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.

  • Article
  • Published:

A prime editor mouse to model a broad spectrum of somatic mutations in vivo

Abstract

Genetically engineered mouse models only capture a small fraction of the genetic lesions that drive human cancer. Current CRISPR–Cas9 models can expand this fraction but are limited by their reliance on error-prone DNA repair. Here we develop a system for in vivo prime editing by encoding a Cre-inducible prime editor in the mouse germline. This model allows rapid, precise engineering of a wide range of mutations in cell lines and organoids derived from primary tissues, including a clinically relevant Kras mutation associated with drug resistance and Trp53 hotspot mutations commonly observed in pancreatic cancer. With this system, we demonstrate somatic prime editing in vivo using lipid nanoparticles, and we model lung and pancreatic cancer through viral delivery of prime editing guide RNAs or orthotopic transplantation of prime-edited organoids. We believe that this approach will accelerate functional studies of cancer-associated mutations and complex genetic combinations that are challenging to construct with traditional models.

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: Quantification of cancer-associated mutations amenable to modeling by base editing or prime editing.
Fig. 2: Design and functional validation of the Rosa26PE2 prime editor allele.
Fig. 3: Ex vivo prime editing and functional testing of Kras and Trp53 mutations.
Fig. 4: PE GEMMs enable autochthonous and orthotopic modeling of lung and pancreatic cancer.

Similar content being viewed by others

Data availability

Rosa26PE2 mice on wild-type (JAX stock JR037953) and Trp53flox/flox (JAX stock JR037954; ref. 98) backgrounds are available from the Jackson Laboratory. Plasmids will be made available through Addgene upon publication. Amplicon sequencing data have been deposited in the SRA repository under accession PRJNA951647 (ref. 99). All other materials and data, including Rosa26PE2 cell and organoid lines, are available from the corresponding author upon reasonable request. Source data are provided in this paper.

Code availability

The pipeline, all related scripts, and intermediate data needed to reproduce our results are available at https://github.com/samgould2/prime-vs-base-editing (2022; ref. 100).

References

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Garraway, L. A. & Lander, E. S. Lessons from the cancer genome. Cell 153, 17–37 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Chang, M. T. et al. Accelerating discovery of functional mutant alleles in cancer. Cancer Discov. 8, 174–183 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Hong, D. S. et al. KRASG12C inhibition with sotorasib in advanced solid tumors. N. Engl. J. Med. 383, 1207–1217 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lynch, T. J. et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 350, 2129–2139 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Hill, W., Caswell, D. R. & Swanton, C. Capturing cancer evolution using genetically engineered mouse models (GEMMs). Trends Cell Biol. 31, 1007–1018 (2021).

    Article  CAS  PubMed  Google Scholar 

  8. 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, 137–153 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Zehir, A. et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 23, 703–713 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. 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 

  11. Annunziato, S. et al. In situ CRISPR–Cas9 base editing for the development of genetically engineered mouse models of breast cancer. EMBO J. 39, e102169 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  15. 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).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  16. Leibowitz, M. L. et al. Chromothripsis as an on-target consequence of CRISPR–Cas9 genome editing. Nat. Genet. 53, 895–905 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Blair, L. M. et al. Oncogenic context shapes the fitness landscape of tumor suppression. Preprint at bioRxiv https://doi.org/10.1101/2022.10.24.511787 (2022).

  18. 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, 420–424 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kurt, I. C. et al. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells. Nat. Biotechnol. 39, 41–46 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Koblan, L. W. et al. Efficient C·G-to-G·C base editors developed using CRISPRi screens, target-library analysis, and machine learning. Nat. Biotechnol. 39, 1414–1425 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lam, D. K. et al. Improved cytosine base editors generated from TadA variants. Nat. Biotechnol. https://doi.org/10.1038/s41587-022-01611-9 (2023).

  22. Tong, H. et al. Programmable A-to-Y base editing by fusing an adenine base editor with an N-methylpurine DNA glycosylase. Nat. Biotechnol. https://doi.org/10.1038/s41587-022-01595-6 (2023).

