A. C. Begg, F. A. Stewart, and C. Vens, Strategies to improve radiotherapy with targeted drugs, Nat Rev Cancer, vol.11, pp.239-53, 2011.

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, Nat Rev Cancer, vol.9, pp.134-176, 2009.

S. M. Bentzen, From cellular to high-throughput predictive assays in radiation oncology: challenges and opportunities, Semin Radiat Oncol, vol.18, pp.75-88, 2008.

C. Bourgier, J. Lacombe, J. Solassol, A. Mange, A. Pèlegrin et al., Late side-effects after curative intent radiotherapy: identification of hypersensitive patients for personalized strategy, Crit Rev Oncol Hematol, vol.93, pp.312-321, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02148578

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.

M. Ozsahin, N. Crompton, S. Gourgou, A. Kramar, L. Li et al., CD4 and CD8 T-lymphocyte apoptosis can predict radiation-induced late toxicity: a prospective study in 399 patients, Clin Cancer Res, vol.11, pp.7426-7459, 2005.

D. Azria, O. Riou, F. Castan, T. D. Nguyen, K. Peignaux et al., Radiation-induced CD8 T-lymphocyte apoptosis as a predictor of breast fibrosis after radiotherapy: results of the prospective multicenter French trial, EBioMedicine, vol.2, pp.1965-73, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02338826

D. Azria, Y. Belkacemi, G. Romieu, S. Gourgou, M. Gutowski et al., Concurrent or sequential adjuvant letrozole and radiotherapy after conservative surgery for early-stage breast cancer (CO-HO-RT): a phase 2 randomised trial, Lancet Oncol, vol.11, pp.258-65, 2010.

K. Schnarr, D. Boreham, J. Sathya, J. Julian, and I. S. Dayes, Radiation-induced lymphocyte apoptosis to predict radiation therapy late toxicity in prostate cancer patients, Int J Radiat Oncol Biol Phys, vol.74, pp.1424-1454, 2009.

P. Foro, M. Algara, J. Lozano, N. Rodriguez, X. Sanz et al., Relationship between radiation-induced apoptosis of T lymphocytes and chronic toxicity in patients with prostate cancer treated by radiation therapy: a prospective study, Int J Radiat Oncol Biol Phys, vol.88, pp.1057-63, 2014.

F. Marchetti, M. A. Coleman, I. M. Jones, and A. J. Wyrobek, Candidate protein biodosimeters of human exposure to ionizing radiation, Int J Radiat Biol, vol.82, pp.605-644, 2006.

X. Feng, Y. H. Li, M. Li, X. Yi, B. Zhang et al., Identification of biomarkers for predicting nasopharyngeal carcinoma response to radiotherapy by proteomics, Cancer Res, vol.70, pp.3450-62, 2010.

B. Zhang, J. Qu, X. L. Yi, H. Zhang, P. Li et al., Identification of heat shock protein 27 as a radioresistance-related protein in nasopharyngeal carcinoma cells, J Cancer Res Clin Oncol, vol.138, pp.2117-2142, 2012.

P. Wu, H. Zhang, L. Qi, Q. Tang, Y. Tang et al., Identification of ERp29 as a biomarker for predicting nasopharyngeal carcinoma response to radiotherapy, Oncol Rep, vol.27, pp.987-94, 2012.

M. Kim, J. Ahn, J. Hwang, S. Woo, H. et al., Quantitative proteomic analysis of single or fractionated radiation-induced proteins in human breast cancer MDA-MB-231 cells, Cell Biosci, vol.5, issue.2, 2015.

L. Guo, Y. Xiao, M. Fan, J. J. Li, and Y. Wang, Profiling global kinome signatures of the radioresistant MCF-7/C6 breast cancer cells using MRM-based targeted proteomics, J Proteome Res, vol.14, pp.193-201, 2015.

R. Wei, Y. Zhang, L. Shen, W. Jiang, C. Li et al., Comparative proteomic and radiobiological analyses in human lung adenocarcinoma cells, Mol Cell Biochem, vol.359, pp.151-160, 2012.

H. S. Yun, J. Baek, J. Yim, H. Um, J. K. Park et al., Radiotherapy diagnostic biomarkers in radioresistant human H460 lung cancer stem-like cells, Cancer Biol Ther, vol.17, pp.208-226, 2016.

J. Hao, P. Graham, L. Chang, J. Ni, V. Wasinger et al., Proteomic identification of the lactate dehydrogenase a in a radioresistant prostate cancer xenograft mouse model for improving radiotherapy, Oncotarget, vol.7, pp.74269-85, 2016.

O. Guipaud, Serum and plasma proteomics and its possible use as detector and predictor of radiation diseases, Adv Exp Med Biol, vol.990, pp.61-86, 2013.

E. Pernot, J. Hall, S. Baatout, M. A. Benotmane, E. Blanchardon et al., Ionizing radiation biomarkers for potential use in epidemiological studies, Mutat Res, vol.751, pp.258-86, 2012.

O. Azimzadeh and S. Tapio, Proteomics approaches to investigate cancer radiotherapy outcome: slow train coming, Transl Cancer Res, vol.6, pp.779-88, 2017.

L. Zhao, L. Wang, J. W. Wang, X. Zhu, X. Hayman et al., Elevation of plasma TGF-beta1 during radiation therapy predicts radiation-induced lung toxicity in patients with non-small-cell lung cancer: a combined analysis from Beijing and Michigan, Int J Radiat Oncol Biol Phys, vol.74, pp.1385-90, 2009.

L. Zhao, K. Sheldon, M. Chen, M. S. Yin, J. A. Hayman et al., The predictive role of plasma TGF-beta1 during radiation therapy for radiationinduced lung toxicity deserves further study in patients with non-small cell lung cancer, Lung Cancer, vol.59, pp.232-241, 2008.

A. Novakova-jiresova, M. M. Van-gameren, R. P. Coppes, H. H. Kampinga, and H. Groen, Transforming growth factor-beta plasma dynamics and post-irradiation lung injury in lung cancer patients, Radiother Oncol, vol.71, pp.183-192, 2004.

S. Siva, M. Macmanus, T. Kron, N. Best, J. Smith et al., A pattern of early radiation-induced inflammatory cytokine expression is Associated with Lung Toxicity in Patients with Non-Small Cell Lung Cancer, PLoS ONE, vol.9, 2014.

D. Jaeger, K. Seppenwoolde, Y. Kampinga, H. H. Boersma, L. J. Belderbos et al., Significance of plasma transforming growth factor-beta levels in radiotherapy for non-small-cell lung cancer, Int J Radiat Oncol Biol Phys, vol.58, pp.1378-87, 2004.

C. E. Rübe, J. Palm, M. Erren, J. Fleckenstein, J. König et al., Cytokine plasma levels: reliable predictors for radiation pneumonitis?, PLoS One, vol.3, p.2898, 2008.

P. A. Riley, Free radicals in biology: oxidative stress and the effects of ionizing radiation, Int J Radiat Biol, vol.65, pp.27-33, 1994.

A. Panday, M. K. Sahoo, D. Osorio, and S. Batra, NADPH oxidases: an overview from structure to innate immunity-associated pathologies, Cell Mol Immunol, vol.12, pp.5-23, 2015.

J. Lacombe, D. Azria, A. Mange, and J. Solassol, Proteomic approaches to identify biomarkers predictive of radiotherapy outcomes, Expert Rev Proteomics, vol.10, pp.33-42, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02372057

Z. Barjaktarovic, D. Schmaltz, A. Shyla, O. Azimzadeh, S. Schulz et al., Radiation-induced signaling results in mitochondrial impairment in mouse heart at 4 weeks after exposure to X-rays, PLoS One, vol.6, p.27811, 2011.

Z. Barjaktarovic, A. Shyla, O. Azimzadeh, S. Schulz, J. Haagen et al., Ionising radiation induces persistent alterations in the cardiac mitochondrial function of C57BL/6 mice 40 weeks after local heart exposure, Radiother Oncol, vol.106, pp.404-414, 2013.

S. J. Kempf, A. Casciati, S. Buratovic, D. Janik, . Toerne-c-von et al., The cognitive defects of neonatally irradiated mice are accompanied by changed synaptic plasticity, adult neurogenesis and neuroinflammation, Mol Neurodegener, vol.9, 2014.

S. I. Chung, J. Seong, Y. N. Park, W. W. Kim, H. J. Oh et al., Identification of proteins indicating radiation-induced hepatic toxicity in cirrhotic rats, J Radiat Res, vol.51, pp.643-50, 2010.

O. Guipaud, V. Holler, V. Buard, G. Tarlet, N. Royer et al., Time-course analysis of mouse serum proteome changes following exposure of the skin to ionizing radiation, Proteomics, vol.7, pp.3992-4002, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00196080

T. Chaze, L. Hornez, C. Chambon, I. Haddad, J. Vinh et al., Serum proteome analysis for profiling predictive protein markers associated with the severity of skin lesions induced by ionizing radiation, Proteomes, vol.1, pp.40-69, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02358352

X. Cai, K. Shedden, X. Ao, M. Davis, X. Fu et al., Plasma proteomic analysis may identify new markers for radiation-induced lung toxicity in patients with non-small-cell lung cancer, Int J Radiat Oncol Biol Phys, vol.77, pp.867-76, 2010.

X. Cai, K. A. Shedden, S. Yuan, M. A. Davis, L. Xu et al., Baseline plasma proteomic analysis to identify biomarkers that predict radiationinduced lung toxicity in patients receiving radiation for non-small cell lung cancer, J Thorac Oncol, vol.6, pp.1073-1081, 2011.

J. H. Oh, J. M. Craft, R. Townsend, J. O. Deasy, and J. D. Bradley, El Naqa I. A bioinformatics approach for biomarker identification in radiationinduced lung inflammation from limited proteomics data, J Proteome Res, vol.10, pp.1406-1421, 2011.

A. Burkart, X. Shi, M. Chouinard, and S. Corvera, Adenylate kinase 2 links mitochondrial energy metabolism to the induction of the unfolded protein response, J Biol Chem, vol.286, pp.4081-4090, 2011.

A. Tanimura, T. Horiguchi, K. Miyoshi, H. Hagita, and T. Noma, Differential expression of adenine nucleotide converting enzymes in mitochondrial intermembrane space: a potential role of adenylate kinase isozyme 2 in neutrophil differentiation, PLoS One, vol.9, p.89916, 2014.

A. Rissone, K. G. Weinacht, G. La-marca, K. Bishop, E. Giocaliere et al., Reticular dysgenesis-associated AK2 protects hematopoietic stem and progenitor cell development from oxidative stress, J Exp Med, vol.212, pp.1185-202, 2015.

E. I. Azzam, J. Jay-gerin, and D. Pain, Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury, Cancer Lett, vol.327, pp.48-60, 2012.

J. F. Turrens, Mitochondrial formation of reactive oxygen species, J Physiol, vol.552, pp.335-379, 2003.

K. Bedard and K. Krause, The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology, Physiol Rev, vol.87, pp.245-313, 2007.

F. Jiang, Y. Zhang, and G. J. Dusting, NADPH oxidase-mediated redox signaling: roles in cellular stress response, stress tolerance, and tissue repair, Pharmacol Rev, vol.63, pp.218-260, 2011.

L. Hecker, R. Vittal, T. Jones, R. Jagirdar, T. R. Luckhardt et al., NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury, Nat Med, vol.15, pp.1077-81, 2009.

J. L. Barnes and Y. Gorin, Myofibroblast differentiation during fibrosis: role of NAD(P)H oxidases, Kidney Int, vol.79, pp.944-56, 2011.

U. Weyemi and C. Dupuy, The emerging role of ROS-generating NADPH oxidase NOX4 in DNA-damage responses, Mutat Res, vol.751, pp.77-81, 2012.

F. Chen, Y. Yu, J. Qian, Y. Wang, B. Cheng et al., Opposing actions of heat shock protein 90 and 70 regulate nicotinamide adenine dinucleotide phosphate oxidase stability and reactive oxygen species production, Arterioscler Thromb Vasc Biol, vol.32, pp.2989-99, 2012.

G. Tell, F. Quadrifoglio, C. Tiribelli, and M. R. Kelley, The many functions of APE1/Ref-1: not only a DNA repair enzyme, Antioxid Redox Signal, vol.11, pp.601-621, 2009.

H. Lee, J. Pyo, Y. Oh, H. Kim, S. Hong et al., AK2 activates a novel apoptotic pathway through formation of a complex with FADD and caspase-10, Nat Cell Biol, vol.9, pp.1303-1313, 2007.

H. Kim, H. Lee, Y. Oh, S. Choi, S. Hong et al., The DUSP26 phosphatase activator adenylate kinase 2 regulates FADD phosphorylation and cell growth, Nat Commun, vol.5, p.3351, 2014.

S. Basuroy, S. Bhattacharya, C. W. Leffler, and H. Parfenova, Nox4 NADPH oxidase mediates oxidative stress and apoptosis caused by TNF-? in cerebral vascular endothelial cells, Am J Physiol Cell Physiol, vol.296, pp.422-454, 2009.

M. Yao, F. Gao, X. Wang, Y. Shi, S. Liu et al., Nox4 is involved in high glucose-induced apoptosis in renal tubular epithelial cells via notch pathway, Mol Med Rep, vol.15, pp.4319-4344, 2017.

S. U. Seo, T. H. Kim, D. E. Kim, K. Min, and T. K. Kwon, NOX4-mediated ROS production induces apoptotic cell death via down-regulation of c-FLIP and Mcl-1 expression in combined treatment with thioridazine and curcumin, Redox Biol, vol.13, pp.608-630, 2017.

S. Kim, K. Kim, Y. Seo, Y. Chun, S. Yu et al., Silibinin induces mitochondrial NOX4-mediated endoplasmic reticulum stress response and its subsequent apoptosis, BMC Cancer, vol.16, p.452, 2016.

E. Six, C. Lagresle-peyrou, S. Susini, C. De-chappedelaine, N. Sigrist et al., AK2 deficiency compromises the mitochondrial energy metabolism required for differentiation of human neutrophil and lymphoid lineages, Cell Death Dis, vol.6, p.1856, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02056595

S. Li, S. S. Tabar, V. Malec, B. G. Eul, W. Klepetko et al., NOX4 regulates ROS levels under normoxic and hypoxic conditions, triggers proliferation, and inhibits apoptosis in pulmonary artery adventitial fibroblasts, Antioxid Redox Signal, vol.10, pp.1687-98, 2008.

G. Chang, L. Chen, H. Lin, Y. Lin, and J. K. Maranchie, Nox4 inhibition enhances the cytotoxicity of cisplatin in human renal cancer cells, J Exp Ther Oncol, vol.10, pp.9-18, 2012.

, Publisher's Note

, Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations