentjens, and K. J. Curran, Toxicity and management in CAR T-cell therapy, Mol. Ther.-Oncolytics, vol.3, p.16011, 2016.

M. Casucci, R. E. Hawkins, G. Dotti, and A. Bondanza, Overcoming the toxicity hurdles of genetically targeted T cells, Cancer Immunol. Immunother, vol.64, pp.123-130, 2015.

M. L. Davila and M. Sadelain, Biology and clinical application of CAR T cells for B cell malignancies, Int. J. Hematol, vol.104, pp.6-17, 2016.

M. Casucci, CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma, Blood, vol.122, pp.3461-3472, 2013.

D. Chakravarti and W. W. Wong, Synthetic biology in cell-based cancer immunotherapy, Trends Biotechnol, vol.33, pp.449-461, 2015.

. Bibliographie-86, V. D. Fedorov, M. Themeli, and M. Sadelain,

, Chimeric Antigen Receptors (iCARs) Divert Off-Target Immunotherapy Responses

, Transl. Med, vol.5, pp.215-172, 2013.

G. L. Beatty, Mesothelin-Specific Chimeric Antigen Receptor mRNA

, Engineered T Cells Induce Antitumor Activity in Solid Malignancies, Cancer Immunol. Res, vol.2, pp.112-120, 2014.

V. Russo, A dual role for genetically modified lymphocytes in cancer immunotherapy, Trends Mol. Med, vol.18, pp.193-200, 2012.

X. Wang, A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells, Blood, vol.118, pp.1255-1263, 2011.

M. Serafini, Characterization of CD20-transduced T lymphocytes as an alternative suicide gene therapy approach for the treatment of graft-versus-host disease

, Gene Ther, vol.15, pp.63-76, 2004.

B. Philip, A highly compact epitope-based marker/suicide gene for easier and safer T-cell therapy, Blood, vol.124, pp.1277-1287, 2014.
DOI : 10.1182/blood-2014-01-545020

URL : http://www.bloodjournal.org/content/124/8/1277.full.pdf

K. Minagawa, X. Zhou, S. Mineishi, and A. Di-stasi, Seatbelts in CAR therapy: How Safe Are CARS?, Pharmaceuticals, vol.8, pp.230-249, 2015.
DOI : 10.3390/ph8020230

URL : http://www.mdpi.com/1424-8247/8/2/230/pdf

M. H. Kershaw, A Phase I Study on Adoptive Immunotherapy Using Gene
DOI : 10.1158/1078-0432.ccr-06-1183

URL : http://clincancerres.aacrjournals.org/content/clincanres/12/20/6106.full.pdf

, Modified T Cells for Ovarian Cancer, Clin. Cancer Res, vol.12, pp.6106-6115, 2006.

D. S. Ritchie, Persistence and Efficacy of Second Generation CAR T Cell Against the LeY Antigen in Acute Myeloid Leukemia, Mol. Ther, vol.21, pp.2122-2129, 2013.

H. Huang, VEGF suppresses T-lymphocyte infiltration in the tumor microenvironment through inhibition of NF-?B-induced endothelial activation, FASEB J, vol.29, pp.227-238, 2015.

N. Ahmed, Human Epidermal Growth Factor Receptor, vol.2, issue.HER2

, Chimeric Antigen Receptor-Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma, J. Clin. Oncol, vol.33, pp.1688-1696, 2015.

, Local Delivery of Interleukin-12 Using T Cells Targeting VEGF Receptor-2

, Eradicates Multiple Vascularized Tumors in Mice

B. D. Choi, Intracerebral delivery of a third generation EGFRvIII-specific chimeric antigen receptor is efficacious against human glioma, J. Clin. Neurosci. Off. J. Neurosurg. Soc. Australas, vol.21, pp.189-190, 2014.
DOI : 10.1016/j.jocn.2013.03.012

URL : http://europepmc.org/articles/pmc3867597?pdf=render

. Bibliographie-99 and G. Dotti, Human cytotoxic T lymphocytes with reduced sensitivity to Fasinduced apoptosis, Blood, vol.105, pp.4677-4684, 2005.

D. Eaton, D. E. Gilham, A. O'neill, and R. E. Hawkins, Retroviral transduction of human peripheral blood lymphocytes with Bcl-X(L) promotes in vitro lymphocyte survival in pro-apoptotic conditions, Gene Ther, vol.9, pp.527-535, 2002.

J. Charo, Bcl-2 Overexpression Enhances Tumor-Specific T-Cell Survival
DOI : 10.1158/0008-5472.can-04-2006

URL : http://cancerres.aacrjournals.org/content/65/5/2001.full.pdf

, Cancer Res, vol.65, 2001.

J. H. Shin, Positive conversion of negative signaling of CTLA4 potentiates antitumor efficacy of adoptive T-cell therapy in murine tumor models, Blood, vol.119, pp.5678-5687, 2012.

L. Gattinoni, C. A. Klebanoff, and N. P. Restifo, Paths to stemness: building the ultimate antitumour T cell, Nat. Rev. Cancer, vol.12, pp.671-684, 2012.
DOI : 10.1038/nrc3322

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6352980

A. Bondanza, Suicide gene therapy of graft-versus-host disease induced by central memory human T lymphocytes, Blood, vol.107, pp.1828-1836, 2006.
DOI : 10.1182/blood-2005-09-3716

URL : http://www.bloodjournal.org/content/107/5/1828.full.pdf

C. A. Klebanoff, Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells, Proc. Natl. Acad. Sci

U. S. , , vol.102, pp.9571-9576, 2005.

C. U. Louis, Antitumor activity and long-term fate of chimeric antigen receptorpositive T cells in patients with neuroblastoma, Blood, vol.118, pp.6050-6056, 2011.

M. E. Dudley, Cancer Regression and Autoimmunity in Patients After Clonal Repopulation with Antitumor Lymphocytes, Science, vol.298, pp.850-854, 2002.

M. E. Dudley, Adoptive Cell Transfer Therapy Following Non-Myeloablative but Lymphodepleting Chemotherapy for the Treatment of Patients With Refractory Metastatic Melanoma, J. Clin. Oncol, vol.23, pp.2346-2357, 2005.

H. Wallen, Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma, PloS One, vol.4, p.4749, 2009.

M. Guimond, Interleukin 7 signaling in dendritic cells regulates the homeostatic proliferation and niche size of CD4+ T cells, Nat. Immunol, vol.10, pp.149-157, 2009.

N. Cieri, Generation of human memory stem T cells after haploidentical Treplete hematopoietic stem cell transplantation, Blood, vol.125, pp.2865-2874, 2015.

A. , A. Innao, V. Gerace, D. Vaddinelli, D. Musolino et al., Adoptive immunotherapy for hematological malignancies: Current status and new insights in chimeric Bibliographie 122 antigen receptor T cells, Blood Cells. Mol. Dis, vol.62, pp.49-63, 2016.

K. Kudo, T Lymphocytes Expressing a CD16 Signaling Receptor Exert Antibody-Dependent Cancer Cell Killing, Cancer Res, vol.74, pp.93-103, 2014.

K. Urbanska, A universal strategy for adoptive immunotherapy of cancer through use of a novel T-cell antigen receptor, Cancer Res, vol.72, pp.1844-1852, 2012.

K. Tamada, Redirecting gene-modified T cells toward various cancer types using tagged antibodies, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.18, pp.6436-6445, 2012.

H. Torikai, A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR, Blood, vol.119, pp.5697-5705, 2012.

L. M. Reyes, Creating Class I MHC-Null Pigs Using Guide RNA and the Cas9

. Endonuclease, J. Immunol, vol.193, pp.5751-5757, 2014.

A. Holzinger, M. Barden, and . Hinrich-abken, The growing world of CAR T cell trials: a systematic review, Cancer Immunol. Immunother. CII, vol.65, pp.1433-1450, 2016.

M. Chmielewski and H. Abken, CAR T cells transform to trucks: chimeric antigen receptor-redirected T cells engineered to deliver inducible IL-12 modulate the tumour stroma to combat cancer, Cancer Immunol. Immunother. CII, vol.61, pp.1269-1277, 2012.

H. J. Pegram, IL-12-secreting CD19-targeted cord blood-derived T cells for the immunotherapy of B-cell acute lymphoblastic leukemia, Leukemia, vol.29, pp.415-422, 2015.

L. Zhang, Improving adoptive T cell therapy by targeting and controlling IL-12 expression to the tumor environment, Mol. Ther. J. Am. Soc. Gene Ther, vol.19, pp.751-759, 2011.

O. O. Yeku and R. J. Brentjens, Armored CAR T-cells: utilizing cytokines and proinflammatory ligands to enhance CAR T-cell anti-tumour efficacy, Biochem. Soc. Trans, vol.44, pp.412-418, 2016.

H. Torikai and L. J. Cooper, Translational Implications for Off-the-shelf Immune Cells Expressing Chimeric Antigen Receptors, Mol. Ther, vol.24, pp.1178-1186, 2016.

H. R. Mirzaei, H. Mirzaei, S. Y. Lee, J. Hadjati, and B. G. Till, Prospects for chimeric antigen receptor (CAR) ?? T cells: A potential game changer for adoptive T cell cancer immunotherapy, Cancer Lett, vol.380, pp.413-423, 2016.

D. C. Deniger, Bispecific T-cells Expressing Polyclonal Repertoire of Endogenous ?? T-cell Receptors and Introduced CD19-specific Chimeric Antigen Receptor

, Mol. Ther, vol.21, pp.638-647, 2013.

M. Rischer, Human ?? T cells as mediators of chimaeric-receptor redirected antiBibliographie 123 tumour immunity, Br. J. Haematol, vol.126, pp.583-592, 2004.

J. S. Miller, Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer, Blood, vol.105, pp.3051-3057, 2005.

J. L. Zakrzewski, Tumor immunotherapy across MHC barriers using allogeneic T-cell precursors, Nat. Biotechnol, vol.26, pp.453-461, 2008.

