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:

Generation of precision preclinical cancer models using regulated in vivo base editing

Abstract

Although single-nucleotide variants (SNVs) make up the majority of cancer-associated genetic changes and have been comprehensively catalogued, little is known about their impact on tumor initiation and progression. To enable the functional interrogation of cancer-associated SNVs, we developed a mouse system for temporal and regulatable in vivo base editing. The inducible base editing (iBE) mouse carries a single expression-optimized cytosine base editor transgene under the control of a tetracycline response element and enables robust, doxycycline-dependent expression across a broad range of tissues in vivo. Combined with plasmid-based or synthetic guide RNAs, iBE drives efficient engineering of individual or multiple SNVs in intestinal, lung and pancreatic organoids. Temporal regulation of base editor activity allows controlled sequential genome editing ex vivo and in vivo, and delivery of sgRNAs directly to target tissues facilitates generation of in situ preclinical cancer 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: Regulatable BE expression across murine tissues.
Fig. 2: Efficient BE in ex vivo derived iBE organoids.
Fig. 3: iBE enables sequential BE in vitro and in vivo.
Fig. 4: In situ BE with iBE drives liver tumors.
Fig. 5: Efficient engineering of missense mutations in pancreatic tumor models.

Similar content being viewed by others

Data availability

All source data (including P values) are available in Supplementary Table 5. Raw FASTQ files have been deposited in the Sequence Read Archive under accession number PRJNA859154. Processed RNA-seq data (transcripts per million values and differentially expressed genes) are available in Supplementary Table 2.

Code availability

Code for analysis and data visualization is available at https://github.com/lukedow/iBE.git.

References

  1. Goodwin, S., McPherson, J. D. & McCombie, W. R. Coming of age: ten years of next-generation sequencing technologies. Nat. Rev. Genet. 17, 333–351 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Landrum, M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 44, D862–D868 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Vivanco, I. et al. Differential sensitivity of glioma- versus lung cancer-specific EGFR mutations to EGFR kinase inhibitors. Cancer Discov. 2, 458–471 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hyman, D. M. et al. AKT inhibition in solid tumors with AKT1 mutations. J. Clin. Oncol. 35, 2251–2259 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vasan, N. et al. Double PIK3CA mutations in cis increase oncogenicity and sensitivity to PI3Kα inhibitors. Science 366, 714–723 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Findlay, G. M. et al. Accurate classification of BRCA1 variants with saturation genome editing. Nature 562, 217–222 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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 

  10. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464–471 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gaudelli, N. M. et al. Directed evolution of adenine base editors with increased activity and therapeutic application. Nat. Biotechnol. 38, 892–900 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Komor, A. C. et al. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity. Sci. Adv. 3, eaao4774 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Rothgangl, T. et al. In vivo adenine base editing of PCSK9 in macaques reduces LDL cholesterol levels. Nat. Biotechnol. 39, 949–957 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Villiger, L. et al. In vivo cytidine base editing of hepatocytes without detectable off-target mutations in RNA and DNA. Nat. Biomed. Eng. 5, 179–189 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Villiger, L. et al. Treatment of a metabolic liver disease by in vivo genome base editing in adult mice. Nat. Med. 24, 1519–1525 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Song, C.-Q. et al. Adenine base editing in an adult mouse model of tyrosinaemia. Nat. Biomed. Eng. 4, 125–130 (2019).

  17. Yeh, W. H., Chiang, H., Rees, H. A., Edge, A. S. B. & Liu, D. R. In vivo base editing of post-mitotic sensory cells. Nat. Commun. 9, 2184 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  18. Banskota, S. et al. Engineered virus-like particles for efficient in vivo delivery of therapeutic proteins. Cell 185, 250–265 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ryu, S. M. et al. Adenine base editing in mouse embryos and an adult mouse model of Duchenne muscular dystrophy. Nat. Biotechnol. 36, 536–539 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Yang, L. et al. Amelioration of an inherited metabolic liver disease through creation of a de novo start codon by cytidine base editing. Mol. Ther. 28, 1673–1683 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Dow, L. E. et al. Conditional reverse tet-transactivator mouse strains for the efficient induction of TRE-regulated transgenes in mice. PLoS ONE 9, e95236 (2014).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  22. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Grunewald, J. et al. CRISPR DNA base editors with reduced RNA off-target and self-editing activities. Nat. Biotechnol. 37, 1041–1048 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yan, N. et al. Cytosine base editors induce off-target mutations and adverse phenotypic effects in transgenic mice. Nat. Commun. 14, 1784 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. 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 

  26. 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 

  27. Schatoff, E. M. et al. Distinct CRC-associated APC mutations dictate response to tankyrase inhibition. Cancer Discov. 9, 1358–1371 (2019).

  28. Katti, A. et al. GO: a functional reporter system to identify and enrich base editing activity. Nucleic Acids Res. 48, 2841–2852 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sanchez-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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mehta, A. & Merkel, O. M. Immunogenicity of Cas9 protein. J. Pharm. Sci. 109, 62–67 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Chew, W. L. et al. A multifunctional AAV–CRISPR–Cas9 and its host response. Nat. Methods 13, 868–874 (2016).

    Article  CAS  PubMed  PubMed Central  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. Ruiz de Galarreta, M. et al. β-catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma. Cancer Discov. 9, 1124–1141 (2019).

  34. Calvisi, D. F. et al. Activation of the canonical Wnt/β-catenin pathway confers growth advantages in c-Myc/E2F1 transgenic mouse model of liver cancer. J. Hepatol. 42, 842–849 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. 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 

  36. Alsner, J. et al. A comparison between p53 accumulation determined by immunohistochemistry and TP53 mutations as prognostic variables in tumours from breast cancer patients. Acta Oncol. 47, 600–607 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. 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 

  38. Bartek, J., Iggo, R., Gannon, J. & Lane, D. P. Genetic and immunochemical analysis of mutant p53 in human breast cancer cell lines. Oncogene 5, 893–899 (1990).

    CAS  PubMed  Google Scholar 

  39. Maresch, R. et al. Multiplexed pancreatic genome engineering and cancer induction by transfection-based CRISPR/Cas9 delivery in mice. Nat. Commun. 7, 10770 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Park, J. S. et al. Pancreatic cancer induced by in vivo electroporation-enhanced sleeping beauty transposon gene delivery system in mouse. Pancreas 43, 614–618 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. 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).

  42. Zhou, C. et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 571, 275–278 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Arbab, M. et al. Determinants of base editing outcomes from target library analysis and machine learning. Cell 182, 463–480 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Marquart, K. F. et al. Predicting base editing outcomes with an attention-based deep learning algorithm trained on high-throughput target library screens. Nat. Commun. 12, 5114 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pallaseni, A. et al. Predicting base editing outcomes using position-specific sequence determinants. Nucleic Acids Res. 50, 3551–3564 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Park, J. & Kim, H. K. Prediction of base editing efficiencies and outcomes using DeepABE and DeepCBE. Methods Mol. Biol. 2606, 23–32 (2023).

    Article  CAS  PubMed  Google Scholar 

  47. Kim, Y. et al. High-throughput functional evaluation of human cancer-associated mutations using base editors. Nat. Biotechnol. 40, 874–884 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Davis, J. R. et al. Efficient prime editing in mouse brain, liver and heart with dual AAVs. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01758-z (2023).

  51. Dow, L. E. et al. A pipeline for the generation of shRNA transgenic mice. Nat. Protoc. 7, 374–393 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. O’Rourke, K. P., Ackerman, S., Dow, L. E. & Lowe, S. W. Isolation, culture, and maintenance of mouse intestinal stem cells. Bio Protoc. 6, e1733 (2016).

    PubMed  Google Scholar 

  53. Huch, M. et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 32, 2708–2721 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  55. Amen, A. M. et al. Endogenous spacing enables co-processing of microRNAs and efficient combinatorial RNAi. Cell Rep. Methods 2, 100239 (2022).

  56. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Finn, J. D. et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell Rep. 22, 2227–2235 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Paffenholz Stella, V. et al. Senescence induction dictates response to chemo- and immunotherapy in preclinical models of ovarian cancer. Proc. Natl Acad. Sci. USA 119, e2117754119 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Leibold, J. et al. Somatic tissue engineering in mouse models reveals an actionable role for WNT pathway alterations in prostate cancer metastasis. Cancer Discov. 10, 1038–1057 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Dow laboratory for advice and comments on the preparation of the paper. We would like to acknowledge K. Tsanov and J. Leibold for assistance and advice in setting up the pancreas EPO protocol. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health (NIH). This work was supported by a project grant from the NIH (R01CA229773), P01 CA087497 (S.W.L.), an MSKCC Functional Genomics Initiative grant (S.W.L.), an Agilent Technologies Thought Leader Award (S.W.L.) and support from Synthego under a Synthego Innovator Award (L.E.D.). A.K. was supported by an F31 Ruth L. Kirschstein Predoctoral Individual National Research Service Award (F31-CA247351-02). A.V. was supported by a Postdoctoral Fellowship from the Human Frontier Scientific Program (LT0011/2023-L). B.J.D. was supported by an F31 Ruth L. Kirschstein Predoctoral Individual National Research Service Award (F31-CA261061-01). E.E.G. is the Kenneth G. and Elaine A. Langone Fellow of the Damon Runyon Cancer Research Foundation (DRG-2343-18). F.J.S.R. was supported by the MSKCC TROT program (5T32CA160001) and a GMTEC Postdoctoral Researcher Innovation Grant and is a Howard Hughes Medical Institute (HHMI) Hanna Gray Fellow. S.W.L. is an HHMI investigator.

Author information

Authors and Affiliations

Authors

Contributions

A.K. and A.V.P. designed and performed experiments, analyzed data and wrote the paper. M.F., J.Z., M.P.Z., S.G., E.G., J.S., W.L, B.J.D., M.C.F., K.H. and F.J.S.R. performed experiments and/or analyzed data. S.W.L. supervised experimental work. L.E.D. designed and supervised experiments, analyzed data and wrote the paper.

Corresponding author

Correspondence to Lukas E. Dow.

Ethics declarations

Competing interests

L.E.D. is a scientific advisor and holds equity in Mirimus, Inc. L.E.D. has received consulting fees and/or honoraria from Volastra Therapeutics, Revolution Medicines, Repare Therapeutics, Fog Pharma and Frazier Healthcare Partners. S.W.L is an advisor for and has equity in the following biotechnology companies: ORIC Pharmaceuticals, Faeth Therapeutics, Blueprint Medicines, Geras Bio, Mirimus, Inc., PMV Pharmaceuticals and Constellation Pharmaceuticals. S.W.L. acknowledges receiving funding and research support from Agilent Technologies for the purposes of massively parallel oligo synthesis. K.H., A.P.K. and J.A.W. are employees and shareholders of Synthego Corporation.

Peer review

Peer review information

Nature Biotechnology thanks the anonymous reviewers 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 Regulatable BE expression in vivo.

a. Calculated BE3RA transgene copy number in iBEhem and iBEhom using a Taqman quantiative PCR assay with genomic DNA from H11-LSL-Cas9 mice as a reference. Data are presented as mean values ± s.e.m. (*p<0.05, Student’s t-test). b. Schematic representation of the targeted RMCE site downstream of the Col1a1 locus. Primers flanking the knock-in cassette and a single primer within the targeted transgene can identify wildtype, hemizygous and homozygous animals, as shown in the example genotyping agarose gel c. Mendelian transmission of Col1a1-targeted iBE knock-in (with and without R26-CAGs-rtTA3 allele) and associated p-value (chi-square test) relative to expected Mendelian inheritance. d. Immunofluorescent detection of Cas9 protein in rtTA3 only and iBEhom mice maintained on normal chow (No dox) or doxycycline chow for 14 days (14D dox). (n=3 mice per genotype and condition). e. Immunofluorescent detection of Cas9 protein in iBEhem (top) or iBEhom (bottom) mice maintained on dox chow for 7 days across four tissues. (n=3 mice per genotype and condition).

Extended Data Fig. 2 Expression of BE3RA across different tissues in mice carrying one or two copy of each allele.

Immunofluorescent detection of Cas9 protein in rtTA3+/- iBE+/- (iBEhem), rtTA3+/+ iBE+/-, rtTA3+/- iBE+/+ and rtTA3+/+iBE+/+ (iBEhom) mice maintained on dox chow for 14 days. Cas9 protein(green), DAPI staining for nuclei (blue) across four tissues analyzed. (n=3 mice per genotype and condition).

Extended Data Fig. 3 Dox treatment does not induce abnormalities in iBEhom mice.

a. Hematoxylin and Eosin (H&E) staining in rtTA3+/-iBE-/- and rtTA3+/+iBE+/+ (iBEhom) on normal chow or doxycycline chow for 14 days. (n=3 mice). b. Flow cytometry analysis of spleen and bone marrow cell suspensions of rtTA3+/-iBE-/- and rtTA3+/+iBE+/+ (iBEhom) on normal chow or maintained on doxycycline chow for 14 days (n=3 mice). Data are presented as mean values ± s.d (*p<0.05, Student’s t-test).

Extended Data Fig. 4 iBE induces low off target RNA editing that is reversed by withdrawal of transgene expression.

a. C to U editing in RNA transcripts detected from RNA sequencing data from intestine and liver from rtTAhem and iBEhom on normal chow (-), dox chow (+) for 14 days, or switched from dox chow for 14 days to normal chow for 6 days (SW). Data in the middle and right panels was derived from re-analysis of published datasets, as indicated under each plot. For experiments with multiple comparisons, p-values were calculated by one-way ANOVA, n=3 mice/condition. For individual pairwise comparisons, Student’s t-test was used. b. A to G editing in RNA transcripts detected from RNA sequencing data from intestine and liver from rtTAhem and iBEhom on normal chow (-), dox chow (+) for 14 days, or switched from dox chow for 14 days to normal chow for 6 days (SW). (n=3 mice). c. Transcript abundance (transcripts per million; TPM) in pancreatic organoids, intestine, and liver from rtTAhem and iBEhom on normal chow (-), dox chow (+) for 14 days, or switched from dox chow for 14 days to normal chow for 6 days (SW). All data shown are presented as mean values +/- s.d., n=3 mice/condition.

Extended Data Fig. 5 iBE has low DNA off target activity.

a. Schematic of experimental set up in mouse embryonic stem cells (ESCs). mESCs containing iBE knock in were transduced with LRT2B-gRNA vector and selected for gRNA expression. sgRNA+ cells were plated with and without dox for 6 days after which cells were plated at low density for clonal outgrowth without dox. 3 pools of 10 clones were picked for each dox conditions across to gRNA targeted cell lines (sgRNAs = Apc.Q1405X and Pik3ca.E545K). In total, 12 pools of 10 clones were sequenced at 800-1000-fold coverage across the MSK-IMPACT cancer gene set. b. Pie chart display of frequency of C>T or C>other SNVs found in pooled clones for each condition (on and off dox) for both sgRNAs. c. Sequencing analysis at cancer gene sites in cell conditions (right) described in a. Solid blue boxes represent on-target activity of the sgRNA, dotted orange boxes signify on-target ‘bystander’ editing within the gRNA window. d. Quantification of C>T and C>other SNVs found across both targets. 2-way ANOVA test for multiple comparisons was used to evaluate statistical significance across conditions. Data are presented as mean values ± s.e.m. p-values are displayed.

Extended Data Fig. 6 iBE does not induce off target RNA editing in organoids.

a. Schematic of experimental set up in iBE derived pancreatic organoids. Organoids were transduced and selected with GFPGO reporter (mScarlet+). Organoids maintained off dox were then split into dox conditions to induce BE expression for 4 days and then split again into + and – dox conditions for an additional day. b. Editing of organoids in each condition (OFF, D4, D8, and D4 sw) was quantified by flow cytometry, calculating the percentage of GFP+ cells within the mScarlet+ population. Data are presented as mean values ± s.e.m. One-way ANOVA with Tukey’s correction c. PCA analysis of RNA sequencing data from OFF, D8, and D4 SW organoids. Colors correspond to dox condition and shape delineates organoid replicate/mouse origin (n=3). d. Volcano plots from RNA-seq data comparing iBE pancreatic organoids culture on dox-containing media vs regular media. e. Off-target RNA editing analysis, processed as described for Supplementary Fig. 4. No significant differences in RNA variants were observed, n=3, one-way ANOVA with Tukey’s correction. Data are presented as mean values ± s.e.m. For all data shown, n=3 independent organoid lines/condition.

Extended Data Fig. 7 Editing dynamics of iBE organoids.

a. Flow cytometry analysis of three independent pancreatic KP mutant organoid lines integrated with GFPGO reporter following dox treatment for 0-8 days (black), transient exposure for 2h or 12h (grey), or transient exposure then re-treatment at 4 days (green). b. Targeted deep sequencing quantification of target C:G to T:A conversion at the ApcQ1405X locus in 2D small intestinal derived iBE cell line following dox addition for 21 days (dark blue), or transient dox treatment for 3 days and withdrawn for 18 days (light blue). c. Targeted deep sequencing quantification of indel conversion of b. Data are presented as mean values ± s.e.m. (*p<0.05, Student’s t-test) (n=3 independently derived line).

Extended Data Fig. 8 Efficient BE in iBE organoids with low collateral editing.

a. Targeted deep sequencing quantification of corresponding target C>T/A/G and indel conversion in small intestinal iBE organoids nucleofected with plasmid (light blue) or synthetic (indigo) gRNAs (ApcQ1405, Trp53Q97, CR8.OS2) as indicated, with and without dox treatment. b. Targeted deep sequencing quantification of target C>T/A/G and indel conversion in small intestinal iBE organoids nucleofected with synthetic gRNAs targeting cancer associated SNVs from Fig. 2f. c-j. Quantification of collateral editing of adjacent cytosines for samples shown in Fig. 2f. Predicted translation of each quantified read is shown below with targeted amino acid substitution (dark grey) and additional amino acid substitution (pink). All data are presented as mean values ± s.e.m.

Extended Data Fig. 9 Analysis of collateral editing before and after functional selection.

a-h. Quantification of collateral editing of adjacent cytosines for data shown in Fig. 2f, unselected (white) and selected (color) in small intestinal iBE organoids nucleofected with various synthetic gRNAs targeting cancer associated SNVs.

Extended Data Fig. 10 In situ base editing with iBE by synthetic gRNA delivery drives liver tumors.

a. HTVI delivery of synthetic gRNAs with SB-Myc as in Fig. 4. BF, H&E images, and IF staining for ß-catenin (green) and glutamine synthetase (GS, red) in livers with tumors. Number of transfected mice with palpable tumors is shown below each column. b. Quantification of target C:G to T:A conversion from tumors described in a). Each point corresponds to an isolated bulk tumor. (n=2-7 mice for a given gRNA target). Individual editing data color-coded by animal in Supplementary Fig. 3. All data are presented as mean values ± s.e.m.

Supplementary information

Supplementary Information

Supplementary Figs. 1–8.

Reporting Summary

Supplementary Table 1

Mendelian transmission of Cola1-targeted iBE knockin.

Supplementary Table 2

RNA-seq results.

Supplementary Table 3

DNA off-target effects using iBE-targeted ESCs expressing ApcQ1405 and Pik3caE545K sgRNAs.

Supplementary Table 4

sgRNAs and primers used in the study.

Supplementary Table 5

Source data P values

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

Katti, A., Vega-Pérez, A., Foronda, M. et al. Generation of precision preclinical cancer models using regulated in vivo base editing. Nat Biotechnol 42, 437–447 (2024). https://doi.org/10.1038/s41587-023-01900-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-023-01900-x

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