Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Cancer in rodents: does it tell us about cancer in humans?

Key Points

  • Whereas laboratory rodents (namely mice and rats) are similar to humans in some aspects, there are important differences among mammalian species that make valid interpretation and extrapolation of the results from rodent cancer experiments to humans problematic.

  • The five most common human cancers are those of the breast (female), the prostate (male), and the lungs, colon, and stomach (both sexes). Mammary tumours are also common in rodents. However, there are no rat or mouse strains that exhibit a high incidence of spontaneous carcinomas of the stomach or colon.

  • A decrease in the overall risk of cancer owing to old age has been recorded in both human and rodent studies. Three important factors could be responsible for this intriguing decline: detection bias, differential selection, and the effects of individual ageing. Studies in rodents argue against a diagnostic bias as a leading cause.

  • The risk of cancer has increased over time in most human populations. Why this is remains unclear, but addressing this problem is crucial for understanding the nature of cancer.

  • Some studies indicate that the differences in cancer incidence rates between males and females are similar in rodents and humans. This is a surprising finding that requires additional explanation.

  • Whereas tumours often grow at a slower rate during old age, the chances for survival of a transplanted tumour in a recipient host often increases with rodent age. This is in agreement with human data indicating that ageing can both decelerate tumour growth and increase the chances of latent tumour survival in older organisms.

  • The spontaneous regression of tumours is a rare phenomenon in adult humans, whereas it is common in mature laboratory rodents. This effect and its implications need further investigation.

  • Few rodent carcinogens were established as clearly carcinogenic to humans. Similarly, some human carcinogens are not carcinogenic to rodents. This creates a significant problem for interpreting the results of animal experiments with carcinogens in relation to humans.

  • These and other differences warn against the simple extrapolation of the results of rodent experiments to humans and call for further investigation of this important problem to reliably predict cancer risks, as well as foster success in treating human cancers based on data from laboratory animal studies.

Abstract

Information obtained from animal models (mostly mice and rats) has contributed substantially to the development of treatments for human cancers. However, important interspecies differences have to be taken into account when considering the mechanisms of cancer development and extrapolating the results from mice to humans. Comparative studies of cancer in humans and animal models mostly focus on genetic factors. This review discusses the bio-epidemiological aspects of cancer manifestation in humans and rodents that have been underrepresented in the literature.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Typical age-patterns of overall cancer incidence and mortality rates in humans in the USA.
Figure 2: Age-patterns of cancer incidence rate in laboratory rodents of different strains.
Figure 3: Time trends in overall cancer incidence rate.
Figure 4: Patterns of sex difference in cancer incidence.

Similar content being viewed by others

References

  1. Hansen, K. & Khanna, C. Spontaneous and genetically engineered animal models; use in preclinical cancer drug development. Eur. J. Cancer 40, 858–880 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Hawk, E. T. et al. Can animal models help us select specific compounds for cancer prevention trials? Recent Results Cancer Res. 166, 71–87 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Hahn, W. C. & Weinberg, R. A. Modelling the molecular circuitry of cancer. Nature Rev. Cancer 2, 331–341 (2002). Basic information on the differences between the genetic events required to induce malignant transformation in human and mouse cells.

    Article  CAS  Google Scholar 

  4. Rangarajan, A. & Weinberg, R. A. Comparative biology of mouse versus human cells: modelling human cancer in mice. Nature Rev. Cancer 3, 952–959 (2003). Important paper addressing differences in how tumorigenesis occurs in mice and humans.

    Article  CAS  Google Scholar 

  5. Maronpot, R. R., Flake, G. & Huff, J. Relevance of animal carcinogenesis findings to human cancer predictions and prevention. Toxicol. Pathol. 32 (Suppl. 1), 40–48 (2004).

    Article  CAS  PubMed  Google Scholar 

  6. DePinho, R. A. The age of cancer. Nature 408, 248–254 (2000). Review paper on the link between advanced age and increased incidence of cancer in mice and humans.

    Article  CAS  PubMed  Google Scholar 

  7. Ward, J. M., Anver, M. R., Mahler, J. F. & Devor-Henneman, D. E. in Pathology of Genetically Engineered Mice (eds J. M. Ward, et al.) 161–179 (Iowa State Univ., Ames, 2000).

    Google Scholar 

  8. Turusov, V. S. et al. Tumors in CF-1 mice exposed for six consecutive generations to DDT. J. Natl Cancer Inst. 51, 983–997 (1973).

