Medical Radiology and Radiation Safety. 2015. Vol. 60. No. 2. P. 66-81

REVIEW

Y.N. Korystov

The Analysis of Radiobiological Data for the Estimation of the Carcinogenic Risk from Low Radiation Doses

Institute of Theoretical and Experimental Biophysics of RAS, Pushchino, Moscow region, Russia, e-mail: This email address is being protected from spambots. You need JavaScript enabled to view it.  

CONTENTS

1. Introduction

2. About experimental basis of linear-no-threshold dose dependence of stochastic effects of radiation

3. Transfer energy absorbed outside cell to target: the factors that can increase target volume and decrease the effective dose

3.1. Effect of irradiated medium

3.2. Bystander effect

4. Radiation induced genome instability

5. DNA reparation and elimination of mutated cells: the factors that can decrease the target volume and increase the effective dose

5.1. Role of DNA reparation in radiation carcinogenesis

5.2. Stimulation of anticancer immunity and apoptosis with low radiation doses

6. Dependence of stochastic effects of radiation on dose rate

7. Influence of low doses of ionizing radiation on carcinogenesis: experimental data

8. Conclusion

Key words: ionizing radiation, low doses, radiation-induced tumors

REFERENCES

  1. Koterov A.N. Ot ochen' malykh do ochen' bol'shikh doz radiatsii: novye dannye po ustanovleniyu diapazonov i ikh eksperimental'no-epidemiologicheskie obosnovaniya. Medical Radiology and Radiation Safety. 2013. Vol. 58. No. 2. P. 5-21.
  2. Dauer L.T., Brooks A.L., Hoel D.G. et. al. Review and evaluation of updated research on the health effects associated with low-dose ionising radiation. Radiat. Prot. Dosimetry. 2010. Vol. 140. No. 2. P. 103-136.
  3. Muller H.J. Radiation and genetics. Amer. Nat. 1930. Vol. 64. P. 220-257.
  4. Brues A.M. A critique of the linear theory of carcinogenesis: present data on human leukemgenesis by radiation indicate that a nonlinear relation is more probable. Science. 1958. Vol. 128. P. 693-699.
  5. Il'in L.A. Realii i mify Chernobylya. Moscow: ALARA limited. 1994. 445 p.
  6. Koterov A.N. Malye dozy radiatsii: fakty i mify. Kniga pervaya. Osnovnye ponyatiya i nestabil'nost' genoma. Moscow. Publ. FMBTs im. A.I. Burnazyana FMBA Rossii. 2010. 283 p.
  7. Petin V.G., Pronkevich M.D. Analiz deistviya malykh doz ioniziruyushchego izlucheniya na onkozabolevaemost' cheloveka. Radiatsiya i risk. 2012. Vol. 21. No. 1. P. 38-56.
  8. Averbeck D. Does scientific evidence support a change from the LNT model for low-dose radiation risk extrapolation? Health Phys. 2009. Vol. 97. No. 5. P. 493-504.
  9. Di Majo V., Rebessi S., Pazzaglia S. et. al. Carcinogenesis in laboratory mice after low doses of ionizing radiation. Radiat. Res. 2003. Vol. 159. No. 1. P. 102-108.
  10. Lacoste-Collin L., Jozan S., Cances-Lauwers V. et. al. Effect of continuous irradiation with a very low dose of gamma rays on life span and the immune system in SJL mice prone to B-cell lymphoma. Radiat. Res. 2007. Vol. 168. No. 6. P. 725-732.
  11. Luckey T.D. Atomic bomb health benefits. Dose-Response. 2008. Vol. 6. No. 4. P. 369-382.
  12. Mitchel R.E.J., Jackson J.S., Mccann R.A. et. al. Adaptive response modification of latency for radiation-induced myeloid leukemia in CBA/H mice. Radiat. Res. 1999. Vol. 152. No. 3. P. 273-279.
