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.

  • Perspective
  • Published:

Translational value of mouse models in oncology drug development

Abstract

Much has been written about the advantages and disadvantages of various oncology model systems, with the overall finding that these models lack the predictive power required to translate preclinical efficacy into clinical activity. Despite assertions that some preclinical model systems are superior to others, no single model can suffice to inform preclinical target validation and molecule selection. This perspective provides a balanced albeit critical view of these claims of superiority and outlines a framework for the proper use of existing preclinical models for drug testing and discovery. We also highlight gaps in oncology mouse models and discuss general and pervasive model-independent shortcomings in preclinical oncology work, and we propose ways to address these issues.

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: Preclinical in vivo efficacy models for oncology drug discovery.

Similar content being viewed by others

References

  1. Hay, M., Thomas, D.W., Craighead, J.L., Economides, C. & Rosenthal, J. Clinical development success rates for investigational drugs. Nat. Biotechnol. 32, 40–51 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Rangarajan, A. & Weinberg, R.A. Opinion: Comparative biology of mouse versus human cells: modelling human cancer in mice. Nat. Rev. Cancer 3, 952–959 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Doudna, J.A. & Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Fellmann, C. & Lowe, S.W. Stable RNA interference rules for silencing. Nat. Cell Biol. 16, 10–18 (2013).

    Article  CAS  Google Scholar 

  5. Xue, W. et al. CRISPR-mediated direct mutation of cancer genes in the mouse liver. Nature 514, 380–384 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yin, H. et al. Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat. Biotechnol. 32, 551–553 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sánchez-Rivera, F.J. et al. Rapid modelling of cooperating genetic events in cancer through somatic genome editing. Nature 516, 428–431 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Hidalgo, M. et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discovery 4, 998–1013 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pantelouris, E.M. Absence of thymus in a mouse mutant. Nature 217, 370–371 (1968).

    Article  CAS  PubMed  Google Scholar 

  10. Rygaard, J. & Povlsen, C.O. Heterotransplantation of a human malignant tumour to 'Nude' mice. Acta Pathol. Microbiol. Scand. 77, 758–760 (1969).

    Article  CAS  PubMed  Google Scholar 

  11. Neve, R.M. et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10, 515–527 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Domcke, S., Sinha, R., Levine, D.A., Sander, C. & Schultz, N. Evaluating cell lines as tumour models by comparison of genomic profiles. Nat. Commun. 4, 2126 (2013).

    Article  PubMed  CAS  Google Scholar 

  13. Klein, C.A. Parallel progression of primary tumours and metastases. Nat. Rev. Cancer 9, 302–312 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Junttila, M.R. & de Sauvage, F.J. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature 501, 346–354 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Lodhia, K.A., Hadley, A., Haluska, P. & Scott, C.L. Prioritizing therapeutic targets using patient-derived xenograft models. Biochim. Biophys. Acta 1855, 223–234 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Hidalgo, M. et al. A pilot clinical study of treatment guided by personalized tumorgrafts in patients with advanced cancer. Mol. Cancer Ther. 10, 1311–1316 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. DeRose, Y.S. et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat. Med. 17, 1514–1520 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, S. et al. Endocrine-therapy-resistant ESR1 variants revealed by genomic characterization of breast-cancer-derived xenografts. Cell Rep. 4, 1116–1130 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Eirew, P. et al. Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature 518, 422–426 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Das Thakur, M. & Stuart, D.D. Molecular Pathways: Response and resistance to BRAF and MEK inhibitors in BRAFV600E tumors. Clin. Cancer Res. 20, 1074–1080 (2013).

    Article  PubMed  CAS  Google Scholar 

  21. Julien, S. et al. Characterization of a large panel of patient-derived tumor xenografts representing the clinical heterogeneity of human colorectal cancer. Clin. Cancer Res. 18, 5314–5328 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Lièvre, A. et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 66, 3992–3995 (2006).

    Article  PubMed  Google Scholar 

  23. Hegde, G.V. et al. Blocking NRG1 and other ligand-mediated Her4 signaling enhances the magnitude and duration of the chemotherapeutic response of non-small cell lung cancer. Sci. Transl. Med. 5, 171ra18 (2013).

    Article  PubMed  CAS  Google Scholar 

  24. Martin, E.S. et al. Development of a colon cancer GEMM-derived orthotopic transplant model for drug discovery and validation. Clin. Cancer Res. 19, 2929–2940 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Westcott, P.M.K. et al. The mutational landscapes of genetic and chemical models of Kras-driven lung cancer. Nature 517, 489–492 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Yadav, M. et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 515, 572–576 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Gubin, M.M. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Walrath, J.C., Hawes, J.J., Van Dyke, T. & Reilly, K.M. Chapter 4: Genetically engineered mouse models in cancer research. In Advances in Cancer Research (Elsevier, 2010).

