M. R. Zalutsky, D. A. Reardon, G. Akabani, R. E. Coleman, A. H. Friedman et al., Clinical experience with alpha-particle emitting 211At: treatment of recurrent brain tumor patients with 211At-labeled chimeric antitenascin monoclonal antibody 81C6, J Nucl Med, vol.49, pp.30-38, 2008.

H. Andersson, E. Cederkrantz, T. Back, C. Divgi, J. Elgqvist et al., Intraperitoneal alpha-particle radioimmunotherapy of ovarian cancer patients: pharmacokinetics and dosimetry of (211)At-MX35 F(ab')2-a phase I study, J Nucl Med, vol.50, pp.1153-60, 2009.

C. Parker, S. Nilsson, D. Heinrich, S. I. Helle, J. M. O'sullivan et al., Alpha emitter radium-223 and survival in metastatic prostate cancer, N Engl J Med, vol.369, pp.213-236, 2013.

J. Strosberg, G. El-haddad, E. Wolin, A. Hendifar, J. Yao et al., Phase 3 trial of (177)Lu-Dotatate for midgut neuroendocrine tumors, N Engl J Med, vol.376, pp.125-160, 2017.

C. Kratochwil, F. Bruchertseifer, H. Rathke, M. Bronzel, C. Apostolidis et al., Targeted alpha-therapy of metastatic castration-resistant prostate cancer with (225)Ac-PSMA-617: dosimetry estimate and empiric dose finding, J Nucl Med, vol.58, pp.1624-1655, 2017.

L. Krolicki, F. Bruchertseifer, J. Kunikowska, H. Koziara, B. Krolicki et al., Prolonged survival in secondary glioblastoma following local injection of targeted alpha therapy with (213)Bi-substance P analogue, Eur J Nucl Med Mol Imaging, vol.45, pp.1636-1680, 2018.

J. P. Pouget, I. Navarro-teulon, M. Bardies, N. Chouin, G. Cartron et al., Clinical radioimmunotherapy-the role of radiobiology, Nat Rev Clin Oncol, vol.8, pp.720-754, 2011.

I. Corre, M. Guillonneau, and F. Paris, Membrane signaling induced by high doses of ionizing radiation in the endothelial compartment. Relevance in radiation toxicity, Int J Mol Sci, vol.14, pp.22678-96, 2013.

G. Stark, The effect of ionizing radiation on lipid membranes, Biochim Biophys Acta, vol.1071, pp.103-125, 1991.

A. Negre-salvayre, C. Coatrieux, C. Ingueneau, and R. Salvayre, Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors, Br J Pharmacol, vol.153, pp.6-20, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00420347

M. Garcia-barros, F. Paris, C. Cordon-cardo, D. Lyden, S. Rafii et al., Tumor response to radiotherapy regulated by endothelial cell apoptosis, Science, vol.300, pp.1155-1164, 2003.

R. Kolesnick and Z. Fuks, Radiation and ceramide-induced apoptosis, Oncogene, vol.22, pp.5897-906, 2003.

J. P. Pouget, A. G. Georgakilas, and J. L. Ravanat, Targeted and off-target (Bystander and Abscopal) effects of radiation therapy: redox mechanisms and risk/ benefit analysis, Antioxid Redox Signal, vol.29, pp.1447-87, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01785294

E. Gulbins and R. Kolesnick, Raft ceramide in molecular medicine, Oncogene, vol.22, pp.7070-7077, 2003.

H. Nagasawa, A. Cremesti, R. Kolesnick, R. Kolesnick, Z. Fuks et al., Involvement of membrane signaling in the bystander effect in irradiated cells, Cancer Res, vol.62, pp.2531-2535, 2002.

F. M. Lyng, P. Maguire, B. Mcclean, C. B. Seymour, and C. Mothersill, The involvement of calcium and MAP kinase signaling pathways in the production of radiation-induced bystander effects, Radiat Res, vol.165, pp.400-409, 2006.

E. I. Azzam, S. M. De-toledo, T. Gooding, and J. B. Little, Intercellular communication is involved in the bystander regulation of gene expression in human cells exposed to very low fluences of alpha particles, Radiat Res, vol.150, pp.497-504, 1998.

E. I. Azzam, S. M. De-toledo, and J. B. Little, Oxidative metabolism, gap junctions and the ionizing radiation-induced bystander effect, Oncogene, vol.22, pp.7050-7057, 2003.