    Article  CAS  PubMed  Google Scholar 

  9. Anisimov, V. N. et al. Dose-dependent effect of melatonin on life span and spontaneous tumor incidence in female SHR mice. Exp. Gerontol. 38, 449–461 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Artandi, S. E. et al. Telomere dysfunction promotes non-reciprocal translocations and epithelial cancers in mice. Nature 406, 641–645 (2000).

    Article  CAS  PubMed  Google Scholar 

  11. Staats, J. Standardized nomenclature for inbred strains of mice: seventh listing. Cancer Res. 40, 2083–2128 (1980).

    CAS  PubMed  Google Scholar 

  12. Storer, J. B. Longevity and gross pathology at death in 22 inbred mouse strains. J. Gerontol. 21, 404–409 (1966).

    Article  CAS  PubMed  Google Scholar 

  13. Ward, J. M. Background data and variations in tumor rates of control rats and mice. Progr. Exp. Tumor Res. 26, 241–263 (1983).

    Article  CAS  PubMed  Google Scholar 

  14. Anisimov, V. N. Carcinogenesis and Aging. Volumes 1 & 2 (CRC, Boca Raton, 1987).

    Google Scholar 

  15. Ferlay, J., Parkin, D. M. & Pisani, P. GLOBOCAN: cancer incidence and mortality worldwide. IARC Cancer Base No. 3 (IARC, Lyon, 1998).

  16. Kaspareit-Rittinghausen, J., Wiese, K., Deerberg, F. & Nitsche, B. Incidence and morphology of spontaneous thyroid tumours in different strains of rats. J. Comp. Pathol. 102, 421–432 (1990).

    Article  CAS  PubMed  Google Scholar 

  17. Vollmer, G. Endometrial cancer: experimental models useful for studies on molecular aspects of endometrial cancer and carcinogenesis. Endocr. Relat. Cancer 10, 23–42 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Anisimov, V. N. Molecular and Physiological Mechanisms of Aging (Nauka, St. Petersburg, 2003).

    Google Scholar 

  19. Kagawa Y. Impact of Westernization on the nutrition of japanese: changes in physique, cancer, longevity and centenarians. Prev. Med. 7, 205–217 (1978).

    Article  CAS  PubMed  Google Scholar 

  20. The International Agency for Research on Cancer. Cancer Incidence in Five Continents. Volumes 1–8 (Lyon, IARC, 1965–2003). Important data source on age-specific cancer incidence rates in humans worldwide.

  21. Ferlay, J., Bray, F., Sankila, R. & Parkin, D. M. EUCAN: Cancer Incidence, Mortality and Prevalence in the European Union in 1996. Version 3.1 (IARC, Lyon, 1999).

    Google Scholar 

  22. National Cancer Institute, Bethesda, Maryland. SEER Cancer Statistics Review, 1975–2002 (eds Ries, L. A. G. et al.) (2005). http://seer.cancer.gov/csr/1975_2002/

  23. WHO Regional Office for Europe. European Health for all database (HFA-DB) (2005). http://www.euro.who.int/hfadb

  24. Center for Disease Control and Prevention. National Center for Health Statistics (2005). http://www.cdc.gov/nchs/

  25. Smith, D. Cancer mortality at very old ages. Cancer 77, 1367–1372 (1996).

    Article  CAS  PubMed  Google Scholar 

  26. Smith, D. in The Paradoxes of Longevity (eds Robine, J. M. et al.) 61–71 (Springer-Verlag, Berlin, 1999). An important study revealing the decline in cancer mortality rate at oldest old ages.

    Book  Google Scholar 

  27. Ukraintseva, S. V. & Yashin, A. I. Individual aging and cancer risk: How are they related? Demogr. Res. 9, 163–196 (2003). A study that establishes a link between somatic ageing and the decline in overall cancer risk at old ages.

    Article  Google Scholar 

  28. Vaupel, J. W. & Yashin, A. I. Cancer rates over age, time, and place: insights from stochastic models of heterogeneous populations. MPIDR Working Papers, WP-1999-06 (1999).

  29. Arbeev, K. G., Ukraintseva, S. V., Arbeeva, L. S. & Yashin A. I. Decline in human cancer incidence rates at old ages: age–period—cohort considerations. Demogr. Res. 12, 272–300 (2005).

    Google Scholar 

  30. Yancik, R. Ovarian cancer. Cancer 73, 517–523 (1993).

    Google Scholar 

  31. Yancik, R. & Ries, L. A. Cancer in older persons. Cancer 74, 1955–2013 (1994).

    Article  Google Scholar 

  32. Pompei, F. & Wilson, R. Age distribution of cancer: the incidence turnover at old age. Human Ecol. Risk Assesment 7, 1619–1650 (2001).

    Article  Google Scholar 

  33. Andersen, S. L. et al. Cancer in the oldest old. Mech. Ageing Dev. 126, 263–267 (2005).

    Article  PubMed  Google Scholar 

  34. Stanta, G. in Comprehensive Geriatric Oncology (eds Balducci, L., G. H. Lyman, W. B. Ershler & M. Extermann) 187–193 (Taylor and Francis Group, London, 2004).

    Google Scholar 

  35. Kuramoto, K., Matsushita, S., Esaki, Y. & Shimada, H. Prevalence, rate of correct clinical diagnosis and mortality of cancer in 4,894 elderly autopsy cases. Nippon Ronen Igakkai Zasshi. 30, 35–40 (1993).

    Article  CAS  PubMed  Google Scholar 

  36. Pompei, F., Polkanov, M. & Wilson, R. Age distribution of cancer in mice: the incidence turnover at old age. Toxicol. Ind. Health 17, 7–16 (2001). Important study demonstrating the decline in cancer incidence rate in old mice.

    Article  CAS  PubMed  Google Scholar 

  37. Anisimov, V. N. et al. Spontaneous tumors in rats bred at animal farm 'Rappolovo' of the USSR Academy of Medical Sciences. Vopr. Onkol. 24, 64–70 (1978).

    CAS  PubMed  Google Scholar 

  38. Dilman, V. M. & Anisimov, V. N. Effect of treatment with phenformin, diphenylhyantoin or L-DOPA on life span and tumor incidence in C3H/Sn mice. Gerontology 26, 241–246 (1980).

    Article  CAS  PubMed  Google Scholar 

  39. Ukraintseva, S. V. & Yashin, A. I. How individual aging may influence human morbidity and mortality patterns. Mech. Ageing Dev. 122, 1447–1460 (2001).

    Article  CAS  PubMed  Google Scholar 

  40. Ershler, W. B. in Comprehensive Geriatric Oncology (eds Balducci, L., Lyman, G. H., Ershler, W. B. & Extermann, M.) 147–157 (London: Taylor and Francis Group, 2004).

    Google Scholar 

  41. Ershler, W. B. Explanations for reduced tumor proliferative capacity with age. Exp. Gerontol. 27, 551–558 (1992).

    Article  CAS  PubMed  Google Scholar 

  42. Ershler, W. B. et al. B16 murine melanoma and aging: slower growth and longer survival in old mice. J. Natl Cancer Inst. 72, 161–164 (1992).

    Article  Google Scholar 

  43. Pili, R. et al. Altered angiogenesis underlying age-dependent changes in tumor growth. J. Natl Cancer Inst. 86, 1303–1314 (1994).

    Article  CAS  PubMed  Google Scholar 

  44. Tanaka, Y. et al. Growth pattern and rate in residual non-functioning pituitary adenomas: correlations among tumor volume doubling time, patient age, and MIB-1 index. J. Neurosurg. 98, 359–365 (2003).

    Article  PubMed  Google Scholar 

  45. Kreisle, R. A., Stebler, B. A. & Ershler, W. B. Effect of host age on tumor-associated angiogenesis in mice. J. Natl Cancer Inst. 82, 44–47 (1990).

    Article  CAS  PubMed  Google Scholar 

  46. Gravekamp, C. et al. Behavior of metastatic and nonmetastatic breast tumors in old mice. Exp. Biol. Med. 229, 665–675 (2004).

    Article  CAS  Google Scholar 

  47. Anisimov, V. N. & Zhukovskaya, N. V. Effect of age on development of transplantable tumors in mice. Vopr. Onkol. 27, 52–59 (1981).

    CAS  PubMed  Google Scholar 

  48. Anisimov, V. N. in Comprehensive Geriatric Oncology (eds Balducci, L., Lyman, G. H., Ershler, W. B. & Extermann, M.) 75–101 (Taylor and Francis Group, London, 2004).

    Google Scholar 

  49. Campisi, J. Cancer, aging and cellular senescence. In Vivo 14, 183–188 (2000).

    CAS  PubMed  Google Scholar 

  50. Holland-Frei Cancer Medicine. 5th edn (eds Bast, R. C. et al.) (BC Decker Inc., Ontario, 2000).

  51. Campisi, J. Cancer and ageing: rival demons. Nature Rev. Cancer 3, 339–349 (2003).

    Article  CAS  Google Scholar 

  52. The International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risk to humans vol. 72. Hormonal contraceptives and post-menopausal hormonal therapy (Lyon, IARC, 1999).

  53. Lipman, R. D., Dallal, G. E. & Bronson, R. T. Effect of genotype and diet on age-related lesions in ad libitum fed and calorie-restricted F344, BN, and BNF3F1 rats. J. Gerontol. Med. Sci. 54A, 478–491 (1999).

    Article  Google Scholar 

  54. Ukraintseva, S. V. & Yashin, A. I. Economic progress as cancer risk factor. I. Puzzling facts of cancer epidemiology. MPIDR Working Papers, WP-2005-021 (2005).

  55. Arbeev, K. G., Ukraintseva, S. V., Arbeeva, L. S. & Yashin, A. I. Mathematical models for human cancer incidence rates. Demogr. Res. 12, 272–300 (2005).

    Google Scholar 

  56. Belitsky, G. A. et al. Concerning the absence of mutagenicity and carcinogenicity in mildronate. Vopr. Oncol. 45, 279–282 (1999).

    CAS  Google Scholar 

  57. Gilbert, C., Gillman, L., Loustalbot, P. & Lutz, W. The modifying influence of diet and the physical environment on spontaneous tumour frequency in rats. Brit. J. Cancer 12, 257–293 (1958).

    Article  Google Scholar 

  58. MacKenzie, W. F. & Garner, F. M. Comparison of neoplasms in six sources of rats. J. Natl Cancer Inst. 50, 1243–1257 (1973).

    Article  CAS  Google Scholar 

  59. Anisimov, V. N. et al. Increase in the longevity and decrease in the tumor incidence in mice given polypeptide thymus and epiphysis factors at different age. Doklady Akad. Nauk SSSR 302, 473–476 (1988).

    CAS  Google Scholar 

  60. Turusov, V. S. et al. Tumors in CF-1 mice exposed for six consecutive generations to DDT. J. Natl Cancer Inst. 51, 983 (1973).

    Article  CAS  PubMed  Google Scholar 

  61. Eiben, R. Frequency and time trends of spontaneous tumors found in B6C3F1 mice oncogenicity studies over 10 years. Exp. Toxicol. Pathol. 53, 399–408 (2001).

    Article  CAS  PubMed  Google Scholar 

  62. Baranova, L. N., Romanov, K. P. & Yamshanov, V. A. Study of levels of benzo(a)pyrene and N-nitrosamines in the food of laboratory animals. Vopr. Onkol. 5, 54–57 (1986).

    Google Scholar 

  63. Tennekes, H., Kaufmann, W., Dammann, M. & van Ravenzwaay, B. The stability of historical control data for common neoplasms in laboratory rats and the implications for carcinogenic assessment. Regul. Toxicol. Pharmacol. 40, 293–304 (2004).

    Article  CAS  PubMed  Google Scholar 

  64. Anisimov, V. N. et al. Spontaneous tumors in outbred LIO rats. J. Exp. Clin. Cancer Res. 8, 254–262 (1989).

    Google Scholar 

  65. McCullough, K. D., Coleman, W. B., Smith, G. J. & Grisham, J. W. Age-dependent regulation of the tumorigenic potential of neoplastically transformed rat liver epithelial cells by the liver microenvironment Cancer Res. 54, 3668–3671 (1994). A study demonstrating an increase in tumour transplantability with the increasing age of recipient rats. These findings implicate that young age provides a more suppressive microenvironment for tumour formation.

    CAS  PubMed  Google Scholar 

  66. Breslow, N. et al. Latent carcinoma of prostate at autopsy in seven areas. Int. J. Cancer 20, 680–688 (1977).

    Article  CAS  PubMed  Google Scholar 

  67. Avetisian, I. L. & Petrova, G. V. Latent thyroid pathology in residents of Kiev, Ukraine. Environ. Pathol. Toxicol. Oncol. 15, 239–243 (1996).

    CAS  Google Scholar 

  68. Barnea, E. R., Barnea, J. D. & Pines, M. Control of cell proliferation by embryonal-origin factors. Am. J. Reprod. Immunol. 35, 318–324 (1996).

    Article  CAS  PubMed  Google Scholar 

  69. Joshi, S. S., Tarantolo, S. R., Kuszynski, C. A. & Kessinger, A. Antitumour therapeutic potential of activated human umbilical cord blood cells against leukemia and breast cancer. Clin. Cancer Res. 11, 4351–4358 (2000).

    Google Scholar 

  70. Staflin, K. et al. Neural progenitor cell lines inhibit rat tumor growth in vivo. Cancer Res. 64, 5347–5354 (2004).

    Article  CAS  PubMed  Google Scholar 

  71. Ukraintseva, S. V. & Yashin, A. I. Treating cancer with embryonic stem cells: rationale comes from aging studies. Frontiers Biosci. 10, 588–595 (2005).

    Article  CAS  Google Scholar 

  72. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000).

    Article  CAS  PubMed  Google Scholar 

  73. Ukraintseva, S. V. & Yashin, A. I. Cancer as 'rejuvenescence'. Ann. N.Y. Acad. Sci. 1019, 200–205 (2004).

    Article  CAS  PubMed  Google Scholar 

  74. McCullough, K. D., Coleman, W. B., Smith, G. J. & Grisham, J. W. Age-dependent induction of hepatic tumor regression by the tissue microenvironment after transplantation of neoplastically transformed rat liver epithelial cells into the liver. Cancer Res. 57, 1807–1813 (1997).

    CAS  PubMed  Google Scholar 

  75. Krtolica, A. & Campisi, J. Integrating epithelial cancer, aging stroma and cellular senescence. Adv. Gerontol. 11, 109–116 (2003).

    CAS  PubMed  Google Scholar 

  76. Krtolica, A. et al. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc. Natl Acad. Sci. USA 98, 12072–12077 (2001). Important study showing that an environment consisting of senescent cells favours the proliferation of pre-malignant cells in culture.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Smith, J. R. & Pereira-Smith, O. M. Replicative senescence: implications for in vivo aging and tumor suppression. Science 273, 63–66 (1996).

    Article  CAS  PubMed  Google Scholar 

  78. Caruso, C., Lio, D. Cavallone, L. & Francerschi, C. Ageing, longevity, inflammation and cancer. Ann. N.Y. Acad. Sci. 1028, 1–13 (2004).

    Article  CAS  PubMed  Google Scholar 

  79. Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860–867 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Dilman, V. M., Ostroumova, M. N. & Tsyrlina, E. V. Hypothalamic mechanisms of ageing and of specific age pathology-II. The sensitivity threshold of hypothalamo-pituitary complex to homeostatic stimuli in adaptive homeostasis. Exp. Gerontol. 14, 175–181 (1979).

    Article  CAS  PubMed  Google Scholar 

  81. Semenchenko, G. V., Anisimov, V. N. & Yashin, A. I. Stressors and antistressors: how do they influence life span in HER-2/neu transgenic mice? Exp. Gerontol. 39, 1499–1511 (2004).

    Article  CAS  PubMed  Google Scholar 

  82. Tulinius, H. Latent malignancies at autopsy: a little used source of information on cancer biology (eds Riboli, E. & Delendi, M.). IARC Sci. Publ. 112, 253–261 (1991).

    Google Scholar 

  83. Mechanisms of carcinogenesis in risk identification (eds Vanio, H., Magee, P. N., McGregor, D. & McMichael, A. J.). IARC Sci. Publ. 116, 9–56 (1992).

  84. Shvemberger, I. N. Conversion of malignant cells into normal ones. Int. Rev. Cytol. 103, 341–86 (1986).

    Article  CAS  PubMed  Google Scholar 

  85. The International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans. Volumes 1–78 (IARC, Lyon, 1972–2001).

  86. Sloan, D., Fleizer, D., Richards, G., Murray, D. & Brown, R. Increased incidence of experimental colon cancer associated with long-term metronidazole therapy. Am. J. Surg. 145, 66–70 (1983).

    Article  CAS  PubMed  Google Scholar 

  87. Chemical Induction of Cancer. Modulation and Combination Effects. An Inventory of the Many Factors which Influence Carcinogenesis (eds Arcos, J. C., Argus, M. F. & Woo, Y.) (Birkhauser, Boston, 1995).

  88. Microbes and Malignancy: Infection as a Cause of Human Cancer (ed. Parsonnet, J.) (Oxford Univ., New York, 1999).

  89. Falk, P. G., Hooper, L. V., Midtvedt, T. & Gordon, J. I. Creating and maintaining the gastrointestinal ecosystem: what we know and need to know from gnotobiology. Microbiol. Mol. Biol. Rev. 62, 1157–1170 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. The International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans vol. 4. Some aromatic amines, hydrazine and related substances, N-nitroso compounds and miscellaneous alkylating agents (IARC, Lyon, 1974).

  91. Anisimov, V. N. Life span extension and cancer risk: myths and reality. Exp. Gerontol. 26, 1101–1137 (2001).

    Article  Google Scholar 

  92. Gart, J. J. et al. Statistical Methods in Cancer Research. Vol. 3 – The Design and Analysis of Long-Term Animal Experiments (IARC, Lyon, 1986).

    Google Scholar 

  93. Davis, S., Mirck, D. K. & Stevens, R. G. Night shift work, light at night, and risk of breast cancer. J. Natl Cancer Inst. 93, 1557–1562 (2001).

    Article  CAS  PubMed  Google Scholar 

  94. Schernhammer, E. S. et al. Rotating night shifts and risk of breast cancer in women participating in the nurses' health study. J. Natl Cancer. Inst. 93, 1563–1568 (2001). This study demonstrates the increased breast cancer risk in night-shift female workers.

    Article  CAS  PubMed  Google Scholar 

  95. Schernhammer, E. S. et al. Night-shift work and risk of colorectal cancer in the nurses' health study. J. Natl Cancer Inst. 5, 825–828 (2003).

    Article  Google Scholar 

  96. Stevens, R. G. & Rea, M. S. Light in the built environment: potential role of circadian disruption in endocrine disruption and breast cancer. Cancer Causes Control 12, 279–287 (2001).

    Article  CAS  PubMed  Google Scholar 

  97. Arendt, J. Melatonin and the Mammalian Pineal Gland (Chapman & Hall, London, 1995).

    Google Scholar 

  98. van den Heiligenberg, S. et al. The tumor promoting effect of constant light exposure on diethylnitrosamine-induced hepatocarcinogenesis in rats. Life Sci. 64, 2523–2534 (1999).

    Article  CAS  PubMed  Google Scholar 

  99. Beniashvili, D. S., Benjamin, S., Baturin, D. A. & Anisimov, V. N. Effect of light/dark regimen on N-nitrosoethylurea-induced transplacental carcinogenesis in rats. Cancer Lett. 163, 51–57 (2001).

    Article  CAS  PubMed  Google Scholar 

  100. Anisimov, V. N. The light–dark regimen and cancer development. Neuroendocrinol. Lett. 23 (Suppl. 2), 28–36 (2002).

    CAS  PubMed  Google Scholar 

  101. Anisimov, V. N. et al. Effect of exposure to light-at-night on life span and spontaneous carcinogenesis in female CBA mice. Int. J. Cancer 111, 475–479 (2004).

    Article  CAS  PubMed  Google Scholar 

  102. Hsieh, C. C., Trichopoulos, D., Katsouyanni, K. & Yuasa, S. Age at menarche, age at menopause, height and obesity as risk factors for breast cancer: associations and interactions in an international case-control study. Int. J. Cancer, 46, 796–800 (1990).

    Article  CAS  PubMed  Google Scholar 

  103. Nagasawa, H & Nakajima, Y. Relationship between incidence and onset age of mammary tumors and reproductive characteristics in mice. Eur. J. Cancer 16, 111–116 (1980).

    Google Scholar 

  104. Sydnor, K. L., Butenandt, O., Brillantes, F. P. & Huggins, C. Race-strain factor related to hydrocarbon-induced mammary cancer in rats. J. Natl Cancer Inst. 29, 805–808 (1962).

    CAS  PubMed  Google Scholar 

  105. DeGraaff, J. & Stolte, L. A. Age at menarche and menopause of uterine cancer patients. Eur. J. Obstet. Gynecol. Reprod. Biol. 8, 187–193 (1978).

    Article  CAS  Google Scholar 

  106. Franceschi, S. et al. Pooled analysis of 3 European case-control studies of ovarian cancer: II. Age at menarche and at menopause. Int. J. Cancer 19, 57–60 (1991).