  13. Pollycove M., Feinendegen L.E. Radiation-induced versus endogenous DNA damage: possible effect of inducible protective responses in mitigating endogenous damage. Hum. Exp. Toxicol. 2003. Vol. 22. No. 6. P. 290-306.
  14. Prise K.M. New advances in radiation biology. Occupat. Medicine. 2006. Vol. 56. No. 3. P. 156-161.
  15. Suzuki K., Yamashita S. Low-dose radiation exposure and carcinogenesis. Jpn. J. Clin. Oncol. 2012. Vol. 42. No. 7. P. 563-568.
  16. Vaiserman A.M. Radiation hormesis: historical perspective and implications for low-dose cancer risk assessment. Dose-Response. 2010. Vol. 8. No. 2. P. 172-191.
  17. Prasad K.N., Cole W.C., Haase G.M. Health risks of low dose ionizing radiation in humans: a review. Exp. Biol. Med. 2004. Vol. 229. No. 5. P. 378-382.
  18. Unated Nations. UNSCEAR 2008. Report to the General Assembly, with Scientific Annexes. Sources and effects of ionizing radiation. Volume II. Annex D. Health effects due to radiation from Chernobyl accident. Unated Nations. New York. 2011. P. 45-220.
  19. Prasad K.N., Cole W.C., Haase G.M. Radiation protection in humans: extending the concept of as low as reasonably achievable (ALARA) from dose to biological damage. Brit. J. Radiol. 2004. Vol. 77. No. 914. P. 97-99.
  20. ICRP Publication 60: 1990 Recommendations of the International Commission on Radiological Protection. Annals of the ICRP 1991. Vol. 21, No. 1-3.
  21. Standards for protection against radiation-Nuclear Regulatory Commission. Final rule. Federal Register. 1991; 56:23360-474.
  22. Biological effects of ionizing radiation BEIR V. National Academic Press: Committee on the Biological Effects of Ionizing Radiation. Washington: DC. 1990.
  23. Kuo S.S., Saad A.H., Koong A.C. et. al. Potassium-channel activation in response to low doses of gamma-irradiation involves reactive oxygen intermediates in nonexcitatory cells. Proc. Acad. Sci. USA. 1993. Vol. 90. No. 3. P. 908-912.
  24. Prasad K.N. Handbook of Radiobiology. 2nd ed. Boca Raton, FL, CRC Press. 1995. 352 p.
  25. Rothkamm K., Lobrich M. From the cover: evidence for a lack of DNA double-strand break repair in human cells exposed to very low x-ray doses. Proc. Natl. Acad. Sci. USA. 2003. Vol. 100. No. 9. P. 5057-5062.
  26. Golfier S., Jost G., Pietsch H. et. al. Dicentric chromosomes and γ-H2AX foci formation in lymphocytes of human blood samples exposed to a CT scanner: a direct comparison of dose response relationships. Radiat. Prot. Dosimetry. 2009. Vol. 134. No. 1. P. 55-61.
  27. Mancuso M., Pasquali E., Leonardi S. et. al. Oncogenic bystander radiation effects in patchedheterozygous mouse cerebellum. Proc. Natl. Acad. Sci. USA. 2008. Vol. 105. No. 34. P. 12445-12450.
  28. Grudzenskia S., Rathsa A., Conrada S. et. al. Inducible response required for repair of low-dose radiation damage in human fibroblasts. Proc. Acad. Sci. USA. 2010. Vol. 107. No. 32. P. 14205-14210.
  29. Neumaier T., Swensonb J., Phamd C. et. al. Evidence for formation of DNA repair centers and dose-response nonlinearity in human cells. Proc. Natl. Acad. Sci. USA. 2011. Vol. 109. No. 2. P. 443-448.
  30. Baure J., Izadi A., Suarez V. et. al. Histone H2AX phosphorylation in response to changes in chromatin structure induced by altered osmolarity. Mutagenesis. 2009. Vol. 24. No. 2. P. 161-167.
  31. de Feraudy S., Revet I., Bezrookove V. et. al. A minority of foci or pan-nuclear apoptotic staining of γ-H2AX in the S phase after UV damage contain DNA double-strand breaks. Proc. Natl. Acad. Sci. USA. 2010. Vol. 107. No. 15. P. 6870-6875.
  32. Korystov Y.N., Shaposhnikova V.V., Korystova A.F. et. al. Detection of reactive oxygen species induced by radiation in cells using the dichlorofluorescein assay. Radiat. Res. 2007. Vol. 168. No. 2. P. 226-232.
  33. Wan X.S., Zhou Z., Kennedy A.R. Adaptation of the dichlorofluorescein assay for detection of radiation induced oxidative stress in cultured cells. Radiat. Res. 2003. Vol. 160. No. 5. P. 622-630.
  34. Wan X.S., Zhou Z., Ware J.H. et. al. Standardization of a fluorometric assay for measuring oxidative stress in irradiated cells. Radiat. Res. 2005. Vol. 163. No. 2. P. 232-240.
  35. Korystov Y.N. About the role of extracellular radiation induced oxidants in cell oxidative stress at irradiation determined with the dichlorofluorescein assay. Radiat. Res. 2008. Vol. 170. No. 3. P. 407-408.
  36. Kumagai J., Nakama M., Miyazaki T. et. al. Scavenging of long-lived radicals by (-)-epigallocatechin-3-O-gallate and simultaneous suppression of mutation in irradiated mammalian cells. Radiat. Phys. Chem. 2002. Vol. 64. No. 4. P. 293-297.
  37. Davies M.J., Fu S., Dean R.T. Protein hydroperoxides can give rise to reactive free radicals. Biochem. J. 1995. Vol. 305. No. 2. P. 643-649.
  38. Dean R.T., Gieseg S., Davies M.J. Reactive species and their accumulation on radical-damaged proteins. Trends Biochem. Sci. 1993. Vol. 18. No. 11. P. 437-441.
  39. Pattison D.I., Dean R.T., Davies M.J. Oxidation of DNA, proteins and lipids by DOPA, protein-bound DOPA, and related catechol(amine)s. Toxicology. 2002. Vol. 177. No. 1. P. 23-37.
  40. Simpson J.A., Narita S., Gieseg S. et. al. Long-lived reactive species on free-radical-damaged proteins. Biochem. J. 1992. Vol. 282. No. 3. P. 621-624.
  41. Bruskov V.I., Karp O.E., Garmash S.A. et. al. Prolongation of oxidative stress by long-lived reactive protein species induced by X-ray radiation and their genotoxic action. Free Radical Res. 2012. Vol. 46. No. 10. P. 1280-1290.
  42. Koterov A.N. Perspektivy ucheta «effekta svidetelya» pri otsenke radiatsionnykh riskov. Mediko-biologicheskie problemy zhiznedeyatel'nosti. 2011. No. 1(5). P. 7-20.
  43. Yang, H., Asaad N., Held K.D. Medium-mediated intercellular communication is involved in bystander responses of X-ray irradiated normal human fibroblasts. Oncogene. 2005. Vol. 24. No. 12. P. 2096-2103.
  44. Hu B., Wu L., Han W. et. al. The time and spatial effects of bystander response in mammalian cells induced by low dose radiation. Carcinogenesis. 2006. Vol. 27. No. 2. P. 245-251.
  45. Lyng F.M., Seymour C.B., Mothersill C. Oxidative stress in cells exposed to low levels of ionizing radiation. Biochem. Soc. Transact. 2001. Vol. 29. No. 2. P. 350-353.
  46. Morgan W.F. Non-targeted and delayed effects of exposure to ionizing radiation: I. Radiation-induced genomic instability and bystander effects in vitro. Radiat. Res. 2003. Vol. 159. No. 5. P. 567-580.
  47. Shao C., Furusawa Y., Kobayashi Y. et. al. Bystander effect induced by counted high-LET particles in confluent human fibroblasts: a mechanistic study. FASEB J. 2003. Vol. 17. No. 11. P. 1422-1427.
  48. Groesser T., Cooper B., Rydberg B. Lack of bystander effects from high-LET radiation for early cytogenetic end points. Radiat. Res. 2008. Vol. 170. No. 6. P. 794-802.
  49. Fournier C., Barberet P., Pouthier T. et. al. No evidence for DNA and early cytogenetic damage in bystander cells after heavy-ion microirradiation at two facilities. Radiat. Res. 2009. Vol. 171. No. 5. P. 530-540.
  50. Sowa M.B., Goetz W., Baulch J.E. et. al. Lack of evidence for low-LET radiation induced bystander response in normal human fibroblasts and colon carcinoma cells. Int. J. Radiat. Biol. 2010. Vol. 86. No. 2. P. 102-113.
  51. Zhou H., Suzuki M., Geard C.R. et. al. Effects of irradiated medium with or without cells on bystander cell responses. Mutat. Res. 2002. Vol. 499. No. 2. P. 135-141.
  52. Eidus L.Kh., Korystov Yu.N. Kislorod v radiobiologii. Moscow. Energoatomizdat. 1984. 176 p.
  53. Petrov R.V. Immunologiya. Moscow. Meditsina. 1982. 368 p.
  54. Coleman W.B., Tsongali G.J. Multiple mechanisms account for genomic instability and molecular mutation in neoplastic transformation. Clin. Chem. 1995. Vol. 41. No. 5. P. 644-657.
  55. Khanna K.K. Cancer risk and the ATM gene: a continuing debate. J. Nat. Cancer Inst. 2000. Vol. 92. No. 10. P. 795-802.
  56. Truong L.N., Wu X. Prevention of DNA re-replication in eukaryotic cells. J. Mol. Cell Biol. 2011. Vol. 3. No. 1. P. 13-22.
  57. Dugan L.C., Bedford J.S. Are chromosomal instabilities induced by exposure of cultured normal human cells to low- or high-LET radiation? Radiat. Res. 2003. Vol. 159. No. 3. P. 301-311.
  58. Koterov A.N. Genomic instability at exposure of low dose radiation with low LET. Mythical mechanism of unproved carcinogenic effects. Int. J. Low Radiation (Paris). 2005. Vol. 1. No. 4. P. 376-451.
  59. Koterov A.N. Otsutstvie faktov nestabil'nosti genoma posle oblucheniya v malykh dozakh radiatsiei s nizkoi LPE kletok bez yavnykh defektov i organizma vne in utero. Radiats. biologiya. Radioekologiya. 2006. Vol. 46. No. 5. P. 585-596.
  60. Koterov A.N. Radiatsionno-indutsirovannaya nestabil'nost' genoma pri deistvii malykh doz radiatsii v nauchnykh publikatsiyakh i v dokumentakh mezhdunarodnykh organizatsii poslednikh let. Medical Radiology and Radiation Safety. 2009. Vol. 54. No. 4. P. 5-13.
  61. Koterov A.N. Istoriya mifa o nestabil'nosti genoma pri malykh dozakh radiatsii. Nauchnaya tochka, veroyatno, postavlena. Medical Radiology and Radiation Safety. 2014. Vol. 59. No. 1. P. 5-19.
  62. Koterov A.N. Novye fakty ob otsutstvii induktsii nestabil'nosti genoma pri malykh dozakh radiatsii s nizkoi LPE i sootvetstvuyushchie vyvody o poroge effekta v soobshchenii NKDAR-2012 (pis'mo v redaktsiyu). Radiats. biologiya. Radioekologiya. 2014. Vol. 54. No. 3. P. 309-312.
  63. Al'ferovich A.L., Gotlib V.Ya., Pelevina I.I. Izmenenie proliferativnoi aktivnosti kletok pri deistvii radiatsii v malykh dozakh. News RAN. Ser. Biologiya. 1995. No. 1. P. 15-18.
  64. Korystov Y.N., Eliseeva N.A., Kublik L.N. et. al. The effect of low-dose irradiation on proliferation of mammalian cells in vitro. Radiat. Res. 1996. Vol. 146, no 3. P. 329-332.
  65. Fialkov P.J. Clonal origin of human tumors. Biochim. Biophys. Acta. 1976. Vol. 458. No. 3. P. 283-321.
  66. Trosko J.E., Chang C.C. The role of mutagenesis in carcinogenesis. Photochem. Photobiol. Rev. 1978. Vol. 3. No. 1. P. 135-168.
  67. Bruner S.D., Norman D.P., Verdine G.L. Structural basis for recognition and repair of the endogenous mutagen 8-oxoguanine in DNA. Nature. 2000. Vol. 403. No. 6772. P. 859-866.
  68. Fleck O., Nielsen O. DNA repair. J. Cell Sci. 2004. Vol. 117. No. 4. P. 515-517.
