J. H. Houtgraaf, J. Versmissen, and W. J. Van-der-giessen, A concise review of DNA damage checkpoints and repair in mammalian cells, Cardiovasc Revascularization Med Mol Interv. sept, vol.7, issue.3, pp.165-72, 2006.

T. Kinoshita, Biosynthesis and deficiencies of glycosylphosphatidylinositol, Proc Jpn Acad Ser B Phys Biol Sci, vol.90, issue.4, p.43, 2014.

J. Takeda, T. Miyata, K. Kawagoe, Y. Iida, Y. Endo et al., Deficiency of the GPI anchor caused by a somatic mutation of the PIG-A gene in paroxysmal nocturnal hemoglobinuria, Cell. 21 mai, vol.73, issue.4, pp.703-714, 1993.

B. B. Gollapudi, A. M. Lynch, R. H. Heflich, S. D. Dertinger, V. N. Dobrovolsky et al., The in vivo Pig-a assay: A report of the International Workshop On Genotoxicity Testing (IWGT) Workgroup

, Mutat Res Genet Toxicol Environ Mutagen. 1 mai, vol.783, pp.23-35, 2015.

R. A. Brodsky, Paroxysmal nocturnal hemoglobinuria. Blood, vol.124, pp.2804-2815, 2014.

D. J. Araten, K. Nafa, K. Pakdeesuwan, and L. Luzzatto, Clonal populations of hematopoietic cells with paroxysmal nocturnal hemoglobinuria genotype and phenotype are present in normal individuals, Proc Natl Acad Sci, vol.96, issue.9, pp.5209-5223, 1999.

R. Hu, G. L. Mukhina, S. Piantadosi, J. P. Barber, R. J. Jones et al., PIG-A mutations in normal hematopoiesis, Blood. 15 mai, vol.105, issue.10, pp.3848-54, 2005.

K. Kawagoe, J. Takeda, Y. Endo, and T. Kinoshita, Molecular cloning of murine pig-a, a gene for GPIanchor biosynthesis, and demonstration of interspecies conservation of its structure, function, and genetic locus, Genomics. oct, vol.23, issue.3, pp.566-74, 1994.

B. Peruzzi, D. J. Araten, R. Notaro, and L. Luzzatto, The use of PIG-A as a sentinel gene for the study of the somatic mutation rate and of mutagenic agents in vivo, Mutat Res. sept, vol.705, issue.1, p.10, 2010.

S. Phonethepswath, S. M. Bryce, J. C. Bemis, and S. D. Dertinger, Erythrocyte-based Pig-a gene mutation assay: demonstration of cross-species potential, Mutat Res. 8 déc, vol.657, issue.2, p.6, 2008.

S. Phonethepswath, D. Franklin, D. K. Torous, S. M. Bryce, J. C. Bemis et al., Pig-a mutation: kinetics in rat erythrocytes following exposure to five prototypical mutagens, Toxicol Sci Off J Soc Toxicol. mars, vol.114, issue.1, p.70, 2010.

J. A. Bhalli, M. G. Pearce, V. N. Dobrovolsky, and R. H. Heflich, Manifestation and persistence of Pig-a mutant red blood cells in C57BL/6 mice following single and split doses of N-ethyl-N-nitrosourea, Environ Mol Mutagen. déc, vol.52, issue.9, pp.766-73, 2011.

D. Miura, V. N. Dobrovolsky, T. Kimoto, Y. Kasahara, and R. H. Heflich, Accumulation and persistence of Pig-A mutant peripheral red blood cells following treatment of rats with single and split doses of Nethyl-N-nitrosourea, Mutat Res. juill, vol.677, issue.1, p.92, 2009.

S. D. Dertinger, S. L. Avlasevich, D. K. Torous, J. C. Bemis, S. Phonethepswath et al., Persistence of cisplatin-induced mutagenicity in hematopoietic stem cells: implications for secondary cancer risk following chemotherapy, Toxicol Sci Off J Soc Toxicol. 1 août, vol.140, issue.2, pp.307-321, 2014.

S. Ohtani, A. Unno, A. Ushiyama, T. Kimoto, D. Miura et al., The in vivo Pig-a gene mutation assay is useful for evaluating the genotoxicity of ionizing radiation in mice, Environ Mol Mutagen, vol.53, issue.8, pp.579-88, 2012.

S. Ohtani, A. Ushiyama, A. Ootsuyama, and N. Kunugita, Persistence of red blood cells with Pig-a mutation in p53 knockout mice exposed to X-irradiation, J Toxicol Sci. févr, vol.39, issue.1, pp.7-14, 2014.

A. Graupner, D. M. Eide, C. Instanes, J. M. Andersen, D. A. Brede et al., Gamma radiation at a human relevant low dose rate is genotoxic in mice, Sci Rep. 6 sept, vol.6, p.32977, 2016.

A. Olsen, S. D. Dertinger, C. T. Krüger, D. M. Eide, C. Instanes et al., The Pig-a Gene Mutation Assay in Mice and Human Cells: A Review, Basic Clin Pharmacol Toxicol. sept, vol.121, 2017.

H. N. Haboubi, R. L. Lawrence, B. Rees, L. Williams, J. M. Manson et al., Developing a blood-based gene mutation assay as a novel biomarker for oesophageal adenocarcinoma, Sci Rep. 26 mars, vol.9, issue.1, p.5168, 2019.

V. N. Dobrovolsky, R. K. Elespuru, C. Bigger, T. W. Robison, and R. H. Heflich, Monitoring humans for somatic mutation in the endogenous PIG-a gene using red blood cells, Environ Mol Mutagen. déc, vol.52, issue.9, pp.784-94, 2011.

M. A. Mcdiarmid, R. J. Albertini, J. D. Tucker, P. M. Vacek, E. W. Carter et al., Measures of genotoxicity in Gulf war I veterans exposed to depleted uranium, Environ Mol Mutagen. août, vol.52, issue.7, p.81, 2011.

T. Rondelli, M. Berardi, B. Peruzzi, L. Boni, R. Caporale et al., The frequency of granulocytes with spontaneous somatic mutations: a wide distribution in a normal human population, PloS One, vol.8, issue.1, p.54046, 2013.

M. Fenech, Cytokinesis-block micronucleus cytome assay, Nat Protoc, vol.2, issue.5, pp.1084-104, 2007.

D. R. Sutherland, M. Keeney, and A. Illingworth, Practical guidelines for the high-sensitivity detection and monitoring of paroxysmal nocturnal hemoglobinuria clones by flow cytometry, Cytometry B Clin Cytom. 1 juill, vol.82, issue.4, pp.195-208, 2012.

J. W. Athens, O. P. Haab, S. O. Raab, A. M. Mauer, H. Ashenbrucker et al., Leukokinetic studies. IV. The total blood, circulating and marginal granulocyte pools and the granulocyte turnover rate in normal subjects, J Clin Invest. juin, vol.40, pp.989-95, 1961.

S. D. Dertinger, S. L. Avlasevich, J. C. Bemis, Y. Chen, and J. T. Macgregor, Human erythrocyte PIG-A assay: an easily monitored index of gene mutation requiring low volume blood samples, Environ Mol Mutagen. mai, vol.56, issue.4, pp.366-77, 2015.

K. Horibata, A. Ukai, S. Ishikawa, A. Sugano, and M. Honma, Monitoring genotoxicity in patients receiving chemotherapy for cancer: application of the PIG-A assay, Mutat Res Genet Toxicol Environ Mutagen. 15 sept, vol.808, pp.20-26, 2016.

T. Lee, R. R. Allison, K. F. O'brien, J. L. Naves, U. L. Karlsson et al., Persistence of Micronuclei in Lymphocytes of Cancer Patients after Radiotherapy, Radiat Res, vol.157, issue.6, pp.678-84, 2002.

E. Minaei, S. Mozdarani, H. Motazakker, M. Mansouri, M. Aghamiri et al., Evaluation of Cytogenetic Alterations in Peripheral Blood Lymphocytes of Esophageal Cancer Patients Treated with Radiotherapy or Chemoradiotherapy using Cytokinesis-Blocked Micronucleus Assay, Acta Med Iran. janv, vol.54, issue.1, pp.9-14, 2016.

H. Thierens, A. Vral, M. Van-eijkeren, F. Speleman, D. Ridder et al., Micronucleus induction in peripheral blood lymphocytes of patients under radiotherapy treatment for cervical cancer or Hodgkin's disease, Int J Radiat Biol. mai, vol.67, issue.5, pp.529-568, 1995.

P. A. Ko?ar, M. Koçer, D. A. Çelik, and N. Özçelik, Cytogenetic findings in patients diagnosed with breast cancer having undergone adjuvant chemotherapy regimens, Genet Mol Res GMR. 18 mars, vol.15, issue.1, 2016.

M. Fenech and S. Bonassi, The effect of age, gender, diet and lifestyle on DNA damage measured using micronucleus frequency in human peripheral blood lymphocytes, Mutagenesis. janv, vol.26, issue.1, pp.43-52, 2011.

B. V. Offersen, L. J. Boersma, C. Kirkove, S. Hol, M. C. Aznar et al., ESTRO consensus guideline on target volume delineation for elective radiation therapy of early stage breast cancer, Radiother Oncol J Eur Soc Ther Radiol Oncol. janv, vol.114, issue.1, p.10, 2015.

K. Verhoeven, C. Weltens, V. Remouchamps, K. Mahjoubi, L. Veldeman et al., Vessel based delineation guidelines for the elective lymph node regions in breast cancer radiation therapy -PROCAB guidelines, Radiother Oncol J Eur Soc Ther Radiol Oncol. janv, vol.114, issue.1, pp.11-17, 2015.

M. Cristy, Active bone marrow distribution as a function of age in humans, Phys Med Biol. mai, vol.26, issue.3, p.400, 1981.

P. Mauch, L. Constine, J. Greenberger, W. Knospe, J. Sullivan et al., Hematopoietic stem cell compartment: acute and late effects of radiation therapy and chemotherapy, Int J Radiat Oncol Biol Phys. 30 mars, vol.31, issue.5, pp.1319-1358, 1995.

J. D. Wright, C. M. St-clair, I. Deutsch, W. M. Burke, P. Gorrochurn et al., Pelvic radiotherapy and the risk of secondary leukemia and multiple myeloma, Cancer. 15 mai, vol.116, issue.10, pp.2486-92, 2010.

J. Cosset, C. Chargari, C. Demoor, G. P. Helfre, S. Mornex et al.,

, Cancer Radiother J Soc Francaise Radiother Oncol. sept, vol.20, pp.61-68, 2016.

L. B. Travis, The epidemiology of second primary cancers, Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol, vol.15, issue.11, pp.2020-2026, 2006.