F. M. Lyng, C. B. Seymour, and C. Mothersill, Initiation of apoptosis in cells exposed to medium from the progeny of irradiated cells: a possible mechanism for bystander-induced genomic instability?, Radiat Res, vol.157, pp.365-70, 2002.

L. Huo, H. Nagasawa, and J. B. Little, HPRT mutants induced in bystander cells by very low fluences of alpha particles result primarily from point mutations, Radiat Res, vol.156, pp.521-526, 2001.

O. V. Belyakov, M. Folkard, C. Mothersill, K. M. Prise, and B. D. Michael, Bystanderinduced differentiation: a major response to targeted irradiation of a urothelial explant model, Mutat Res, vol.597, pp.43-52, 2006.

E. I. Azzam, G. P. Raaphorst, and R. E. Mitchel, Radiation-induced adaptive response for protection against micronucleus formation and neoplastic transformation in C3H 10T1/2 mouse embryo cells, Radiat Res, vol.138, pp.28-31, 1994.

J. Poleszczuk, A. Krzywon, U. Forys, and M. Widel, Connecting radiation-induced bystander effects and senescence to improve radiation response prediction, Radiat Res, vol.183, pp.571-578, 2015.

W. F. Morgan, Is there a common mechanism underlying genomic instability, bystander effects and other nontargeted effects of exposure to ionizing radiation?, Oncogene, vol.22, pp.7094-7103, 2003.

W. F. Morgan, Non-targeted and delayed effects of exposure to ionizing radiation: II. Radiation-induced genomic instability and bystander effects in vivo, clastogenic factors and transgenerational effects, Radiat Res, vol.159, pp.581-96, 2003.

W. F. Morgan, Non-targeted and delayed effects of exposure to ionizing radiation: I. Radiation-induced genomic instability and bystander effects in vitro, Radiat Res, vol.159, pp.567-80, 2003.

C. Mothersill and C. B. Seymour, Radiation-induced bystander effects-implications for cancer, Nat Rev Cancer, vol.4, pp.158-64, 2004.

K. A. Vallis, R. M. Reilly, D. Scollard, P. Merante, A. Brade et al., Phase I trial to evaluate the tumor and normal tissue uptake, radiation dosimetry and safety of (111)In-DTPA-human epidermal growth factor in patients with metastatic EGFR-positive breast cancer, Am J Nucl Med Mol Imaging, vol.4, pp.181-92, 2014.

L. Santoro, S. Boutaleb, V. Garambois, C. Bascoul-mollevi, V. Boudousq et al., Noninternalizing monoclonal antibodies are suitable candidates for 125I radioimmunotherapy of small-volume peritoneal carcinomatosis, J Nucl Med, vol.50, pp.2033-2074, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00442929

A. Hallqvist, K. Bergmark, T. A. Back, H. Andersson, P. Dahm-kahler et al., Intraperitoneal alpha-emitting radio immunotherapy with Astatine-211 in relapsed ovarian cancer; long-term follow-up with individual absorbed dose estimations, J Nucl Med, 2019.

E. Deshayes, R. Ladjohounlou, L. Fur, P. Pichard, A. Lozza et al., Anti-MISRII radiolabeled antibodies: new tools for a theranostic approach in ovarian cancer, J Nucl Med, vol.59, pp.1234-1276, 2018.

G. Kohler, S. C. Howe, and C. Milstein, Fusion between immunoglobulin-secreting and nonsecreting myeloma cell lines, Eur J Immunol, vol.6, pp.292-297, 1976.

V. Boudousq, L. Bobyk, M. Busson, V. Garambois, M. Jarlier et al., Comparison between internalizing anti-HER2 mAbs and noninternalizing anti-CEA mAbs in alpha-radioimmunotherapy of small volume peritoneal carcinomatosis using 212Pb, PLoS One, vol.8, p.69613, 2013.

S. Paillas, R. Ladjohounlou, C. Lozza, A. Pichard, V. Boudousq et al., Localized irradiation of cell membrane by Auger electrons is cytotoxic through oxidative stress-mediated nontargeted effects, Antioxid Redox Signal, vol.25, pp.467-84, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01905223

J. P. Pouget, L. Santoro, L. Raymond, N. Chouin, M. Bardies et al., Cell membrane is a more sensitive target than cytoplasm to dense ionization produced by auger electrons, Radiat Res, vol.170, pp.192-200, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00311037

K. Rothkamm and M. Lobrich, Evidence for a lack of DNA double-strand break repair in human cells exposed to very low x-ray doses, Proc Natl Acad Sci U S A, vol.100, pp.5057-62, 2003.