  23. Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Liu, P. et al. Improved prime editors enable pathogenic allele correction and cancer modelling in adult mice. Nat. Commun. 12, 2121 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gainor, J. F. et al. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer. Cancer Discov. 6, 1118–1133 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gainor, J. F. et al. Patterns of metastatic spread and mechanisms of resistance to crizotinib in ROS1-positive non-small-cell lung cancer. JCO Precis. Oncol. 2017, PO.17.00063 (2017).

  28. Kobayashi, S. et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 352, 786–792 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Chen, P. J. et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 184, 5635–5652 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Canté-Barrett, K. et al. Lentiviral gene transfer into human and murine hematopoietic stem cells: size matters. BMC Res. Notes 9, 312 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Zinn, E. & Vandenberghe, L. H. Adeno-associated virus: fit to serve. Curr. Opin. Virol. 8, 90–97 (2014).

    Article  PubMed  Google Scholar 

  32. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Böck, D. et al. In vivo prime editing of a metabolic liver disease in mice. Sci. Transl. Med. 14, eabl9238 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Sánchez-Rivera, F. J. et al. Base editing sensor libraries for high-throughput engineering and functional analysis of cancer-associated single nucleotide variants. Nat. Biotechnol. 40, 862–873 (2022).

  36. Erwood, S. et al. Saturation variant interpretation using CRISPR prime editing. Nat. Biotechnol. 40, 885–895 (2022).

  37. Chen, L. et al. Programmable C:G to G:C genome editing with CRISPR-Cas9-directed base excision repair proteins. Nat. Commun. 12, 1384 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Grünewald, J. et al. A dual-deaminase CRISPR base editor enables concurrent adenine and cytosine editing. Nat. Biotechnol. 38, 861–864 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Yuan, T. et al. Optimization of C-to-G base editors with sequence context preference predictable by machine learning methods. Nat. Commun. 12, 4902 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Anzalone, A. V. et al. Programmable deletion, replacement, integration and inversion of large DNA sequences with twin prime editing. Nat. Biotechnol. 40, 731–740 (2022).

    Article  CAS  PubMed  Google Scholar 

  41. Jiang, T., Zhang, X.-O., Weng, Z. & Xue, W. Deletion and replacement of long genomic sequences using prime editing. Nat. Biotechnol. 40, 227–234 (2022).

    Article  CAS  PubMed  Google Scholar 

  42. Bult, C. J. et al. Mouse Genome Database (MGD) 2019. Nucleic Acids Res. 47, D801–D806 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Mouse Genome Informatics. Mouse Genome Database (MGD). https://www.informatics.jax.org/index.shtml (2022).

  44. Shaner, N. C. et al. A bright monomeric green fluorescent protein derived from Branchiostoma lanceolatum. Nat. Methods 10, 407–409 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Ng, S. R. et al. CRISPR-mediated modeling and functional validation of candidate tumor suppressor genes in small cell lung cancer. Proc. Natl Acad. Sci. USA 117, 513–521 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  47. Freed-Pastor, W. A. et al. The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer. Cancer Cell 39, 1342–1360 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Schlake, T. & Bode, J. Use of mutated FLP recognition target (FRT) sites for the exchange of expression cassettes at defined chromosomal loci. Biochemistry 33, 12746–12751 (1994).

    Article  CAS  PubMed  Google Scholar 

  49. Bindels, D. S. et al. mScarlet: a bright monomeric red fluorescent protein for cellular imaging. Nat. Methods 14, 53–56 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Vassilev, L. T. et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303, 844–848 (2004).

    Article  ADS  CAS  PubMed  Google Scholar 

  51. Nelson, J. W. et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 40, 402–410 (2022).

    Article  CAS  PubMed  Google Scholar 

  52. Bae, S., Park, J. & Kim, J.-S. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 30, 1473–1475 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Qiu, M. et al. Lipid nanoparticle-mediated codelivery of Cas9 mRNA and single-guide RNA achieves liver-specific in vivo genome editing of Angptl3. Proc. Natl Acad. Sci. USA 118, e2020401118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hingorani, S. R. et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4, 437–450 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. el Marjou, F. et al. Tissue-specific and inducible Cre-mediated recombination in the gut epithelium. Genesis 39, 186–193 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Kim, H. K. et al. Predicting the efficiency of prime editing guide RNAs in human cells. Nat. Biotechnol. 39, 198–206 (2021).

