Skip to main content

Advertisement

Log in

From replica to instruments: animal models in biomedical research

  • Original Paper
  • Published:
History and Philosophy of the Life Sciences Aims and scope Submit manuscript

Abstract

The ways in which other animal species can be informative about human biology are not exhausted by the traditional picture of the animal model. In this paper, I propose to distinguish two roles which laboratory organisms can have in biomedical research. In the more traditional case, organisms act as surrogates for human beings, and as such are expected to be more manageable replicas of humans. However, animal models can inform us about human biology in a much less straightforward way, by being used as measuring devices—what I call their instrumental role. I first characterize this role and provide criteria for it, before illustrating it with some examples from biomedical research, especially cancer research. In such an instrumental role, phenotypes are not expected to phenocopy human phenomena, but instead have the purely instrumental value of detecting or measuring differences. I argue that the instrumental role is more prevalent than might first be suspected, and that some characteristics of contemporary biomedical research are increasingly shifting the use of laboratory organisms to the instrumental role. Finally, in light of the distinction proposed, I discuss the meaning of the expression “animal model”.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Notes

  1. The 1998 report of the Committee of the Institute for Laboratory Animal Research on New and Emerging Models in Biomedical and Behavioral Research writes begins with the following definition: "A biomedical model is a surrogate for a human being, or a human biologic system, that can be used to understand normal and abnormal function from gene to phenotype and to provide a basis for preventive or therapeutic intervention in human diseases." (Committee on New and Emerging Models in Biomedical and Behavioral Research 1998, p.10).

  2. In fact, Keuck (2012) noted that in many cases, complete recapitulation of a disease, for instance, might not even be desirable.

  3. Maugeri and Blasimme (2011) discuss how scientists actively correct these disanalogies.

  4. Animal models are models which have the property of being animal, i.e. of involving animals, or parts of animals. In contrast, model organisms are organisms with the additional feature of being models, generally because they are expected to be representative of a broader class of organisms (see especially Ankeny and Leonelli 2011, as well as Gayon 2006). The present paper is not about model organisms, but about animals used in the study of human diseases—about animal models and their frontiers.

  5. Discussing the intricacies of these distinctions is not the purpose of this paper, and readers are invited to refer to Meunier (2011) for a detailed discussion in the context of experimental biology.

  6. For a scientific scientific review of the CSC model, see Visvader and Lindeman (2012) or Valent et al. (2012). For philosophical discussions, see Blasimme (2013).

  7. See for instance the debate of melanoma-initiating cells: Schatton et al. (2008), Quintana et al. (2008). Both examples are discussed in more detail in Germain (2014).

  8. The information on the drug screening aspects of this zebrafish model comes from the few months I could spent under precious tutoring provided by Cristina Santoriello, at the IFOM-IEO Campus, Milano. The results of the screen have not yet been published, but very similar screens have been published on other mutant larval phenotypes, and they are reviewed in White et al. (2013).

  9. “The experimental conditions ‘contain’ the scientific objects in the double sense of this expression: they embed them, and through that very embracement, they restrict and constrain them.” (Rheinberger 1997, p. 29).

References

  • Ankeny, R. A., & Leonelli, S. (2011). What’s so special about model organisms? Studies In History and Philosophy of Science Part A, 42(2), 313–323.

    Article  Google Scholar 

  • Blasimme, A., Maugeri, P., & Germain, P.-L. (2013). What mechanisms can’t do: Explanatory frameworks and the function of the p53 gene in molecular oncology. Studies in History and Philosophy of Biological and Biomedical Sciences, 44(3), 374–384.

    Article  Google Scholar 

  • Bolker, J. A. (2009). Exemplary and surrogate models: Two modes of representation in biology. Perspectives in Biology and Medicine, 52(4), 485–499. doi:10.1353/pbm.0.0125.

  • Committee on Models for Biomedical Research. (1985). Models for Biomedical Research: A New Perspective. Washington: National Academy Press.

  • Committee on New and Emerging Models in Biomedical and Behavioral Research, Institute for Laboratory Animal Research. (1998). Biomedical models and resources: Current needs and future opportunities. Washington: National Academy Press.