    Google Scholar 

  30. Mestas, J. & Hughes, C.C.W. Of mice and not men: differences between mouse and human immunology. J. Immunol. 172, 2731–2738 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Platzer, B., Stout, M. & Fiebiger, E. Antigen cross-presentation of immune complexes. Front Immunol 5, 140 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Hsieh, C.S., Macatonia, S.E., O'Garra, A. & Murphy, K.M. T cell genetic background determines default T helper phenotype development in vitro. J. Exp. Med. 181, 713–721 (1995).

    Article  CAS  PubMed  Google Scholar 

  33. Ito, R., Takahashi, T., Katano, I. & Ito, M. Current advances in humanized mouse models. Cell. Mol. Immunol. 9, 208–214 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Shultz, L.D., Brehm, M.A., Garcia-Martinez, J.V. & Greiner, D.L. Humanized mice for immune system investigation: progress, promise and challenges. Nat. Rev. Immunol. 12, 786–798 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Vatakis, D.N. et al. Antitumor activity from antigen-specific CD8 T cells generated in vivo from genetically engineered human hematopoietic stem cells. Proc. Natl. Acad. Sci. USA 108, E1408–E1416 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Singh, M., Murriel, C.L. & Johnson, L. Genetically engineered mouse models: closing the gap between preclinical data and trial outcomes. Cancer Res. 72, 2695–2700 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Feldser, D.M. et al. Stage-specific sensitivity to p53 restoration during lung cancer progression. Nature 468, 572–575 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ventura, A. et al. Restoration of p53 function leads to tumour regression in vivo. Nature 445, 661–665 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Martins, C.P., Brown-Swigart, L. & Evan, G.I. Modeling the therapeutic efficacy of p53 restoration in tumors. Cell 127, 1323–1334 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Junttila, M.R. et al. Selective activation of p53-mediated tumour suppression in high-grade tumours. Nature 468, 567–571 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, Y. et al. Chimeric mouse tumor models reveal differences in pathway activation between ERBB family- and KRAS-dependent lung adenocarcinomas. Nat. Biotechnol. 28, 71–78 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Huijbers, I.J. et al. Rapid target gene validation in complex cancer mouse models using re-derived embryonic stem cells. EMBO Mol. Med. 6, 212–225 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tran, C. et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 324, 787–790 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Forbes, S.A. et al. COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res. 43, D805–D811 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Klijn, C. et al. A comprehensive transcriptional portrait of human cancer cell lines. Nat. Biotechnol. 33, 306–312 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Wilson, T.R. et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 487, 505–509 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Utama, F.E. et al. Insensitivity of human prolactin receptors to nonhuman prolactins: relevance for experimental modeling of prolactin receptor-expressing human cells. Endocrinology 150, 1782–1790 (2009).

    Article  CAS  PubMed  Google Scholar 

  49. Singh, M. et al. Assessing therapeutic responses in Kras mutant cancers using genetically engineered mouse models. Nat. Biotechnol. 28, 585–593 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Ott, P.A., Hodi, F.S. & Robert, C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin. Cancer Res. 19, 5300–5309 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Hodi, F.S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Grosso, J.F. & Jure-Kunkel, M.N. CTLA-4 blockade in tumor models: an overview of preclinical and translational research. Cancer Immun. 13, 5 (2013).

    PubMed  PubMed Central  Google Scholar 

  53. Herbst, R.S. et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515, 563–567 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Junttila, T.T. et al. Antitumor efficacy of a bispecific antibody that targets HER2 and activates T cells. Cancer Res. 74, 5561–5571 (2014).

    Article  CAS  PubMed  Google Scholar 

  55. Saxena, M. & Christofori, G. Rebuilding cancer metastasis in the mouse. Mol. Oncol. 7, 283–296 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Francia, G., Cruz-Munoz, W., Man, S., Xu, P. & Kerbel, R.S. Mouse models of advanced spontaneous metastasis for experimental therapeutics. Nat. Rev. Cancer 11, 135–141 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bos, P.D. et al. Genes that mediate breast cancer metastasis to the brain. Nature 459, 1005–1009 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Minn, A.J. et al. Genes that mediate breast cancer metastasis to lung. Nature 436, 518–524 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Enquist, I.B. et al. Lymph node-independent liver metastasis in a model of metastatic colorectal cancer. Nat. Commun. 5, 3530 (2014).

    Article  PubMed  CAS  Google Scholar 

  60. Eisenhauer, E.A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).

    Article  CAS  PubMed  Google Scholar 

  61. Johnson, J.I. et al. Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. Br. J. Cancer 84, 1424–1431 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Voskoglou-Nomikos, T., Pater, J.L. & Seymour, L. Clinical predictive value of the in vitro cell line, human xenograft, and mouse allograft preclinical cancer models. Clin. Cancer Res. 9, 4227–4239 (2003).

    PubMed  Google Scholar 

  63. Wong, H. et al. Antitumor activity of targeted and cytotoxic agents in murine subcutaneous tumor models correlates with clinical response. Clin. Cancer Res. 18, 3846–3855 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Yauch, R. et al. A paracrine requirement for hedgehog signalling in cancer. Nature 455, 406–410 (2008).

    Article  CAS  PubMed  Google Scholar 

  65. Gajjar, A. et al. Phase-I study of vismodegib in children with recurrent or refractory medulloblastoma: a Pediatric Brain Tumor Consortium (PBTC) study. Clin. Cancer Res. doi:10.1158/1078-0432.CCR-13-1425 (27 September 2013).

  66. Rudin, C. et al. Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N. Engl. J. Med. 361, 1173–1178 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wong, H. et al. Pharmacokinetic-pharmacodynamic analysis of vismodegib in preclinical models of mutational and ligand-dependent hedgehog pathway activation. Clin. Cancer Res. 17, 4682–4692 (2011).

    Article  CAS  PubMed  Google Scholar 

  68. Romer, J.T. et al. Suppression of the Shh pathway using a small molecule inhibitor eliminates medulloblastoma in Ptc1(+/−)p53(−/−) mice. Cancer Cell 6, 229–240 (2004).

    Article  CAS  PubMed  Google Scholar 

  69. Berlin, J.D. et al. A randomized phase II trial of vismodegib versus placebo with FOLFOX or FOLFIRI and bevacizumab in patients with previously untreated Metastatic colorectal cancer. Clin. Cancer Res. 19, 258–267 (2012).

    Article  PubMed  CAS  Google Scholar 

  70. Wong, H. et al. Bridging the gap between preclinical and clinical studies using pharmacokinetic-pharmacodynamic modeling: an analysis of GDC-0973, a MEK inhibitor. Clin. Cancer Res. 18, 3090–3099 (2012).

    Article  CAS  PubMed  Google Scholar 

  71. Wong, H. et al. Pharmacodynamics of 2-{4-[(1E)-1-(Hydroxyimino)-2,3-dihydro-1H-inden-5-yl]-3-(pyridine-4-yl)-1H-pyrazol-1-yl}ethan-1-ol (GDC-0879), a potent and selective B-Raf kinase inhibitor: understanding relationships between systemic concentrations, phosphorylated mitogen-activated protein kinase kinase 1 inhibition, and efficacy. J. Pharmacol. Exp. Ther. 329, 360–367 (2009).

    Article  CAS  PubMed  Google Scholar 

  72. Workman, P. et al. Guidelines for the welfare and use of animals in cancer research. Br. J. Cancer 102, 1555–1577 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Perrin, S. Preclinical research: Make mouse studies work. Nature 507, 423–425 (2014).

    Article  PubMed  Google Scholar 

  74. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Bedard, P.L., Hansen, A.R., Ratain, M.J. & Siu, L.L. Tumour heterogeneity in the clinic. Nature 501, 355–364 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Marusyk, A. et al. Non-cell-autonomous driving of tumour growth supports sub-clonal heterogeneity. Nature 514, 54–58 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Couzin-Frankel, J. Hope in a mouse. Science 346, 28–29 (2014).

    Article  CAS  PubMed  Google Scholar 

  78. McFadden, D.G. et al. Genetic and clonal dissection of murine small cell lung carcinoma progression by genome sequencing. Cell 156, 1298–1311 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Gersbach, C.A. Genome engineering: the next genomic revolution. Nat. Methods 11, 1009–1011 (2014).

    Article  CAS  PubMed  Google Scholar 

  80. Platt, R.J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Chen, S. et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell 160, 1246–1260 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Lancaster, M.A. & Knoblich, J.A. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345, 1247125 (2014).

    Article  PubMed  CAS  Google Scholar 

  83. Matano, M. et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat. Med. 21, 256–262 (2015).

    Article  CAS  PubMed  Google Scholar 

  84. Li, X. et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat. Med. 20, 769–777 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Gao, D. et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 159, 176–187 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Sato, T. et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology 141, 1762–1772 (2011).

    Article  CAS  PubMed  Google Scholar 

  87. Boj, S.F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank A. Bruce for her contributions to figure design.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Frederic J de Sauvage.

Ethics declarations

Competing interests

The authors are employees of Genentech, Inc. and own shares of Roche.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gould, S., Junttila, M. & de Sauvage, F. Translational value of mouse models in oncology drug development. Nat Med 21, 431–439 (2015). https://doi.org/10.1038/nm.3853

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3853

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer