J. Ferlay, H. Shin, F. Bray, D. Forman, C. Mathers et al., Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008, International Journal of Cancer, vol.8, issue.19, pp.2893-917, 2008.
DOI : 10.1002/ijc.25516

M. Trakatelli, C. Ulrich, V. Del-marmol, S. Euvrard, S. Euvard et al., Epidemiology of nonmelanoma skin cancer (NMSC) in Europe: accurate and comparable data are needed for effective public health monitoring and interventions, British Journal of Dermatology, vol.26, issue.s3, pp.1-7, 2007.
DOI : 10.1001/archderm.127.7.1029

L. Hollestein, E. De-vries, and T. Nijsten, Trends of cutaneous squamous cell carcinoma in the Netherlands: Increased incidence rates, but stable relative survival and mortality 1989???2008, European Journal of Cancer, vol.48, issue.13, pp.2046-53, 1990.
DOI : 10.1016/j.ejca.2012.01.003

G. Guy and D. Ekwueme, Years of potential life lost and indirect costs of melanoma and nonmelanoma skin cancer: a systematic review of the literature, Pharmacoeconomics, 2011.

J. Bonerandi and S. Monestier, Carcinome épidermoïde (spinocellulaire) et ses précurseurs. Emc -Dermatol, pp.1-18, 2011.
DOI : 10.1016/s0246-0319(11)56345-9

K. Frenkel, Carcinogenesis: Role of Reactive Oxygen and Nitrogen Species
DOI : 10.1016/B0-12-227555-1/00038-1

K. Johnson, T. Wilgus, D. Buggy, M. Kerin, R. Watson et al., Multiple roles for VEGF in non-melanoma skin cancer: angiogenesis and beyond Inhibition of the stress response to breast cancer surgery by regional anesthesia and analgesia does not affect vascular endothelial growth factor and prostaglandin E2, J Skin Cancer. Anesth Analg, vol.2012100, issue.1, pp.244-253, 2005.

J. Talmadge and I. Fidler, AACR Centennial Series: The Biology of Cancer Metastasis: Historical Perspective, Cancer Research, vol.70, issue.14, pp.5649-69, 2010.
DOI : 10.1158/0008-5472.CAN-10-1040

I. Fidler, Timeline: The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited, Nature Reviews Cancer, vol.3, issue.6, pp.453-461, 2003.
DOI : 10.1038/nrc1098

P. Forget, V. Collet, P. Lavand-'homme, D. Kock, and M. , Does analgesia and condition influence immunity after surgery? Effects of fentanyl, ketamine and clonidine on natural killer activity at different ages, European Journal of Anaesthesiology, vol.27, issue.3, pp.233-273, 2010.
DOI : 10.1097/EJA.0b013e32832d540e

C. Jose and R. Rossignol, Rationale for mitochondria-targeting strategies in cancer bioenergetic therapies, The International Journal of Biochemistry & Cell Biology, vol.45, issue.1, pp.123-132
DOI : 10.1016/j.biocel.2012.07.005

T. Soga, Cancer metabolism: Key players in metabolic reprogramming, Cancer Science, vol.10, issue.3, p.2013
DOI : 10.1111/cas.12085

P. Bendriss, P. Dabadie, J. Mazat, L. Letellier, and E. P. , Molecular mechanism of action of local anesthetics]. Ann Françaises Anesthèsie Rèanimation, pp.189-97, 1988.

K. Nouette-gaulain, N. Bellance, B. Prévost, E. Passerieux, C. Pertuiset et al., Erythropoietin Protects against Local Anesthetic Myotoxicity during Continuous Regional Analgesia, Anesthesiology, vol.110, issue.3, pp.648-59, 2009.
DOI : 10.1097/ALN.0b013e3181974f7a

K. Nouette-gaulain, A. Quinart, T. Letellier, and F. Sztark, [Mitochondria in anaesthesia and intensive care]. Ann Françaises Anesthèsie Rèanimation, pp.319-352, 2007.

