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.

  • Review Article
  • Published:

Towards quantitative and multiplexed in vivo functional cancer genomics

A Correction to this article was published on 16 October 2018

This article has been updated

Abstract

Large-scale sequencing of human tumours has uncovered a vast array of genomic alterations. Genetically engineered mouse models recapitulate many features of human cancer and have been instrumental in assigning biological meaning to specific cancer-associated alterations. However, their time, cost and labour-intensive nature limits their broad utility; thus, the functional importance of the majority of genomic aberrations in cancer remains unknown. Recent advances have accelerated the functional interrogation of cancer-associated alterations within in vivo models. Specifically, the past few years have seen the emergence of CRISPR–Cas9-based strategies to rapidly generate increasingly complex somatic alterations and the development of multiplexed and quantitative approaches to ascertain gene function in vivo.

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

Access options

Buy this article

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

Fig. 1: Diversity and complexity of somatic mutations in human cancer genomes.
Fig. 2: Autochthonous mouse models of cancer recapitulate complex and multifaceted genetic and microenvironmental inputs that affect tumorigenesis.
Fig. 3: Strategies to model somatic cancer-associated alterations in mice.
Fig. 4: Increasing the throughput and quantitative nature of functional genomics in vivo through multiplexed approaches.
Fig. 5: Integration of genome engineering with DNA barcoding enables barcode sequencing-based quantitative analysis of tumour suppression and oncogenicity.
Fig. 6: Generating combinatorial alterations in vivo.
Fig. 7: The next frontier: accurately reconstructing and characterizing all aspects of carcinogenesis.

Similar content being viewed by others

Change history

  • 16 October 2018

    The originally published article failed to acknowledge the equal first authorship contribution of I. P. Winters and C. W. Murray. The article has now been corrected online. The editors apologize for this error.

References

  1. Forbes, S. A. et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 45, D777–D783 (2017).

    CAS  PubMed  Google Scholar 

  2. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    PubMed  Google Scholar 

  3. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    PubMed  PubMed Central  Google Scholar 

  4. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Stratton, M. R., Campbell, P. J. & Futreal, P. A. The cancer genome. Nature 458, 719–724 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Watson, I. R., Takahashi, K., Futreal, P. A. & Chin, L. Emerging patterns of somatic mutations in cancer. Nat. Rev. Genet. 14, 703 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Yi, S. et al. Functional variomics and network perturbation: connecting genotype to phenotype in cancer. Nat. Rev. Genet. 18, 395 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gordon, D. J., Resio, B. & Pellman, D. Causes and consequences of aneuploidy in cancer. Nat. Rev. Genet. 13, 189 (2012).

    CAS  PubMed  Google Scholar 

  9. Hyman, D. M., Taylor, B. S. & Baselga, J. Implementing genome-driven oncology. Cell 168, 584–599 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Berger, M. F. & Mardis, E. R. The emerging clinical relevance of genomics in cancer medicine. Nat. Rev. Clin. Oncol. 15, 353–365 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Greenman, C. et al. Patterns of somatic mutation in human cancer genomes. Nature 446, 153–158 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Rogers, Z. N. et al. Mapping the in vivo fitness landscape of lung adenocarcinoma tumor suppression in mice. Nat. Genet. 50, 483–486 (2018). This article presents a multiplexed, quantitative analysis of tumour suppressor function in the context of pairwise combinations of tumour suppressor alterations in vivo.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Mina, M. et al. Conditional selection of genomic alterations dictates cancer evolution and oncogenic dependencies. Cancer Cell 32, 155–168 (2017).

    CAS  PubMed  Google Scholar 

  14. Schneider, G., Schmidt-Supprian, M., Rad, R. & Saur, D. Tissue-specific tumorigenesis: context matters. Nat. Rev. Cancer 17, 239 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    CAS  PubMed  Google Scholar 

  16. Lyssiotis, C. A. & Kimmelman, A. C. Metabolic interactions in the tumor microenvironment. Trends Cell Biol. 27, 863–875 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. McAllister, S. S. & Weinberg, R. A. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat. Cell Biol. 16, 717–727 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Quail, D. F. & Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Tabassum, D. P. & Polyak, K. Tumorigenesis: it takes a village. Nat. Rev. Cancer 15, 473–483 (2015).

    CAS  PubMed  Google Scholar 

  20. Northey, J. J., Przybyla, L. & Weaver, V. M. Tissue force programs cell fate and tumor aggression. Cancer Discov. 7, 1224 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Gengenbacher, N., Singhal, M. & Augustin, H. G. Preclinical mouse solid tumour models: status quo, challenges and perspectives. Nat. Rev. Cancer 17, 751 (2017).

    CAS  PubMed  Google Scholar 

  22. Frese, K. K. & Tuveson, D. A. Maximizing mouse cancer models. Nat. Rev. Cancer 7, 645–658 (2007).

    CAS  PubMed  Google Scholar 

  23. Kersten, K., de Visser, K. E., van Miltenburg, M. H. & Jonkers, J. Genetically engineered mouse models in oncology research and cancer medicine. EMBO Mol. Med. 9, 137 (2017).

    CAS  PubMed  Google Scholar 

  24. Heyer, J., Kwong, L. N., Lowe, S. W. & Chin, L. Non-germline genetically engineered mouse models for translational cancer research. Nat. Rev. Cancer 10, 470 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Chiou, S.-H. et al. Pancreatic cancer modeling using retrograde viral vector delivery and in vivo CRISPR/Cas9-mediated somatic genome editing. Genes Dev. 29, 1576–1585 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Dow, L. E. et al. Inducible in vivo genome editing with CRISPR-Cas9. Nat. Biotechnol. 33, 390–394 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Platt, R. J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014). References 25–27 represent the first reports of genome editing in vivo using genetically engineered mice harbouring germline-encoded Cas9 alleles.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Chow, R. D. et al. AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma. Nat. Neurosci. 20, 1329 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Maresch, R. et al. Multiplexed pancreatic genome engineering and cancer induction by transfection-based CRISPR/Cas9 delivery in mice. Nat. Commun. 7, 10770 (2016). This study first demonstrated the use of the CRISPR–Cas9 platform in a high-complexity, multiplexed format within an autochthonous mouse model.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Rogers, Z. N. et al. A quantitative and multiplexed approach to uncover the fitness landscape of tumor suppression in vivo. Nat. Methods 14, 737–742 (2017). This article presents an initial high-resolution, multiplexed survey of tumour suppressor function within genetically engineered mice via integration of genome editing, tumour barcoding and high-throughput sequencing.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, G. et al. Mapping a functional cancer genome atlas of tumor suppressors in mouse liver using AAV-CRISPR–mediated direct in vivo screening. Sci. Adv. 4, eaao5508 (2018).

    PubMed  PubMed Central  Google Scholar 

  32. Weber, J. et al. CRISPR/Cas9 somatic multiplex-mutagenesis for high-throughput functional cancer genomics in mice. Proc. Natl Acad. Sci. USA 112, 13982–13987 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Winters, I. P. et al. Multiplexed in vivo homology-directed repair and tumor barcoding enables parallel quantification of Kras variant oncogenicity. Nat. Commun. 8, 2053 (2017). This study is a quantitative analysis of oncogenicity across a series of common variants in Kras introduced via HDR within multiple tissues of genetically engineered mice.

    PubMed  PubMed Central  Google Scholar 

  34. Xu, C. et al. piggyBac mediates efficient in vivo CRISPR library screening for tumorigenesis in mice. Proc. Natl Acad. Sci. USA 114, 722 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Zuckermann, M. et al. Somatic CRISPR/Cas9-mediated tumour suppressor disruption enables versatile brain tumour modelling. Nat. Commun. 6, 7391 (2015).

    CAS  PubMed  Google Scholar 

  36. Balani, S., Nguyen, L. V. & Eaves, C. J. Modeling the process of human tumorigenesis. Nat. Commun. 8, 15422 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Drost, J. & Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 18, 407–418 (2018).

    CAS  PubMed  Google Scholar 

  38. Mohr, S. E., Smith, J. A., Shamu, C. E., Neumüller, R. A. & Perrimon, N. RNAi screening comes of age: improved techniques and complementary approaches. Nat. Rev. Mol. Cell Biol. 15, 591 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Shalem, O., Sanjana, N. E. & Zhang, F. High-throughput functional genomics using CRISPR–Cas9. Nat. Rev. Genet. 16, 299 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Johannessen, C. M. & Boehm, J. S. Progress towards precision functional genomics in cancer. Curr. Opin. Syst. Biol. 2, 74–83 (2017).

    Google Scholar 

  41. Frampton, G. M. et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 5, 859–859 (2015).

    Google Scholar 

  42. Khurana, E. et al. Role of non-coding sequence variants in cancer. Nat. Rev. Genet. 17, 93 (2016).

    CAS  PubMed  Google Scholar 

  43. Bunting, S. F. & Nussenzweig, A. End-joining, translocations and cancer. Nat. Rev. Cancer 13, 443 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. McGranahan, N. & Swanton, C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell 168, 613–628 (2017).

    CAS  PubMed  Google Scholar 

  45. Chalmers, Z. R. et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 9, 34 (2017).

    PubMed  PubMed Central  Google Scholar 

  46. Miller, M. L. et al. Pan-cancer analysis of mutation hotspots in protein domains. Cell Syst. 1, 197–209 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Chen, W., Li, Y. & Wang, Z. Evolution of oncogenic signatures of mutation hotspots in tyrosine kinases supports the atavistic hypothesis of cancer. Sci. Rep. 8, 8256 (2018).

    PubMed  PubMed Central  Google Scholar 

  48. Baeissa, H., Benstead-Hume, G., Richardson, C. J. & Pearl, F. M. G. Identification and analysis of mutational hotspots in oncogenes and tumour suppressors. Oncotarget 8, 21290–21304 (2017).

    PubMed  PubMed Central  Google Scholar 

  49. Zack, T. I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Esteller, M. et al. Epigenetic inactivation of LKB1 in primary tumors associated with the Peutz-Jeghers syndrome. Oncogene 19, 164 (2000).

    CAS  PubMed  Google Scholar 

  51. Herman, J. G. et al. Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc. Natl Acad. Sci. USA 91, 9700 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Beroukhim, R. et al. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899–905 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumor types. Nature 505, 495–501 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Bailey, M. H. et al. Comprehensive characterization of cancer driver genes and mutations. Cell 173, 371–385 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Weir, B. A. et al. Characterizing the cancer genome in lung adenocarcinoma. Nature 450, 893–898 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Hollstein, M., Alexandrov, L. B., Wild, C. P., Ardin, M. & Zavadil, J. Base changes in tumour DNA have the power to reveal the causes and evolution of cancer. Oncogene 36, 158 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  59. Stewart, T. A., Pattengale, P. K. & Leder, P. Spontaneous mammary adenocarcinomas in transgenic mice that carry and express MTV/myc fusion genes. Cell 38, 627–637 (1984).

    CAS  PubMed  Google Scholar 

  60. Quaife, C. J., Pinkert, C. A., Ornitz, D. M., Palmiter, R. D. & Brinster, R. L. Pancreatic neoplasia induced by ras expression in acinar cells of transgenic mice. Cell 48, 1023–1034 (1987).

    CAS  PubMed  Google Scholar 

  61. Andres, A. C. et al. Ha-ras oncogene expression directed by a milk protein gene promoter: tissue specificity, hormonal regulation, and tumor induction in transgenic mice. Proc. Natl Acad. Sci. USA 84, 1299 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Muller, W. J., Sinn, E., Pattengale, P. K., Wallace, R. & Leder, P. Single-step induction of mammary adenocarcinoma in transgenic mice bearing the activated c-neu oncogene. Cell 54, 105–115 (1988).

    CAS  PubMed  Google Scholar 

  63. Rüther, U., Komitowski, D., Schubert, F. R. & Wagner, E. F. c-Fos expression induces bone tumors in transgenic mice. Oncogene 4, 861–865 (1989).

    PubMed  Google Scholar 

  64. Tsukamoto, A. S., Grosschedl, R., Guzman, R. C., Parslow, T. & Varmus, H. E. Expression of the int-1 gene in transgenic mice is associated with mammary gland hyperplasia and adenocarcinomas in male and female mice. Cell 55, 619–625 (1988).

    CAS  PubMed  Google Scholar 

  65. Capecchi, M. R. The new mouse genetics: altering the genome by gene targeting. Trends Genet. 5, 70–76 (1989).

    CAS  PubMed  Google Scholar 

  66. Donehower, L. A. et al. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 356, 215 (1992).

    CAS  PubMed  Google Scholar 

  67. Jacks, T. et al. Effects of an Rb mutation in the mouse. Nature 359, 295 (1992).

    CAS  PubMed  Google Scholar 

  68. Jacks, T. et al. Tumor spectrum analysis in p53-mutant mice. Curr. Biol. 4, 1–7 (1994).

    CAS  PubMed  Google Scholar 

  69. Purdie, C. A. et al. Tumour incidence, spectrum and ploidy in mice with a large deletion in the p53 gene. Oncogene 9, 603–609 (1994).

    CAS  PubMed  Google Scholar 

  70. Dankort, D. et al. A new mouse model to explore the initiation, progression, and therapy of BRAF(V600E)-induced lung tumors. Genes Dev. 21, 379–384 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Gu, H., Marth, J. D., Orban, P. C., Mossmann, H. & Rajewsky, K. Deletion of a DNA polymerase beta gene segment in T cells using cell type-specific gene targeting. Science 265, 103 (1994).

    CAS  PubMed  Google Scholar 

  72. Jackson, E. L. et al. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev. 15, 3243–3248 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Jonkers, J. et al. Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat. Genet. 29, 418–425 (2001).

    CAS  PubMed  Google Scholar 

  74. Zhu, Y. et al. Early inactivation of p53 tumor suppressor gene cooperating with NF1 loss induces malignant astrocytoma. Cancer Cell 8, 119–130 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Chin, L. et al. Essential role for oncogenic Ras in tumour maintenance. Nature 400, 468 (1999).

    CAS  PubMed  Google Scholar 

  76. Furth, P. A. et al. Temporal control of gene expression in transgenic mice by a tetracycline-responsive promoter. Proc. Natl Acad. Sci. USA 91, 9302 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Moody, S. E. et al. Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis. Cancer Cell 2, 451–461 (2002).

    CAS  PubMed  Google Scholar 

  78. Premsrirut, P. K. et al. A rapid and scalable system for studying gene function in mice using conditional rna interference. Cell 145, 145–158 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Shockett, P., Difilippantonio, M., Hellman, N. & Schatz, D. G. A modified tetracycline-regulated system provides autoregulatory, inducible gene expression in cultured cells and transgenic mice. Proc. Natl Acad. Sci. USA 92, 6522 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Vooijs, M., Jonkers, J. & Berns, A. A highly efficient ligand-regulated Cre recombinase mouse line shows that LoxP recombination is position dependent. EMBO Rep. 2, 292–297 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Sánchez-Rivera, F. J. & Jacks, T. Applications of the CRISPR–Cas9 system in cancer biology. Nat. Rev. Cancer 15, 387 (2015).

    PubMed  PubMed Central  Google Scholar 

  82. Ventura, A. & Dow, L. E. Modeling cancer in the CRISPR era. Annu. Rev. Cancer Biol. 2, 111–131 (2018).

    Google Scholar 

  83. Annunziato, S. et al. Modeling invasive lobular breast carcinoma by CRISPR/Cas9-mediated somatic genome editing of the mammary gland. Genes Dev. 30, 1470–1480 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Wang, D. et al. Adenovirus-mediated somatic genome editing of Pten by CRISPR/Cas9 in mouse liver in spite of Cas9-specific immune responses. Hum. Gene Ther. 26, 432–442 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Xue, W. et al. CRISPR-mediated direct mutation of cancer genes in the mouse liver. Nature 514, 380–384 (2014). This research first demonstrated the feasibility of conducting CRISPR–Cas9-mediated genome editing in vivo.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Roper, J. et al. In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis. Nat. Biotechnol. 35, 569 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Wu, Q. et al. In vivo CRISPR screening unveils histone demethylase UTX as an important epigenetic regulator in lung tumorigenesis. Proc. Natl Acad. Sci. USA 115, E3978–E3986 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Huang, J. et al. Generation and comparison of CRISPR-Cas9 and Cre-mediated genetically engineered mouse models of sarcoma. Nat. Commun. 8, 15999 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Walter, D. M. et al. Systematic in vivo inactivation of chromatin regulating enzymes identifies Setd2 as a potent tumor suppressor in lung adenocarcinoma. Cancer Res. 77, 1719–1729 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Mikuni, T., Nishiyama, J., Sun, Y., Kamasawa, N. & Yasuda, R. High-throughput, high-resolution mapping of protein localization in mammalian brain by in vivo genome editing. Cell 165, 1803–1817 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Chung, W.-J. et al. Kras mutant genetically engineered mouse models of human cancers are genomically heterogeneous. Proc. Natl Acad. Sci. USA 114, E10947–E10955 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Hsu, P. D., Lander, E. S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Adams, J. M. et al. The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice. Nature 318, 533 (1985).

    CAS  PubMed  Google Scholar 

  99. Heisterkamp, N. et al. Acute leukaemia in bcr/abl transgenic mice. Nature 344, 251 (1990).

    CAS  PubMed  Google Scholar 

  100. Maddalo, D. et al. In vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 system. Nature 516, 423 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Buchholz, F., Refaeli, Y., Trumpp, A. & Bishop, J. M. Inducible chromosomal translocation of AML1 and ETO genes through Cre/loxP-mediated recombination in the mouse. EMBO Rep. 1, 133–139 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Collins, E. C., Pannell, R., Simpson, E. M., Forster, A. & Rabbitts, T. H. Inter-chromosomal recombination of Mll and Af9 genes mediated by cre-loxP in mouse development. EMBO Rep. 1, 127–132 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Yu, Y. & Bradley, A. Engineering chromosomal rearrangements in mice. Nat. Rev. Genet. 2, 780–790 (2001).

    CAS  PubMed  Google Scholar 

  104. Forster, A. et al. Engineering de novo reciprocal chromosomal translocations associated with Mll to replicate primary events of human cancer. Cancer Cell 3, 449–458 (2003).

    CAS  PubMed  Google Scholar 

  105. Lobato, M. N. et al. Modeling chromosomal translocations using conditional alleles to recapitulate initiating events in human leukemias. J. Natl Cancer Inst. Monogr. 2008, 58–63 (2008).

    Google Scholar 

  106. Blasco, R. B. et al. Simple and rapid in vivo generation of chromosomal rearrangements using CRISPR/Cas9 technology. Cell Rep. 9, 1219–1227 (2014).

    CAS  PubMed  Google Scholar 

  107. Cook, P. J. et al. Somatic chromosomal engineering identifies BCAN-NTRK1 as a potent glioma driver and therapeutic target. Nat. Commun. 8, 15987 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Lagutina, I. V. et al. Modeling of the human alveolar rhabdomyosarcoma Pax3-Foxo1 chromosome translocation in mouse myoblasts using CRISPR-Cas9 nuclease. PLOS Genet. 11, e1004951 (2015).

    PubMed  PubMed Central  Google Scholar 

  109. Han, T. et al. R-Spondin chromosome rearrangements drive Wnt-dependent tumour initiation and maintenance in the intestine. Nat. Commun. 8, 15945 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Li, Y. et al. A versatile reporter system for CRISPR-mediated chromosomal rearrangements. Genome Biol. 16, 111 (2015).

    PubMed  PubMed Central  Google Scholar 

  111. Grimm, S. The art and design of genetic screens: mammalian culture cells. Nat. Rev. Genet. 5, 179 (2004).

    CAS  PubMed  Google Scholar 

  112. Boehm, J. S. & Golub, T. R. An ecosystem of cancer cell line factories to support a cancer dependency map. Nat. Rev. Genet. 16, 373 (2015).

    CAS  PubMed  Google Scholar 

  113. Gillet, J. P., Varma, S. & Gottesman, M. M. The clinical relevance of cancer cell lines. J. Natl Cancer Inst. 105, 452–458 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Bric, A. et al. Functional identification of tumor suppressor genes through an in vivo RNA interference screen in a mouse lymphoma model. Cancer Cell 16, 324–335 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Meacham, C. E., Ho, E. E., Dubrovsky, E., Gertler, F. B. & Hemann, M. T. In vivo RNAi screening identifies regulators of actin dynamics as key determinants of lymphoma progression. Nat. Genet. 41, 1133 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Zender, L. et al. An oncogenomics-based in vivo RNAi screen identifies tumor suppressors in liver cancer. Cell 135, 852–864 (2008). This paper presents one of the first demonstrations of a multiplexed genetic screen in vivo through delivery of pooled genetic perturbations ex vivo and subsequent orthotopic transplant.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Xue, W. et al. A cluster of cooperating tumor-suppressor gene candidates in chromosomal deletions. Proc. Natl Acad. Sci. USA 109, 8212 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Malina, A. et al. Repurposing CRISPR/Cas9 for in situ functional assays. Genes Dev. 27, 2602–2614 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. McFadden, D. G. et al. Mutational landscape of EGFR-, MYC-, and Kras-driven genetically engineered mouse models of lung adenocarcinoma. Proc. Natl Acad. Sci. USA 113, 6409–6417 (2016).

    Google Scholar 

  120. Beronja, S. et al. RNAi screens in mice identify physiological regulators of oncogenic growth. Nature 501, 185–190 (2013). This article presents an initial demonstration of the feasibility of multiplexed, high-complexity genetic screens within autochthonous models.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Schramek, D. et al. Direct in vivo RNAi screen unveils myosin IIa as a tumor suppressor of squamous cell carcinomas. Science 343, 309–313 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Rudalska, R. et al. In vivo RNAi screening identifies a mechanism of sorafenib resistance in liver cancer. Nat. Med. 20, 1138 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Boutros, M. & Ahringer, J. The art and design of genetic screens: RNA interference. Nat. Rev. Genet. 9, 554 (2008).

    CAS  PubMed  Google Scholar 

  124. Birmingham, A. et al. 3’ UTR seed matches, but not overall identity, are associated with RNAi off-targets. Nat. Methods 3, 199 (2006).

    CAS  PubMed  Google Scholar 

  125. Jackson, A. L. et al. Expression profiling reveals off-target gene regulation by RNAi. Nat. Biotechnol. 21, 635 (2003).

    CAS  PubMed  Google Scholar 

  126. Jackson, A. L. & Linsley, P. S. Recognizing and avoiding siRNA off-target effects for target identification and therapeutic application. Nat. Rev. Drug Discov. 9, 57 (2010).

    CAS  PubMed  Google Scholar 

  127. Adamson, B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Michlits, G. et al. CRISPR-UMI: single-cell lineage tracing of pooled CRISPR–Cas9 screens. Nat. Methods 14, 1191–1197 (2017).

    CAS  PubMed  Google Scholar 

  129. Schmierer, B. et al. CRISPR/Cas9 screening using unique molecular identifiers. Mol. Syst. Biol. 13, 945 (2017).References 128 and 129 are early demonstrations of the utility of diversifying sgRNAs with DNA barcodes to enable analyses of individual clones within pooled formats, thereby increasing sensitivity in genetic screens.

    PubMed  PubMed Central  Google Scholar 

  130. Macaulay, I. C., Ponting, C. P. & Voet, T. Single-cell multiomics: multiple measurements from single cells. Trends Genet. 33, 155–168 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Gawad, C., Koh, W. & Quake, S. R. Single-cell genome sequencing: current state of the science. Nat. Rev. Genet. 17, 175 (2016).

    CAS  PubMed  Google Scholar 

  132. Zhang, X. et al. Single-cell sequencing for precise cancer research: progress and prospects. Cancer Res. 76, 1305 (2016).

    CAS  PubMed  Google Scholar 

  133. Tanay, A. & Regev, A. Scaling single-cell genomics from phenomenology to mechanism. Nature 541, 331 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Heath, J. R., Ribas, A. & Mischel, P. S. Single-cell analysis tools for drug discovery and development. Nat. Rev. Drug Discov. 15, 204 (2015).

    PubMed  PubMed Central  Google Scholar 

  135. Han, K. et al. Synergistic drug combinations for cancer identified in a CRISPR screen for pairwise genetic interactions. Nat. Biotechnol. 35, 463 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Najm, F. J. et al. Orthologous CRISPR–Cas9 enzymes for combinatorial genetic screens. Nat. Biotechnol. 36, 179 (2017).

    PubMed  PubMed Central  Google Scholar 

  137. Wong, A. S. L. et al. Multiplexed barcoded CRISPR-Cas9 screening enabled by CombiGEM. Proc. Natl Acad. Sci. USA 113, 2544 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Dominguez, A. A., Lim, W. A. & Qi, L. S. Beyond editing: repurposing CRISPR–Cas9 for precision genome regulation and interrogation. Nat. Rev. Mol. Cell Biol. 17, 5–15 (2016).

    CAS  PubMed  Google Scholar 

  139. Komor, A. C., Badran, A. H. & Liu, D. R. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell 168, 20–36 (2017).

    CAS  PubMed  Google Scholar 

  140. Gilbert, L. A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Gaudelli, N. M. et al. Programmable base editing of A·T to G·C in genomic DNA without DNA cleavage. Nature 551, 464 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Gerhke, J. M., Cervantes, O. R., Clement, M. K., Pinello, L. & Joung, J. K. An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nat. Biotechnol. https://doi.org/10.1038/nbt.4199 (2018).

  143. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Zafra, M. P. et al. Optimized base editors enable efficient editing in cells, organoids and mice. Nat. Biotechnol. 36, 888–893 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Tai, D. J. C. et al. Engineering microdeletions and microduplications by targeting segmental duplications with CRISPR. Nat. Neurosci. 19, 517 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Maciejowski, J., Li, Y., Bosco, N., Campbell, Peter, J. & de Lange, T. Chromothripsis and kataegis induced by telomere crisis. Cell 163, 1641–1654 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Dagogo-Jack, I. & Shaw, A. T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 15, 81 (2017).

    PubMed  Google Scholar 

  148. Maley, C. C. et al. Classifying the evolutionary and ecological features of neoplasms. Nat. Rev. Cancer 17, 605 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Meacham, C. E. & Morrison, S. J. Tumour heterogeneity and cancer cell plasticity. Nature 501, 328 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Alizadeh, A. A. et al. Toward understanding and exploiting tumor heterogeneity. Nat. Med. 21, 846 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Chen, C. et al. MLL3 is a haploinsufficient 7q tumor suppressor in acute myeloid leukemia. Cancer Cell 25, 652–665 (2014).

    PubMed  PubMed Central  Google Scholar 

  152. Lok, B. H. et al. PARP inhibitor activity correlates with SLFN11 expression and demonstrates synergy with temozolomide in small cell lung cancer. Clin. Cancer Res. 23, 523–535 (2016).

    PubMed  PubMed Central  Google Scholar 

  153. Quick, L. et al. Jak1-STAT3 signals are essential effectors of the USP6/TRE17 oncogene in tumorigenesis. Cancer Res. 76, 5337–5347 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Romero, R. et al. Keap1 loss promotes Kras-driven lung cancer and results in dependence on glutaminolysis. Nat. Med. 23, 1362 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Hong, A. L. et al. Integrated genetic and pharmacologic interrogation of rare cancers. Nat. Commun. 7, 11987 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    CAS  PubMed  Google Scholar 

  157. Shi, J. et al. Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains. Nat. Biotechnol. 33, 661–667 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Woodworth, M. B., Girskis, K. M. & Walsh, C. A. Building a lineage from single cells: genetic techniques for cell lineage tracking. Nat. Rev. Genet. 18, 230 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Caswell, D. R. et al. Obligate progression precedes lung adenocarcinoma dissemination. Cancer Discov. 4, 781 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Livet, J. et al. Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature 450, 56 (2007).

    CAS  PubMed  Google Scholar 

  161. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    CAS  PubMed  Google Scholar 

  162. Dick, J. E., Magli, M. C., Huszar, D., Phillips, R. A. & Bernstein, A. Introduction of a selectable gene into primitive stem cells capable of long-term reconstitution of the hemopoietic system of W/Wv mice. Cell 42, 71–79 (1985).

    CAS  PubMed  Google Scholar 

  163. Keller, G., Paige, C., Gilboa, E. & Wagner, E. F. Expression of a foreign gene in myeloid and lymphoid cells derived from multipotent haematopoietic precursors. Nature 318, 149 (1985).

    CAS  PubMed  Google Scholar 

  164. Lemischka, I. R., Raulet, D. H. & Mulligan, R. C. Developmental potential and dynamic behavior of hematopoietic stem cells. Cell 45, 917–927 (1986).

