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, pp.1821-1828, 2008.
DOI : 10.1093/annonc/mdn386

S. Hofer, G. Molema, R. Hermens, H. Wanebo, J. Reichner et al., The effect of surgical wounding on tumour development, European Journal of Surgical Oncology (EJSO), vol.25, issue.3, pp.231-243, 1999.
DOI : 10.1053/ejso.1998.0634

R. Abramovitch, M. Marikovsky, G. Meir, and M. Neeman, Stimulation of tumour growth by wound-derived growth factors, British Journal of Cancer, vol.79, issue.9/10, pp.1392-1398, 1999.
DOI : 10.1038/sj.bjc.6690223

P. Paraskeva, P. Ridgway, S. Olsen, C. Isacke, D. Peck et al., A surgically induced hypoxic environment causes changes in the metastatic behaviour of tumours in vitro, Clinical & Experimental Metastasis, vol.227, issue.3, pp.149-157, 2006.
DOI : 10.1007/s10585-006-9028-8

O. Leary, D. Wang, J. Cotter, T. Redmond, and H. , Less stress, more success? Oncological implications of surgery-induced oxidative stress, Gut, vol.62, issue.3, pp.461-470, 2013.
DOI : 10.1136/gutjnl-2011-300948

M. Delavary, B. Van-der-veer, W. Van-egmond, M. Niessen, F. Beelen et al., Macrophages in skin injury and repair, Immunobiology, vol.216, issue.7, pp.753-762, 2011.
DOI : 10.1016/j.imbio.2011.01.001

P. Allavena and A. Mantovani, Immunology in the clinic review series; focus on cancer: tumour-associated macrophages: undisputed stars of the inflammatory tumour microenvironment, Clinical & Experimental Immunology, vol.441, issue.2, pp.195-205, 2012.
DOI : 10.1111/j.1365-2249.2011.04515.x

M. Tham, K. Tan, J. Keeble, X. Wang, S. Hubert et al., Melanoma-initiating cells exploit M2 macrophage TGF?? and arginase pathway for survival and proliferation, Oncotarget, vol.5, issue.23, pp.12027-12042, 2014.
DOI : 10.18632/oncotarget.2482

J. Predina, J. B. Fridlender, Z. Aliperti, L. Madajewski, B. Kapoor et al., A positive-margin resection model recreates the postsurgical tumor microenvironment and is a reliable model for adjuvant therapy evaluation, Cancer Biology & Therapy, vol.142, issue.9, pp.745-755, 2012.
DOI : 10.1172/JCI0215324

B. Toh, X. Wang, J. Keeble, W. Sim, K. Khoo et al., Mesenchymal Transition and Dissemination of Cancer Cells Is Driven by Myeloid-Derived Suppressor Cells Infiltrating the Primary Tumor, PLoS Biology, vol.11, issue.9, p.1001162, 2011.
DOI : 10.1371/journal.pbio.1001162.s012

H. Ohno, Y. Uemura, H. Murooka, H. Takanashi, T. Tokieda et al., The orally-active and selective c-Fms tyrosine kinase inhibitor Ki20227 inhibits disease progression in a collagen-induced arthritis mouse model, European Journal of Immunology, vol.44, issue.1, pp.283-291, 2008.
DOI : 10.1002/eji.200737199

R. Casero, . Jr, and L. Marton, Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases, Nature Reviews Drug Discovery, vol.272, issue.5, pp.373-390, 2007.
DOI : 10.1038/nrd2243

G. Palmieri, P. Ascierto, F. Perrone, S. Satriano, A. Ottaiano et al., Prognostic Value of Circulating Melanoma Cells Detected by Reverse Transcriptase???Polymerase Chain Reaction, Journal of Clinical Oncology, vol.21, issue.5, pp.767-773, 2003.
DOI : 10.1200/JCO.2003.01.128

S. Vinokurova, N. Wentzensen, J. Einenkel, R. Klaes, C. Ziegert et al., Clonal History of Papillomavirus-Induced Dysplasia in the Female Lower Genital Tract, JNCI Journal of the National Cancer Institute, vol.97, issue.24, pp.1816-1821, 2005.
DOI : 10.1093/jnci/dji428

W. Tseng, N. Fadaki, and S. Leong, Metastatic Tumor Dormancy in Cutaneous Melanoma: Does Surgery Induce Escape?, Cancers, vol.3, issue.4, pp.730-746, 2011.
DOI : 10.3390/cancers3010730

E. Suzuki, S. Kim, H. Cheung, M. Corbley, X. Zhang et al., A Novel Small-Molecule Inhibitor of Transforming Growth Factor ?? Type I Receptor Kinase (SM16) Inhibits Murine Mesothelioma Tumor Growth In vivo and Prevents Tumor Recurrence after Surgical Resection, Cancer Research, vol.67, issue.5, pp.2351-2359, 2007.
DOI : 10.1158/0008-5472.CAN-06-2389

A. Bonde, V. Tischler, S. Kumar, A. Soltermann, and R. Schwendener, Intratumoral macrophages contribute to epithelial-mesenchymal transition in solid tumors, BMC Cancer, vol.27, issue.1, p.35, 2012.
DOI : 10.1038/sj.onc.1210915

J. Wyckoff, Y. Wang, E. Lin, J. Li, S. Goswami et al., Direct Visualization of Macrophage-Assisted Tumor Cell Intravasation in Mammary Tumors, Cancer Research, vol.67, issue.6, pp.2649-2656, 2007.
DOI : 10.1158/0008-5472.CAN-06-1823

B. Qian, Y. Deng, J. Im, R. Muschel, Y. Zou et al., A Distinct Macrophage Population Mediates Metastatic Breast Cancer Cell Extravasation, Establishment and Growth, PLoS ONE, vol.11, issue.8, p.6562, 2009.
DOI : 10.1371/journal.pone.0006562.s012

K. Hoek, O. Eichhoff, N. Schlegel, U. Dobbeling, N. Kobert et al., In vivo Switching of Human Melanoma Cells between Proliferative and Invasive States, Cancer Research, vol.68, issue.3, pp.650-656, 2008.
DOI : 10.1158/0008-5472.CAN-07-2491

V. Battaglia, D. Shields, C. Murray-stewart, T. Casero, R. et al., Polyamine catabolism in carcinogenesis: potential targets for chemotherapy and chemoprevention, Amino Acids, vol.185, issue.10, pp.511-520, 2014.
DOI : 10.1007/s00726-013-1529-6

Q. Li, L. Liu, Q. Zhang, S. Liu, D. Ge et al., Interleukin-17 Indirectly Promotes M2 Macrophage Differentiation through Stimulation of COX-2/PGE2 Pathway in the Cancer Cells, Cancer Research and Treatment, vol.46, issue.3, pp.297-306, 2014.
DOI : 10.4143/crt.2014.46.3.297

L. Liu, D. Ge, L. Ma, J. Mei, S. Liu et al., Interleukin-17 and Prostaglandin E2 Are Involved in Formation of an M2 Macrophage-Dominant Microenvironment in Lung Cancer, Journal of Thoracic Oncology, vol.7, issue.7, pp.1091-1100, 2012.
DOI : 10.1097/JTO.0b013e3182542752

L. Shaashua, E. Rosenne, E. Neeman, L. Sorski, L. Sominsky et al., Plasma IL-12 levels are suppressed in vivo by stress and surgery through endogenous release of glucocorticoids and prostaglandins but not catecholamines or opioids, Psychoneuroendocrinology, vol.42, pp.11-23, 2014.
DOI : 10.1016/j.psyneuen.2013.12.001

H. Hill, T. Conway, . Jr, M. Sabel, Y. Jong et al., Cancer immunotherapy with interleukin 12 and granulocyte-macrophage colony-stimulating factor-encapsulated microspheres: coinduction of innate and adaptive antitumor immunity and cure of disseminated disease, Cancer Res, vol.62, pp.7254-7263, 2002.

W. Jarnagin, J. Zager, D. Klimstra, K. Delman, S. Malhotra et al., Neoadjuvant treatment of hepatic malignancy: an oncolytic herpes simplex virus expressing IL-12 effectively treats the parent tumor and protects against recurrence-after resection, Cancer Gene Therapy, vol.10, issue.3, pp.215-223, 2003.
DOI : 10.1038/sj.cgt.7700558

Y. Goldfarb, L. Sorski, M. Benish, B. Levi, R. Melamed et al., Improving Postoperative Immune Status and Resistance to Cancer Metastasis, Annals of Surgery, vol.253, issue.4, pp.798-810, 2011.
DOI : 10.1097/SLA.0b013e318211d7b5

L. Tai, C. De-souza, S. Belanger, L. Ly, A. Alkayyal et al., Preventing Postoperative Metastatic Disease by Inhibiting Surgery-Induced Dysfunction in Natural Killer Cells, Cancer Research, vol.73, issue.1, pp.97-107, 2013.
DOI : 10.1158/0008-5472.CAN-12-1993

S. Pyonteck, L. Akkari, A. Schuhmacher, R. Bowman, L. Sevenich et al., CSF-1R inhibition alters macrophage polarization and blocks glioma progression, Nature Medicine, vol.102, issue.10, pp.1264-1272, 2013.
DOI : 10.1038/nm.3337

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3840724

C. Ries, M. Cannarile, S. Hoves, J. Benz, K. Wartha et al., Targeting Tumor-Associated Macrophages with Anti-CSF-1R Antibody Reveals a Strategy for Cancer Therapy, Cancer Cell, vol.25, issue.6, pp.846-859, 2014.
DOI : 10.1016/j.ccr.2014.05.016

R. Lengagne, S. Graff-dubois, M. Garcette, L. Renia, M. Kato et al., Distinct Role for CD8 T Cells toward Cutaneous Tumors and Visceral Metastases, The Journal of Immunology, vol.180, issue.1, pp.130-137, 2008.
DOI : 10.4049/jimmunol.180.1.130