F. Sztark, M. Malgat, P. Dabadie, and J. Mazat, Comparison of the Effects of Bupivacaine and Ropivacaine on Heart Cell Mitochondrial Bioenergetics, Anesthesiology, vol.88, issue.5, pp.1340-1349, 1998.
DOI : 10.1097/00000542-199805000-00026

O. Cela, C. Piccoli, R. Scrima, G. Quarato, A. Marolla et al., Bupivacaine uncouples the mitochondrial oxidative phosphorylation, inhibits respiratory chain complexes I and III and enhances ROS production: Results of a study on cell cultures, Mitochondrion, vol.10, issue.5, pp.487-96, 2010.
DOI : 10.1016/j.mito.2010.05.005

K. Nouette-gaulain, P. Sirvent, M. Canal-raffin, D. Morau, M. Malgat et al., Effects of Intermittent Femoral Nerve Injections of Bupivacaine, Levobupivacaine, and Ropivacaine on Mitochondrial Energy Metabolism and Intracellular Calcium Homeostasis in Rat Psoas Muscle, Anesthesiology, vol.106, issue.5
DOI : 10.1097/01.anes.0000265164.29630.b4

R. Demicheli, M. Retsky, W. Hrushesky, M. Baum, and I. Gukas, The effects of surgery on tumor growth: a century of investigations, Annals of Oncology, vol.19, issue.11, 2008.
DOI : 10.1093/annonc/mdn386

Y. Goldfarb and S. Ben-eliyahu, Surgery as a Risk Factor for Breast Cancer Recurrence and Metastasis: Mediating Mechanisms and Clinical Prophylactic Approaches, Breast Disease, vol.26, issue.1, pp.99-114, 2006.
DOI : 10.3233/BD-2007-26109

E. Neeman and S. Ben-eliyahu, Surgery and stress promote cancer metastasis: New outlooks on perioperative mediating mechanisms and immune involvement, Brain, Behavior, and Immunity, vol.30, 2013.
DOI : 10.1016/j.bbi.2012.03.006

A. Heaney and D. Buggy, Can anaesthetic and analgesic techniques affect cancer recurrence or metastasis?, British Journal of Anaesthesia, vol.109, issue.suppl 1, pp.17-28
DOI : 10.1093/bja/aes421

URL : http://bja.oxfordjournals.org/cgi/content/short/109/suppl_1/i17

J. Lloyd, C. Mciver, S. Stephenson, P. Hewett, N. Rieger et al., Identification of Early-Stage Colorectal Cancer Patients at Risk of Relapse Post-Resection by Immunobead Reverse Transcription-PCR Analysis of Peritoneal Lavage Fluid for Malignant Cells, Clinical Cancer Research, vol.12, issue.2, pp.417-440, 2006.
DOI : 10.1158/1078-0432.CCR-05-1473

J. Kundu and Y. Surh, Inflammation: Gearing the journey to cancer, Mutation Research/Reviews in Mutation Research, vol.659, issue.1-2, pp.15-30, 2008.
DOI : 10.1016/j.mrrev.2008.03.002

S. Grivennikov, F. Greten, and K. M. , Immunity, inflammation, and cancer. Cell, pp.883-99, 2010.

Y. Tsuchiya, S. Sawada, I. Yoshioka, Y. Ohashi, M. Matsuo et al., Increased surgical stress promotes tumor metastasis, Surgery, vol.133, issue.5, pp.547-55, 2003.
DOI : 10.1067/msy.2003.141

F. Vittimberga, . Jr, D. Foley, W. Meyers, and M. Callery, Laparoscopic Surgery and the Systemic Immune Response, Annals of Surgery, vol.227, issue.3, pp.326-360, 1998.
DOI : 10.1097/00000658-199803000-00003

G. Snyder and S. Greenberg, Effect of anaesthetic technique and other perioperative factors on cancer recurrence, British Journal of Anaesthesia, vol.105, issue.2, pp.106-121, 2010.
DOI : 10.1093/bja/aeq164

H. Beloeil and K. Nouette-gaulain, The perioperative period in cancer surgery: a critical moment! Is there a role for regional anesthesia in preventing cancer recurrence?]. Ann Françaises Anesthèsie Rèanimation, pp.528-564, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01134832

O. Reilly, M. Boehm, T. Shing, Y. Fukai, N. Vasios et al., Endostatin: An Endogenous Inhibitor of Angiogenesis and Tumor Growth, Cell, vol.88, issue.2, pp.277-85, 1997.
DOI : 10.1016/S0092-8674(00)81848-6

M. Pascual, S. Alonso, D. Parés, R. Courtier, M. Gil et al., Randomized clinical trial comparing inflammatory and angiogenic response after open versus laparoscopic curative resection for colonic cancer, British Journal of Surgery, vol.31, issue.1, pp.50-59, 2011.
DOI : 10.1002/bjs.7258

G. Page, W. Blakely, and S. Ben-eliyahu, Evidence that postoperative pain is a mediator of the tumor-promoting effects of surgery in rats, Pain, vol.90, issue.1, pp.191-200, 2001.
DOI : 10.1016/S0304-3959(00)00403-6

Y. Shavit, F. Martin, R. Yirmiya, S. Ben-eliyahu, G. Terman et al., Effects of a single administration of morphine or footshock stress on natural killer cell cytotoxicity, Brain, Behavior, and Immunity, vol.1, issue.4, pp.318-346, 1987.
DOI : 10.1016/0889-1591(87)90034-1

J. Lewis, Y. Shavit, G. Terman, L. Nelson, R. Gale et al., Apparent involvement of opioid peptides in stress-induced enhancement of tumor growth, Peptides, vol.4, issue.5, pp.635-643, 1983.
DOI : 10.1016/0196-9781(83)90010-4

G. Page and S. Ben-eliyahu, Indomethacin attenuates the immunosuppressive and tumor-promoting effects of surgery, The Journal of Pain, vol.3, issue.4, pp.301-309, 2002.
DOI : 10.1054/jpai.2002.125184

S. Ben-eliyahu, G. Shakhar, E. Rosenne, Y. Levinson, and B. Beilin, Hypothermia in Barbiturate-anesthetized Rats Suppresses Natural Killer Cell Activity and Compromises Resistance to Tumor Metastasis, Anesthesiology, vol.91, issue.3, pp.732-772, 1999.
DOI : 10.1097/00000542-199909000-00026

A. Kurz, D. Sessler, and R. Lenhardt, Perioperative Normothermia to Reduce the Incidence of Surgical-Wound Infection and Shorten Hospitalization, New England Journal of Medicine, vol.334, issue.19, pp.1209-1224, 1996.
DOI : 10.1056/NEJM199605093341901

B. Beilin, Y. Shavit, J. Razumovsky, Y. Wolloch, A. Zeidel et al., Effects of Mild Perioperative Hypothermia on Cellular Immune Responses, Anesthesiology, vol.89, issue.5, pp.1133-1173, 1998.
DOI : 10.1097/00000542-199811000-00013

L. Ydy, N. Slhessarenko, and J. De-aguilar-nascimento, Effect of Perioperative Allogeneic Red Blood Cell Transfusion on the Immune-Inflammatory Response After Colorectal Cancer Resection, World Journal of Surgery, vol.35, issue.Suppl 7A, pp.2044-51, 2007.
DOI : 10.1007/s00268-007-9159-3

J. Kaplan, S. Sarnaik, J. Gitlin, and J. Lusher, Diminished helper/suppressor lymphocyte ratios and natural killer activity in recipients of repeated blood transfusions. Blood, pp.308-318, 1984.

G. Chen, F. Zhang, M. Gong, and M. Yan, Effect of perioperative autologous versus allogeneic blood transfusion on the immune system in gastric cancer patients, Journal of Zhejiang University SCIENCE B, vol.8, issue.8, 2007.
DOI : 10.1631/jzus.2007.B0560

M. Blajchman, L. Bardossy, C. R. Sastry, A. Singal, and D. , Allogeneic blood transfusioninduced enhancement of tumor growth: two animal models showing amelioration by leukodepletion and passive transfer using spleen cells, Blood, vol.81, issue.7, pp.1880-1882, 1993.

A. Amato and M. Pescatori, Perioperative blood transfusions for the recurrence of colorectal cancer, Cochrane Database Syst Rev Online, issue.1, p.5033, 2006.

R. Weber, N. Jabbour, and R. Martin, Anemia and Transfusions in Patients Undergoing Surgery for Cancer, Annals of Surgical Oncology, vol.5, issue.Suppl 2, pp.34-45, 2008.
DOI : 10.1245/s10434-007-9502-9

J. Caro, M. Salas, A. Ward, and G. Goss, Anemia as an independent prognostic factor for survival in patients with cancer: a systemic, quantitative review. Cancer, Jun, vol.1591, issue.12, pp.2214-2235, 2001.

R. Melamed, S. Bar-yosef, G. Shakhar, K. Shakhar, and S. Ben-eliyahu, Suppression of Natural Killer Cell Activity and Promotion of Tumor Metastasis by Ketamine, Thiopental, and Halothane, but Not by Propofol: Mediating Mechanisms and Prophylactic Measures, Anesthesia & Analgesia, vol.97, issue.5, pp.1331-1340, 2003.
DOI : 10.1213/01.ANE.0000082995.44040.07

E. Monzani, A. Shtil, L. Porta, and C. , The water channels, new druggable targets to combat cancer cell survival, invasiveness and metastasis. Curr Drug Targets, pp.1132-1139, 2007.

H. Niwa, D. Rowbotham, D. Lambert, D. Buggy, B. Beilin et al., Can anesthetic techniques or drugs affect cancer recurrence in patients undergoing cancer surgery? J Anesth Low-dose ketamine affects immune responses in humans during the early postoperative period, Br J Anaesth, vol.5699, issue.4, pp.522-529, 2007.

M. Tsuchiya, A. Asada, K. Arita, T. Utsumi, T. Yoshida et al., Induction and mechanism of apoptotic cell death by propofol in HL-60 cells, Acta Anaesthesiologica Scandinavica, vol.20, issue.9, pp.1068-74, 2002.
DOI : 10.1016/S0168-8227(98)00044-8

T. Inada, K. Kubo, K. Shingu, T. Inada, and K. Shingu, Possible link between cyclooxygenase-inhibiting and antitumor properties of propofol Enhancement of antitumor immunity after propofol treatment in mice, J Anesth Immunopharmacol Immunotoxicol, vol.2529, issue.4, pp.569-753, 2007.

T. Mammoto, M. Mukai, A. Mammoto, Y. Yamanaka, Y. Hayashi et al., Intravenous anesthetic, propofol inhibits invasion of cancer cells. Cancer Lett, pp.165-70, 2002.

Y. Miao, Y. Zhang, H. Wan, L. Chen, and F. Wang, GABA-receptor agonist, propofol inhibits invasion of colon carcinoma cells, Biomedicine & Pharmacotherapy, vol.64, issue.9, 2010.
DOI : 10.1016/j.biopha.2010.03.006

V. Garib, K. Lang, B. Niggemann, K. Zänker, L. Brandt et al., Propofol-induced calcium signalling and actin reorganization within breast carcinoma cells, European Journal of Anaesthesiology, vol.22, issue.8, 2005.
DOI : 10.1017/S026502150500102X

V. Garib, B. Niggemann, K. Zänker, L. Brandt, and B. Kubens, Influence of non-volatile anesthetics on the migration behavior of the human breast cancer cell line MDA-MB-468, Acta Anaesthesiologica Scandinavica, vol.20, issue.7, pp.836-880, 2002.
DOI : 10.1023/A:1006548902592

T. Inada, Y. Yamanouchi, S. Jomura, S. Sakamoto, M. Takahashi et al., Effect of propofol and isoflurane anaesthesia on the immune response to surgery, Anaesthesia, vol.171, issue.10, 2004.
DOI : 10.1016/0167-5699(94)90220-8

R. Wu, K. Wu, J. Yang, S. Chiou, C. Yu et al., Etomidate induces cytotoxic effects and gene expression in a murine leukemia macrophage cell line

E. Fleischmann, C. Marschalek, K. Schlemitz, J. Dalton, T. Gruenberger et al., Nitrous oxide may not increase the risk of cancer recurrence after colorectal surgery: a follow-up of a randomized controlled trial Different apoptosis ratios and gene expressions in two human cell lines after sevoflurane anaesthesia, Bmc Anesth. Acta Anaesthesiol Scand, vol.953, issue.9, pp.1192-1201, 2009.

S. Kvolik, L. Glavas-obrovac, V. Bares, and I. Karner, Effects of inhalation anesthetics halothane, sevoflurane, and isoflurane on human cell lines, Life Sciences, vol.77, issue.19, pp.2369-83, 2005.
DOI : 10.1016/j.lfs.2004.12.052

T. Loop, D. Dovi-akue, M. Frick, M. Roesslein, L. Egger et al., Volatile Anesthetics Induce Caspase-dependent, Mitochondria-mediated Apoptosis in Human T Lymphocytes In Vitro, Anesthesiology, vol.102, issue.6
DOI : 10.1097/00000542-200506000-00014

H. Wada, S. Seki, T. Takahashi, N. Kawarabayashi, H. Higuchi et al., Combined spinal and general anesthesia attenuates liver metastasis by preserving TH1

M. Tylman, R. Sarbinowski, J. Bengtson, A. Kvarnström, and A. Bengtsson, Inflammatory response in patients undergoing colorectal cancer surgery: the effect of two different anesthetic techniques

B. Afsharimani, P. Cabot, and M. Parat, Morphine and tumor growth and metastasis, Cancer and Metastasis Reviews, vol.579, issue.1???3, pp.225-263, 2011.
DOI : 10.1007/s10555-011-9285-0

URL : http://espace.library.uq.edu.au/view/UQ:229184/CancerMetRev_Morphine_and_tumor_growth.pdf

B. Beilin, Y. Shavit, J. Hart, B. Mordashov, S. Cohn et al., Effects of anesthesia based on large versus small doses of fentanyl on natural killer cell cytotoxicity in the perioperative period

C. Gavériaux-ruff, H. Matthes, J. Peluso, and B. Kieffer, Abolition of morphine-immunosuppression in mice lacking the ??-opioid receptor gene, Proceedings of the National Academy of Sciences, vol.95, issue.11, pp.6326-6356, 1998.
DOI : 10.1073/pnas.95.11.6326

M. Yeager, T. Colacchio, C. Yu, L. Hildebrandt, A. Howell et al., Morphine Inhibits Spontaneous and Cytokine-enhanced Natural Killer Cell Cytotoxicity in Volunteers, Anesthesiology, vol.83, issue.3, 1995.
DOI : 10.1097/00000542-199509000-00008

B. Mathew, F. Lennon, J. Siegler, T. Mirzapoiazova, N. Mambetsariev et al., The Novel Role of the Mu Opioid Receptor in Lung Cancer Progression, Anesthesia & Analgesia, vol.112, issue.3, pp.558-67, 2011.
DOI : 10.1213/ANE.0b013e31820568af

A. Cronin, N. Aucutt-walter, T. Budinetz, C. Bonafide, N. Divittore et al., Low-dose remifentanil infusion does not impair natural killer cell function in healthy volunteers, British Journal of Anaesthesia, vol.91, issue.6, pp.805-814, 2003.
DOI : 10.1093/bja/aeg273

K. Gupta, S. Kshirsagar, L. Chang, R. Schwartz, P. Law et al., Morphine stimulates angiogenesis by activating proangiogenic and survival-promoting signaling and promotes breast tumor growth, Cancer Res, vol.62, issue.15, pp.4491-4499, 2002.

P. Singleton, J. Garcia, and J. Moss, Synergistic effects of methylnaltrexone with 5-fluorouracil and bevacizumab on inhibition of vascular endothelial growth factor-induced angiogenesis, Molecular Cancer Therapeutics, vol.7, issue.6, pp.1669-79, 2008.
DOI : 10.1158/1535-7163.MCT-07-2217

Y. Harimaya, K. Koizumi, T. Andoh, H. Nojima, Y. Kuraishi et al., Potential ability of morphine to inhibit the adhesion, invasion and metastasis of metastatic colon 26-L5 carcinoma cells. Cancer Lett, pp.121-128, 2002.

I. Hatsukari, N. Hitosugi, R. Ohno, K. Hashimoto, S. Nakamura et al., Induction of apoptosis by morphine in human tumor cell lines in vitro, Anticancer Res, vol.27, issue.2, pp.857-64, 2007.

G. Page, S. Ben-eliyahu, R. Yirmiya, and J. Liebeskind, Morphine attenuates surgery-induced enhancement of metastatic colonization in rats, Pain, vol.54, issue.1, pp.21-29, 1993.
DOI : 10.1016/0304-3959(93)90095-7

G. Page, J. Mcdonald, and S. Ben-eliyahu, Pre-operative versus postoperative administration of morphine: impact on the neuroendocrine, behavioural, and metastatic-enhancing effects of surgery, British Journal of Anaesthesia, vol.81, issue.2, pp.216-239, 1998.
DOI : 10.1093/bja/81.2.216

T. Sasamura, S. Nakamura, Y. Iida, H. Fujii, J. Murata et al., Morphine analgesia suppresses tumor growth and metastasis in a mouse model of cancer pain produced by orthotopic tumor inoculation, European Journal of Pharmacology, vol.441, issue.3, pp.185-91, 2002.
DOI : 10.1016/S0014-2999(02)01450-4

F. Sinicrope and S. Gill, Role of cyclooxygenase-2 in colorectal cancer, Cancer and Metastasis Reviews, vol.23, issue.1/2, pp.63-75, 2004.
DOI : 10.1023/A:1025863029529

D. Wang and R. Dubois, Eicosanoids and cancer, Nature Reviews Cancer, vol.133, issue.3, pp.181-93, 2010.
DOI : 10.1038/nrc2809

D. Holt, X. Ma, N. Kundu, F. A. Prostaglandin, and E. , PGE (2)) suppresses natural killer cell function primarily through the PGE(2) receptor EP4, Cancer Immunol Immunother Cii, issue.2, 2011.

R. C. Harris, A. Laiyemo, B. Graubard, A. Hollenbeck, A. Schatzkin et al., cox-2) blockade in the chemoprevention of cancers of the colon, breast, prostate, and lung Non-steroidal anti-inflammatory drugs and colorectal cancer risk in a large, prospective cohort, Inflammopharmacology. Am J Gastroenterol, vol.17106, issue.227, pp.55-671340, 2009.

Y. Zhao, S. Zhu, X. Li, F. Wang, F. Hu et al., Association between NSAIDs use and breast cancer risk: a systematic review and meta-analysis, Breast Cancer Research and Treatment, vol.365, issue.9472, pp.141-50, 2009.
DOI : 10.1007/s10549-008-0228-6

P. Renzi and L. Ginns, Effect of Lidocaine on Natural Killer Activity: Rapid Inhibition of Lysis, Immunopharmacology and Immunotoxicology, vol.129, issue.97
DOI : 10.1016/S0065-2776(08)60241-2

P. Renzi and L. Ginns, Natural Killer Activity is Present in Rat Lung Lavage and Inhibited by Lidocaine, Immunopharmacology and Immunotoxicology, vol.129, issue.3, pp.389-415, 1990.
DOI : 10.1378/chest.95.2.383

S. Takagi, S. Kitagawa, K. Oshimi, F. Takaku, and Y. Miura, Effect of local anaesthetics on human natural killer cell activity, Clin Exp Immunol, vol.53, issue.2, pp.477-81, 1983.

J. Krog, M. Hokland, P. Ahlburg, E. Parner, and E. Tønnesen, Lipid solubility- and concentration-dependent attenuation of in vitro natural killer cell cytotoxicity by local anesthetics, Acta Anaesthesiologica Scandinavica, vol.7, issue.7, pp.875-81, 2002.
DOI : 10.1056/NEJM200007063430107

T. Martinsson, Ropivacaine inhibits serum-induced proliferation of colon adenocarcinoma cells in vitro, J Pharmacol Exp Ther. Feb, vol.288, issue.2, pp.660-664, 1999.

E. Lucchinetti, A. Awad, M. Rahman, J. Feng, P. Lou et al., Antiproliferative Effects of Local Anesthetics on Mesenchymal Stem Cells, Anesthesiology, vol.116, issue.4, pp.841-56
DOI : 10.1097/ALN.0b013e31824babfe

M. Sakaguchi, Y. Kuroda, and M. Hirose, The Antiproliferative Effect of Lidocaine on Human Tongue Cancer Cells with Inhibition of the Activity of Epidermal Growth Factor Receptor, Anesthesia & Analgesia, vol.102, issue.4, 2006.
DOI : 10.1213/01.ane.0000198330.84341.35

T. Mammoto, S. Higashiyama, M. Mukai, A. Mammoto, M. Ayaki et al., Infiltration anesthetic lidocaine inhibits cancer cell invasion by modulating ectodomain shedding of heparin-binding epidermal growth factor-like growth factor (HB-EGF), Journal of Cellular Physiology, vol.232, issue.3, pp.351-359, 2002.
DOI : 10.1002/jcp.10145

Y. Arai, T. Kondo, K. Tanabe, Q. Zhao, F. Li et al., Enhancement of hyperthermiainduced apoptosis by local anesthetics on human histiocytic lymphoma U937 cells, J Biol Chem, 2002.

P. Lirk, R. Berger, M. Hollmann, and H. Fiegl, Lidocaine time- and dose-dependently demethylates deoxyribonucleic acid in breast cancer cell lines in vitro, British Journal of Anaesthesia, vol.109, issue.2, pp.200-207, 2012.
DOI : 10.1093/bja/aes128

M. Karniel, R. R. Beitner, G. Page, G. Shakhar, K. Shakhar et al., Local anesthetics induce a decrease in the levels of glucose 1, 6- bisphosphate, fructose 1,6-bisphosphate, and ATP, and in the viability of melanoma cells Attenuation of the tumor-promoting effect of surgery by spinal blockade in rats, Mol Genet Metab. Anesthesiology, vol.6994, issue.1056, pp.40-51066, 2000.

M. Salo and M. Nissilä, Cell-mediated and humoral immune responses to total hip replacement under spinal or general anaesthesia, Acta Anaesthesiologica Scandinavica, vol.75, issue.Suppl. 550, pp.241-249, 1990.
DOI : 10.1111/j.1399-6576.1990.tb03078.x

W. Koltun, M. Bloomer, A. Tilberg, J. Seaton, O. Ilahi et al., Awake epidural anesthesia is associated with improved natural killer cell cytotoxicity and a reduced stress response, The American Journal of Surgery, vol.171, issue.1
DOI : 10.1016/S0002-9610(99)80076-2

C. Deegan, D. Murray, P. Doran, P. Ecimovic, D. Moriarty et al., Effect of anaesthetic technique on oestrogen receptor-negative breast cancer cell function in vitro, British Journal of Anaesthesia, vol.103, issue.5, 2009.
DOI : 10.1093/bja/aep261

I. Conrick-martin, M. Kell, and D. Buggy, Meta-analysis of the effect of central neuraxial regional anesthesia compared with general anesthesia on postoperative natural killer T lymphocyte function, Journal of Clinical Anesthesia, vol.24, issue.1, pp.3-7, 2012.
DOI : 10.1016/j.jclinane.2011.09.001

B. Schlagenhauff, U. Ellwanger, H. Breuninger, W. Stroebel, G. Rassner et al., Prognostic impact of the type of anaesthesia used during the excision of primary cutaneous melanoma, Melanoma Research, vol.10, issue.2
DOI : 10.1097/00008390-200004000-00009

A. Exadaktylos, D. Buggy, D. Moriarty, E. Mascha, and D. Sessler, Can anesthetic technique for primary breast cancer surgery affect recurrence or metastasis? Anesthesiology, pp.660-664, 2006.

B. Biki, E. Mascha, D. Moriarty, J. Fitzpatrick, D. Sessler et al., Anesthetic Technique for Radical Prostatectomy Surgery Affects Cancer Recurrence, Anesthesiology, vol.109, issue.2, pp.180-187, 2008.
DOI : 10.1097/ALN.0b013e31817f5b73

B. Tsui, S. Rashiq, D. Schopflocher, A. Murtha, S. Broemling et al., Anesth??sie p??ridurale et taux de r??cidive du cancer apr??s une prostatectomie radicale, Canadian Journal of Anesthesia/Journal canadien d'anesth??sie, vol.294, issue.2, pp.107-119, 2010.
DOI : 10.1007/s12630-009-9214-7

P. Wuethrich, H. Schmitz, S. Kessler, T. Thalmann, G. Studer et al., Potential Influence of the Anesthetic Technique Used during Open Radical Prostatectomy on Prostate Cancer-Related Outcome, Anesthesiology, vol.113, issue.3, pp.570-576, 2010.
DOI : 10.1097/ALN.0b013e3181e4f6ec

H. Ismail, K. Ho, K. Narayan, and S. Kondalsamy-chennakesavan, Effect of neuraxial anaesthesia on tumour progression in cervical cancer patients treated with brachytherapy: a retrospective cohort study, British Journal of Anaesthesia, vol.105, issue.2, pp.145-154, 2010.
DOI : 10.1093/bja/aeq156

L. Lin, C. Liu, H. Tan, H. Ouyang, Y. Zhang et al., Anaesthetic technique may affect prognosis for ovarian serous adenocarcinoma: a retrospective analysis, British Journal of Anaesthesia, vol.106, issue.6, pp.814-836, 2011.
DOI : 10.1093/bja/aer055

R. Christopherson, K. James, M. Tableman, P. Marshall, and F. Johnson, Long-term survival after colon cancer surgery: a variation associated with choice of anesthesia, Anesth Analg, 2008.

A. Gottschalk, J. Ford, C. Regelin, J. You, E. Mascha et al., Association between Epidural Analgesia and Cancer Recurrence after Colorectal Cancer Surgery, Anesthesiology, vol.113, issue.1, 2010.
DOI : 10.1097/ALN.0b013e3181de6d0d

A. Gupta, A. Björnsson, M. Fredriksson, O. Hallböök, and C. Eintrei, Reduction in Mortality After Epidural Anaesthesia and Analgesia in Patients Undergoing Rectal But Not Colonic Cancer Surgery, Survey of Anesthesiology, vol.56, issue.4, 2011.
DOI : 10.1097/01.SA.0000415549.45668.24

P. Myles, P. Peyton, B. Silbert, J. Hunt, J. Rigg et al., Perioperative epidural analgesia for major abdominal surgery for cancer and recurrence-free survival: randomised trial Can regional analgesia reduce the risk of recurrence after breast cancer? Methodology of a multicenter randomized trial, BMJ. Contemp Clin Trials, vol.34229, issue.4, pp.1491-122517, 2008.

X. Capdevila, M. Ponrouch, and O. Choquet, Continuous peripheral nerve blocks in clinical practice, Current Opinion in Anaesthesiology, vol.21, issue.5
DOI : 10.1097/ACO.0b013e32830c66c2

M. Andreae and D. Andreae, Local anaesthetics and regional anaesthesia for preventing chronic pain after surgery, Cochrane Database Syst Rev. @@@@@@@@@@@@@@@@@@@@@@@@@@@@@@@ @@@@@@@@@@@@@@@@@@@@@@@ @@@@@@@@@@@@@@@@@@@@@@@@@, vol.10, p.7105, 2012.
DOI : 10.1002/14651858.cd007105