D. De-ruysscher, G. Niedermann, N. G. Burnet, S. Siva, A. Lee et al.,

, Radiotherapy toxicity, Nat Rev Dis Primers, vol.5, p.13, 2019.

P. Rubin and G. W. Casarett, Clinical radiation pathology as applied to curative radiotherapy, Cancer, vol.22, pp.767-78, 1968.

H. B. Stone, C. N. Coleman, M. S. Anscher, and W. H. Mcbride, Effects of radiation on normal tissue: consequences and mechanisms, Lancet Oncol, vol.4, pp.529-565, 2003.

C. Chargari, K. M. Kirov, M. A. Bollet, N. Magné, L. Védrine et al., Cardiac toxicity in breast cancer patients: from a fractional point of view to a global assessment, Cancer Treat Rev, vol.37, pp.321-351, 2011.

A. Vallard, C. Rancoule, L. Floch, H. Guy, J. Espenel et al.,

, prevention and treatment of radiation

, Cancer Radiother, vol.21, pp.411-434, 2017.

D. Schaue, E. L. Kachikwu, and W. H. Mcbride, Cytokines in radiobiological responses: a review, Radiat Res, vol.178, pp.505-528, 2012.

D. E. Hallahan, L. Geng, and Y. Shyr, Effects of intercellular adhesion molecule 1 (ICAM-1) null mutation on radiation-induced pulmonary fibrosis and respiratory insufficiency in mice, J Natl Cancer Inst, vol.94, pp.733-774, 2002.

T. A. Wynn and T. R. Ramalingam, Mechanisms of fibrosis: therapeutic translation for fibrotic disease, Nat Med, vol.18, pp.1028-1068, 2012.

C. Strup-perrot, M. Vozenin-brotons, M. Vandamme, C. Linard, and D. Mathé, Expression of matrix metalloproteinases and tissue inhibitor metalloproteinases increases in X-irradiated rat ileum despite the disappearance of CD8a T cells, World J Gastroenterol, vol.11, pp.6312-6333, 2005.

J. P. Chute, To survive radiation injury, remember your aPCs, Nat Med, vol.18, pp.1013-1017, 2012.

D. Greene-schloesser, M. E. Robbins, A. M. Peiffer, E. G. Shaw, K. T. Wheeler et al., Radiation-induced brain injury: A review, Front Oncol, vol.2, p.73, 2012.

K. M. Joo, J. J. Kang, B. G. Lee, S. J. Kim, K. H. Yang et al., Trans-differentiation of neural stem cells: a therapeutic mechanism against the radiation induced brain damage, PLoS ONE, vol.7, 2012.

S. Balentova and A. M. Molecular, Cellular and Functional Effects of Radiation-Induced Brain Injury: A Review, Int J Mol Sci, vol.16, pp.27796-815, 2015.

L. Meziani, M. Mondini, B. Petit, A. Boissonnas, T. De-montpreville et al., CSF1R inhibition prevents radiation pulmonary fibrosis by depletion of interstitial macrophages, Eur Respir J, vol.51, 2018.

M. R. Ferreira, A. Muls, D. P. Dearnaley, and H. Andreyev, Microbiota and radiationinduced bowel toxicity: lessons from inflammatory bowel disease for the radiation oncologist

, Lancet Oncol, vol.15, issue.13, pp.70504-70511, 2014.

C. Vale, A. Nightingale, N. Spera, A. Whelan, B. Hanley et al., Late complications from chemoradiotherapy for cervical cancer: reflections from cervical cancer survivors 10 years after the national cancer institute alert, Clin Oncol (R Coll Radiol), vol.22, pp.588-597, 2010.

M. J. Resnick, T. Koyama, K. Fan, P. C. Albertsen, M. Goodman et al., Long-term functional outcomes after treatment for localized prostate cancer, N Engl J Med, vol.368, pp.436-481, 2013.

P. Hopwood, J. S. Haviland, G. Sumo, J. Mills, J. M. Bliss et al., Comparison of patient-reported breast, arm, and shoulder symptoms and body image after radiotherapy for early breast cancer: 5-year follow-up in the randomised Standardisation of Breast Radiotherapy (START) trials, Lancet Oncol, vol.11, issue.09, pp.70382-70383, 2010.