    CAS  PubMed  Google Scholar 

  165. Gerrits, A. et al. Cellular barcoding tool for clonal analysis in the hematopoietic system. Blood 115, 2610 (2010).

    CAS  PubMed  Google Scholar 

  166. Walsh, C. & Cepko, C. L. Widespread dispersion of neuronal clones across functional regions of the cerebral cortex. Science 255, 434 (1992).

    CAS  PubMed  Google Scholar 

  167. Schepers, K. et al. Dissecting T cell lineage relationships by cellular barcoding. J. Exp. Med. 205, 2309 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. van Heijst, J. W. J. et al. Recruitment of antigen-specific CD8+ T cells in response to infection is markedly efficient. Science 325, 1265 (2009).

    PubMed  Google Scholar 

  169. Chuang, C.-H. et al. Molecular definition of a metastatic lung cancer state reveals a targetable CD109–Janus kinase–Stat axis. Nat. Med. 23, 291 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Bhang, H.-e. C. et al. Studying clonal dynamics in response to cancer therapy using high-complexity barcoding. Nat. Med. 21, 440 (2015).

    CAS  PubMed  Google Scholar 

  171. Grüner, B. M. et al. An in vivo multiplexed small-molecule screening platform. Nat. Methods 13, 883–889 (2016).

    PubMed  PubMed Central  Google Scholar 

  172. Levy, S. F. et al. Quantitative evolutionary dynamics using high-resolution lineage tracking. Nature 519, 181–186 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Lu, R., Neff, N. F., Quake, S. R. & Weissman, I. L. Tracking single hematopoietic stem cells in vivo using high-throughput sequencing in conjunction with viral genetic barcoding. Nat. Biotechnol. 29, 928 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Naik, S. H. et al. Diverse and heritable lineage imprinting of early haematopoietic progenitors. Nature 496, 229 (2013).

    CAS  PubMed  Google Scholar 

  175. Nguyen, L. V. et al. Barcoding reveals complex clonal dynamics of de novo transformed human mammary cells. Nature 528, 267 (2015).

    CAS  PubMed  Google Scholar 

  176. Sun, J. et al. Clonal dynamics of native haematopoiesis. Nature 514, 322 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Venkataram, S. et al. Development of a comprehensive genotype-to-fitness map of adaptation-driving mutations in yeast. Cell 166, 1585–1596 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Frieda, K. L. et al. Synthetic recording and in situ readout of lineage information in single cells. Nature 541, 107 (2016).

    PubMed  PubMed Central  Google Scholar 

  179. McKenna, A. et al. Whole organism lineage tracing by combinatorial and cumulative genome editing. Science 353, aaf7907 (2016).

    PubMed  PubMed Central  Google Scholar 

  180. Pei, W. et al. Polylox barcoding reveals haematopoietic stem cell fates realized in vivo. Nature 548, 456 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Spanjaard, B. et al. Simultaneous lineage tracing and cell-type identification using CRISPR–Cas9-induced genetic scars. Nat. Biotechnol. 36, 469–473 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Kalhor, R., Mali, P. & Church, G. M. Rapidly evolving homing CRISPR barcodes. Nat. Methods 14, 195–200 (2017).

    CAS  PubMed  Google Scholar 

  183. Schmidt, S. T., Zimmerman, S. M., Wang, J., Kim, S. K. & Quake, S. R. Quantitative analysis of synthetic cell lineage tracing using nuclease barcoding. ACS Synth. Biol. 6, 936–942 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Alemany, A., Florescu, M., Baron, C. S., Peterson-Maduro, J. & van Oudenaarden, A. Whole-organism clone tracing using single-cell sequencing. Nature 556, 108 (2018).

    CAS  PubMed  Google Scholar 

  185. Farzadfard, F. & Lu, T. K. Genomically encoded analog memory with precise in vivo DNA writing in living cell populations. Science 346, 1256272 (2014).

    PubMed  PubMed Central  Google Scholar 

  186. Tang, W. & Liu, D. R. Rewritable multi-event analog recording in bacterial and mammalian cells. Science 360, eaap8992 (2018).

    PubMed  PubMed Central  Google Scholar 

  187. Ledford, H. Big science: the cancer genome challenge. Nature 464, 972–974 (2010).

    CAS  PubMed  Google Scholar 

  188. Zhu, Y., Ghosh, P., Charnay, P., Burns, D. K. & Parada, L. F. Neurofibromas in NF1: Schwann cell origin and role of tumor environment. Science 296, 920–922 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Dow, L. E. et al. Apc restoration promotes cellular differentiation and reestablishes crypt homeostasis in colorectal cancer. Cell 161, 1539–1552 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Yates, L. R. & Campbell, P. J. Evolution of the cancer genome. Nat. Rev. Genet. 13, 795–806 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank the vibrant and innovative genome editing, cancer genetics and cancer modelling communities for their commitment to data sharing and collaboration. The authors apologize to those authors whose important work they could not highlight owing to space limitations. The authors thank D. Feldser and all members of the Winslow laboratory for helpful comments. I.P.W. and C.W.M. were supported by the US National Science Foundation Graduate Research Fellowship Program. I.P.W was additionally supported by US National Institutes of Health (NIH) grants F31-CA210627 and T32-HG000044. C.W.M. was additionally supported by an Anne T. and Robert M. Bass Stanford Graduate Fellowship. M.M.W. was supported by NIH grants R01-CA175336 and R01-CA207133. The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of the various funding bodies or Stanford University.

Reviewer information

Nature Reviews Genetics thanks R. Rad and the other, anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

I.P.W. and C.W.M. contributed equally to all aspects of this manuscript. M.M.W. reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Monte M. Winslow.

Ethics declarations

Competing interests

Stanford University has filed a patent on related work on which I.P.W. and M.M.W. are co- inventors. I.P.W. and M.M.W. are co-founders of and hold equity in D2G Oncology, Inc. The authors declare no additional competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Autochthonous mouse models of cancer

Mouse models in which tumours are initiated de novo from somatic cells and progress within the native in vivo environment.

Organoids

3D cultures of a tissue that recapitulate biological features of a specific organ in a more experimentally tractable in vitro setting.

Allografts

Transplants of cells from a donor mouse into a recipient mouse.

Xenografts

Transplants of cells or bulk tissue from one species into a different species; typically refers to the transplant of human cells or tissue into an immunodeficient mouse.

Driver mutations

Mutations that confer a selective advantage to the cell in which they occur and are therefore causally implicated in tumorigenesis.

Proto-oncogenes

Genes with a normal cellular function that promote tumorigenesis when altered.

Tumour suppressor genes

Genes for which the normal function is to restrain or inhibit tumorigenesis and the loss of which promotes tumorigenesis.

Somatic cancer alterations

Genetic changes in a cancer cell relative to the constitutional genome.

Substitutions

Mutations resulting from the replacement of one nucleotide for another.

Insertion and deletions

(Indels). Mutations resulting from an insertion or deletion of one or more — typically <1,000 — nucleotides.

Structural alterations

Alterations that affect the linear architecture or content of the genome, typically spanning a region of DNA at least 1 kb in length.

Silent

A mutation within a coding region that does not change the amino acid sequence of the encoded protein. It is also used to refer to mutations that have no detectable impact on cellular phenotypes.

Missense

A single-nucleotide substitution in a coding region that results in an amino acid substitution in the encoded protein.

Nonsense

A single-nucleotide substitution in a coding region that introduces a stop codon, creating a truncated protein.

Frameshift

An insertion or deletion in a coding region that alters the triplicate open reading frame of the encoded gene, therefore altering the downstream amino acid sequence of the encoded protein.

Copy number variants

Alterations in which there is a gain or loss of genomic material spanning ≥1 kb.

Translocations

Alterations in which genomic material is transferred from one chromosome to another (non-reciprocal) or exchanged between two chromosomes (reciprocal).

Inversions

Structural alterations in which the orientation of a chromosomal region is inverted.

Oncogenic fusion proteins

The products of the in-frame juxtaposition of two distinct coding sequences as a consequence of a structural alteration, such as a translocation or inversion.

Aneuploidy

An abnormal number of chromosomes in a cell, generally not including multiples of chromosomal complements.

Polyploidy

The presence of more than two complete sets of chromosomes in a cell.

Passenger mutations

Mutations that have no functional effect on the selective fitness of the neoplastic cell in which they occur.

Non-synonymous

A single-nucleotide substitution within a coding region that changes an amino acid in the encoded protein.

Hotspot

A term to describe a localized genomic region that frequently incurs mutations in cancer.

Transgenic mice

Mouse models in which an exogenous genetic element is stably engineered into the mouse germ line.

Single guide RNA

(sgRNA). A synthetic chimeric RNA that encodes a desired CRISPR RNA (crRNA) and the trans-activating CRISPR RNA (tracrRNA) of the CRISPR type II system to direct mammalian genome editing.

Germline alleles

Genetic alterations that are heritable.

Synonymous mutations

Single-nucleotide substitutions within a coding region that do not change the amino acid sequence of the encoded protein.

Chromothripsis

A pattern of extensive chromosomal rearrangements and copy number variants typically confined to one or several chromosomes; thought to result from the random repair of a catastrophic chromosome-shattering event.

Chromoplexy

Large chains of structural alterations that affect multiple chromosomes of the cancer genome; most commonly observed in prostate cancer genomes.

Kataegis

A pattern of localized hypermutation that typically coincides with chromothripsis.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Winters, I.P., Murray, C.W. & Winslow, M.M. Towards quantitative and multiplexed in vivo functional cancer genomics. Nat Rev Genet 19, 741–755 (2018). https://doi.org/10.1038/s41576-018-0053-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-018-0053-7

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