    Article  Google Scholar 

  107. Miccozzi, M. S. Functional consequences from varying patterns of growth and maturation during adolescence. Horm. Res. 39 (Suppl. 3), 49–58 (1993).

    Article  Google Scholar 

  108. Harvard report on cancer prevention: causes of human cancer. Smoking (eds Monson, R. & Wals, J.). Cancer Causes Control 1 (Suppl. 1), S5–S6 (1996).

  109. Giovannucci, E. Insulin, insulin-like growth factors and colon cancer: a review of the evidence. J. Nutr. 131 (Suppl. 11), 3109–3120 (2001).

    Article  Google Scholar 

  110. Anisimov, V. N. Insulin/IGF-1 signaling pathway driving aging and cancer as a target for pharmacological intervention. Exp. Gerontol. 38, 1041–1049 (2003).

    Article  CAS  PubMed  Google Scholar 

  111. Ikeno, Y et al. Delayed occurrence of fatal neoplastic diseases in Ames dwarf mice: correlation to extended longevity. J. Gerontol. Biol. Med. Sci. 58, 291–296 (2003).

    Article  Google Scholar 

  112. Kenyon, C. A conserved regulatory system for aging. Cell 105, 165–168 (2001).

    Article  CAS  PubMed  Google Scholar 

  113. Bartke, A. et al. Insulin-like growth factor 1 (IGF-1) and aging: controverses and new insights. Biogerontology 4, 1–8 (2003).

    Article  CAS  PubMed  Google Scholar 

  114. Gries, C. L. & Young, S. S. Positive correlation of body weight with pituitary tumor incidence in rats. Fund. Appl. Toxicol. 2, 145–148 (1982).

    Article  CAS  Google Scholar 

  115. Anisimov, V. N. et al. Body weight is not always a good predictor of longevity in mice. Exp. Gerontol. 39, 305–319 (2004).

    Article  PubMed  Google Scholar 

  116. Anisimov, V. N. et al. Is early life body weight a predictor of longevity and tumor risk in rats? Exp Gerontol. 39, 807–816 (2004).

    Article  PubMed  Google Scholar 

  117. Inoue, M., Sobue, T. & Tsugane, S. JPHC Study Group. Impact of body mass index on the risk of total cancer incidence and mortality among middle-aged Japanese: data from a large-scale population-based cohort study — the JPHC study. Cancer Causes Control 15, 671–680 (2004).

    Article  PubMed  Google Scholar 

  118. Engeland, A., Tretli, S. & Bjorge, T. Height and body mass index in relation to esophageal cancer; 23-year follow-up of two million Norwegian men and women. Cancer Causes Control 8, 837–43 (2004).

    Article  Google Scholar 

  119. Ford, E. S. Body mass index and colon cancer in a national sample of adult US men and women. Am. J. Epidemiol. 150, 390–398 (1999).

    Article  CAS  PubMed  Google Scholar 

  120. Hu, G. et al. The effects of physical activity and body mass index on cardiovascular, cancer and all-cause mortality among 47212 middle-aged Finnish men and women. Int. J. Obes. Relat. Metab. Disord. 29, 894–902 (2005).

    Article  CAS  Google Scholar 

  121. Eichholzer, M., Bernasconi, F., Jordan, P. & Stahelin, H. B. Body mass index and the risk of male cancer mortality of various sites: 17-year follow-up of the Basel cohort study. Swiss Med. Wkly. 135, 27–33 (2005)

    PubMed  Google Scholar 

  122. Dilman, V. M. Ageing, metabolic immunodepression and carcinogenesis. Mech. Ageing Dev. 8, 153–173 (1978).

    Article  CAS  PubMed  Google Scholar 

  123. Dilman, V. M. Development, Aging and Disease. A New Rationale for an Intervention (Harwood Academic, Pennsylvania, 1994).

    Google Scholar 

  124. Colangelo, L. A. et al. Colorectal cancer mortality and factors related to the insulin resistance syndrome. Cancer Epidemiol. Biomarkers Prev. 11, 385–391 (2002).

    PubMed  Google Scholar 

  125. Gupta, K., Krishnaswamy, G., Karnad, A. & Peiris, A. N. Insulin: a novel factor in carcinogenesis. Am. J. Med. Sci. 323, 140–145 (2002).

    Article  CAS  PubMed  Google Scholar 

  126. Pollak, M. N., Schernhammer, E. S. & Hankinson, S. E. Insulin-like growth factors and neoplasia. Nature Rev. Cancer 4, 505–518 (2004).

    Article  CAS  Google Scholar 

  127. Popovich, I. G. et al. Insulin in aging and cancer: new antidiabetic drug Diabenol as geroprotector and anticarcinogen. Int. J. Biochem. Cell. Biol. 37, 1117–1129 (2005).

    Article  CAS  PubMed  Google Scholar 

  128. Berstein, L. M. Clinical usage of hypolipidemic and antidiabetic drugs in the prevention and treatment of cancer. Cancer Lett. 224, 203–212 (2005).

    Article  CAS  PubMed  Google Scholar 

  129. Dupont, W. D. & Page, D. L. Breast cancer risk associated with proliferative disease, age at first birth, and a family history of breast cancer. Am. J. Epidemiol. 125, 769–779 (1987).

    Article  CAS  PubMed  Google Scholar 

  130. Hemminki, K., Kyyronen, P. & Vaittinen, P. Parental age as a risk factor of childhood leukemia and brain cancer in offspring. Epidemiology 10, 271–275(1999). A large-scale study showing that both maternal and paternal old age increase cancer risk in children.

    Article  CAS  PubMed  Google Scholar 

  131. Hemminki, K. & Kyyronen, P. Parental age and risk of sporadic and familial cancer in offspring; implications for germ cell mutagenesis. Epidemiology 10, 747–751 (1999).

    Article  CAS  PubMed  Google Scholar 

  132. Hemminki, K., Vaittinen, P. & Kyyronen, P. Age-specific risks in common cancers of the offspring. Int. J. Cancer 78, 172–175 (1998).

    Article  CAS  PubMed  Google Scholar 

  133. Anisimov, V. N. & Gvardina, O. E. N-nitrosomethylurea-induced carcinogenesis in the progeny of male rats of different ages. Mutat. Res. 316, 139–145 (1995).

    Article  CAS  PubMed  Google Scholar 

  134. Evan, G. I. & Voudsen, K. H. Proliferation, cell cycle and apoptosis in cancer. Nature 411, 342–348 (2001).

    Article  CAS  PubMed  Google Scholar 

  135. Weber, B. L. Cancer genomics. Cancer Cell 1, 37–47 (2002).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors wish to thank James W. Vaupel for the opportunity to conduct substantial parts of this work at the Max Planck Institute for Demographic Research, Germany. We are thankful to Mark A. Zabezhinski and Igor Akushevich for their comments on the paper. We are also thankful to Virginia Lewis for help in preparing this manuscript. This research was partly supported by NIH/NIA grants and by a grant from the President of the Russian Federation.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Vladimir N. Anisimov or Svetlana V. Ukraintseva.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

Entrez Gene

Trp53

National Cancer Institute

bladder tumours

colon cancer

endometrial carcinomas

leukaemia

lymphomas

skin carcinoma

stomach cancer

Glossary

CANCER INCIDENCE RATE

A proportion of new cancer cases (registered for the first time) in a population of a given age.

PREVALENCE OF CANCER

A proportion of individuals with a diagnosed cancer (no matter when the diagnosis was made) in a population of a given age. The prevalence characterizes the cancer burden.

CANCER MORTALITY RATE

A proportion of cancer deaths in a population of a given age.

POLYCYCLIC AROMATIC HYDROCARBONS

Their metabolites (diol epoxides) bind DNA and induce point-mutations in oncogenes (for example, HRAS).

NITROSO COMPOUNDS

Potent alkylating mutagens and carcinogens. The most important target in DNA is guanine at the O6 position.

2-NAPHTHYLAMINE

The oxidation of 2-naphthylamine at the amine group leads to the formation of hydroxylamine, which binds DNA in the target tissue.

ONTOGENY

The total of the stages of an organism's life history.

HYPERINSULINAEMIA

An increased level of insulin in the serum.

ANTIDIABETIC MEDICINES

Antidiabetic drugs, phenformin (1-phenylethylbiguanide), buformin (1-butylbiguanide hydrochloride) and metformin (N,N-dimethylbiguanide) decrease the blood glucose level and increase the susceptibility of tissues to insulin.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Anisimov, V., Ukraintseva, S. & Yashin, A. Cancer in rodents: does it tell us about cancer in humans?. Nat Rev Cancer 5, 807–819 (2005). https://doi.org/10.1038/nrc1715

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc1715

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing