D. M. Pardoll, The blockade of immune checkpoints in cancer immunotherapy, Nat Rev Cancer, vol.12, issue.4, pp.252-64, 2012.

J. Brozek, F. Grande, J. T. Anderson, and A. Keys, DENSITOMETRIC ANALYSIS OF BODY COMPOSITION: REVISION OF SOME QUANTITATIVE ASSUMPTIONS, Ann N Y Acad Sci, vol.110, pp.113-153, 1963.

D. A. Jones, J. M. Round, A. Haan, and . De, Physiologie du muscle squelettique: de la structure au mouvement, 2005.

W. Shen, M. Punyanitya, Z. Wang, D. Gallagher, M. St-onge et al., Total body skeletal muscle and adipose tissue volumes: estimation from a single abdominal crosssectional image, J Appl Physiol, vol.97, issue.6, pp.2333-2341, 2004.

N. Mitsiopoulos, R. N. Baumgartner, S. B. Heymsfield, W. Lyons, D. Gallagher et al., Cadaver validation of skeletal muscle measurement by magnetic resonance imaging and computerized tomography, J Appl Physiol, vol.85, issue.1, pp.115-137, 1998.

R. N. Baumgartner, K. M. Koehler, D. Gallagher, L. Romero, S. B. Heymsfield et al., Epidemiology of sarcopenia among the elderly in New Mexico, Am J Epidemiol, vol.147, issue.8, pp.755-63, 1998.

A. J. Cruz-jentoft, J. P. Baeyens, J. M. Bauer, Y. Boirie, T. Cederholm et al., European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People, vol.39, pp.412-435, 2010.

R. A. Fielding, B. Vellas, W. J. Evans, S. Bhasin, J. E. Morley et al., Sarcopenia: an undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences. International working group on sarcopenia, J Am Med Dir Assoc, vol.12, issue.4, pp.249-56, 2011.

L. Chen, L. Liu, J. Woo, P. Assantachai, T. Auyeung et al.,

, Sarcopenia in Asia: consensus report of the Asian Working Group for Sarcopenia, vol.15, pp.95-101, 2014.

A. Y. Bijlsma, C. Meskers, C. Ling, M. Narici, S. E. Kurrle et al., Defining sarcopenia: the impact of different diagnostic criteria on the prevalence of sarcopenia in a large middle aged cohort, Age (Dordr), vol.35, issue.3, pp.871-81, 2013.

I. Janssen, S. B. Heymsfield, Z. M. Wang, and R. Ross, Skeletal muscle mass and distribution in 468 men and women aged 18-88 yr, J Appl Physiol, vol.89, issue.1, pp.81-89, 2000.

P. Ritz, Modifications du métabolisme énergétique et de la composition corporelle au cours du vieillissement, Traité de nutrition de la personne âgée

W. Evans, Functional and metabolic consequences of sarcopenia, J Nutr, vol.127, issue.5, pp.998-1003, 1997.

A. Cessot, X. Hebuterne, R. Coriat, J. Durand, O. Mir et al., Defining the clinical condition of cancer patients: it is time to switch from performance status to nutritional status. Support Care Cancer, vol.19, pp.869-70, 2011.

L. Martin, L. Birdsell, N. Macdonald, T. Reiman, M. T. Clandinin et al., Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index, Fadila et al. -Coordonnateurs ateliers J2R 2013. Referentiel_AFSOS_Nutrition_chez_le_patient_adulte_atteint_de_cancer.pdf 17, vol.31, pp.489-95, 2011.

K. Fearon, D. J. Glass, and D. C. Guttridge, Cancer cachexia: mediators, signaling, and metabolic pathways, Cell Metab, vol.16, issue.2, pp.153-66, 2012.

P. Collins, C. Bing, P. Mcculloch, and G. Williams, Muscle UCP-3 mRNA levels are elevated in weight loss associated with gastrointestinal adenocarcinoma in humans, Br J Cancer, vol.86, issue.3, pp.372-377, 2002.

C. Vazeille, A. Jouinot, J. Durand, N. Neveux, P. Boudou-rouquette et al., Relation between hypermetabolism, cachexia, and survival in cancer patients: a prospective study in 390 cancer patients before initiation of anticancer therapy, Am J Clin Nutr, vol.105, issue.5, pp.1139-1186, 2017.