  • Gaudillière, J.-P. (2006). “Produire et utiliser les souris inbred: complexe biomédical, cancer et obésité aux États-Unis d’Amérique après 1945”, dans Les Organismes Modèles dans la Recherche Médicale, sous la direction de Gachelin G., Presses Universitaires de France, pp. 163–180.

  • Gayon, J. (2006). “Les organismes modèles en biologie et en médecine”, dans Les Organismes Modèles dans la Recherche Médicale, sous la direction de Gachelin G., Presses Universitaires de France, pp. 9–44.

  • Germain, P.-L. (2014). Living instruments and theoretical terms. In M. C. Galavotti, S. Hartmann, M. Weber, W. Gonzalez, D. Dieks, & T. Uebel (Eds.), New Directions in the Philosophy of Science. Berlin: Springer.

  • Goodman, N. (1968). Languages of art: An approach to a theory of symbols (2nd ed.). Indianapolis, IN: Bobbs-Merrill.

    Google Scholar 

  • Greene, H. S. N. (1948). Identification of malignant tissues. JAMA, the Journal of the American Medical Association, 137(16), 1364–1366.

    Article  Google Scholar 

  • Keller, E. F. (2000). Models of and models for: Theory and practice in contemporary biology. Philosophy of Science, 67, S72–S86.

    Article  Google Scholar 

  • Keller, E. F. (2002). Making sense of life: explaining biological development with models, metaphors, and machines. Cambridge, MA: Harvard University Press.

    Google Scholar 

  • Keuck, L. K. (2012). Relevant similarity in the light of biomedical experimentation. In K. Hagen, F. Schnieke, Angelika, & Thiele (Eds.), Large animals as biomedical models: Ethical, societal, legal and biological aspects (pp. 69–83). Bad Neuenahr-Ahrweiler: Europäische Akademie.

    Google Scholar 

  • Knight, A. (2011) The costs and benefits of animal experiments. Basingstoke: Palgrave Macmillan.

  • Kraus, M. H., Yuasa, Y., & Aaronson, S. A. (1984). A position 12-activated H-ras oncogene in all HS578T mammary carcinosarcoma cells but not normal mammary cells of the same patient. Proceedings of the National Academy Sciences, 81(17), 5384–5388.

    Article  Google Scholar 

  • LaFollette, H., & Shanks, N. (1996). Brute science. Dilemmas of animal experimentation. London: Routledge.

    Google Scholar 

  • Landecker, H. (2007). Culturing life: How cells became technologies. Cambridge: Harvard University Press.

    Book  Google Scholar 

  • Maugeri, P., & Blasimme, A. (2011). Humanised models of cancer in molecular medicine: The experimental control of disanalogy. History and Philosophy of the Life Sciences, 33, 603–622.

    Google Scholar 

  • Meunier, R. (2011). Thick and thin characters: Organismal form and representational practice in embryology and genetics. Ph.D. thesis, Università degli Studi di Milano.

  • Meunier, R. (2012). Stages in the development of a model organism as a platform for mechanistic models in developmental biology: Zebrafish, 1970–2000. Studies in History and Philosophy of Biological and Biomedical Sciences, 43(2), 522–531.

    Article  Google Scholar 

  • Morange, M. (1993). The discovery of cellular oncogenes. History and Philosophy of the Life Sciences, 13(1), 45–58.

    Google Scholar 

  • Olszynko-Gryn, J. (2013). The demand for pregnancy testing: The Aschheim–Zondek reaction, diagnostic versatility, and laboratory services in 1930s Britain. Studies in History and Philosophy of Biological and Biomedical Sciences. doi:10.1016/j.shpsc.2013.12.002.

  • Piotrowska, M. (2012). From humanized mice to human disease: Guiding extrapolation from model to target. Biology and Philosophy, 28(3), 439–455.

    Article  Google Scholar 

  • Quintana, E., Shackleton, M., Sabel, M. S., Fullen, D. R., Johnson, T. M., & Morrison, S. J. (2008). Efficient tumour formation by single human melanoma cells. Nature, 456(7222), 593–598.