    Article  CAS  PubMed  Google Scholar 

  57. Drost, J. et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 521, 43–47 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  58. Zafra, M. P. et al. An in vivo Kras allelic series reveals distinct phenotypes of common oncogenic variants. Cancer Discov. 10, 1654–1671 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Strickler, J. H. et al. First data for sotorasib in patients with pancreatic cancer with KRAS p.G12C mutation: a phase I/II study evaluating efficacy and safety. J. Clin. Oncol. 40, 360490 (2022).

    Article  Google Scholar 

  60. Hofmann, M. H., Gerlach, D., Misale, S., Petronczki, M. & Kraut, N. Expanding the reach of precision oncology by drugging all KRAS mutants. Cancer Discov. 12, 924–937 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Waters, A. M. & Der, C. J. KRAS: the critical driver and therapeutic target for pancreatic cancer. Cold Spring Harb. Perspect. Med. 8, a031435 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Wang, X. et al. Identification of MRTX1133, a noncovalent, potent, and selective KRASG12D inhibitor. J. Med. Chem. 65, 3123–3133 (2022).

    Article  CAS  PubMed  Google Scholar 

  63. Awad, M. M. et al. Acquired resistance to KRASG12C inhibition in cancer. N. Engl. J. Med. 384, 2382–2393 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Pao, W. et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2, e73 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Freed-Pastor, W. A. & Prives, C. Mutant p53: one name, many proteins. Genes Dev. 26, 1268–1286 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  68. Schulz-Heddergott, R. et al. Therapeutic ablation of gain-of-function mutant p53 in colorectal cancer inhibits Stat3-mediated tumor growth and invasion. Cancer Cell 34, 298–314 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Barta, J. A., Pauley, K., Kossenkov, A. V. & McMahon, S. B. The lung-enriched p53 mutants V157F and R158L/P regulate a gain of function transcriptome in lung cancer. Carcinogenesis 41, 67–77 (2020).

    Article  CAS  PubMed  Google Scholar 

  70. Shakya, R. et al. Mutant p53 upregulates alpha-1 antitrypsin expression and promotes invasion in lung cancer. Oncogene 36, 4469–4480 (2017).

    Article  CAS  PubMed  Google Scholar 

  71. Jackson, E. L. et al. The differential effects of mutant p53 alleles on advanced murine lung cancer. Cancer Res. 65, 10280–10288 (2005).

    Article  CAS  PubMed  Google Scholar 

  72. Winslow, M. M. et al. Suppression of lung adenocarcinoma progression by Nkx2-1. Nature 473, 101–104 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  73. LaFave, L. M. et al. Epigenomic state transitions characterize tumor progression in mouse lung adenocarcinoma. Cancer Cell 38, 212–228 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Aoyama, N. et al. Transgenic mice that accept luciferase- or GFP-expressing syngeneic tumor cells at high efficiencies. Genes Cells 23, 580–589 (2018).

    Article  CAS  PubMed  Google Scholar 

  76. Hingorani, S. R. et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 (2005).

    Article  CAS  PubMed  Google Scholar 

  77. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Liu, B. et al. A split prime editor with untethered reverse transcriptase and circular RNA template. Nat. Biotechnol. https://doi.org/10.1038/s41587-022-01255-9 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Hunter, J. C. et al. Biochemical and structural analysis of common cancer-associated KRAS mutations. Mol. Cancer Res. 13, 1325–1335 (2015).

    Article  CAS  PubMed  Google Scholar 

  80. Amodio, V. et al. EGFR blockade reverts resistance to KRASG12C inhibition in colorectal cancer. Cancer Discov. 10, 1129–1139 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Canon, J. et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 575, 217–223 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  82. Kemp, S. B. et al. Efficacy of a small-molecule inhibitor of KrasG12D in immunocompetent models of pancreatic cancer. Cancer Discov. 13, 298–311 (2023).

    Article  CAS  PubMed  Google Scholar 

  83. Escobar-Hoyos, L. F. et al. Altered RNA splicing by mutant p53 activates oncogenic RAS signaling in pancreatic cancer. Cancer Cell 38, 198–211 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Klemke, L. et al. The gain-of-function p53 R248W mutant promotes migration by STAT3 deregulation in human pancreatic cancer cells. Front. Oncol. 11, 642603 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Schoenfeld, A. J. et al. The genomic landscape of SMARCA4 alterations and associations with outcomes in patients with lung cancer. Clin. Cancer Res. 26, 5701–5708 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Gibson, D. G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 6, 343–345 (2009).

    Article  CAS  PubMed  Google Scholar 

  87. Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol. 37, 224–226 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Hsu, J. Y. et al. PrimeDesign software for rapid and simplified design of prime editing guide RNAs. Nat. Commun. 12, 1034 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  89. Engler, C., Kandzia, R. & Marillonnet, S. A one pot, one step, precision cloning method with high throughput capability. PLoS ONE 3, e3647 (2008).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  90. Akama-Garren, E. H. et al. A modular assembly platform for rapid generation of DNA constructs. Sci. Rep. 6, 16836 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  91. Naranjo, S. et al. Modeling diverse genetic subtypes of lung adenocarcinoma with a next-generation alveolar type 2 organoid platform. Genes Dev. 36, 936–949 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Zettler, J., Schütz, V. & Mootz, H. D. The naturally split Npu DnaE intein exhibits an extraordinarily high rate in the protein trans-splicing reaction. FEBS Lett. 583, 909–914 (2009).

    Article  CAS  PubMed  Google Scholar 

  93. DuPage, M., Dooley, A. L. & Jacks, T. Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nat. Protoc. 4, 1064–1072 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  95. Owczarzy, R. et al. IDT SciTools: a suite for analysis and design of nucleic acid oligomers. Nucleic Acids Res. 36, W163–W169 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Jamal-Hanjani, M. et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 376, 2109–2121 (2017).

    Article  CAS  PubMed  Google Scholar 

  97. Campbell, J. D. et al. Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas. Nat. Genet. 48, 607–616 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Marino, S., Vooijs, M., van Der Gulden, H., Jonkers, J. & Berns, A. Induction of medulloblastomas in p53-null mutant mice by somatic inactivation of Rb in the external granular layer cells of the cerebellum. Genes Dev. 14, 994–1004 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Ely, Z. A. et. al. A prime-editor mouse to model a broad spectrum of somatic mutations in vivo. Sequence Read Archive. Datasets. https://www.ncbi.nlm.nih.gov/sra/?term=PRJNA951647 (2023).

  100. Gould, S. Prime-vs-base-editing. Github. https://github.com/samgould2/prime-vs-base-editing (2022).

Download references

Acknowledgements

We thank the entire Jacks Laboratory with particular thanks to D. Sandel, C. Concepcion, M. Burger, W. Hwang, M. Heileman and A. Jaeger for useful discussions. We also thank K. Yee, J. Teixeira, K. Anderson and M. Magendantz for administrative support. We thank S. Murray, R. Bronson, G. Johnson, J. Kuhn and M. Cornwall-Brady for helpful advice. S.I.G. and F.J.S.R. thank E. Kastenhuber for useful advice on human and mouse ortholog analyses, as well as D. Solit, N. Schultz, M. Berger and B. Gross (MSKCC) for access to MSK-IMPACT data. Z.A.E. acknowledges support from the Ludwig Center at MIT and the National Cancer Institute of the National Institutes of Health under award F31CA268835. S.I.G. was supported by T32GM136540. F.J.S.R. is an HHMI Hanna Gray Fellow and was supported by the V Foundation for Cancer Research (V2022-028), NCI Cancer Center Support Grant P30-CA1405, the Ludwig Center at MIT (2036636), Koch Institute Frontier Awards (2036648 and 2036642) and the MIT Research Support Committee (3189800). G.A.N. was supported by a Helen Hay Whitney Postdoctoral Fellowship and K99 award HL163805. We also thank the Koch Institute Swanson Biotechnology Center for technical support, especially the Histology core facility. This study was also supported by the Howard Hughes Medical Institute, David H. Koch Graduate Fellowship Fund, NCI Cancer Center Support Grant P30-CA1405, NIH Pre-Doctoral Training Grant T32GM007287, NIH grants U01 AI142756, RM1 HG009490 and R35 GM118062 and the Lustgarten Foundation for Pancreatic Cancer Research.

Author information

Authors and Affiliations

Authors

Contributions

Z.A.E. and T.J. conceived of the study. Z.A.E., N.M.A. and T.J. designed the experiments. N.M.A. conducted all autochthonous lung modeling. S.N. and Z.A.E. designed and constructed all DNA vectors described, including the transgene cassette. Z.A.E. designed pegRNAs, with contributions from N.M.A., P.B.R., J.R.D. and K.H.; G.A.N., S.I.G. and F.J.S.R. designed the computational pipeline to analyze patient mutation data; and S.I.G. conducted related bioinformatics analyses. W.A.F.-P., G.C.J. and Z.K. conducted pancreatic orthotopic transplant experiments. L.L. and G.C.J. conducted autochthonous pancreatic modeling. N.M.A., J.M.K., B.L.H. and G.A.N. conducted LNP experiments. C.M.C. and S.N. conducted experiments involving lung organoids. N.M.A., Z.A.E., N.B.P. and W.A.F.-P. conducted in vitro experiments using pancreatic organoids. W.M.R. conducted mESC targeting and chimera generation. K.L.M. provided animal husbandry expertise and conceptual advice. S.N., G.A.N., P.B.R., A.V.A., W.A.F.-P., P.M.K.W., K.L.M., S.L.S., F.J.S.R. and D.R.L. provided conceptual advice. Z.A.E., N.M.A., S.I.G., F.J.S.R. and T.J. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Tyler Jacks.

Ethics declarations

Competing interests

T.J. is a member of the Board of Directors of Amgen and Thermo Fisher Scientific, and a co-founder of Dragonfly Therapeutics and T2 Biosystems. T.J. serves on the Scientific Advisory Board of Dragonfly Therapeutics, SQZ Biotech, and Skyhawk Therapeutics. T.J. is also the President of Break Through Cancer. None of these affiliations represent a conflict of interest with respect to the design or execution of this study or the interpretation of data presented in this manuscript. His laboratory currently receives funding from the Johnson & Johnson Lung Cancer Initiative, but this funding did not support the research described in this manuscript. D.R.L. is a consultant for Prime Medicine, Beam Therapeutics, Pairwise Plants, Chroma Medicine, and Nvelop Therapeutics, companies that use or deliver genome editing or genome engineering agents, and owns equity in these companies. K.H. and A.V.A. are currently employees of Prime Medicine. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Andrea Ventura and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Prime editing has a greater capacity to model recurrent cancer-associated mutations than base editing.

a Quantification of mutations that are detected in multiple patients in cancer-associated genes, depicted for each mutant variant type (Single nucleotide variants = SNV, deletions = DEL, insertions = INS, di-nucleotide variants = DNV, oligo-nucleotide variants = ONV). The y-axis in all plots indicates the total number of unique mutations per variant type. b Quantification of recurrent mutations potentially amenable to modeling by a base editor with an NGG PAM (top) or a prime editor with an NGG PAM and a 30 base pair RT template (bottom). The columns show results considering mutations that occur in ≥5 patients (left) or ≥10 patients (right). ‘CBE/ABE high’ indicates that the SNV falls in the high efficiency editing window (position +4 to +8 in the protospacer), while ‘CBE/ABE low’ indicates the SNV falls within the protospacer but outside the high efficiency window. The data include SNVs (blue outer circle; 78.8% of mutations) and other mutation types (gray outer circle; 20.2% of mutations). All calculations assume a base or prime editor that recognizes only NGG PAMs. c Total percentage of recurrent mutations amenable to modeling by a base editor with an NGG PAM, quantified at multiple thresholds of mutation frequency, from mutations that occur ≥1 patient to those that occur ≥10 patients. d Total percentage of recurrent mutations amenable to modeling by a prime editor with an NGG PAM and a 30 base pair RT template at multiple thresholds of mutation frequency, from mutations that occur ≥1 patient to those that occur ≥10 patients. e Capabilities of prime versus base editing to model orthologous mutations in mice at multiple thresholds of mutation frequency, from ≥1 to ≥10 patients with each mutation. Prime editing can model approximately double the number of orthologous mutations in mice at all thresholds of mutation frequency. Base editing (BE) shown in red. Prime editing (PE) shown in blue. f Quantification of the ability of base editing (left) and prime editing (right) to model orthologous mutations in mice for mutations that occur in ≥5 or ≥ 10 patients.