J. M. Argilés, S. Busquets, B. Stemmler, and F. J. López-soriano, Cancer cachexia: understanding the molecular basis, Nat Rev Cancer, vol.14, issue.11, pp.754-62, 2014.

A. Bonetto, F. Penna, Z. Aversa, P. Mercantini, F. M. Baccino et al., Early changes of muscle insulin-like growth factor-1 and myostatin gene expression in gastric cancer patients, Muscle Nerve, vol.48, issue.3, pp.387-92, 2013.

P. Todorov, P. Cariuk, T. Mcdevitt, B. Coles, K. Fearon et al., Characterization of a cancer cachectic factor, Nature, vol.379, issue.6567, pp.739-781, 1996.

J. V. White, P. Guenter, G. Jensen, A. Malone, and M. Schofield, Academy Malnutrition Work Group, et al. Consensus statement: Academy of Nutrition and Dietetics and American Society for Parenteral and Enteral Nutrition: characteristics recommended for the identification and documentation of adult malnutrition (undernutrition), JPEN J Parenter Enteral Nutr, vol.36, issue.3, pp.275-83, 2012.

C. Prado, J. R. Lieffers, L. J. Mccargar, T. Reiman, M. B. Sawyer et al.,

, Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study, Lancet Oncol, vol.9, issue.7, pp.629-664, 2008.

S. Cousin, A. Hollebecque, S. Koscielny, O. Mir, A. Varga et al., Low skeletal muscle is associated with toxicity in patients included in phase I trials, Invest New Drugs, vol.32, issue.2, pp.382-389, 2014.

M. Massicotte, I. Borget, S. Broutin, V. E. Baracos, S. Leboulleux et al., Body composition variation and impact of low skeletal muscle mass in patients with advanced medullary thyroid carcinoma treated with vandetanib: results from a placebo-controlled study, J Clin Endocrinol Metab, vol.98, issue.6, pp.2401-2409, 2013.

D. Sebastiano, K. M. , Y. L. Zbuk, K. Wong, R. K. Chow et al., Accelerated muscle and adipose tissue loss may predict survival in pancreatic cancer patients: the relationship with diabetes and anaemia, Br J Nutr, vol.109, issue.2, pp.302-314, 2013.

F. Pamoukdjian, T. Bouillet, V. Lévy, M. Soussan, L. Zelek et al., Prevalence and predictive value of pre-therapeutic sarcopenia in cancer patients: A systematic review, Clin Nutr, vol.37, issue.4, pp.1101-1114, 2018.

F. Bozzetti, Forcing the vicious circle: sarcopenia increases toxicity, decreases response to chemotherapy and worsens with chemotherapy, Ann Oncol, vol.28, issue.9, pp.2107-2125, 2017.

V. E. Baracos, T. Reiman, M. Mourtzakis, I. Gioulbasanis, and S. Antoun, Body composition in patients with non-small cell lung cancer: a contemporary view of cancer cachexia with the use of computed tomography image analysis, Am J Clin Nutr, vol.91, issue.4, pp.1133-1137, 2010.

G. B. Stene, J. L. Helbostad, T. Amundsen, S. Sørhaug, H. Hjelde et al., Changes in skeletal muscle mass during palliative chemotherapy in patients with advanced lung cancer, Acta Oncol, vol.54, issue.3, pp.340-348, 2015.

E. Y. Kim, Y. S. Kim, I. Park, H. K. Ahn, E. K. Cho et al., Prognostic Significance of CT-Determined Sarcopenia in Patients with Small-Cell Lung Cancer, J Thorac Oncol, vol.10, issue.12, pp.1795-1804, 2015.

S. Antoun, L. Birdsell, M. B. Sawyer, P. Venner, B. Escudier et al., Association of skeletal muscle wasting with treatment with sorafenib in patients with advanced renal cell carcinoma: results from a placebo-controlled study, J Clin Oncol, vol.28, issue.6, pp.1054-60, 2010.

S. P. Psutka, S. A. Boorjian, M. R. Moynagh, G. D. Schmit, B. A. Costello et al., Decreased Skeletal Muscle Mass is Associated with an Increased Risk of Mortality after Radical Nephrectomy for Localized Renal Cell Cancer, J Urol, vol.195, issue.2, pp.270-276, 2016.

O. Huillard, O. Mir, M. Peyromaure, C. Tlemsani, J. Giroux et al.,

, Sarcopenia and body mass index predict sunitinib-induced early dose-limiting toxicities in renal cancer patients, Br J Cancer, vol.108, issue.5, pp.1034-1075, 2013.

A. B. Smith, A. M. Deal, H. Yu, B. Boyd, J. Matthews et al., Sarcopenia as a predictor of complications and survival following radical cystectomy, J Urol, issue.6, pp.1714-1734, 2014.

H. Fukushima, M. Yokoyama, Y. Nakanishi, K. Tobisu, F. Koga et al., Sarcopenia as a prognostic biomarker of advanced urothelial carcinoma, PLoS ONE, vol.10, issue.1, 2015.

, Mortality after radical cystectomy: impact of obesity versus adiposity after adjusting for skeletal muscle wasting, J Urol, vol.193, issue.5, pp.1507-1520, 2015.

A. W. Wendrich, J. E. Swartz, S. I. Bril, I. Wegner, A. De-graeff et al., Low skeletal muscle mass is a predictive factor for chemotherapy dose-limiting toxicity in patients with locally advanced head and neck cancer, Oral Oncol, vol.71, pp.26-33, 2017.

D. Fabbro, E. Parsons, H. Warneke, C. L. Pulivarthi, K. Litton et al., The relationship between body composition and response to neoadjuvant chemotherapy in women with operable breast cancer, Oncologist, vol.17, issue.10, pp.1240-1245, 2012.

C. Prado, V. E. Baracos, L. J. Mccargar, T. Reiman, M. Mourtzakis et al.,

, Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment, Clin Cancer Res, vol.15, issue.8, pp.2920-2926, 2009.

S. Dalal, D. Hui, L. Bidaut, K. Lem, D. Fabbro et al., Relationships among body mass index, longitudinal body composition alterations, and survival in patients with locally advanced pancreatic cancer receiving chemoradiation: a pilot study, J Pain Symptom Manage, vol.44, issue.2, pp.181-91, 2012.

P. Peng, O. Hyder, A. Firoozmand, P. Kneuertz, R. D. Schulick et al., Impact of sarcopenia on outcomes following resection of pancreatic adenocarcinoma, J Gastrointest Surg, vol.16, issue.8, pp.1478-86, 2012.

J. R. Lieffers, O. F. Bathe, K. Fassbender, M. Winget, and V. E. Baracos, Sarcopenia is associated with postoperative infection and delayed recovery from colorectal cancer resection surgery, Br J Cancer, vol.107, issue.6, pp.931-937, 2012.

S. S. Shachar, G. R. Williams, H. B. Muss, and T. F. Nishijima, Prognostic value of sarcopenia in adults with solid tumours: A meta-analysis and systematic review, Eur J Cancer, vol.57, pp.58-67, 2016.

E. Deluche, S. Leobon, J. C. Desport, L. Venat-bouvet, J. Usseglio et al., Impact of body composition on outcome in patients with early breast cancer, Support Care Cancer, vol.26, issue.3, pp.861-869, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02019336

A. Bye, B. Sjøblom, T. Wentzel-larsen, B. H. Grønberg, V. E. Baracos et al., Muscle mass and association to quality of life in non-small cell lung cancer patients. J Cachexia Sarcopenia Muscle, vol.8, pp.759-67, 2017.

C. Prado, V. E. Baracos, L. J. Mccargar, M. Mourtzakis, K. E. Mulder et al., Body composition as an independent determinant of 5-fluorouracil-based chemotherapy toxicity, Clin Cancer Res, vol.13, issue.11, pp.3264-3272, 2007.

D. J. Morgan and K. M. Bray, Lean body mass as a predictor of drug dosage. Implications for drug therapy, Clin Pharmacokinet, vol.26, issue.4, pp.292-307, 1994.

R. J. Freedman, N. Aziz, D. Albanes, T. Hartman, D. Danforth et al., Weight and body composition changes during and after adjuvant chemotherapy in women with breast cancer, J Clin Endocrinol Metab, vol.89, issue.5, pp.2248-53, 2004.

C. Prado, I. Lima, V. E. Baracos, R. R. Bies, L. J. Mccargar et al., An exploratory study of body composition as a determinant of epirubicin pharmacokinetics and toxicity, Cancer Chemother Pharmacol, vol.67, issue.1, pp.93-101, 2011.

S. Antoun, V. E. Baracos, L. Birdsell, B. Escudier, and M. B. Sawyer, Low body mass index and sarcopenia associated with dose-limiting toxicity of sorafenib in patients with renal cell carcinoma, Ann Oncol, vol.21, issue.8, pp.1594-1602, 2010.

O. Mir, R. Coriat, B. Blanchet, J. Durand, P. Boudou-rouquette et al., Sarcopenia predicts early dose-limiting toxicities and pharmacokinetics of sorafenib in patients with hepatocellular carcinoma, PLoS ONE, vol.7, issue.5, p.37563, 2012.

C. M. Prado, J. R. Lieffers, L. J. Mccargar, T. Reiman, M. B. Sawyer et al.,

, Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study, The Lancet Oncology, vol.56

D. Hanahan and R. A. Weinberg, The hallmarks of cancer. Cell, vol.100, pp.57-70, 2000.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, issue.5, pp.646-74, 2011.

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, issue.11, pp.991-999, 2002.

R. L. Strausberg, Tumor microenvironments, the immune system and cancer survival

, Genome Biol, vol.6, issue.3, p.211, 2005.

D. S. Chen and I. Mellman, Oncology meets immunology: the cancer-immunity cycle, Immunity, vol.39, issue.1, pp.1-10, 2013.

F. S. Hodi, S. J. O'day, D. F. Mcdermott, R. W. Weber, J. A. Sosman et al., Improved survival with ipilimumab in patients with metastatic melanoma, N Engl J Med, vol.363, issue.8, pp.711-734, 2010.

M. Reck, D. Rodríguez-abreu, A. G. Robinson, R. Hui, T. Cs?szi et al., Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer, N Engl J Med, vol.375, issue.19, pp.1823-1856, 201610.

L. Gandhi, D. Rodríguez-abreu, S. Gadgeel, E. E. Felip, E. et al., Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer, N Engl J Med, vol.378, issue.22, pp.2078-92, 2018.

L. Paz-ares, A. Luft, D. Vicente, A. Tafreshi, M. Gümü? et al., Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer, N Engl J Med, vol.379, issue.21, pp.2040-51, 201822.
URL : https://hal.archives-ouvertes.fr/hal-02345238

M. A. Socinski, R. M. Jotte, F. Cappuzzo, F. Orlandi, D. Stroyakovskiy et al., Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC, N Engl J Med, vol.378, issue.24, pp.2288-301, 2018.

M. D. Hellmann, T. Ciuleanu, A. Pluzanski, J. S. Lee, G. A. Otterson et al., Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden, N Engl J Med, vol.378, issue.22, pp.2093-104, 2018.

R. S. Herbst, P. Baas, D. Kim, E. Felip, J. L. Pérez-gracia et al.,

, Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-smallcell lung cancer (KEYNOTE-010): a randomised controlled trial, Lancet, vol.387, pp.1540-50, 2016.

E. E. Vokes, N. Ready, E. Felip, L. Horn, M. A. Burgio et al., Nivolumab versus docetaxel in previously treated advanced non-small-cell lung cancer (CheckMate 017 and CheckMate 057): 3-year update and outcomes in patients with liver metastases, Ann Oncol, vol.29, issue.4, pp.959-65, 2018.

H. Borghaei, L. Paz-ares, L. Horn, D. R. Spigel, M. Steins et al., Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer, N Engl J Med, vol.373, issue.17, pp.1627-1666, 2015.

A. Rittmeyer, F. Barlesi, D. Waterkamp, K. Park, F. Ciardiello et al.,

, Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial, Lancet, vol.389, pp.255-65, 10066.

R. J. Motzer, N. M. Tannir, D. F. Mcdermott, A. Frontera, O. Melichar et al., Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma, N Engl J Med, vol.378, issue.14, pp.1277-90, 2018.

R. J. Motzer, T. Powles, M. B. Atkins, B. Escudier, D. F. Mcdermott et al., IMmotion151: A Randomized Phase III Study of Atezolizumab Plus Bevacizumab vs Sunitinib in Untreated Metastatic Renal Cell Carcinoma (mRCC), Journal of Clinical Oncology, vol.36, issue.6_suppl, pp.578-578, 2018.

T. K. Choueiri, B. I. Rini, J. Larkin, G. A. Bjarnason, G. Gravis et al.,

, Avelumab plus axitinib vs sunitinib as first-line treatment of advanced renal cell carcinoma: Phase 3 study (JAVELIN Renal 101), Journal of Clinical Oncology, vol.35, issue.15_suppl, pp.4594-4594, 2017.

B. Escudier, C. Porta, M. Schmidinger, N. Rioux-leclercq, A. Bex et al., Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up, Ann Oncol, vol.27, issue.5, pp.58-68, 2016.

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

B. Burtness, K. J. Harrington, R. Greil, D. Soulières, M. Tahara et al., LBA8_PRKEYNOTE-048: Phase III study of first-line pembrolizumab (P) for recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC), Annals of Oncology, vol.77, pp.1856-67, 2016.

T. Y. Seiwert, B. Burtness, R. Mehra, J. Weiss, R. Berger et al., Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial, Lancet Oncol, vol.17, issue.7, pp.956-65, 2016.

E. E. Cohen, K. J. Harrington, L. Tourneau, C. Dinis, J. Licitra et al.,

, LBA45_PRPembrolizumab (pembro) vs standard of care (SOC) for recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC): Phase 3 KEYNOTE-040 trial, Annals of Oncology, vol.80, issue.31, pp.3541-3544, 2015.

S. Champiat, L. Dercle, S. Ammari, C. Massard, A. Hollebecque et al.,

, Hyperprogressive Disease Is a New Pattern of Progression in Cancer Patients Treated by Anti-PD-1/PD-L1, Clin Cancer Res, vol.23, issue.8, pp.1920-1928, 201715.

L. Seymour, J. Bogaerts, A. Perrone, R. Ford, L. H. Schwartz et al., iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics, Lancet Oncol, vol.18, issue.3, pp.143-52, 2017.

J. B. Haanen, .. G. Carbonnel, F. Robert, C. Kerr, K. M. Peters et al., Multi-institutional, Pathologist-Based Assessment of 4 Immunohistochemistry Assays for PD-L1 Expression in Non-Small Cell Lung Cancer, 85. Blank CU, Haanen JB, Ribas A, Schumacher TN. CANCER IMMUNOLOGY. The 'cancer immunogram'. Science, vol.28, pp.58-66, 2016.

C. Chang, J. Qiu, D. O'sullivan, M. D. Buck, T. Noguchi et al., Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression, Cell, vol.162, issue.6, pp.1229-1270, 2015.

S. Cousin, A. Crombe, G. A. , L. Moulec, S. Toulmonde et al., Can body composition (BC) be predictive for outcomes and severe toxicities (ST) in metastatic solid tumors patients (pts) treated with checkpoint inhibitor (CPI)? An analysis of 145 patients, Journal of Clinical Oncology, vol.36, issue.15_suppl, pp.3069-3069, 2018.

M. Revel, B. Raynard, F. Pigneur, D. Palma, M. Toledano et al.,

, Sarcopenia and toxicity of the anti-PD1 inhibitors in real-life lung cancer patients: Results from the French Nationwide SCAN study. JCO, vol.36, pp.21066-21066, 2018.

S. Shachar, S. Fried, R. Shafran, I. Moskovitz, M. T. Williams et al., Body composition as predictor of toxicity and outcomes in patients with metastatic non-small cell lung cancer (mNSCLC) receiving nivolumab (Nivo), Journal of Clinical Oncology, vol.36, issue.15_suppl, pp.21010-21010, 2018.

V. Heidelberger, F. Goldwasser, N. Kramkimel, A. Jouinot, and O. Huillard,

P. Rouquette, Sarcopenic overweight is associated with early acute limiting toxicity of anti-PD1 checkpoint inhibitors in melanoma patients, Invest New Drugs, vol.35, issue.4, pp.436-477, 2017.

F. Fadila, A. Jérôme, B. Frédéric, C. Ferrand, B. Alyssia et al.,

, Haute Autorité de Santé -OPDIVO (nivolumab), anticorps anti-PD1

A. Krebber, L. M. Buffart, G. Kleijn, I. C. Riepma, R. De-bree et al., Prevalence of depression in cancer patients: a meta-analysis of diagnostic interviews and selfreport instruments, Psychooncology, vol.23, issue.2, pp.121-151, 2014.

B. B. Barone, Long-term All-Cause Mortality in Cancer Patients With Preexisting Diabetes Mellitus: A Systematic Review and Meta-analysis, JAMA, vol.300, issue.23, p.2754, 2008.

B. Raynard, F. Pigneur, H. Simon, V. Tissot, D. Vansteene et al., High prevalence but low recognition of sarcopenia in metastatic cancer patients: Results of the scan study, Clinical Nutrition, vol.37, pp.37-45, 2018.

H. A. Parsons, V. E. Baracos, N. Dhillon, D. S. Hong, and R. Kurzrock, Body composition, symptoms, and survival in advanced cancer patients referred to a phase I service, PLoS ONE, vol.7, issue.1, p.29330, 2012.

R. D. Nipp, G. Fuchs, A. El-jawahri, M. J. Troschel, F. M. Greer et al., Sarcopenia Is Associated with Quality of Life and Depression in Patients with Advanced Cancer, Oncologist, vol.23, issue.1, pp.97-104, 2018.

J. Brahmer, K. L. Reckamp, P. Baas, L. Crinò, W. Eberhardt et al., Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer, N Engl J Med, vol.373, issue.2, pp.123-158, 2009.

T. F. Nishijima, H. B. Muss, S. S. Shachar, and S. J. Moschos, Comparison of efficacy of immune checkpoint inhibitors (ICIs) between younger and older patients: A systematic review and meta-analysis, Cancer Treat Rev, vol.45, pp.30-37, 2016.

A. V. Balar, M. D. Galsky, J. E. Rosenberg, T. Powles, D. P. Petrylak et al., Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial, Lancet, vol.389, pp.67-76, 10064.

C. Boutros, A. Tarhini, E. Routier, O. Lambotte, F. L. Ladurie et al., Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination, Nat Rev Clin Oncol, vol.13, issue.8, pp.473-86, 2016.

C. Robert, J. Schachter, G. V. Long, A. Arance, J. J. Grob et al., Pembrolizumab versus Ipilimumab in Advanced Melanoma, N Engl J Med, vol.372, issue.26, pp.2521-2553, 2015.

M. Kostine, L. Rouxel, T. Barnetche, R. Veillon, F. Martin et al., Rheumatic disorders associated with immune checkpoint inhibitors in patients with cancer-clinical aspects and relationship with tumour response: a single-centre prospective cohort study, Ann Rheum Dis, vol.77, issue.3, pp.393-401, 2018.

D. B. Johnson, J. M. Balko, M. L. Compton, S. Chalkias, J. Gorham et al., Fulminant Myocarditis with Combination Immune Checkpoint Blockade, N Engl J Med, vol.375, issue.18, pp.1749-55, 2016.

. Serment-d'hippocrate,

, Au moment d'être admise à exercer la médecine, je promets et je jure d'être fidèle aux lois de l'honneur et de la probité

, Mon premier souci sera de rétablir, de préserver et de promouvoir la santé dans tous ses éléments, physiques et mentaux, individuels et sociaux

, Je respecterai toutes les personnes, leur autonomie et leur volonté, sans aucune discrimination selon leur état ou leurs convictions. J'interviendrai pour les protéger si elles sont affaiblies, vulnérables ou menacées dans leur intégrité ou leur dignité. Même sous la contrainte

, Je ne tromperai jamais leur confiance et n'exploiterai pas le pouvoir hérité des circonstances pour forcer les consciences

, Je ne me laisserai pas influencer par la soif du gain ou la recherche de la gloire

, Admise dans l'intimité des personnes, je respecterai les foyers et ma conduite ne servira pas à corrompre les moeurs

, Je ne prolongerai pas abusivement les agonies. Je ne provoquerai jamais la mort délibérément

, Je n'entreprendrai rien qui dépasse mes compétences. Je les entretiendrai et les perfectionnerai pour assurer au mieux les services qui me seront demandés

, J'apporterai mon aide à mes confrères ainsi qu'à leurs familles dans l'adversité

, Que les hommes et mes confrères m'accordent leur estime si je suis fidèle à mes promesses