C. Holohan, S. Van-schaeybroeck, D. B. Longley, and P. G. Johnston, Cancer drug resistance: An evolving paradigm, Nature Reviews Cancer, p.24060863, 2013.
DOI : 10.1038/nrc3599

A. Persidis, Cancer multidrug resistance. Nature Biotechnology, p.9920278, 1999.

L. A. Garraway and P. A. Jänne, Circumventing cancer drug resistance in the era of personalized medicine. Cancer Discovery, p.22585993, 2012.

G. Housman, S. Byler, S. Heerboth, K. Lapinska, M. Longacre et al., Drug resistance in cancer: An overview. Cancers, p.25198391, 2014.

R. A. Gatenby, A. S. Silva, R. J. Gillies, and B. R. Frieden, Adaptive therapy, Cancer Research, vol.69, p.19487300, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01887358

J. Zhang, J. J. Cunningham, J. S. Brown, and R. A. Gatenby, Integrating evolutionary dynamics into treatment of metastatic castrate-resistant prostate cancer, Nature Communications, p.29180633, 2017.

K. Bacevic, R. Noble, A. Soffar, W. Ammar, O. Boszonyik et al., Spatial competition constrains resistance to targeted cancer therapy, Nature Communications, vol.8, p.29222471, 1995.
URL : https://hal.archives-ouvertes.fr/hal-02120922

C. A. Aktipis, A. M. Boddy, R. A. Gatenby, J. S. Brown, and C. C. Maley, Life history trade-offs in cancer evolution, Nature Reviews Cancer, p.24213474, 2013.

C. Jacqueline, P. A. Biro, C. Beckmann, A. P. Moller, F. Renaud et al., Cancer: A disease at the crossroads of trade-offs, Evolutionary Applications, vol.10, p.28250806, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01418002

P. M. Enriquez-navas and R. A. Gatenby, Chapter 14-Applying Tools From Evolutionary Biology to Cancer Research. Ecology and Evolution of Cancer, pp.193-200, 2017.

H. Ledford, Cancer treatment: The killer within, Nature, vol.508, p.24695297, 2014.

G. Darrasse-jèze, A. S. Bergot, A. Durgeau, F. Billiard, B. L. Salomon et al., Tumor emergence is sensed by self-specific CD44hi memory Tregs that create a dominant tolerogenic environment for tumors in mice, Journal of Clinical Investigation, vol.119, p.19652360, 2009.

A. M. Georgoudaki, K. E. Prokopec, V. F. Boura, E. Hellqvist, S. Sohn et al., Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis, Cell Reports, vol.15, p.27210762, 2000.

V. K. Harris, J. D. Schiffman, and A. M. Boddy, Evolution of Cancer Defense Mechanisms Across Species. Ecology and Evolution of Cancer, pp.99-110, 2017.

C. A. Aktipis and R. M. Nesse, Evolutionary foundations for cancer biology, Evolutionary Applications, p.23396885, 2013.

J. Degregori, Evolved tumor suppression: Why are we so good at not getting cancer? Cancer Research, p.21610109, 2011.

W. Who-|-cancer,

T. Madsen, A. Arnal, M. Vittecoq, F. Bernex, J. Abadie et al., Cancer Prevalence and Etiology in Wild and Captive Animals. Ecology and Evolution of Cancer, pp.11-46, 2017.

M. J. Bissell and W. C. Hines, Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression, Nature Medicine, p.21383745, 2011.

J. Folkman and R. Kalluri, Cancer without disease, Nature, vol.427, p.14985739, 2004.

F. Thomas, C. Jacqueline, T. Tissot, M. Henard, S. Blanchet et al., The importance of cancer cells for animal evolutionary ecology. Nature Ecology and Evolution, p.29066812, 2017.
URL : https://hal.archives-ouvertes.fr/cea-01887606

R. M. Nesse, D. Ganten, T. R. Gregory, and G. S. Omenn, Evolutionary molecular medicine, Journal of Molecular Medicine, p.22544168, 2012.

S. C. Stearns, Evolutionary medicine: its scope, interest and potential, Proceedings of the Royal Society B: Biological Sciences, vol.279, p.22933370, 2012.
DOI : 10.1098/rspb.2012.1326

URL : http://rspb.royalsocietypublishing.org/content/royprsb/279/1746/4305.full.pdf

W. D. Hamilton, The moulding of senescence by natural selection, Journal of Theoretical Biology, vol.12, p.6015424, 1966.