D. T. Goodhead, Initial events in the cellular effects of ionizing radiations: clustered damage in DNA, Int J Radiat Biol, vol.65, pp.7-17, 1994.

A. Haimovitz-friedman, C. C. Kan, D. Ehleiter, R. S. Persaud, M. Mcloughlin et al., Ionizing radiation acts on cellular membranes to generate ceramide and initiate apoptosis, J Exp Med, vol.180, pp.525-560, 1994.

A. Catala, Lipid peroxidation of membrane phospholipids generates hydroxy-alkenals and oxidized phospholipids active in physiological and/or pathological conditions, Chem Phys Lipids, vol.157, pp.1-11, 2009.

K. Simons and E. Ikonen, Functional rafts in cell membranes, Nature, vol.387, pp.569-72, 1997.

D. A. Brown and E. London, Functions of lipid rafts in biological membranes, Annu Rev Cell Dev Biol, vol.14, pp.111-147, 1998.

C. E. Chalfant, K. Kishikawa, M. C. Mumby, C. Kamibayashi, A. Bielawska et al., Long chain ceramides activate protein phosphatase-1 and protein phosphatase-2A. Activation is stereospecific and regulated by phosphatidic acid, J Biol Chem, vol.274, pp.20313-20320, 1999.

N. A. Bourbon, J. Yun, and M. Kester, Ceramide directly activates protein kinase C zeta to regulate a stress-activated protein kinase signaling complex, J Biol Chem, vol.275, pp.35617-35640, 2000.

Y. Zhang, B. Yao, S. Delikat, S. Bayoumy, X. H. Lin et al., Kinase suppressor of Ras is ceramide-activated protein kinase, Cell, vol.89, pp.63-72, 1997.

J. Cadet, T. Delatour, T. Douki, D. Gasparutto, J. P. Pouget et al., Hydroxyl radicals and DNA base damage, Mutat Res, vol.424, pp.9-21, 1999.

H. Qiu, T. Edmunds, J. Baker-malcolm, K. P. Karey, S. Estes et al., Activation of human acid sphingomyelinase through modification or deletion of C-terminal cysteine, J Biol Chem, vol.278, pp.32744-52, 2003.

P. Santana, L. A. Pena, A. Haimovitz-friedman, S. Martin, D. Green et al., Acid sphingomyelinase-deficient human lymphoblasts and mice are defective in radiation-induced apoptosis, Cell, vol.86, pp.189-99, 1996.

S. S. Castillo, M. Levy, J. V. Thaikoottathil, and T. Goldkorn, Reactive nitrogen and oxygen species activate different sphingomyelinases to induce apoptosis in airway epithelial cells, Exp Cell Res, vol.313, pp.2680-2686, 2007.

M. Sathekge, F. Bruchertseifer, O. Knoesen, F. Reyneke, I. Lawal et al., Ac-PSMA-617 in chemotherapy-naive patients with advanced prostate cancer: a pilot study, Eur J Nucl Med Mol Imaging, vol.46, issue.225, pp.129-167, 2019.

A. Morgenstern, C. Apostolidis, C. Kratochwil, M. Sathekge, L. Krolicki et al., An overview of targeted alpha therapy with (225)Actinium and (213)Bismuth, Curr Radiopharm, vol.11, pp.200-208, 2018.

S. Tubin, H. H. Popper, and L. Brcic, Novel stereotactic body radiation therapy (SBRT)-based partial tumor irradiation targeting hypoxic segment of bulky tumors (SBRT-PATHY): improvement of the radiotherapy outcome by exploiting the bystander and abscopal effects, Radiat Oncol, vol.14, p.21, 2019.

S. Demaria, B. Ng, M. L. Devitt, J. S. Babb, N. Kawashima et al., Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated, Int J Radiat Oncol Biol Phys, vol.58, pp.862-70, 2004.

J. B. Gorin, J. Menager, S. Gouard, C. Maurel, Y. Guilloux et al., Antitumor immunity induced after alpha irradiation, Clin Cancer Res Riad Ladjohounlou, vol.16, pp.4775-4790, 2014.