Extended Data Fig. 2 Prime editing enables modeling a broader scope of cancer-associated mutations from residues conserved in mice at various homology stringencies.

a The percentage of mutations, categorized by variant type, that fall in a region of homology as a function of flank size. Flank size is defined as the number of amino acids on either side of the mutant codon that must match between the human and mouse orthologs for the mutated codon to be considered orthologous (for example, a flank size of two would mean that a total of five amino acids–two upstream and two downstream of the mutant codon–would need to match in the human and mouse protein to be considered orthologous.). b Capabilities of prime editing (blue) and base editing (red) to model mutations that derive from a wild-type amino acid residue conserved in mice as a function of conserved flank size (that is, the stringency of homology). All calculations assume a base or prime editor that recognizes only NGG PAMs. c Quantification of the ability of base editing (top) and prime editing (bottom) to model orthologous mutations in mice at different flank size values (2, 3, 5, and 10 from left to right). These plots correspond with the data points in panel (b).

Extended Data Fig. 3 Prime editing in Trp53-proficient cells and off-target activity at computationally predicted loci.

a Prime editing in Trp53flox/flox;Rosa26PE2/+ versus Trp53+/+;Rosa26PE2/+ TTFs across several indicated loci. Data and error bars indicate the mean and standard deviation of three independent transductions. b Prime editing in Trp53flox/flox;Rosa26PE2/+ versus Trp53+/+;Rosa26PE2/+ pancreatic across several indicated loci. Note that the editing data for Dnmt1+GGG in Trp53flox/flox organoids are the same depicted in Fig. 2f. Data and error bars indicate the mean and standard deviation of three independent transductions. c Quantification of off-target prime editing for a pegRNA targeted to Dnmt1. Off-targets represent three computationally predicted loci for tail-tip fibroblasts cultured for more than one month in the presence of the Dnmt1+GGG pegRNA or a control pegRNA. Data and error bars indicate the mean and standard deviation of three independent transductions. Off-target locus details are described in Supplementary Table 4. d Quantification of off-target prime editing at four computationally predicted loci for tail-tip fibroblasts cultured with a KrasG12A epegRNA or a control pegRNA. Data and error bars indicate the mean and standard deviation of three independent transductions.

Extended Data Fig. 4 Mismatch repair-evasive edits amplify prime editing efficiency across multiple cancer-associated loci.

a Design of epegRNAs encoding EgfrL860R (right) or KrasG12C (left) with (bottom) or without (top) silent mutations intended to promote MMR evasion. The black line on the sequence schematics indicates the Cas9 nicking site. b Editing efficiency and indel byproduct frequency of epegRNAs encoding KrasG12C with or without silent mutations in tail tip-derived fibroblasts. Note that the data for KrasG12C without silent edits is also featured in Fig. 3. Data and error bars indicate the mean and standard deviation of three independent transductions. c Editing efficiency and indel byproduct frequency of epegRNAs encoding EgfrL860R with or without silent mutations. Data and error bars indicate the mean and standard deviation of three independent transductions. d Editing efficiency and indel byproduct frequency of epegRNAs encoding KrasG12C with or without silent mutations in pancreatic organoids. Note that the data for KrasG12C without silent edits is also featured in Fig. 3. Data and error bars indicate the mean and standard deviation of three independent transductions.

Extended Data Fig. 5 Treatment of prime edited KrasG12D and KrasG12C pancreatic organoids with EGFR and KRAS inhibitors in the presence or absence of a secondary resistance mutation.

a Bright-field images of unedited Rosa26PE2/+ pancreatic organoids treated with gefitinib after infection with the template UPEC vector (that is, lacking a pegRNA). Scale bars: 100 µm. b Additional bright-field images supplementing Fig. 3d and depicting prime edited KrasG12D and KrasG12C organoids treated either with DMSO, gefitinib, or sotorasib. Scale bars: 100 µm. c Allele frequencies of KrasG12R, KrasG12A, or KrasG12C+silent edits mutations in pancreatic organoids before and after two passages of treatment with gefitinib (1 µM). Data and error bars indicate the mean and standard deviation of three independent transductions pre- and post-treatment. Note that pre-treatment replicate data is also represented in Extended Data Fig. 4 (KrasG12C + silent edits) and Fig. 3 (KrasG12R and KrasG12A). d Editing efficiency and indel frequencies of epegRNAs templating the KrasY96C transversion in Rosa26PE2/+ TTFs. Data and error bars indicate the mean and standard deviation of three independent transductions. e Nucleotide percentage quilts and allele frequency tables output by CRISPResso2 depicting sequencing reads derived from prime edited KrasG12C and KrasG12D pancreatic organoids. Prime edited alleles were previously selected with gefitinib, and these sequencing data derive from organoids after treatment with gefitinib in combination with mutant KRAS inhibitors.

Extended Data Fig. 6 On- and off-target prime editing at Trp53 in pancreatic organoids.

a Editing efficiency and indel byproduct frequency of epegRNAs encoding Trp53M240FS-14nt (n = 4 independent transductions) or Trp53M240S* (n = 4 independent transductions) in Trp53flox/+ pancreatic organoids. Data and error bars indicate the mean and standard deviation across independent transductions. b Design of the epegRNAs for Trp53M240FS-14nt or Trp53M240S*. The black line below the sequence schematics indicates the Cas9 nicking site. These epegRNAs were designed to both evade MMR and modify the seed and PAM sequences. c CRISPResso allele frequency plots of prime edited reads for Trp53M240FS-14nt before and after nutlin selection in pancreatic organoids. d Quantification of off-target prime editing for an epegRNA encoding Trp53M240FS-14nt. Off-targets represent four computationally predicted loci for pancreatic organoids cultured for more than one month in the presence of the Trp53M240FS-14nt epegRNA and nutlin-3a or a control pegRNA. Data and error bars indicate the mean and standard deviation of three independent transductions. e Quantification of off-target prime editing for an epegRNA encoding Trp53R245Q. Off-targets represent four computationally predicted loci for pancreatic organoids cultured for more than one month in the presence of the Trp53R245Q epegRNA and nutlin-3a or a control pegRNA. Data and error bars indicate the mean and standard deviation of three independent transductions. f Quantification of off-target prime editing for an epegRNA encoding Trp53R245W. Off-targets represent four computationally predicted loci for pancreatic organoids cultured for more than one month in the presence of the Trp53R245W epegRNA and nutlin-3a or a control pegRNA. Data and error bars indicate the mean and standard deviation of three independent transductions. Statistical significance was calculated using an unpaired, one-tailed Welch’s t-test (P = 0.0045 for the leftmost off-target site).

Extended Data Fig. 7 Prime edited tumors recapitulate histological features of gold standard GEMMs of lung adenocarcinoma.

a Aiforia grade analysis of 16-week tumor-bearing lungs initiated by in vivo prime editing (red = Grade 1, green = Grade 2, blue = Grade 3, orange = Grade 4). b H&E and immunohistochemical (IHC) staining of a representative Grade 2 tumor 16 weeks post-initiation. IHC staining was performed with antibodies specific to Nkx2.1, a lung lineage transcription factor, or Surfactant Protein C (Sftpc), a marker of AT2 cells. Scale bars: 200 µm. c H&E and IHC staining of a Grade 3/4 tumor 16 weeks post-initiation. IHC staining was performed with antibodies specific to NKX2-1 or HMGA2, a lung embryonic chromatin regulator. Scale bars: 200 µm.