M. Themeli, I. Rivière, and M. Sadelain, New Cell Sources for T Cell Engineering and Adoptive Immunotherapy. Cell Stem Cell, vol.16, pp.357-366, 2015.

L. A. Johnson and C. H. June, Driving gene-engineered T cell immunotherapy of cancer, Cell Res, vol.27, pp.38-58, 2017.

H. Zhang, New Strategies for the Treatment of Solid Tumors with CAR-T Cells

, Int. J. Biol. Sci, vol.12, pp.718-729, 2016.

R. H. Carter and D. T. Fearon, CD19: lowering the threshold for antigen receptor stimulation of B lymphocytes, Science, vol.256, pp.105-107, 1992.

R. H. Scheuermann and E. Racila, CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy, Leuk. Lymphoma, vol.18, pp.385-397, 1995.

R. J. Brentjens, Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias, Blood, vol.118, pp.4817-4828, 2011.

R. Brentjens, CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia, Sci. Transl. Med, vol.5, pp.177-215, 2013.

S. L. Maude, Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia, N. Engl. J. Med, vol.371, pp.1507-1517, 2014.

J. N. Kochenderfer, Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19, Blood, vol.116, pp.4099-4102, 2010.

J. N. Kochenderfer, B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptortransduced T cells, Blood, vol.119, pp.2709-2720, 2012.

C. S. Sauter, Phase I trial of 19-28z chimeric antigen receptor modified T cells (19-28z CAR-T) post-high dose therapy and autologous stem cell transplant (HDT-ASCT) for relapsed and refractory (rel/ref) aggressive B-cell non-Hodgkin lymphoma (B-NHL), J. Clin

, Oncol, vol.33, 2015.

. Bibliographie,

A. L. Garfall, Chimeric Antigen Receptor T Cells against CD19 for Multiple Myeloma, N. Engl. J. Med, vol.373, pp.1040-1047, 2015.

K. E. De-visser, L. V. Korets, and L. M. Coussens, De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent, Cancer Cell, vol.7, pp.411-423, 2005.

M. Ammirante, J. Luo, S. Grivennikov, S. Nedospasov, and M. Karin, B-cellderived lymphotoxin promotes castration-resistant prostate cancer, Nature, vol.464, pp.302-305, 2010.

F. Balkwill, A. Montfort, and M. Capasso, B regulatory cells in cancer, Trends Immunol, vol.34, pp.169-173, 2013.

A. Ronson, A. Tvito, and J. M. Rowe, Treatment of Relapsed/Refractory Acute Lymphoblastic Leukemia in Adults, Curr. Oncol. Rep, vol.18, 2016.

R. A. Morgan,

, Mol. Ther. J. Am. Soc. Gene Ther, vol.18, pp.843-851, 2010.

K. Greish, Enhanced permeability and retention (EPR) effect for anticancer nanomedicine drug targeting, Methods Mol. Biol. Clifton NJ, vol.624, pp.25-37, 2010.

G. Rahir and M. Moser, Tumor microenvironment and lymphocyte infiltration, Cancer Immunol. Immunother. CII, vol.61, pp.751-759, 2012.

C. Y. Slaney, M. H. Kershaw, and P. K. Darcy, Trafficking of T cells into tumors

, Cancer Res, vol.74, pp.7168-7174, 2014.

N. Karin and G. Wildbaum, The Role of Chemokines in Shaping the Balance Between CD4+ T Cell Subsets and Its Therapeutic Implications in Autoimmune and Cancer Diseases

, Front. Immunol, vol.6, 2015.

P. S. Adusumilli, Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity, Sci. Transl. Med, vol.6, pp.261-151, 2014.

M. Chmielewski, C. Kopecky, and A. A. Hombach, & Abken, H. IL-12 Res, vol.71, pp.5697-5706, 2011.

T. F. Gajewski, Immune resistance orchestrated by the tumor microenvironment

, Immunol. Rev, vol.213, pp.131-145, 2006.

, Lack of B7 expression, not human leukocyte antigen expression, facilitates immune Bibliographie 125 evasion by human malignant gliomas (PDF Download Available), ResearchGate Available at

E. F. Tewalt, Lymphatic endothelial cells induce tolerance via PD-L1 and lack of costimulation leading to high-level PD-1 expression on CD8 T cells, Blood, vol.120, pp.4772-4782, 2012.

M. E. Prosser, C. E. Brown, A. F. Shami, S. J. Forman, and M. C. Jensen, Tumor PDL1 co-stimulates primary human CD8(+) cytotoxic T cells modified to express a PD1:CD28 chimeric receptor, Mol. Immunol, vol.51, pp.263-272, 2012.
DOI : 10.1016/j.molimm.2012.03.023

V. Hillerdal and M. Essand, Chimeric Antigen Receptor-Engineered T Cells for the Treatment of Metastatic Prostate Cancer, Biodrugs, vol.29, pp.75-89, 2015.

A. D. Posey, Engineered CAR T Cells Targeting the Cancer-Associated TnGlycoform of the Membrane Mucin MUC1 Control Adenocarcinoma, Immunity, vol.44, pp.1444-1454, 2016.