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.

In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis

A Corrigendum to this article was published on 08 December 2017

This article has been updated

Abstract

In vivo interrogation of the function of genes implicated in tumorigenesis is limited by the need to generate and cross germline mutant mice. Here we describe approaches to model colorectal cancer (CRC) and metastasis, which rely on in situ gene editing and orthotopic organoid transplantation in mice without cancer-predisposing mutations. Autochthonous tumor formation is induced by CRISPR-Cas9-based editing of the Apc and Trp53 tumor suppressor genes in colon epithelial cells and by orthotopic transplantation of Apc-edited colon organoids. ApcΔ/Δ;KrasG12D/+;Trp53Δ/Δ (AKP) mouse colon organoids and human CRC organoids engraft in the distal colon and metastasize to the liver. Finally, we apply the orthotopic transplantation model to characterize the clonal dynamics of Lgr5+ stem cells and demonstrate sequential activation of an oncogene in established colon adenomas. These experimental systems enable rapid in vivo characterization of cancer-associated genes and reproduce the entire spectrum of tumor progression and metastasis.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: CRISPR-Cas9-based in situ Apc editing in the colon epithelium induces adenoma formation.
Figure 2: Orthotopic transplantation models of mouse- and patient-derived colorectal cancer.
Figure 3: Lgr5 cell lineage tracing and sequential mutagenesis in established orthotopic colon adenomas.

Accession codes

Primary accessions

BioProject

Change history

  • 06 July 2017

    In the version of this article initially published, the initial “J” was omitted from an author's name, which should appear as Francisco J Sánchez-Rivera. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

  2. Seshagiri, S. et al. Recurrent R-spondin fusions in colon cancer. Nature 488, 660–664 (2012).

    Article  CAS  Google Scholar 

  3. Moser, A.R., Pitot, H.C. & Dove, W.F. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science 247, 322–324 (1990).

    Article  CAS  Google Scholar 

  4. Su, L.K. et al. Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science 256, 668–670 (1992).

    Article  CAS  Google Scholar 

  5. Roper, J. & Hung, K.E. Priceless GEMMs: genetically engineered mouse models for colorectal cancer drug development. Trends Pharmacol. Sci. 33, 449–455 (2012).

    Article  CAS  Google Scholar 

  6. Golovko, D., Kedrin, D., Yilmaz, Ö.H. & Roper, J. Colorectal cancer models for novel drug discovery. Expert Opin. Drug Discov. 10, 1217–1229 (2015).

    Article  Google Scholar 

  7. Hinoi, T. et al. Mouse model of colonic adenoma-carcinoma progression based on somatic Apc inactivation. Cancer Res. 67, 9721–9730 (2007).

    Article  CAS  Google Scholar 

  8. Haigis, K.M. et al. Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat. Genet. 40, 600–608 (2008).

    Article  CAS  Google Scholar 

  9. Xue, Y., Johnson, R., Desmet, M., Snyder, P.W. & Fleet, J.C. Generation of a transgenic mouse for colorectal cancer research with intestinal cre expression limited to the large intestine. Mol. Cancer Res. MCR 8, 1095–1104 (2010).

    Article  CAS  Google Scholar 

  10. Shibata, H. et al. Rapid colorectal adenoma formation initiated by conditional targeting of the Apc gene. Science 278, 120–123 (1997).

    Article  CAS  Google Scholar 

  11. Hung, K.E. et al. Development of a mouse model for sporadic and metastatic colon tumors and its use in assessing drug treatment. Proc. Natl. Acad. Sci. USA 107, 1565–1570 (2010).

    Article  CAS  Google Scholar 

  12. Hadac, J.N. et al. Colon tumors with the simultaneous induction of driver mutations in APC, KRAS, and PIK3CA still progress through the adenoma-to-carcinoma sequence. Cancer Prev. Res. (Phila.) 8, 952–961 (2015).

    Article  CAS  Google Scholar 

  13. Fu, X.Y., Besterman, J.M., Monosov, A. & Hoffman, R.M. Models of human metastatic colon cancer in nude mice orthotopically constructed by using histologically intact patient specimens. Proc. Natl. Acad. Sci. USA 88, 9345–9349 (1991).

    Article  CAS  Google Scholar 

  14. Takahashi, T., Morotomi, M. & Nomoto, K. A novel mouse model of rectal cancer established by orthotopic implantation of colon cancer cells. Cancer Sci. 95, 514–519 (2004).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  16. Bhullar, J.S. et al. A novel nonoperative orthotopic colorectal cancer murine model using electrocoagulation. J. Am. Coll. Surg. 213, 54–60, discussion 60–61 (2011).

    Article  Google Scholar 

  17. Zigmond, E. et al. Utilization of murine colonoscopy for orthotopic implantation of colorectal cancer. PLoS One 6, e28858 (2011).

    Article  CAS  Google Scholar 

  18. Sánchez-Rivera, F.J. & Jacks, T. Applications of the CRISPR-Cas9 system in cancer biology. Nat. Rev. Cancer 15, 387–395 (2015).

    Article  Google Scholar 

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

    Article  Google Scholar 

  20. Maddalo, D. & Ventura, A. Somatic engineering of oncogenic chromosomal rearrangements: a perspective. Cancer Res. 76, 4918–4923 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. Matano, M. et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat. Med. 21, 256–262. http://dx.doi.org/10.1038/nm.3802 (2015).

    Article  CAS  Google Scholar 

  23. Beyaz, S. et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature 531, 53–58 (2016).

    Article  CAS  Google Scholar 

  24. Pinto, D., Robine, S., Jaisser, F., El Marjou, F.E. & Louvard, D. Regulatory sequences of the mouse villin gene that efficiently drive transgenic expression in immature and differentiated epithelial cells of small and large intestines. J. Biol. Chem. 274, 6476–6482 (1999).

    Article  CAS  Google Scholar 

  25. Barker, N. et al. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457, 608–611 (2009).

    Article  CAS  Google Scholar 

  26. Schepers, A.G. et al. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science 337, 730–735 (2012).

    Article  CAS  Google Scholar 

  27. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  CAS  Google Scholar 

  28. Schwank, G. et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 13, 653–658 (2013).

    Article  CAS  Google Scholar 

  29. Heckl, D. et al. Generation of mouse models of myeloid malignancy with combinatorial genetic lesions using CRISPR-Cas9 genome editing. Nat. Biotechnol. 32, 941–946 (2014).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  32. Yui, S. et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5+ stem cell. Nat. Med. 18, 618–623 (2012).

    Article  CAS  Google Scholar 

  33. Fukuda, M. et al. Small intestinal stem cell identity is maintained with functional Paneth cells in heterotopically grafted epithelium onto the colon. Genes Dev. 28, 1752–1757 (2014).

    Article  CAS  Google Scholar 

  34. O'Rourke, K.P. et al. Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer. Nat. Biotechnol. http://dx.doi.org/10.1038/nbt.3837 (2017).

  35. Le, D.T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015).

    Article  CAS  Google Scholar 

  36. Li, Y. et al. Induction of functional human macrophages from bone marrow promonocytes by M-CSF in humanized mice. J. Immunol. 191, 3192–3199 (2013).

    Article  CAS  Google Scholar 

  37. Sharpless, N.E. & Depinho, R.A. The mighty mouse: genetically engineered mouse models in cancer drug development. Nat. Rev. Drug Discov. 5, 741–754 (2006).

    Article  CAS  Google Scholar 

  38. Roper, J. et al. Combination PI3K/MEK inhibition promotes tumor apoptosis and regression in PIK3CA wild-type, KRAS mutant colorectal cancer. Cancer Lett. 347, 204–211 (2014).

    Article  CAS  Google Scholar 

  39. van de Wetering, M. et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 161, 933–945 (2015).

    Article  CAS  Google Scholar 

  40. Fujii, M. et al. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell 18, 827–838 (2016).

    Article  CAS  Google Scholar 

  41. Kuraguchi, M. et al. Adenomatous polyposis coli (APC) is required for normal development of skin and thymus. PLoS Genet. 2, e146 (2006).

    Article  Google Scholar 

  42. Johnson, L. et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature 410, 1111–1116 (2001).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  44. Huch, M. et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 494, 247–250 (2013).

    Article  CAS  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  Google Scholar 

  46. Shultz, L.D. et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. J. Immunol. 174, 6477–6489 (2005).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  49. Vidigal, J.A. & Ventura, A. Rapid and efficient one-step generation of paired gRNA CRISPR-Cas9 libraries. Nat. Commun. 6, 8083 (2015).

    Article  CAS  Google Scholar 

  50. Miyoshi, H. & Stappenbeck, T.S. In vitro expansion and genetic modification of gastrointestinal stem cells in spheroid culture. Nat. Protoc. 8, 2471–2482 (2013).

    Article  CAS  Google Scholar 

  51. Koo, B.-K., Sasselli, V. & Clevers, H. Retroviral gene expression control in primary organoid cultures. Curr. Protoc. Stem Cell Biol. 27, 5A.6 (2013).

    Article  Google Scholar 

  52. Roper, J. et al. The dual PI3K/mTOR inhibitor NVP-BEZ235 induces tumor regression in a genetically engineered mouse model of PIK3CA wild-type colorectal cancer. PLoS One 6, e25132 (2011).

    Article  CAS  Google Scholar 

  53. Yilmaz, Ö.H. et al. mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake. Nature 486, 490–495 (2012).

    Article  CAS  Google Scholar 

  54. Tammela, T. et al. Angiopoietin-1 promotes lymphatic sprouting and hyperplasia. Blood 105, 4642–4648 (2005).

    Article  CAS  Google Scholar 

  55. Chen, D. et al. Rapid discovery of potent siRNA-containing lipid nanoparticles enabled by controlled microfluidic formulation. J. Am. Chem. Soc. 134, 6948–6951 (2012).

    Article  CAS  Google Scholar 

  56. Dong, Y. et al. Lipopeptide nanoparticles for potent and selective siRNA delivery in rodents and nonhuman primates. Proc. Natl. Acad. Sci. USA 111, 3955–3960 (2014).

    Article  CAS  Google Scholar 

  57. Abouelhoda, M.I., Kurtz, S. & Ohlebusch, E. Replacing suffix trees with enhanced suffix arrays. J. Discrete Algorithms 2, 53–86 (2004).

    Article  Google Scholar 

  58. Smith, T.F. & Waterman, M.S. Identification of common molecular subsequences. J. Mol. Biol. 147, 195–197 (1981).

    Article  CAS  Google Scholar 

  59. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  Google Scholar 

  60. Döring, A., Weese, D., Rausch, T. & Reinert, K. SeqAn an efficient, generic C+ library for sequence analysis. BMC Bioinformatics 9, 11 (2008).

    Article  Google Scholar 

  61. Zhao, M., Lee, W.-P., Garrison, E.P. & Marth, G.T. SSW library: an SIMD Smith-Waterman C/C. library for use in genomic applications. PLoS One 8, e82138 (2013).

    Article  Google Scholar 

  62. Thorvaldsdóttir, H., Robinson, J.T. & Mesirov, J.P. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the Howard Hughes Medical Institute (T.J., R.O.H.), NIH (K08 CA198002, J.R.; K99 CA187317, T.T.; Ö.H.Y.; R01CA211184, Ö.H.Y.; U54-CA163109, R.O.H.), the Sigrid Juselius Foundation (Ö.H.Y), the Maud Kuistila Foundation and the Hope Funds for Cancer Research (Ö.H.Y.), Department of Defense (PRCRP Career Development Award CA120198; J.R.), and the V Foundation V Scholar Award (J.R. and Ö.H.Y.), the Sidney Kimmel Scholar Award (Ö.H.Y.), the Pew-Stewart Trust Scholar Award (Ö.H.Y.), the Koch Institute Frontier Research Program through the Kathy and Curt Marble Cancer Research Fund (Ö.H.Y.), American Federation of Aging Research (AFAR, Ö.H.Y.), the Hope Funds for Cancer Research (T.T.), the Metastasis/Cancer Research Postdoc fellowship from the MIT Ludwig Center for Molecular Oncology Research (S.R.), the Bloodwise (UK) Visiting Fellowship Grant 14043 (A.R.), and by the Koch Institute Support (core) Grant P30-CA14051 from the National Cancer Institute. We thank T. Papagiannakopoulos for helpful discussions; Y. Soto-Feliciano for help and expertise with massively parallel sequencing; K. Bedrossian for assistance with human colorectal cancer sample collection at Tufts Medical Center; the Swanson Biotechnology Center at the Koch Institute for technical support, specifically K. Cormier and C. Condon at the Hope Babette Tang (1983) Histology Facility; S. Holder for histology support; and Y.D. Soo and the Peterson (1957) Nanotechnology Materials Core Facility for assistance with electron microscopy. L-WRN cells were a kind gift from T. Stappenbeck, Washington University.

Author information

Authors and Affiliations

Authors

Contributions

J.R. and T.T. performed all experiments and participated in their design and interpretation with T.J. and Ö.H.Y. J.R. and Ö.H.Y. developed and optimized the colonoscopy mucosal injection technique, with assistance from D.K. and P.K. J.R. wrote the paper with support from T.T. and Ö.H.Y. N.M.C. contributed to study design, plasmid design, lentivirus production, and mucosal injections. F.J.S.-R. contributed to plasmid and study design, and performed massively parallel sequencing. M.A., Y.K.P., R.N., R.A., X.L., D.K., K.W., S.R., and A.A. assisted with mucosal injections, mouse and human organoid derivation, molecular biology, and immunohistochemistry. A.R. assisted with humanized mouse experiments. M.A.O. designed and synthesized lipid nanoparticles for mRNA encapsulation. G.E., E.T.S., M.S.T., A.J.B., Y.S., J.Y., L.C., V.D., and L.Z. assisted with human CRC specimen collection. S.B. performed organoid qRT-PCR. A.B. performed bioinformatics analysis. R.L., J.L., J.C., P.N.T., R.O.H., and T.J. participated in interpretation of results. Ö.H.Y. supervised all aspects of the study.

Corresponding author

Correspondence to Ömer H Yilmaz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Mucosal injection of PGK::Cre lentivirus results in recombination in crypt base stem cells and tumorigenesis in Apcfl/fl mice.

(a) Schematic and matching optical colonoscopy images of colonoscopy-guided mucosal injection. Injection of 50 μl of liquid results in a “bubble” between the epithelium and lamina propria. (b) Bioluminescence (IVIS) and tdTomato fluorescence imaging of Rosa26LSL-tdTomato/LSL-Luciferase mice (N=2) two days after mucosal delivery of adenovirus expressing Cre-recombinase (Ad5CMV::Cre, titer 300,000 TU/μl). Arrowheads indicate recombined areas. (c) EpCAM and tdTomato immunofluorescence imaging of colon sections from Rosa26LSL-tdTomato/+ mice seven days following colonoscopy-guided mucosal injection of a lentiviral vector expressing Cre recombinase (lenti-PGK::Cre, titer 100,000 TU/μl). Arrows indicate recombination in crypt base stem cells. Arrowhead shows recombination in stromal cells (N=5 mice). (d) PGK::Cre-induced tumors in Apcfl/fl mice (titer 30,000 TU/μl). Tumors are visualized with colonoscopy (dotted line), hematoxylin and eosin histology, and β-catenin immunohistochemistry. Histology images are 20X and insets are 60X (Scale bar: 200 μm). (R26: Rosa26; PGK: human phosphoglycerate kinase-1 promoter; H&E: hematoxylin and eosin; T: tumor; N: normal).

Supplementary Figure 2 In situ deletion of Apc using floxed alleles results in tumorigenesis.

(a) Tumors in Apcfl/fl mice after colonoscopy-guided mucosal injection of Ad5CMV::Cre (titer: 300,000 TU/μl). Tumorigenesis is indicated by colonoscopy, necropsy, hematoxylin and eosin (H&E) histology, and β-catenin immunohistochemistry. (b) tdTomato immunofluorescence imaging of Rosa26LSL-tdTomato;VillinCreER mice injected via colonoscopy with 100 μM 4-hydroxytamoxifen. Arrows indicate recombination in epithelial cells. (c) Tumors in the distal colons of Apcfl/fl;VillinCreER mice following injection of 100 μM 4-hydroxytamoxifen (colonoscopy, H&E staining, and β-catenin immunohistochemistry). H&E staining, biopsy of an in vivo tumor. (d) Tumorigenesis in Apcfl/fl;Lgr5eGFP-creER/+ mice after mucosal delivery of 100 μM 4-hydroxytamoxifen (colonoscopy, brightfield/GFP necropsy, H&E staining, and GFP immunofluorescence). Arrows indicate Lgr5+/GFP+ adenoma cells. Tumors are indicated with dotted lines. Histology images are 20X and insets are 60X (Scale bar: 200 μm) (tdT or tdTom:tTdTomato; R26: Rosa26; N: normal; T: tumor).

Supplementary Figure 3 Mucosal delivery of lipid nanoparticle-encapsulated Cre mRNA induces tumor formation in Apcfl/fl mice

(a) Lipid nanoparticles are composed of mRNA and an amine-containing ionizable lipid that electrostatically complexes with the negatively charged mRNA and can both help facilitate cellular uptake and endosomal escape of the mRNA to the cytoplasm. In addition, a phospholipid is added that provides structure to the lipid nanoparticle and can assist in endosomal escape. Cholesterol is added to enhance the stability and promote membrane fusion. A lipid-anchored polyethylene glycol is also added to stabilize the particles and reduce nonspecific interactions. As represented in the schematic, the LNPs assume a multi-laminar spherical shape. (b) Cryogenic transmission electron microscopy image of lipid nanoparticles (arrowhead) in a buffered solution on a lacey copper grid coated with a continuous carbon film (Scale bar: 200 nm). (c) Bioluminescence (IVIS) imaging two days after colonoscopy-guided mucosal delivery into wild-type mice with luciferase mRNA alone (CL) or lipid nanoparticle-encapsulated luciferase mRNA (LNP). Arrowhead denotes luminescence signal at mucosal injection sites. (d) EpCAM and tdTomato immunofluorescence imaging of colon sections from Rosa26LSL-tdTomato/+ mice seven days after colonoscopy-guided mucosal injection of lipid nanoparticle-encapsulated Cre mRNA (N=3). Arrows indicate recombined crypt cells, and arrowheads point to recombination in stromal cells. Injection of Cre mRNA alone did not induce recombination in Rosa26LSL-tdTomato/+ recipient colons (N=3 mice, not shown). (e) Tumors following delivery of lipid nanoparticle-encapsulated Cre mRNA to the colon mucosa of Apcfl/fl mice are demonstrated by colonoscopy (dotted line), H&E staining, and β-catenin immunohistochemistry. (PEG: polyethylene glycol; TEM: transmission electron microscopy; TdTom: TdTomato; R26: Rosa26). Histology images are 20X and insets are 60X (Scale bar: 200 μm).

Supplementary Figure 4 Analysis of mutations introduced in vivo by CRISPR-Cas9 at the Apc locus.

Tumors initiated by disruption of Apc in vivo in the colon epithelium were subjected to massively parallel sequencing of a 200 base pair (bp) genomic region comprising 100 bp on either side of the sgApc binding site. (a) Tumor containing 4 different types of Apc mutations induced by U6::sgApc-EFS::Cas9-P2A-GFP lentivirus infection in a wild-type mouse (representative results from eight analyzed tumors); (b) Representative 6-week-old and 1-year-old tumors containing multiple types of Apc mutations induced by U6::sgApc-EFS::turboRFP in Rosa26LSL-Cas9-eGFP/+;VillinCreER mice. Blue in the pie charts represents the fraction of wild-type reads, most likely arising from wild-type stroma present in the tumors. Alignments of the wild-type locus sequence and the most common mutated alleles are shown below the pie charts. The 10 most common mutant alleles among 25 six-week-old samples and five one-year-old samples are described. Locations of the guide RNA target sequence and PAM sequence (light orange highlight) are indicated. Each mutant allele is characterized by its frequency in the total pool of mutant reads or samples, the type of event (DEL: deletion, INS: insertion), event size (in bp), and impact on the coding sequence (FS: frameshift mutation; NFS: non-frameshift mutation). Deletions are indicated by red dashes and insertions by red triangles. Locations of the guide RNA target sequence, PAM sequence, and mutant allele frequencies are described.

Supplementary Figure 5 CRISPR-Cas9-based in situ Apc and Trp53 editing in the colonic epithelium induces adenoma formation.

(a) One-year-old tumors in tamoxifen-treated Rosa26LSL-Cas9-eGFP/+;VillinCreER mice that were injected under colonoscopy guidance with U6::sgApc-EFS::turboRFP lentivirus (titer: 10,000 TU/μl). Tumors were seen with white light/GFP/turboRFP fluorescence colonoscopy and necropsy. Immunofluorescence for GFP, turboRFP, and β-catenin demonstrate GFP+ adenoma (arrows) and limited turboRFP expression in stromal cells (arrowheads). (b) H&E staining of six-week-old vs. one-year-old tumors from tamoxifen-treated Rosa26LSL-Cas9-eGFP/+;VillinCreER mice that were injected with U6::sgApc-EFS::turboRFP lentivirus. (c) Colon tumors from Apcfl/fl;Rosa26LSL-Cas9-eGFP/+ mice injected under colonoscopy guidance with U6::sgTrp53-CMV::Cre lentivirus (titer: 10,000 TU/μl). Tumors were imaged by white light/GFP fluorescence colonoscopy and necropsy as well as hematoxylin and eosin (H&E) histology. (d) Tumors in tamoxifen-treated Rosa26LSL-Cas9-eGFP/+;VillinCreER mice that received mucosal delivery of hU6::sgApc-sU6::sgTrp53-EFS::turboRFP lentivirus (titer: 10,000 TU/μl). Tumors were seen with white light/GFP/turboRFP fluorescence colonoscopy and necropsy and examined with H&E staining. Histology images are 20X and insets are 60X (Scale bar: 200 μm). (tRFP: turboRFP, R26: Rosa26).

Supplementary Figure 6 Analysis of mutations introduced in vivo by CRISPR-Cas9 at the Apc and Trp53 loci.

(a) Tumors were induced in Apcfl/fl;R26LSL-Cas9-eGFP/+ mice by mucosal injection of U6::sgTrp53-CMV::Cre lentivirus, then subjected to massively parallel sequencing of a 200 base pair (bp) genomic region consisting of 100 bp on either side of the sgTrp53 binding site. (b) Mucosal delivery of hU6::sgApc-sU6::sgTrp53-EFS::turboRFP lentivirus into Rosa26LSL-Cas9-eGFP/+;VillinCreER mice treated with tamoxifen generated tumors that were then subjected to massively parallel sequencing of a 200 bp genomic region consisting of 100 bp on either side of the loci in Apc and Trp53 targeted by the respective sgRNAs. Blue in the pie charts represents the fraction of wild-type reads, most likely arising from wild-type stroma present in the tumors. Alignments of the wild-type locus sequence and the 10 most common mutated alleles in a representative sample and in three tumors are shown. Locations of the guide RNA target sequence and PAM sequence (light orange highlight) are indicated. Each mutant allele is characterized by its frequency in the total pool of mutant reads or samples, the type of event (DEL: deletion, INS: insertion), event size (in bp), and impact on the coding sequence (FS: frameshift mutation; NFS: non-frameshift mutation). Deletions are indicated by red dashes and insertions by red triangles. Locations of the guide RNA target sequence, PAM sequence, and mutant allele frequencies are described.

Supplementary Figure 7 Orthotopic engraftment of intestinal organoids.

(a) Orthotopic transplantation of small intestinal organoids derived from tamoxifen-treated Rosa26LSL-tdTomato/+;VillincreER mice. tdTomato+ organoids are visualized in vivo with white light colonoscopy and tdTomato fluorescence colonoscopy. Mice received a pulse of 5-ethynyl-2’-deoxyuridine (EdU) four hours before sacrifice. Arrowheads on immunofluorescence images indicate tdTomato+ intestinal organoids in the lamina propria of the recipient mice. Arrows point to lysozyme positive Paneth cells and EdU positive proliferating cells. Orthotopic transplantation was successful in all three mice receiving two organoid injections each. (b) Tumors in mice transplanted with Apcfl/flVillincreER intestinal organoids and treated with tamoxifen. Tumors are imaged with colonoscopy, necropsy, hematoxylin and eosin histology, and β-catenin immunohistochemistry (N=2). Tumors are indicated with dotted lines. (Scale bar: 200 μm). Dotted lines indicate tumors. (EdU: 5-ethynyl-2’-deoxyuridine; NSG: nod SCID gamma; H&E: hematoxylin and eosin).

Supplementary Figure 8 Orthotopic transplantation of murine colorectal cancer cells and organoids results in tumor formation.

(a) Tumors in C57BL/6 mice following orthotopic transplantation of a murine CRC cell line derived from a Cre-excised ApcΔ/Δ;KrasG12D/+Trp53Δ/Δ (AKP) genetically engineered tumor. Tumors are visualized with colonoscopy, hematoxylin and eosin (H&E) stain, and β-catenin immunohistochemistry. Arrow indicates invasion of the muscularis propria. (b) Wild-type C57BL/6 intestinal organoids infected with U6::sgApc-EFS::Cas9-P2A-GFP lentivirus, then selected in media without Wnt agonists. (c) sgApc intestinal organoids were subjected to massively parallel sequencing of a 200 base pair (bp) genomic region comprising 100 bp on either side of the sgRNA binding site. Locations of the guide RNA target sequence and PAM sequence (light orange highlight) are indicated. Each mutant allele is characterized by its frequency in the total pool of mutant reads, the type of event (DEL: deletion, INS: insertion), event size (in bp), and impact on the coding sequence (FS: frameshift mutation; NFS: non-frameshift mutation). Deletions are indicated by red dashes and insertions by red triangles. The ten most common mutated sequences are listed. (d) RT-PCR for selected Wnt target genes in sgApc small intestinal and colon organoids. (e) Tumors in C57BL/6 mice following orthotopic engraftment of sgApc organoids (colonoscopy, necropsy, H&E stain, and β-catenin immunohistochemistry). (f) Tumors in C57BL/6 mice transplanted with AKP colon organoids (colonoscopy, necropsy, H&E stain, β-catenin immunohistochemistry, and trichrome stain). Arrows indicate invasion of the muscularis propria. Arrowheads show desmoplastic reaction. (g) Tumors following orthotopic transplantation of Apcfl/fl;Rosa26LSL-Cas9-eGFP/+ colon organoids infected with U6::sgTrp53-CMV::Cre lentivirus (white light/GFP fluorescence colonoscopy, GFP/β-catenin immunofluorescence). Histology images are 20X and insets are 60X (Scale bar: 200 μm). Dotted lines indicate tumors. (R26: Rosa26; N: normal; T: tumor).

Supplementary Figure 9 Analysis of mutations introduced by CRISPR-Cas9 at the sgApc locus in wild-type colon organoids and organoid orthotopic transplant tumors.

(a) Wild-type colon organoids were infected with U6::sgApc-EFS::Cas9-P2A-GFP lentivirus, then subjected to massively parallel sequencing of a 200 base pair (bp) genomic region comprising 100 bp on either side of the sgApc binding site. Alignments of the wild-type locus sequence and all 18 mutant reads are shown next to the pie chart. (b) These Apc-null organoids were orthotopically transplanted into recipient NSG mice. Eight weeks after transplantation, three samples each from two tumors were sequenced at the sgApc locus, as described above. Alignments of the wild-type locus sequence and the 10 most common mutant reads are shown. (c) Summary of the 10 most common Apc mutations in the six orthotopic transplant samples from Tumors 1 and 2. Locations of the guide RNA target sequence and PAM sequence (light orange highlight) are indicated. Each mutant allele is characterized by its frequency in the total pool of mutant reads or samples, the type of event (DEL: deletion, INS: insertion), event size (in bp), and impact on the coding sequence (FS: frameshift mutation; NFS: non-frameshift mutation). Deletions are indicated by red dashes and insertions by red triangles.

Supplementary Figure 10 Analysis of mutations introduced by CRISPR-Cas9 at the Trp53 locus in Apcfl/fl;R26LSL-Cas9-eGFP/+ colon organoids and organoid orthotopic transplant tumors.

(a) Apcfl/fl;R26LSL-Cas9-eGFP/+ colon organoids were infected with U6::sgTrp53-CMV::Cre lentivirus, and then subjected to massively parallel sequencing of a 200 bp genomic region comprising 100 bp on either side of the sgTrp53 binding site. (b) ApcΔ/Δ, Trp53-edited organoids were then orthotopically transplanted into recipient NSG mice to form tumors that were similarly sequenced at the sgTrp53 locus (N=3; data from a representative tumor are shown). Blue in the pie charts represents the fraction of wild-type reads, most likely arising from wild-type stroma present in the tumors. Alignments of the wild-type locus sequence and the 10 most commonly mutated sequences are shown below the pie charts. Locations of the guide RNA target sequence and PAM sequence (light orange highlight) are described. Each mutant allele is characterized by its frequency in the total pool of mutant reads, the type of event (DEL: deletion, INS: insertion), event size (in bp), and impact on the coding sequence (FS: frameshift mutation; NFS: non-frameshift mutation). Deletions are indicated by red dashes and insertions by red triangles.

Supplementary Figure 11 Orthotopic engraftment of human colorectal cancer cell lines and patient-derived tumors.

Tumors initiated by orthotopic engraftment of (a) LS174 human CRC cells, (b) HT29 human CRC cells, and (c) patient-derived CRC (colonoscopy, necropsy, hematoxylin and eosin staining, and β-catenin immunohistochemistry). Arrows indicate invasion invasion into the muscularis propria. Histology images are 20X and insets are 60X (Scale bar: 200 μm). Dotted lines indicate tumors. (N: normal; T: tumor; NSG: nod SCID gamma; H&E: hematoxylin and eosin).

Supplementary Figure 12 Orthotopic engraftment of patient-derived organoids.

(a) Patient A colorectal cancer hematoxylin and eosin (H&E) histology and corresponding patient-derived organoids. (b) Human CDX2 in situ hybridization in tumors derived from orthotopic transplantation of Patient A CRC organoids. (c) Lynch Syndrome colorectal cancer H&E histology (characterized by a high level of microsatellite instability and tumor lymphocytic infiltration; Patient B) and corresponding tumor organoids engrafted into the flanks of recipient NSG mice (H&E stain, β-catenin immunohistochemistry). (d) Tumors derived from orthotopic transplantation of Patient B organoids (H&E histology, β-catenin immunohistochemistry). Arrows indicate invasion of the muscularis propria. (e) Liver metastasis after orthotopic transplantation of Patient B tumor organoids (H&E staining, CDX2/human Keratin20 immunohistochemistry). (f) Analysis of human CD3 T cells in Patient B-derived tumors orthotopically transplanted into NSG mice with a reconstituted human immune system (humanized NSG). These tumors and their exhibited human CD3 T cell infiltration by immunohistochemistry and flow cytometry, with enrichment for human memory T cells (CD45RO memory / CD45 RA naive ratio = 26) compared to spleen control (memory / naive ratio = 2.2). Histology images are 20X and insets are 60X (Scale bar: 200 μm). Dotted lines indicate tumors. (ISH: in situ hybridization; N: normal; T: tumor; S: stromal lymphocytes; NSG: nod SCID gamma; hKeratin20: human Keratin20; MSI-H, microsatellite instability-high).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 and Supplementary Tables 1 and 2 (PDF 6327 kb)

Colonoscopy-guided mucosal injection.

During optical colonoscopy, a 33-gauge needle with 45-degree bevel is inserted into the working channel of the endoscopy and directed to the colonic mucosa without passing through the muscularis propria or serosa. 50-100 μl of liquid (containing virus or organoids, for example) is then rapidly injected to produce a mucosal bubble. (MPG 20682 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roper, J., Tammela, T., Cetinbas, N. et al. In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis. Nat Biotechnol 35, 569–576 (2017). https://doi.org/10.1038/nbt.3836

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3836

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