Browse > Article
http://dx.doi.org/10.5487/TR.2018.34.4.281

Thresholds of Genotoxic and Non-Genotoxic Carcinogens  

Nohmi, Takehiko (Division of Pathology, Biological Safety Research Center, National Institute of Health Sciences)
Publication Information
Toxicological Research / v.34, no.4, 2018 , pp. 281-290 More about this Journal
Abstract
Exposure to chemical agents is an inevitable consequence of modern society; some of these agents are hazardous to human health. The effects of chemical carcinogens are of great concern in many countries, and international organizations, such as the World Health Organization, have established guidelines for the regulation of these chemicals. Carcinogens are currently categorized into two classes, genotoxic and non-genotoxic carcinogens, which are subject to different regulatory policies. Genotoxic carcinogens are chemicals that exert carcinogenicity via the induction of mutations. Owing to their DNA interaction properties, there is thought to be no safe exposure threshold or dose. Genotoxic carcinogens are regulated under the assumption that they pose a cancer risk for humans, even at very low doses. In contrast, non-genotoxic carcinogens, which induce cancer through mechanisms other than mutations, such as hormonal effects, cytotoxicity, cell proliferation, or epigenetic changes, are thought to have a safe exposure threshold or dose; thus, their use in society is permitted unless the exposure or intake level would exceed the threshold. Genotoxicity assays are an important method to distinguish the two classes of carcinogens. However, some carcinogens have negative results in in vitro bacterial mutation assays, but yield positive results in the in vivo transgenic rodent gene mutation assay. Non-DNA damage, such as spindle poison or topoisomerase inhibition, often leads to positive results in cytogenetic genotoxicity assays such as the chromosome aberration assay or the micronucleus assay. Therefore, mechanistic considerations of tumor induction, based on the results of the genotoxicity assays, are necessary to distinguish genotoxic and non-genotoxic carcinogens. In this review, the concept of threshold of toxicological concern is introduced and the potential risk from multiple exposures to low doses of genotoxic carcinogens is also discussed.
Keywords
Genotoxic carcinogens; Non-genotoxic carcinogens; Threshold; Threshold of toxicological concern; TTC;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Roberts, R.A., Goodman, J.I., Shertzer, H.G., Dalton, T.P. and Farland, W.H. (2003) Rodent toxicity and nongenotoxic carcinogenesis: knowledge-based human risk assessment based on molecular mechanisms. Toxicol. Mech. Methods, 13, 21-29.   DOI
2 Ramirez, T., Eastmond, D.A. and Herrera, L.A. (2007) Nondisjunction events induced by albendazole in human cells. Mutat. Res., 626, 191-195.   DOI
3 Asanami, S. and Shimono, K. (1999) The effect of hyperthermia on micronucleus induction by mutagens in mice. Mutat. Res., 446, 149-154.   DOI
4 Armstrong, M.J., Gara, J.P., Gealy, R., III, Greenwood, S.K., Hilliard, C.A., Laws, G.M. and Galloway, S.M. (2000) Induction of chromosome aberrations in vitro by phenolphthalein: mechanistic studies. Mutat. Res., 457, 15-30.   DOI
5 Galloway, S.M., Miller, J.E., Armstrong, M.J., Bean, C.L., Skopek, T.R. and Nichols, W.W. (1998) DNA synthesis inhibition as an indirect mechanism of chromosome aberrations: comparison of DNA-reactive and non-DNA-reactive clastogens. Mutat. Res., 400, 169-186.   DOI
6 Elhajouji, A., Van, H.P. and Kirsch-Volders, M. (1995) Indications for a threshold of chemically-induced aneuploidy in vitro in human lymphocytes. Environ. Mol. Mutagen., 26, 292-304.   DOI
7 Nohmi, T., Toyoda-Hokaiwado, N., Yamada, M., Masumura, K., Honma, M. and Fukushima, S. (2008) International symposium on genotoxic and carcinogenic thresholds. Genes Environ., 30, 101-107.   DOI
8 Lovell, D.P. (2000) Dose-response and threshold-mediated mechanisms in mutagenesis: statistical models and study design. Mutat. Res., 464, 87-95.   DOI
9 Butterworth, B.E. (1990) Consideration of both genotoxic and nongenotoxic mechanisms in predicting carcinogenic potential. Mutat. Res., 239, 117-132.   DOI
10 Fukushima, S., Kinoshita, A., Puatanachokchai, R., Kushida, M., Wanibuchi, H. and Morimura, K. (2005) Hormesis and dose-response-mediated mechanisms in carcinogenesis: evidence for a threshold in carcinogenicity of non-genotoxic carcinogens. Carcinogenesis, 26, 1835-1845.   DOI
11 Fukushima, S., Wei, M., Kakehashi, A. and Wanibuchi, H. (2012) Threshold for genotoxic carcinogens: the central concern in carcinogenic risk assessment. Genes Environ., 32, 153-156.
12 Jenkins, G.J., Zair, Z., Johnson, G.E. and Doak, S.H. (2010) Genotoxic thresholds, DNA repair, and susceptibility in human populations. Toxicology, 278, 305-310.   DOI
13 Nohmi, T. and Tsuzuki, T. (2016) Possible mechanisms underlying genotoxic thresholds: DNA repair and translesion DNA synthesis in Thresholds of Genotoxic Carcinogens: from Mechanisms to Regulation (Nohmi, T. and Fukushima, S. Eds.) Elsevier, pp. 49-66.
14 Suzuki, M., Matsui, K., Yamada, M., Kasai, H., Sofuni, T. and Nohmi, T. (1997) Construction of mutants of Salmonella typhimurium deficient in 8-hydroxyguanine DNA glycosylase and their sensitivities to oxidative mutagens and nitro compounds. Mutat. Res., 393, 233-246.   DOI
15 Kim, S.R., Kokubo, K., Matsui, K., Yamada, N., Kanke, Y., Fukuoka, M., Yamada, M. and Nohmi, T. (2005) Lightdependent mutagenesis by benzo[a]pyrene is mediated via oxidative DNA damage. Environ. Mol. Mutagen., 46, 141-149.   DOI
16 Kroes, R., Renwick, A.G., Cheeseman, M., Kleiner, J., Mangelsdorf, I., Piersma, A., Schilter, B., Schlatter, J., van, S.F., Vos, J.G. and Wurtzen, G. (2004) Structure-based thresholds of toxicological concern (TTC): guidance for application to substances present at low levels in the diet. Food Chem. Toxicol., 42, 65-83.   DOI
17 Lynch, A., Harvey, J., Aylott, M., Nicholas, E., Burman, M., Siddiqui, A., Walker, S. and Rees, R. (2003) Investigations into the concept of a threshold for topoisomerase inhibitorinduced clastogenicity. Mutagenesis, 18, 345-353.   DOI
18 Elhajouji, A., Lukamowicz, M., Cammerer, Z. and Kirsch-Volders, M. (2011) Potential thresholds for genotoxic effects by micronucleus scoring. Mutagenesis, 26, 199-204.   DOI
19 Umemura, T., Kuroiwa, Y., Tasaki, M., Okamura, T., Ishii, Y., Kodama, Y., Nohmi, T., Mitsumori, K., Nishikawa, A. and Hirose, M. (2007) Detection of oxidative DNA damage, cell proliferation and in vivo mutagenicity induced by dicyclanil, a non-genotoxic carcinogen, using gpt delta mice. Mutat. Res., 633, 46-54.   DOI
20 Federal Register (1995) Toxicological principles for the safety assessment of direct food additives and color additives used in food in Fed. Regist. pp. 36582-36596.
21 Europena Food Safety Authority (EFSA) (2016) Review of the Threshold of Toxicological Concern (TTC) approach and development of new TTC decision tree. EFSA Support. Publ., 13, 1-50.
22 Munro, I.C., Ford, R.A., Kennepohl, E. and Sprenger, J.G. (1996) Correlation of structural class with no-observedeffect levels: a proposal for establishing a threshold of concern. Food Chem. Toxicol., 34, 829-867.   DOI
23 Ohta, T. (2006) Mutagenic activity of a mixture of heterocyclic amines at doses below the biological threshold level of each. Genes Environ., 28, 181-184.   DOI
24 Nohmi, T., Katoh, M., Suzuki, H., Matsui, M., Yamada, M., Watanabe, M., Suzuki, M., Horiya, N., Ueda, O., Shibuya, T., Ikeda, H. and Sofuni, T. (1996) A new transgenic mouse mutagenesis test system using Spi- and 6-thioguanine selections. Environ. Mol. Mutagen., 28, 465-470.   DOI
25 Maron, D.M. and Ames, B.N. (1983) Revised methods for the Salmonella mutagenicity test. Mutat. Res., 113, 173-215.   DOI
26 Ashby, J. and Tennant, R.W. (1991) Definitive relationships among chemical structure, carcinogenicity and mutagenicity for 301 chemicals tested by the U.S. NTP. Mutat. Res., 257, 229-306.   DOI
27 Nohmi, T., Masumura, K. and Toyoda-Hokaiwado, N. (2017) Transgenic rat models for mutagenesis and carcinogenesis. Genes Environ., 39, 11. doi:10.1186/s41021-016-0072-6.   DOI
28 Nohmi, T., Suzuki, T. and Masumura, K. (2000) Recent advances in the protocols of transgenic mouse mutation assays. Mutat. Res., 455, 191-215.   DOI
29 MacGregor, J.T., Frotschl, R., White, P.A., Crump, K.S., Eastmond, D.A., Fukushima, S., Guerard, M., Hayashi, M., Soeteman-Hernandez, L.G., Johnson, G.E., Kasamatsu, T., Levy, D.D., Morita, T., Muller, L., Schoeny, R., Schuler, M.J. and Thybaud, V. (2015) IWGT report on quantitative approaches to genotoxicity risk assessment II. Use of pointof-departure (PoD) metrics in defining acceptable exposure limits and assessing human risk. Mutat. Res., 783, 66-78.   DOI
30 Kasper, P., Uno, Y., Mauthe, R., Asano, N., Douglas, G., Matthews, E., Moore, M., Mueller, L., Nakajima, M., Singer, T. and Speit, G. (2007) Follow-up testing of rodent carcinogens not positive in the standard genotoxicity testing battery: IWGT workgroup report. Mutat. Res., 627, 106-116.   DOI
31 National Toxicology Program (NTP) (1989) Toxicology and carcinogenesis studies of ochratoxin A (CAS No. 303-47-9) in F344/N rats (gavage studies). Natl. Toxicol. Program. Tech. Rep. Ser., 358, 1-142.
32 Malir, F., Ostry, V., Pfohl-Leszkowicz, A., Malir, J. and Toman, J. (2016) Ochratoxin A: 50 years of research. Toxins. (Basel), 8, E191. doi:10.3390/toxins8070191.   DOI
33 International Agency for Research on Cancer (IARC) (1993) Ochratoxin A. IARC Monogr. Eval. Carcinog. Risks Hum., 56, 489-521.
34 Hibi, D., Suzuki, Y., Ishii, Y., Jin, M., Watanabe, M., Sugita-Konishi, Y., Yanai, T., Nohmi, T., Nishikawa, A. and Umemura, T. (2011) Site-specific in vivo mutagenicity in the kidney of gpt delta rats given a carcinogenic dose of ochratoxin A. Toxicol. Sci., 122, 406-414.   DOI
35 Nohmi, T., Suzuki, M., Masumura, K., Yamada, M., Matsui, K., Ueda, O., Suzuki, H., Katoh, M., Ikeda, H. and Sofuni, T. (1999) Spi- selection: An efficient method to detect gammaray-induced deletions in transgenic mice. Environ. Mol. Mutagen., 34, 9-15.   DOI
36 Fessard, V., Godard, T., Huet, S., Mourot, A. and Poul, J.M. (1999) Mutagenicity of malachite green and leucomalachite green in in vitro tests. J. Appl. Toxicol., 19, 421-430.   DOI
37 European Food Safety Authority (EFSA) (2016) Malachite Green in Food. EFSA J., 14, 1-80.
38 Mittelstaedt, R.A., Mei, N., Webb, P.J., Shaddock, J.G., Dobrovolsky, V.N., McGarrity, L.J., Morris, S.M., Chen, T., Beland, F.A., Greenlees, K.J. and Heflich, R.H. (2004) Genotoxicity of malachite green and leucomalachite green in female Big Blue B6C3F1 mice. Mutat. Res., 561, 127-138.   DOI
39 Manjanatha, M.G., Shelton, S.D., Bishop, M., Shaddock, J.G., Dobrovolsky, V.N., Heflich, R.H., Webb, P.J., Blankenship, L.R., Beland, F.A., Greenlees, K.J. and Culp, S.J. (2004) Analysis of mutations and bone marrow micronuclei in Big Blue rats fed leucomalachite green. Mutat. Res., 547, 5-18.   DOI
40 Culp, S.J., Blankenship, L.R., Kusewitt, D.F., Doerge, D.R., Mulligan, L.T. and Beland, F.A. (1999) Toxicity and metabolism of malachite green and leucomalachite green during short-term feeding to Fischer 344 rats and B6C3F1 mice. Chem. Biol. Interact., 122, 153-170.   DOI
41 World Health Organization (WHO) (2000) Toxicological evaluation of certain veterinary drug residues in food, fiftyfourth meeting of the Joint FAO/WHO Expert Committee on Food Additives. WHO Food Additive Series, 45, 75-89.
42 Srivastava, S., Sinha, R. and Roy, D. (2004) Toxicological effects of malachite green. Aquat. Toxicol., 66, 319-329.   DOI
43 Culp, S.J., Mellick, P.W., Trotter, R.W., Greenlees, K.J., Kodell, R.L. and Beland, F.A. (2006) Carcinogenicity of malachite green chloride and leucomalachite green in B6C3F1 mice and F344 rats. Food Chem. Toxicol., 44, 1204-1212.   DOI
44 Phillips, D.H., Miller, J.A., Miller, E.C. and Adams, B. (1981) Structures of the DNA adducts formed in mouse liver after administration of the proximate hepatocarcinogen 1'-hydroxyestragole. Cancer Res., 41, 176-186.
45 Onami, S., Cho, Y.M., Toyoda, T., Horibata, K., Ishii, Y., Umemura, T., Honma, M., Nohmi, T., Nishikawa, A. and Ogawa, K. (2014) Absence of in vivo genotoxicity of 3-monochloropropane-1,2-diol and associated fatty acid esters in a 4-week comprehensive toxicity study using F344 gpt delta rats. Mutagenesis, 29, 295-302.   DOI
46 Kuroiwa, Y., Umemura, T., Nishikawa, A., Kanki, K., Ishii, Y., Kodama, Y., Masumura, K., Nohmi, T. and Hirose, M. (2007) Lack of in vivo mutagenicity and oxidative DNA damage by flumequine in the livers of gpt delta mice. Arch. Toxicol., 81, 63-69.   DOI
47 Wiseman, R.W., Miller, E.C., Miller, J.A. and Liem, A. (1987) Structure-activity studies of the hepatocarcinogenicities of alkenylbenzene derivatives related to estragole and safrole on administration to preweanling male C57BL/6J x C3H/HeJ F1 mice. Cancer Res., 47, 2275-2283.
48 Sekizawa, J. and Shibamoto, T. (1982) Genotoxicity of safrole-related chemicals in microbial test systems. Mutat. Res., 101, 127-140.   DOI
49 Zeiger, E., Anderson, B., Haworth, S., Lawlor, T., Mortelmans, K. and Speck, W. (1987) Salmonella mutagenicity tests: III. Results from the testing of 255 chemicals. Environ. Mutagen., 9 Suppl 9, 1-109.
50 Suzuki, Y., Umemura, T., Ishii, Y., Hibi, D., Inoue, T., Jin, M., Sakai, H., Kodama, Y., Nohmi, T., Yanai, T., Nishikawa, A. and Ogawa, K. (2012) Possible involvement of sulfotransferase 1A1 in estragole-induced DNA modification and carcinogenesis in the livers of female mice. Mutat. Res., 749, 23-28.   DOI
51 Alnouti, Y. and Klaassen, C.D. (2006) Tissue distribution and ontogeny of sulfotransferase enzymes in mice. Toxicol. Sci., 93, 242-255.   DOI
52 Masumura, K., Sakamoto, Y., Kumita, W., Honma, M., Nishikawa, A. and Nohmi, T. (2015) Genomic integration of lambda EG10 transgene in gpt delta transgenic rodents. Genes Environ., 37, 24. doi:10.1186/s41021-015-0024-6.   DOI
53 Nohmi, T. (2015) Past, present and future directions of gpt delta rodent gene mutation assays. Food Safety, 4, 1-13.
54 Ashby, J. and Tennant, R.W. (1988) Chemical structure, Salmonella mutagenicity and extent of carcinogenicity as indicators of genotoxic carcinogenesis among 222 chemicals tested in rodents by the U.S. NCI/NTP. Mutat. Res., 204, 17-115.   DOI
55 Fukushima, S., Gi, M., Kakehashi, A. and Wanibuchi, H. (2016) Qualitative and quantitative assessments on low-dose carcinogenicity of genotoxic hepatocarcinogens: dose-response for key events in rat hapatocarcinogenesis in Thresholds of Genotoxic Carcinogens: from Mechanisms to Regulation (Nohmi, T. and Fukushima, S. Eds.). Elsevier, pp. 1-17.
56 Boice, J.D., Jr. (2017) The linear nonthreshold (LNT) model as used in radiation protection: an NCRP update. Int. J. Radiat. Biol., 93, 1079-1092.   DOI
57 Auerbach, C. (1958) Radiomimetic substances. Radiat. Res., 9, 33-47.   DOI
58 The Food and Agricultural Organization of the United Nations and the World Health Organization (FAO/WHO) (2009) Chapter 7: risk characterization in Environmental Health Criteria 240: Principles and Methods for Risk Assessment of Chemicals in Food. pp. 1-18.
59 Bolt, H.M. (2008) The concept of "practical thresholds" in the derivation of occupational exposure limits for carcinogens by the scientific committee on occupatinal exposure limits (SCOEL) of the European Union. Genes Environ., 30, 114-119.   DOI
60 The Food and Agricultural Organization of the United Nations and the World Health Organization (FAO/WHO) (2009) Chapter 5: dose-response assessment and derivation of healthbased guidance values in Environmental Health Criteria 240. pp. 2-55.
61 Kirsch-Volders, M., Aardema, M. and Elhajouji, A. (2000) Concepts of threshold in mutagenesis and carcinogenesis. Mutat. Res., 464, 3-11.   DOI
62 Hayashi, Y. (1992) Overview of genotoxic carcinogens and non-genotoxic carcinogens. Exp. Toxicol. Pathol., 44, 465-471.   DOI
63 Hayashi, M. (2016) The micronucleus test-most widely used in vivo genotoxicity test. Genes Environ., 38, 18. doi:10.1186/s41021-016-0044-x.   DOI
64 Suzuki, Y., Umemura, T., Hibi, D., Inoue, T., Jin, M., Ishii, Y., Sakai, H., Nohmi, T., Yanai, T., Nishikawa, A. and Ogawa, K. (2012) Possible involvement of genotoxic mechanisms in estragole-induced hepatocarcinogenesis in rats. Arch. Toxicol., 86, 1593-1601.   DOI
65 Ishii, Y., Takasu, S., Kuroda, K., Matsushita, K., Kijima, A., Nohmi, T., Ogawa, K. and Umemura, T. (2014) Combined application of comprehensive analysis for DNA modification and reporter gene mutation assay to evaluate kidneys of gpt delta rats given madder color or its constituents. Anal. Bioanal. Chem., 406, 2467-2475.   DOI
66 Boobis, A., Brown, P., Cronin, M.T.D., Edwards, J., Galli, C.L., Goodman, J., Jacobs, A., Kirkland, D., Luijten, M., Marsaux, C., Martin, M., Yang, C. and Hollnagel, H.M. (2017) Origin of the TTC values for compounds that are genotoxic and/or carcinogenic and an approach for their re-evaluation. Crit Rev. Toxicol., 47, 705-727.
67 Jin, M., Kijima, A., Hibi, D., Ishii, Y., Takasu, S., Matsushita, K., Kuroda, K., Nohmi, T., Nishikawa, A. and Umemura, T. (2013) In vivo genotoxicity of methyleugenol in gpt delta transgenic rats following medium-term exposure. Toxicol. Sci., 131, 387-394.   DOI
68 Kuroda, K., Ishii, Y., Takasu, S., Kijima, A., Matsushita, K., Watanabe, M., Takahashi, H., Sugita-Konishi, Y., Sakai, H., Yanai, T., Nohmi, T., Ogawa, K. and Umemura, T. (2013) Cell cycle progression, but not genotoxic activity, mainly contributes to citrinin-induced renal carcinogenesis. Toxicology, 311, 216-224.   DOI
69 Maeda, J., Kijima, A., Inoue, K., Ishii, Y., Ichimura, R., Takasu, S., Kuroda, K., Matsushita, K., Kodama, Y., Saito, N., Umemura, T. and Yoshida, M. (2014) In vivo genotoxicity of Ginkgo biloba extract in gpt delta mice and constitutive androstane receptor knockout mice. Toxicol. Sci., 140, 298-306.   DOI
70 Kirsch-Volders, M., Gonzalez, L., Carmichael, P. and Kirkland, D. (2009) Risk assessment of genotoxic mutagens with thresholds: a brief introduction. Mutat. Res., 678, 72-75.   DOI