A. I. Rozhok and J. Degregori, The Evolution of Lifespan and Age-Dependent Cancer Risk. Trends in Cancer, p.28439564, 2016.

S. A. Frank, Age-Specific Acceleration of Cancer, Current Biology, vol.14, p.14761658, 2004.
DOI : 10.1016/s0960-9822(03)00937-0

M. E. Hochberg, F. Thomas, E. Assenat, and U. Hibner, Preventive Evolutionary Medicine of Cancers. Evolutionary Applications, p.23396860, 2013.
DOI : 10.1111/eva.12033

URL : http://onlinelibrary.wiley.com/doi/10.1111/eva.12033/pdf

M. Y. Mapara and M. Sykes, Tolerance and cancer: Mechanisms of tumor evasion and strategies for breaking tolerance, Journal of Clinical Oncology, p.15020616, 2004.

J. Campisi, . Ph75ch30-campisi, and . Introduction, AGING AND CANCER. Annu Rev Physiol, vol.75, pp.685-705, 2013.

F. Thomas, R. M. Nesse, R. Gatenby, C. Gidoin, F. Renaud et al., Evolutionary Ecology of Organs: A Missing Link in Cancer Development? Trends in Cancer, p.28741494, 2016.

M. Greaves, A causal mechanism for childhood acute lymphoblastic leukaemia, Nature Reviews Cancer, p.29784935, 2018.
DOI : 10.1038/s41568-018-0015-6

J. S. Brown and C. A. Aktipis, Inclusive fitness effects can select for cancer suppression into old age, Philosophical transactions of the Royal Society of London Series B, vol.370, p.26056358, 2015.
DOI : 10.1098/rstb.2015.0160

URL : http://rstb.royalsocietypublishing.org/content/370/1673/20150160.full.pdf

A. Corthay, Does the immune system naturally protect against cancer? Frontiers in Immunology, p.24860567, 2014.

G. P. Dunn, A. T. Bruce, H. Ikeda, L. J. Old, and R. D. Schreiber, Cancer immunoediting: From immunosurveillance to tumor escape, Nature Immunology, p.12407406, 2002.
DOI : 10.1038/ni1102-991

D. Mittal, M. M. Gubin, R. D. Schreiber, and M. J. Smyth, New insights into cancer immunoediting and its three component phases-elimination, equilibrium and escape. Current Opinion in Immunology, p.24531241, 2014.
DOI : 10.1016/j.coi.2014.01.004

URL : http://europepmc.org/articles/pmc4388310?pdf=render

E. Peranzoni, A. Rivas-caicedo, H. Bougherara, H. Salmon, and E. Donnadieu, Positive and negative influence of the matrix architecture on antitumor immune surveillance, Cellular and molecular life sciences: CMLS, vol.70, p.23649148, 2013.

A. Górski, V. Castronovo, B. Stepie?sopniewska, P. Grieb, M. Ryba et al., Depressed immune surveillance against cancer: Role of deficient t cell: Extracellular matrix interactions, Cell Communication and Adhesion, vol.2, pp.225-233, 1994.

S. K. Biswas, A. Sica, and C. E. Lewis, Plasticity of macrophage function during tumor progression: regulation by distinct molecular mechanisms, Journal of immunology, vol.180, pp.2011-2017, 1950.

N. B. Hao, M. H. Lü, Y. H. Fan, Y. L. Cao, Z. R. Zhang et al., Macrophages in tumor microenvironments and the progression of tumors. Clinical and Developmental Immunology, p.22778768, 2012.

H. Salmon, K. Franciszkiewicz, D. Damotte, M. C. Dieu-nosjean, P. Validire et al., Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors, Journal of Clinical Investigation, vol.122, p.22293174, 2012.

A. Mantovani, P. Allavena, A. Sica, and F. Balkwill, Cancer-related inflammation, Nature, p.18650914, 2008.

B. F. Zamarron and W. Chen, Dual roles of immune cells and their factors in cancer development and progression, International Journal of Biological Sciences, vol.7, p.21647333, 2011.

K. G. Anderson, I. M. Stromnes, and P. D. Greenberg, Obstacles Posed by the Tumor Microenvironment to T cell Activity: A Case for Synergistic Therapies, p.28292435, 2017.

S. B. Willingham, J. Volkmer, A. J. Gentles, D. Sahoo, P. Dalerba et al., The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors, Proceedings of the National Academy of Sciences, vol.109, p.22451913, 2012.

H. F. Dvorak, Similarities between tumor stroma generation and wound healing, vol.315, p.3537791, 1986.

B. Rybinski, J. Franco-barraza, and E. Cukierman, The wound healing, chronic fibrosis, and cancer progression triad, Physiological Genomics, vol.46, p.24520152, 2014.

J. Carreras, A. Lopez-guillermo, B. C. Fox, L. Colomo, A. Martinez et al., High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma, Blood, vol.108, p.16825494, 2006.

A. Tzankov, C. Meier, P. Hirschmann, P. Went, S. A. Pileri et al., Correlation of high numbers of intratumoral FOXP3+ regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin's lymphoma, Haematologica, vol.93, p.18223287, 2008.

G. P. Dunn, L. J. Old, and R. D. Schreiber, The immunobiology of cancer immunosurveillance and immunoediting, p.15308095, 2004.

`. Cal?`bcal?`cal?, B. Molon, and A. Viola, Tuning cancer fate: the unremitting role of host immunity, Open Biology, vol.7, p.28404796, 2017.

C. M. Koebel, W. Vermi, J. B. Swann, N. Zerafa, S. J. Rodig et al., Adaptive immunity maintains occult cancer in an equilibrium state, Nature, vol.450, p.18026089, 2007.
DOI : 10.1038/nature06309

M. H. Manjili, Tumor dormancy and relapse: From a natural byproduct of evolution to a disease state, p.28507050, 2017.

C. M. Blatteis, PMID: 3090790 multi:fever is a natural immune response to infection. . .;fever is a natural immune response to infection, Yale Journal of Biology and Medicine, vol.59, pp.107-116, 1986.

Q. Wang, J. Gao, and X. Wu, Pseudoprogression and hyperprogression after checkpoint blockade, Int. Immunopharmacol, vol.58, p.29579717, 2018.
DOI : 10.1016/j.intimp.2018.03.018

S. Kato, A. Goodman, V. Walavalkar, D. A. Barkauskas, A. Sharabi et al., Hyperprogressors after immunotherapy: Analysis of genomic alterations associated with accelerated growth rate. Clinical Cancer Research, p.28351930, 2017.

N. P. Restifo, M. J. Smyth, and A. Snyder, Acquired resistance to immunotherapy and future challenges, Nature Reviews Cancer, p.26822578, 2016.
DOI : 10.1038/nrc.2016.2

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6330026

. Couzin-frankel, J. Cancer Immunotherapy. Science, vol.342, p.24357284, 2013.

C. Jacqueline, N. Bonnefoy, G. M. Charrière, and F. Thomas, Personal history of infections and immunotherapy: unexpected links and possible therapeutic opportunities, OncoImmunology, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01887524

H. Westdorp, S. Kolders, N. Hoogerbrugge, I. De-vries, M. Jongmans et al., Immunotherapy holds the key to cancer treatment and prevention in constitutional mismatch repair deficiency (CMMRD) syndrome. Cancer Letters, p.28645564, 2017.

Z. Ye, Q. Qian, H. Jin, and Q. Qian, Cancer vaccine: learning lessons from immune checkpoint inhibitors, Journal of Cancer, p.29344272, 2018.
DOI : 10.7150/jca.20059

URL : http://www.jcancer.org/v09p0263.pdf

Y. Zhao, L. Wu, X. Yue, C. Zhang, J. Wang et al., A polymorphism in the tumor suppressor p53 affects aging and longevity in mouse models, eLife, p.29557783, 2018.

R. R. Langley and I. J. Fidler, The seed and soil hypothesis revisited-The role of tumor-stroma interactions in metastasis to different organs, International Journal of Cancer, p.21365651, 2011.

K. C. Valkenburg, D. Groot, A. E. Pienta, and K. J. , Targeting the tumour stroma to improve cancer therapy, Nature Reviews Clinical Oncology, p.29651130, 2018.

X. Liu, S. Jiang, C. Fang, S. Yang, D. Olalere et al., Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice, Cancer Research, p.26330166, 2015.