Extended Data Fig. 8 Autochthonous modeling of diverse Kras variants in the lung.

a Tumor burden quantification using Aiforia grading algorithm for KrasG12D (n = 5 mice), KrasG12A (n = 9 mice), and KrasG12R (n = 9 mice), tumors at 16 weeks. Tumor burden is defined as the ratio of tumor area (Grades 1–4) over total lung area. Data and error bars indicate the mean and standard deviation of five to nine biological replicates. b Representative fluorescence imaging of lungs from Trp53flox/flox;Rosa26PE2/+ and Trp53flox/flox;Rosa26PE2/PE2 mice infected with UPEC-KrasG12A (n = 10), UPEC-KrasG12R (n = 9), UPEC-KrasG12D (n = 20), all harvested at 16 weeks, as well as mice infected with UPEC-KrasG12C+silent edits harvested at 12 weeks. c H&E staining of representative tissue from a mouse infected with UPEC-KrasG12C+silent edits. Tissue was collected at 12 weeks. Scale bars indicate 2 mm, 100 µm, and 20 µm respectively. Grading analysis indicates proportion of Grade 1, 2, and 3 lesions from four representative animals. d CRISPResso allele frequency plots depicting prime edited reads derived from lung tumors from a Trp53flox/flox;Rosa26PE2/PE2 mouse infected with UPEC-KrasG12C (top plot) and three different Trp53flox/flox;Rosa26PE2/PE2 mice infected with UPEC-KrasG12C+silent edits (bottom three plots). The black line on the sequence schematics indicates the Cas9 nicking site. The intended mutations are indicated by black boxes. The red box in the top plot indicates an unintended, silent KrasA11A substitution. The percentages indicated represent the percentage of all analyzed reads (edited + unedited reads) that bear this sequence. e H&E staining of representative tissue from a Trp53+/+;Rosa26PE2/+ mouse infected with UPEC-KrasG12D. H&E indicates histopathology consistent with a low grade adenoma. Scale bars indicate 2 mm (left) and 100 µm (right). f CRISPResso allele frequency plot depicting prime edited reads derived from an adenoma from a Trp53+/+;Rosa26PE2/+ mouse infected with UPEC-KrasG12D. The intended mutation is indicated by a black box. Note that the percentage indicated represents the percentage of all analyzed reads (including unedited reads) that bear this sequence. g Representative bright-field and fluorescence imaging of adenomas from two Trp53+/+;Rosa26PE2/+ mice infected with UPEC-KrasG12D (harvested at 10 weeks). White arrows indicate smaller fluorescent adenomas.

Extended Data Fig. 9 Modeling of pancreatic cancer through organoid transplantation and lentiviral initiation.

a Stacked bar plots indicating incidence of tumors and metastases in animals orthotopically transplanted with UPEC-KrasG12D (n = 9) and UPEC-KrasG12C (n = 9) prime-edited pancreatic organoids. b Bright-field and fluorescent imaging of an omental metastasis and surrounding tissue in an animal transplanted with UPEC-KrasG12D prime-edited pancreatic organoids. c H&E staining of a representative omental metastasis. Scale bar indicates 100 µm. d Experimental workflow for autochthonous pancreatic tumor modeling through retrograde pancreatic ductal infection of lentivirus templating KrasG12D (n = 4) or KrasY96C (n = 3) pegRNAs. e Representative H&E staining of autochthonous pancreatic adenocarcinoma initiated through pancreatic ductal infection of UPEC-KrasG12D. Scale bars indicate 1 mm and 50 µm, respectively.

Supplementary information

Supplementary Information

Supplementary Figs. 1–8, Supplementary Data (Flow Cytometry Gating Strategy) and Supplementary Note.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–5.

Source data

Source Data Fig. 3

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ely, Z.A., Mathey-Andrews, N., Naranjo, S. et al. A prime editor mouse to model a broad spectrum of somatic mutations in vivo. Nat Biotechnol 42, 424–436 (2024). https://doi.org/10.1038/s41587-023-01783-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-023-01783-y

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