  69. Lindahl T. Instability and decay of the primary structure of DNA. Nature. 1993. Vol. 362. No. 6422. P. 709-715.
  70. Fortini P., Dogliotti E. Base damage and single-strand break repair: echanisms and functional significance of short- and long-patch repair subpathways. DNA Repair. 2007. Vol. 6. No. 4. P. 398-409.
  71. Lieber M.R. The mechanism of human nonhomologous DNA end joining. J. Biol. Chem. 2008. Vol. 283. No. 1. P. 1-5.
  72. Le X.C., Xing J.Z., Lee J. et. al. Inducible repair of thymine glycol detected by an ultra sensitive assay for DNA damage. Science. 1998. Vol. 280. No. 5366. P. 1066-1069.
  73. Shadley J.D., Afzal V., Wolff. S. Characterization of the adaptive response to ionizing radiation induced by low doses of X-rays to human lymphocytes. Radiat. Res. 1987. Vol. 111. No. 3. P. 511-517.
  74. Wiencke J.K., Afzal V., Olivieri G. et. al. Evidence that the [3H] thymidine-induced adaptive response of human lymphocytes to subsequent doses of X-rays involves the induction of a chromosomal repair mechanism. Mutagenesis. 1986. Vol. 1. No. 5. P. 375-380.
  75. Wolf S. The adaptive response in radiobiology: evolving insights and implications. Environ. Health Persp. 1998. Vol. 106. No. 5. P. 277-283.
  76. Redpath J.L. Radiation induced neoplastic transformation in vitro: evident for a protective effect at low doses of low LET radiation. Cancer Metastasis Rev. 2004. Vol. 23. No. 3-4. P. 333-339.
  77. Azzam E.I., Raaohorst G.P., Mitchel R.E.J. Radiation-induced adaptive response for protection against micronucleus formation and neoplastic transformation in C3H 10t1/2 mouse embryo cells. Radiat. Res. 1994. Vol. 138. No. 1s. P. S28-S31.
  78. Rigaud O., Papadopoulo D., Moustacchi E. Decreased deletion mutation in radioadapted human lymphoblasts. Radiat. Res. 1993. Vol. 133. No. 1. P. 94-101.
  79. Zhou P.K., Liu X.Y., Sun W.Z. et. al. Cultured mouse SR-1 cells exposed to low dose of y-rays become less susceptible to the induction of mutagenesis by radiation as well as bleomycin. Mutagenesis. 1993. Vol. 8. No. 2. P. 109-111.
  80. Dunn G.P., Bruce A.T., Ikeda H. et. al. Cancer immunoediting: from immunosurveillance to tumor escape. Nature Immunol., 2002. Vol. 3. No. 11. P. 991-998.
  81. Schreiber R.D., Old L.J., Smyth M.J. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011. Vol. 331. No. 6034. P. 1565-1570.
  82. Liu S.Z. Nonlinear dose-response relationship in the immune system following exposure to ionizing radiation: mechanisms and implications. Nonlinearity Biol. Toxicol. Med. 2003. Vol. 1. No. 1. P. 71-92.
  83. Liu S.Z. Biological effects of low level exposures to ionizing radiation: Theory and practice. Hum. Exp. Toxicol. 2010. Vol. 29. No. 4. P. 275-281.
  84. Pollycove M. Radiobiological basis of low-dose irradiation in prevention and therapy of cancer. Dose-Response. 2007. Vol. 5. No. 1. P. 26-38.
  85. Mitchel R.E.J. Low doses of radiation reduce risk in vivo. Dose-Response. 2007. Vol. 5. No. 1. P. 1-10.
  86. Li X.Y., Li X.J., He R.H. et. al. Influence of low dose radiation on the carcinogenic effect of high dose radiation. Chin. J. Radiol. Med. Protect. 2003. Vol. 23. No. 2. P. 411-413. (in Chinese).
  87. Liu S.Z. Cancer control related to stimulation of immunity by low-dose radiation. Dose-Response. 2007. Vol. 5. No. 1. P. 39-47.
  88. Hashimoto S., Shirato H., Hosokawa M. et. al. The suppression of metastases and the change in host immune response after low-dose total-body irradiation in tumor-bearing rats. Radiat. Res. 1999. Vol. 151. No. 6. P. 717-724.
  89. Jin S.Z., Pan X.N., Wu N. et. al. Whole-body low dose irradiation promotes the efficacy of conventional radiotherapy for cancer and possible mechanisms. Dose-Response. 2007. Vol. 5. No. 4. P. 349-558.
  90. Sakamoto K., Myogin M., Hosoi Y. Fundamental and clinical studies on cancer control with total or upper half body irradiation. J. Jpn. Soc. Ther. Oncol. 1997. Vol. 9. No. 1. P. 161-175.
  91. Potten C.S. Extreme sensitivity of some intestinal crypt cells to X and γ-irradiation. Nature. 1977. Vol. 269. No. 5628. P. 518-521.
  92. Li D.E. Deistvie radiatsii na zhivye kletki. Moscow. Gosatomizdat. 1963. 288 p.
  93. Jaruga P., Dizdaroglu M. Repair of products of oxidative DNA base damage in human cells. Nucleic Acid Res. 1996. Vol. 24. No. 8. P. 1389-1394.
  94. Frankenberg-Schwager M. Induction, repair and biological relevance of radiation-induced DNA lesions in eukaryotic cells. Radiat. Environ. Biophys. 1990. Vol. 29. No. 4. P. 273-292.
  95. Lobrich M., Rief N., Kuhne M. et. al. In vivo formation and repair of DNA double-strand breaks after computed tomography examinations. Proc. Acad. Sci. USA. 2005. Vol. 102. No. 25. P. 8984-8989.
  96. Yarmonenko S.P., Vainson A.A. Radiobiologiya cheloveka i zhivotnykh. Moscow. Vyssh. Shk. 2004. 549 p.
  97. Liu S.Z. On radiation hormesis expressed in the immune system. Crit. Rev. Toxicol. 2003. Vol. 33. No. 3-4. P. 431-441.
  98. Ishii-Ohba H., Kobayashi S., Nishimura M. et. al. Existence of a threshold-like dose for gamma-ray induction of thymic lymphomas and no susceptibility to radiation-induced solid tumors in SCID mice. Mutat. Res. 2007. Vol. 619. No. 1-2. P. 124-133.
  99. Tanooka H. Threshold dose-response in radiation carcinogenesis: an approach from chronic beta-irradiation experiments and a review of non tumor doses. Int. J. Radiat. Biol. 2001. Vol. 77. No. 5. P. 541-551.
  100. Makinodan T. Cellular and subcellular alteration in immune cells induced by chronic, intermittent exposure in vivo to very low dose of ionizing radiation (ldr) and its ameliorating effects on progression of autoimmune disease and mammary tumor growth. In: Low Dose Irradiation and Biological Defense Mechanisms. In Sugahara T., Sagan L.A., Aoyama T. (eds.). Amsterdam, Exerpta Medica. 1992. P. 233-237.
  101. Crump K.S., Duport P., Jiang H. et. al. A meta-analysis of evidence for hormesis in animal radiation carcinogenesis, including a discussion of potential pitfalls in statistical analyses to detect hormesis. J. Toxicol. Environ. Health B. Crit. Rev. 2012. Vol. 15. No. 3. P. 210-231.
  102. Upton A.C., Randolph M L., Conklin, J.W. et. al. Late effects of fast neutrons and gamma-rays in mice as influenced by the dose rate of irradiation: Induction of neoplasia. Radiat. Res. 1970. Vol. 41. No. 3. P. 467-491.
  103. Benjamin S.A., Lee A.C., Angleton G.M. et. al. Mortality in beagles irradiated during prenatal and postnatal development. II. Contribution of benign and malignant neoplasia. Radiat. Res. 1998. Vol. 150. No. 3. P. 330-348.

For citation: Zakharov IS. The Analysis of Radiobiological Data for the Estimation of the Carcinogenic Risk from Low Radiation Doses. Medical Radiology and Radiation Safety. 2015;60(2):66-81. Russian.

PDF (RUS) Full-text article (in Russian)