H. Holthusen, Erfahrungen über die Verträglichkeitsgrenze für Röntgenstrahlen and deren Nutzanwendung zur Verhütung von Schäden, Strahlentherapie, vol.1936, pp.254-69

N. G. Burnet, J. Johansen, I. Turesson, J. Nyman, and J. H. Peacock, Describing patients' normal tissue reactions: concerning the possibility of individualising radiotherapy dose prescriptions based on potential predictive assays of normal tissue radiosensitivity. Steering Committee of the BioMed2 European Union Concerted Action Programme on the Development of Predictive Tests of Normal Tissue Response to Radiation Therapy, Int J Cancer, vol.79, pp.606-619, 1998.

H. Edvardsen, H. Landmark-høyvik, K. V. Reinertsen, X. Zhao, G. I. Grenaker-alnaes et al., SNP in TXNRD2 associated with radiation-induced fibrosis: a study of genetic variation in reactive oxygen species metabolism and signaling, Int J Radiat Oncol Biol Phys, vol.86, pp.791-800, 2013.

S. L. Tucker, F. B. Geara, L. J. Peters, and W. A. Brock, How much could the radiotherapy dose be altered for individual patients based on a predictive assay of normal-tissue radiosensitivity?, Radiother Oncol, vol.38, pp.103-116, 1996.

N. G. Burnet, J. Nyman, I. Turesson, R. Wurm, J. R. Yarnold et al., Prediction of normal-tissue tolerance to radiotherapy from in-vitro cellular radiation sensitivity, Lancet, vol.339, pp.1570-1571, 1992.

J. Johansen, S. M. Bentzen, J. Overgaard, and M. Overgaard, Relationship between the in vitro radiosensitivity of skin fibroblasts and the expression of subcutaneous fibrosis, telangiectasia, and skin erythema after radiotherapy, Radiother Oncol, vol.40, pp.101-110, 1996.

J. Peacock, A. Ashton, J. Bliss, C. Bush, J. Eady et al., Cellular radiosensitivity and complication risk after curative radiotherapy, Radiother Oncol, vol.55, pp.173-181, 2000.

F. B. Geara, L. J. Peters, K. K. Ang, J. L. Wike, and W. A. Brock, Prospective comparison of in vitro normal cell radiosensitivity and normal tissue reactions in radiotherapy patients, Int J Radiat Oncol Biol Phys, vol.27, pp.1173-1182, 1993.

N. Somaiah, M. Chua, S. Bourne, F. Daley, A. '-hern et al., Correlation between DNA damage responses of skin to a test dose of radiation and late adverse effects of earlier breast radiotherapy, Radiotherapy and Oncology, vol.119, pp.244-253, 2016.

,

C. West, S. E. Davidson, S. Elyan, H. Valentine, S. A. Roberts et al.,

, Lymphocyte radiosensitivity is a significant prognostic factor for morbidity in carcinoma of the cervix, International Journal of Radiation Oncology*Biology*Physics, vol.51, pp.1575-1584, 2001.

M. Ozsahin, H. Ozsahin, Y. Shi, B. Larsson, F. E. Würgler et al., Rapid assay of intrinsic radiosensitivity based on apoptosis in human CD4 and CD8 T-lymphocytes, Int J Radiat Oncol Biol Phys, vol.38, pp.429-469, 1997.

D. Azria, B. S. Rosenstein, and M. Ozsahin, Radiation-Induced Side Effects With or Without Systemic Therapies: Prime Time for Prediction of Individual Radiosensitivity, International Journal of Radiation Oncology*Biology*Physics, vol.71, pp.1293-1297, 2008.

P. L. Olive, J. P. Banáth, and M. Keyes, Residual gammaH2AX after irradiation of human lymphocytes and monocytes in vitro and its relation to late effects after prostate brachytherapy, Radiother Oncol, vol.86, pp.336-382, 2008.

J. B. Barber, W. Burrill, A. R. Spreadborough, E. Levine, C. Warren et al., Relationship between in vitro chromosomal radiosensitivity of peripheral blood lymphocytes and the expression of normal tissue damage following radiotherapy for breast cancer

, Radiother Oncol, vol.55, pp.158-167, 2000.

U. Hoeller, K. Borgmann, M. Bonacker, A. Kuhlmey, A. Bajrovic et al., Individual radiosensitivity measured with lymphocytes may be used to predict the risk of fibrosis after radiotherapy for breast cancer, Radiotherapy and Oncology, vol.69, pp.137-181, 2003.

G. I. Terzoudi, V. I. Hatzi, K. Barszczewska, K. N. Manola, C. Stavropoulou et al., G2-checkpoint abrogation in irradiated lymphocytes: A new cytogenetic approach to assess individual radiosensitivity and predisposition to cancer, Int J Oncol, vol.35, pp.1223-1253, 2009.

. Copernic-project-investigators, A. Granzotto, M. A. Benadjaoud, G. Vogin, C. Devic et al., Influence of Nucleoshuttling of the ATM Protein in the Healthy Tissues Response to Radiation Therapy: Toward a Molecular Classification of Human Radiosensitivity, Int J Radiat Oncol Biol Phys, vol.94, pp.450-60, 2016.

C. M. West and G. C. Barnett, Genetics and genomics of radiotherapy toxicity: towards prediction, Genome Med, vol.3, 2011.

B. S. Rosenstein, Identification of SNPs associated with susceptibility for development of adverse reactions to radiotherapy, Pharmacogenomics, vol.12, pp.267-75, 2011.

G. C. Barnett, C. E. Coles, R. M. Elliott, C. Baynes, C. Luccarini et al., Independent validation of genes and polymorphisms reported to be associated with radiation toxicity: a prospective analysis study, The Lancet Oncology, vol.13, pp.65-77, 2012.

S. L. Kerns, H. Ostrer, R. Stock, W. Li, J. Moore et al., Genome-wide association study to identify single nucleotide polymorphisms (SNPs) associated with the development of erectile dysfunction in African-American men after radiotherapy for prostate cancer, Int J Radiat Oncol Biol Phys, vol.78, pp.1292-300, 2010.

,

S. L. Kerns, N. N. Stone, R. G. Stock, L. Rath, H. Ostrer et al., A 2-stage genomewide association study to identify single nucleotide polymorphisms associated with development of urinary symptoms after radiotherapy for prostate cancer, J Urol, vol.190, pp.102-110, 2013.

S. L. Kerns, L. Fachal, L. Dorling, G. C. Barnett, A. Baran et al.,

, Radiogenomics Consortium Genome-Wide Association Study Meta-Analysis of Late Toxicity After Prostate Cancer Radiotherapy, J Natl Cancer Inst, vol.112, pp.179-90, 2020.

T. Wang, G. Shen, M. Chen, J. Zhang, Y. Sun et al., Genome-Wide Association Study of Susceptibility Loci for Radiation-Induced Brain Injury, JNCI: Journal of the National Cancer Institute, vol.111, pp.620-628, 2019.

B. S. Rosenstein, Radiogenomics: Identification of Genomic Predictors for Radiation Toxicity, Semin Radiat Oncol, vol.27, pp.300-309, 2017.

C. West, D. Azria, J. Chang-claude, S. Davidson, P. Lambin et al., The REQUITE project: validating predictive models and biomarkers of radiotherapy toxicity to reduce side-effects and improve quality of life in cancer survivors, Clin Oncol (R Coll Radiol), vol.26, pp.739-781, 2014.

G. C. Barnett, C. West, A. M. Dunning, R. M. Elliott, C. E. Coles et al., Normal tissue reactions to radiotherapy: towards tailoring treatment dose by genotype, Nature Reviews Cancer, vol.9, pp.134-176, 2009.

S. Delanian and J. Lefaix, The radiation-induced fibroatrophic process: therapeutic perspective via the antioxidant pathway, Radiother Oncol, vol.73, pp.119-150, 2004.

J. Yarnold and M. Brotons, Pathogenetic mechanisms in radiation fibrosis, Radiother Oncol, vol.97, pp.149-61, 2010.

S. Delanian, R. Porcher, S. Balla-mekias, and J. Lefaix, Randomized, placebo-controlled trial of combined pentoxifylline and tocopherol for regression of superficial radiation-induced fibrosis, J Clin Oncol, vol.21, pp.2545-50, 2003.

C. Bourgier, A. Auperin, S. Rivera, P. Boisselier, B. Petit et al., Pravastatin Reverses Established Radiation-Induced Cutaneous and Subcutaneous Fibrosis in Patients With Head and Neck Cancer: Results of the Biology-Driven Phase 2 Clinical Trial Pravacur
URL : https://hal.archives-ouvertes.fr/hal-02452478

, Int J Radiat Oncol Biol Phys, vol.104, pp.365-73, 2019.

A. N. Hanania, W. Mainwaring, Y. T. Ghebre, N. A. Hanania, and M. Ludwig, Radiation-Induced Lung Injury: Assessment and Management, Chest, vol.156, pp.150-62, 2019.

C. Chung, A. Bryant, and P. D. Brown, Interventions for the treatment of brain radionecrosis after radiotherapy or radiosurgery, Cochrane Database Syst Rev, vol.7, 2018.

I. M. Paquette, J. D. Vogel, M. A. Abbas, D. L. Feingold, and S. R. Steele, Surgeons O behalf of the CPGC of TAS of C and R. The American Society of Colon and Rectal Surgeons Clinical Practice Guidelines for the Treatment of Chronic Radiation Proctitis, Diseases of the Colon & Rectum, vol.61, pp.1135-1140, 2018.

C. Pascoe, C. Duncan, B. W. Lamb, N. F. Davis, T. H. Lynch et al., Current management of radiation cystitis: a review and practical guide to clinical management, BJU Int, vol.123, pp.585-94, 2019.

A. Rehailia-blanchard, M. Y. He, C. Rancoule, A. Vallard, S. Espenel et al.,

, Cancer Radiother, vol.23, pp.240-247, 2019.

H. Andreyev, A. Wotherspoon, J. W. Denham, and M. Hauer-jensen, Pelvic radiation disease": new understanding and new solutions for a new disease in the era of cancer survivorship, Scand J Gastroenterol, vol.46, pp.389-97, 2011.

C. Linard, E. Busson, V. Holler, C. Strup-perrot, J. Lacave-lapalun et al., Repeated autologous bone marrow-derived mesenchymal stem cell injections improve radiation-induced proctitis in pigs, Stem Cells Transl Med, vol.2, pp.916-943, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02647101

S. François, B. Usunier, M. Forgue-lafitte, L. 'homme, B. Benderitter et al., Mesenchymal Stem Cell Administration Attenuates Colon Cancer Progression by Modulating the Immune Component within the Colorectal Tumor Microenvironment, Stem Cells Transl Med, vol.8, pp.285-300, 2019.

J. J. Lataillade, C. Doucet, E. Bey, H. Carsin, C. Huet et al., New approach to radiation burn treatment by dosimetry-guided surgery combined with autologous mesenchymal stem cell therapy, Regen Med, vol.2, pp.785-94, 2007.

G. Rigotti, A. Marchi, M. Galiè, G. Baroni, D. Benati et al., Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells, Plast Reconstr Surg, vol.119, pp.1409-1431, 2007.

L. Maione, V. Vinci, F. Caviggioli, F. Klinger, B. Banzatti et al., Autologous fat graft in postmastectomy pain syndrome following breast conservative surgery and radiotherapy, Aesthetic Plast Surg, vol.38, pp.528-560, 2014.

D. Peiffert, J. Simon, and F. Eschwege,

, Cancer Radiother, vol.11, pp.309-321, 2007.

J. Voswinkel, S. Francois, J. Simon, M. Benderitter, N. Gorin et al., Use of mesenchymal stem cells (MSC) in chronic inflammatory fistulizing and fibrotic diseases: a comprehensive review, Clin Rev Allergy Immunol, vol.45, pp.180-92, 2013.

M. R. Irhimeh and J. Cooney, Management of Inflammatory Bowel Disease Using Stem Cell Therapy, Curr Stem Cell Res Ther, vol.11, pp.72-79, 2016.

O. Ringdén, M. Uzunel, B. Sundberg, L. Lönnies, S. Nava et al., Tissue repair using allogeneic mesenchymal stem cells for hemorrhagic cystitis, pneumomediastinum and perforated colon, Leukemia, vol.21, pp.2271-2277, 2007.

M. M. Lalu, L. Mcintyre, C. Pugliese, D. Fergusson, B. W. Winston et al., Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials, PLoS ONE, vol.7, p.47559, 2012.