    Article  Google Scholar 

  • Rader, K. (2004). Making mice: Standardizing animals for American biomedical research, 1900–1955. Princeton: Princeton University Press.

    Google Scholar 

  • Rheinberger, H.-J. (1997). Toward a history of epistemic things: Synthesizing proteins in the test tube. Stanford: Stanford University Press.

  • Santoriello, C., Deflorian, G., Pezzimenti, F., Kawakami, K., Lanfrancone, L., d’Adda di Fagagna, F., et al. (2009). Expression of H-RASV12 in a zebrafish model of Costello syndrome causes cellular senescence in adult proliferating cells. Disease Models & Mechanisms, 2(1–2), 56–67.

    Article  Google Scholar 

  • Santoriello, C., Gennaro, E., Anelli, V., Distel, M., Kelly, A., Köster, R. W., Hurlstone, A., Mione, M. (2010). Kita driven expression of oncogenic HRAS leads to early onset and highly penetrant melanoma in zebrafish. PloS One, 5(12), e15170.

  • Schatton, T., Murphy, G. F., Frank, N. Y., Yamaura, K., Waaga-Gasser, A. M., Gasser, M., et al. (2008). Identification of cells initiating human melanomas. Nature, 451(7176), 345–349.

    Article  Google Scholar 

  • Shanks, N., Greek, R., & Greek, J. (2009). Are animal models predictive for humans? Philosophy, Ethics, and Humanities in Medicine, 4, 2.

    Article  Google Scholar 

  • Shelley, C. (2010). Why test animals to treat humans? On the validity of animal models. Studies in History and Philosophy of Biological and Biomedical Sciences, 41(3), 292–299.

    Article  Google Scholar 

  • Steel, D. P. (2008). Across the boundaries: Extrapolation in biology and social science. New York: Cambridge University Press.

    Google Scholar 

  • Suarez, M. (2004). An inferential conception of scientific representation. Philosophy of Science, 71(5), 767–779.

    Article  Google Scholar 

  • Valent, P., Eaves, C., Bonnet, D., De Maria, R., Lapidot, T., Copland, M., et al. (2012). Cancer stem cell definitions and terminology: The devil is in the details. Nature Reviews Cancer, 12(11), 767–775.

    Article  Google Scholar 

  • Visvader, J., & Lindeman, G. (2012). Cancer stem cells: Current status and evolving complexities. Cell Stem Cell, 10(6), 717–728.

    Article  Google Scholar 

  • Weber, M. (2005). The philosophy of experimental biology. Cambridge, MA: Cambridge University Press.

    Google Scholar 

  • Weisberg, M. (2007). Who is a modeler? The British Journal for the Philosophy of Science, 58(2), 207–233.

    Article  Google Scholar 

  • White, R., Rose, K., & Zon, L. (2013). Zebrafish cancer: The state of the art and the path forward. Nature Reviews Cancer, 13(9), 624–636.

    Article  Google Scholar 

  • Zondek, B. (1928). Die Schwangerschaftsdiagnose aus dem Harn durch Nachweis des Hypophysenvorderlappenhormons. Die Naturwissenschaften, 51, 1088–1090.

    Article  Google Scholar 

Download references

Acknowledgments

In addition to the participants of the Second European Advanced Seminar in the Philosophy of the Life Sciences (EASPLS 2012), whose contributions are partly found in the present issue, I would like to thank the participants of the workshop “Animal Models, Model Animals? Meanings and Practices in the Biomedical Sciences” (Centre for the History of Science, Technology and Medicine, of the University of Manchester, 2012) at which this paper was also presented. I also wish to thank Cristina Santoriello for her precious tutoring, and Marina Mione for welcoming me in her lab. Finally, I am grateful to Maël Lemoine, Jean Gayon, Giuseppe Testa and my FOLSATEC colleagues for interesting discussions on these topics.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Pierre-Luc Germain.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Germain, PL. From replica to instruments: animal models in biomedical research. HPLS 36, 114–128 (2014). https://doi.org/10.1007/s40656-014-0007-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40656-014-0007-0

Keywords

Navigation