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:

Sequential cancer mutations in cultured human intestinal stem cells

Subjects

Abstract

Crypt stem cells represent the cells of origin for intestinal neoplasia. Both mouse and human intestinal stem cells can be cultured in medium containing the stem-cell-niche factors WNT, R-spondin, epidermal growth factor (EGF) and noggin over long time periods as epithelial organoids that remain genetically and phenotypically stable. Here we utilize CRISPR/Cas9 technology for targeted gene modification of four of the most commonly mutated colorectal cancer genes (APC, P53 (also known as TP53), KRAS and SMAD4) in cultured human intestinal stem cells. Mutant organoids can be selected by removing individual growth factors from the culture medium. Quadruple mutants grow independently of all stem-cell-niche factors and tolerate the presence of the P53 stabilizer nutlin-3. Upon xenotransplantation into mice, quadruple mutants grow as tumours with features of invasive carcinoma. Finally, combined loss of APC and P53 is sufficient for the appearance of extensive aneuploidy, a hallmark of tumour progression.

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: Inactivation of APC and P53 in human intestinal organoids.
Figure 2: KRASG12D/APCKO/P53KO/SMAD4KO organoids grow in the absence of stem-cell-niche factors in vitro.
Figure 3: Quadruple-mutant organoids grow as invasive carcinomas in vivo.
Figure 4: Progressive CIN and aneuploidy upon introduction of CRC mutations.

Similar content being viewed by others

Accession codes

Primary accessions

European Nucleotide Archive

Data deposits

Sequencing data have been deposited in the EMBL European Nucleotide Archive under accession number ERP009240.

References

  1. Fearon, E. R. & Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 61, 759–767 (1990)

    Article  CAS  Google Scholar 

  2. Fearon, E. R. Molecular genetics of colorectal cancer. Annu. Rev. Pathol. 6, 479–507 (2011)

    Article  CAS  Google Scholar 

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

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

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

  6. Zhu, L. et al. Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation. Nature 457, 603–607 (2009)

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  Google Scholar 

  8. Sato, T. et al. Single Lgr5 stem cell build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009)

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  Google Scholar 

  10. Sato, T. & Clevers, H. Primary mouse small intestinal epithelial cell cultures. Methods Mol. Biol. 945, 319–328 (2013)

    Article  Google Scholar 

  11. Jung, P. et al. Isolation and in vitro expansion of human colonic stem cells. Nature Med. 17, 1225–1227 (2011)

    Article  CAS  Google Scholar 

  12. 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  Google Scholar 

  13. 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  ADS  Google Scholar 

  14. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013)

    Article  CAS  ADS  Google Scholar 

  15. Cho, S. W., Kim, S., Kim, J. M. & Kim, J. S. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nature Biotechnol. 31, 230–232 (2013)

    Article  CAS  Google Scholar 

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

    Article  CAS  ADS  Google Scholar 

  17. 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 

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

    Article  CAS  ADS  Google Scholar 

  19. Shi, Y., Hata, A., Lo, R. S., Massaqué, J. & Pavletich, N. P. A structural basis for mutational inactivation of the tumour suppressor Smad4. Nature 388, 87–93 (1997)

    Article  CAS  ADS  Google Scholar 

  20. Chacko, B. M. et al. Structural basis of heteromeric smad protein assembly in TGF-β signaling. Mol. Cell 15, 813–823 (2004)

    Article  CAS  Google Scholar 

  21. Fleming, N. I. et al. SMAD2, SMAD3 and SMAD4 mutations in colorectal cancer. Cancer Res. 73, 725–735 (2013)

    Article  CAS  Google Scholar 

  22. Rajagopalan, H., Nowak, M. A., Vogelstein, B. & Lengauer, C. The significance of unstable chromosomes in colorectal cancer. Nature Rev. Cancer 3, 695–701 (2003)

    Article  CAS  Google Scholar 

  23. Fodde, R. et al. Mutations in the APC tumour suppressor gene cause chromosomal instability. Nature Cell Biol. 3, 433–438 (2001)

    Article  CAS  Google Scholar 

  24. Bomme, L. et al. Clonal karyotypic abnormalities in colorectal adenomas: clues to the early genetic events in the adenoma-carcinoma sequence. Genes Chromosom. Cancer 10, 190–196 (1994)

    Article  CAS  Google Scholar 

  25. Herbergs, J. et al. Chromosome aberrations in adenomas of the colon. Proof of trisomy 7 in tumor cells by combined interphase cytogenetics and immunocytochemistry. Int. J. Cancer 57, 781–785 (1994)

    Article  CAS  Google Scholar 

  26. Ried, T. et al. Comparative genomic hybridization reveals a specific pattern of chromosomal gains and losses during the genesis of colorectal tumors. Genes Chromosom. Cancer 15, 234–245 (1996)

    Article  CAS  Google Scholar 

  27. Van de Wetering, M. et al. Prospective derivation of a Living Organoid Biobank of colorectal cancer patients. Cellhttp://dx.doi.org/10.1016/j.cell.2015.03.053 (in the press)

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

    Article  CAS  Google Scholar 

  29. Kim, K. A. et al. Mitogenic influence of human R-spondin1 on the intestinal epithelium. Science 309, 1256–1259 (2005)

    Article  CAS  ADS  Google Scholar 

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

    Article  CAS  Google Scholar 

  31. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009)

    Article  CAS  Google Scholar 

  32. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nature Genet. 43, 491–498 (2011)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank H. M. Rodermond for help with in vivo transplantation assays and members of the contributing laboratories for support. We thank A. Pronk, W. van Houdt and J. van Gorp for facilitating human colon tissue. We are grateful for support from the following: The Netherlands Organisation for Scientific Research (NWO-ZonMw) VENI grant to J.D. (91614138); University of Amsterdam (2012-5735) and The Dutch Digestive Diseases Foundation (MLDS) (FP13-07) to C.Z. and J.P.M.; Netherlands Institute of Regenerative Medicine (N.S. and G.S.); Dutch Cancer Society (KWF) (KWF/PF-HUBR 2007-3956 for H.B.; KWF Fellowship UU2013-6070 for H.J.S.); Stand Up to Cancer/Stichting Vrienden van het Hubrecht (M.v.d.W.); NWO-ZonMw (116.005.002 for R.v.B.); and the CancerGenomics.nl (NWO Gravitation) program.

Author information

Authors and Affiliations

Authors

Contributions

J.D. and H.C. conceived the project and wrote the manuscript. J.D. engineered and characterized all mutant organoid lines. R.H.v.J., B.P., H.J.S., R.M.O. and G.J.P.L.K. designed and performed live-cell imaging experiments. C.Z. and J.P.M. performed in vivo transplantation assays. R.v.B. and E.C. performed off-target analyses. J.D. performed karyotyping. A.B. made karyograms. G.J.O. staged subcutaneous tumours. H.B. and J.K. performed immunohistochemistry. N.S. optimized matrix for organoid growth. G.S. designed APC sgRNAs. M.v.d.W. established normal human colon organoid line. M.L. helped genotype the mutant small intestinal organoids.

Corresponding author

Correspondence to Hans Clevers.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Introducing inactivating mutations in the APC and P53 genes in human intestinal organoids using CRISPR/Cas9.

a, Schematic representation of the targeted exon of the human APC (left) and P53 (right) loci and sequences of the designed sgRNAs. b, c, PCR amplification products of the mutated alleles of APC (b) and P53 (c) were obtained using primers flanking the targeted exon. Subsequent sequencing revealed indels at the expected locations. PAM sequences are underlined in red in wild-type sequences. Of note, the curved lines bridging the gaps in deleted alleles are drawn by the alignment software.

Extended Data Figure 2 KRASG12D, APCKO, P53KO and SMAD4KO mutation combinations in human intestinal organoids.

ac, PCR amplification products of the indicated genes of KRASG12D/APCKO (a), KRASG12D/APCKO/P53KO (b) and KRASG12D/APCKO/P53KO/SMAD4KO (c) organoids were obtained using primers flanking the targeted exon. Subsequent sequencing revealed indels at the expected locations. PAM sequences are underlined in red. Of note, the curved lines bridging the gaps in deleted alleles are drawn by the alignment software. d, Schematic representation of the targeted exon of the human SMAD4 locus and sequences of the designed sgRNAs. e, qRT–PCR for AXIN2 (top) and P21 (bottom) in the indicated organoid cultures. Top, the indicated organoid lines were cultured in the presence (WENR) or absence (EN) of WNT/R-spondin. Bottom, the indicated organoid lines were cultured in the presence or absence of nutlin-3 for 24 h. Expression was normalized to GAPDH. Horizontal bars represent mean of n = 3 independent experiments.

Extended Data Figure 3 Using CRISPR/Cas9-mediated genome editing to introduce APC, P53, KRASG12D and SMAD4 mutations in human colonic organoids.

ad, Using the strategies depicted in Figs 1a and 2a, d, APCKO, APCKO/P53KO (a), KRASG12D (b), KRASG12D/APCKO, KRASG12D/APCKO/P53KO, KRASG12D/APCKO/P53KO/SMAD4KO (c) and P53KO (d) mutant human colon organoids were generated. Experiment was performed at least three independent times for each mutation. e, qRT–PCR for AXIN2 in the indicated organoid lines cultured in the presence (WENR) or absence (EN) of WNT/R-spondin. Expression was normalized to GAPDH. Horizontal bars represent mean of n = 3 independent experiments. f, Western blot analysis of P53 and P21 expression in the indicated human colon organoid lines cultured in the presence or absence of nutlin-3. GAPDH, loading control. g, qRT–PCR for AXIN2 in the indicated organoid lines cultured in the presence (WENR) or absence (EN) of WNT/R-spondin. Expression was normalized to GAPDH. Horizontal bars represent mean of n = 3 independent experiments. h, Western blot analysis of SMAD4 and P53 expression in the indicated human colon organoid lines. Please note that quadruple-mutant clone 1 contains SMAD4 frameshift-inducing indels in both alleles whereas clone 2 contains a frameshift-inducing indel in one and an in-frame deletion in the other allele (reduced SMAD4 expression). GAPDH, loading control. Scale bars, 100 µm.

Extended Data Figure 4 Quadruple-mutant human intestinal organoids grow as tumours with features of invasive carcinoma in vivo.

a, Wild-type and all engineered human intestinal organoid lines were injected subcutaneously in immunodeficient mice. Mice injected with KRASG12D/APCKO/P53KO (triple) and KRASG12D/APCKO/P53KO/SMAD4KO (quadruple) organoids developed visible nodules. b, Tumour sizes were examined 8 weeks after transplantation. c, d, H&E (top left, bottom left), hKRT (top right, bottom middle) and Ki67 (bottom right) immunostainings on nodules isolated from triple- (c) and quadruple-mutant (d) injected mice. Triple-mutant organoids did engraft but remained small, showed only weak proliferation and had adenoma features (n = 3 mice). Quadruple-mutant-derived tumours were highly proliferative with features of invasive carcinoma (n = 13 mice). See Fig. 3 for more details. Scale bars, 100 µm.

Extended Data Figure 5 Histological analysis of triple- and quadruple-mutant organoids reveals morphological changes in vitro.

a, Representative H&E and Ki67 immunostainings on the indicated human small intestinal organoid lines (n = 4 independent experiments). b, Representative H&E and Ki67 immunostainings on the indicated human colon organoid lines (n = 3 independent experiments). c, Representative E-cadherin immunostainings on wild-type and quadruple-mutant human small intestinal organoids (n = 4 independent experiments). Asterisk indicates residual Matrigel. Scale bars, 100 µm.

Extended Data Figure 6 Progressive aneuploidy upon introduction of CRC mutations.

ad, Karyograms of APCKO (a), APCKO/P53KO (b), KRASG12D/APCKO/P53KO (c) and KRASG12D/APCKO/P53KO/SMAD4KO (d) organoids, showing extensive aneuploidy in organoids harbouring CRC mutations (20 spreads were analysed per line). Note the occurrence of trisomy 7 in APCKO and APCKO/P53KO (independent clones) organoids. M, marker chromosomes.

Extended Data Figure 7 Loss of both APC and P53 results in extensive CIN and aneuploidy.

a, APCKO, P53KO and APCKO/P53KO mutations were introduced in a second independent human intestinal organoid line. PCR amplification products of the mutated alleles of APC and P53 were obtained using primers flanking the targeted exon. Subsequent sequencing revealed frameshift-inducing indels at the expected locations. Left, APC genotyping; right, P53 genotyping. PAM sequences are underlined in red. Of note, the curved lines bridging the gaps in deleted alleles are drawn by the alignment software. b, Western blot analysis for P53 and P21 expression in the second human intestinal organoid line cultured in the presence or absence of nutlin-3. GAPDH, loading control. c, qRT–PCR for AXIN2 in the second human intestinal organoid line cultured in the presence (WENR) or absence (EN) of WNT/R-spondin. Expression was normalized to GAPDH. Horizontal bar represents mean of n = 3 independent experiments. d, Chromosome numbers were counted in the second human intestinal organoid lines. Graphs plot the percentage of cells with chromosome counts <44, 44–48 (normal) and >48 (at least 50 spreads were counted). e, As in d, but for indicated human colon organoid lines. f, Live-cell imaging was performed to monitor chromosome segregations in the indicated human small intestinal organoid lines. Graph shows the percentage of erroneous mitoses. Each dot represents the percentage of errors in one organoid. Horizontal bars represent median of all dots. A video is included of organoids depicted as dots with green outline (Supplementary Video 6). WT, wild type; KO, knockout. g, As in f, but for indicated human colon organoid lines. h, Western blot analysis of phospho-CHK1 and P53 expression in the indicated organoid lines treated with the DNA-damaging drug doxorubicin, or left untreated. GAPDH, loading control.

Extended Data Figure 8 Engineered mutant human colon organoids grow as invasive carcinomas in vivo.

a, Wild-type, triple- and quadruple-mutant human colon organoids were injected subcutaneously in immunodeficient mice. Nodules were counted 7 weeks after transplantation. b, Tumour sizes were examined 7 weeks after transplantation. c, Representative pictures of a ‘cystic’ triple-mutant (left) and ‘solid’ quadruple-mutant (right) tumour in immunodeficient mice. d, H&E (top left, bottom left), hKRT (top middle, bottom middle) and Ki67 (top right, bottom right) immunostainings on nodules isolated from triple-mutant-injected mice. Representative pictures of a well-differentiated carcinoma with limited invasive growth. The invasive growth has an expansive growth pattern with little tumour budding. n = 6 mice. e, As in d but for quadruple-mutant-derived tumours. Representative pictures of a poorly differentiated invasive carcinoma with frequent tumour budding at the invasive front (invasion of isolated or small aggregates of cells into the stroma is frequently observed (black arrowheads)). Invasive character is confirmed by the invasive growth into the underlying muscle tissue (asterisk, muscle tissue). n = 8 mice. Scale bars, 100 µm.

Extended Data Table 1 Introducing oncogenic mutations in human intestinal organoids using CRISPR/Cas9

Supplementary information

Supplementary Figure

This file contains Supplementary Figure 1, uncropped images of western blots. (PDF 1147 kb)

Supplementary Table 1

This table shows off-target analysis of KRAS, APC, P53 and SMAD4 sgRNAs. Off-targets were assessed by amplicon-based NGS sequencing of candidate off-target sites in the indicated organoid cultures. The columns in the table indicate the position of the candidate off-target sites (chromosome, start and end position), the alignment of the off-target sequence with the sequence of the sgRNA (indicated are bases that differ), the off-target score (a score of 1 indicates complete sequence similarity, the lower the score the more sequence differences), the mean base coverage per sample over the indicated off-target sequence, coordinates of identified small insertions and deletions (indels) in the indicated candidate off-target site and the variant allele frequencies (VAF) for the identified indels in the different samples, respectively. Only indels at the target sites for which the sgRNAs were designed (KRAS, APC, P53 and SMAD4; marked in yellow) were identified. One indel was observed at a candidate off-target site of the APC sgRNA in all samples (non-coding region). However, as the KRASG12D organoid line was not transfected with this sgRNA, this indel is not considered as an off-target effect. No additional indels were observed. (XLSX 45 kb)

Live cell imaging of chromosome segregations in human intestinal stem cell organoids

Representative live cell imaging experiments of wild-type (1), APCKO (2), APCKO/P53KO (3), KRASG12D/APCKO/P53KO (4) and RASG12D/APCKO/P53KO/SMAD4KO (5) organoids and P53KO in the second human intestinal organoid line (6). Organoids were subjected to confocal imaging for 16 – 20 hours. Shown are representative parts of this time window and organoids presented are depicted as green-lined dots in quantification plot Figure 4a. Upper right quadrant shows H2B-mNeon fluorescence after maximum-projection of 3D z-stacks; upper left represents same data, color-coded for depth (z), facilitating tracking of individual events; lower quadrants include transmitted light images with and without merged H2B-mNeon (green). Scale bars as indicated in video. (MP4 16929 kb)

Live cell imaging of chromosome segregations in human intestinal stem cell organoids

Representative live cell imaging experiments of wild-type (1), APCKO (2), APCKO/P53KO (3), KRASG12D/APCKO/P53KO (4) and KRASG12D/APCKO/P53KO/SMAD4KO (5) organoids and P53KO in the second human intestinal organoid line (6). Organoids were subjected to confocal imaging for 16 – 20 hours. Shown are representative parts of this time window and organoids presented are depicted as green-lined dots in quantification plot Figure 4a. Upper right quadrant shows H2B-mNeon fluorescence after maximum-projection of 3D z-stacks; upper left represents same data, color-coded for depth (z), facilitating tracking of individual events; lower quadrants include transmitted light images with and without merged H2B-mNeon (green). Scale bars as indicated in video. (MP4 18060 kb)

Live cell imaging of chromosome segregations in human intestinal stem cell organoids

Representative live cell imaging experiments of wild-type (1), APCKO (2), APCKO/P53KO (3), KRASG12D/APCKO/P53KO (4) and KRASG12D/APCKO/P53KO/SMAD4KO (5) organoids and P53KO in the second human intestinal organoid line (6). Organoids were subjected to confocal imaging for 16 – 20 hours. Shown are representative parts of this time window and organoids presented are depicted as green-lined dots in quantification plot Figure 4a. Upper right quadrant shows H2B-mNeon fluorescence after maximum-projection of 3D z-stacks; upper left represents same data, color-coded for depth (z), facilitating tracking of individual events; lower quadrants include transmitted light images with and without merged H2B-mNeon (green). Scale bars as indicated in video. (MP4 15960 kb)

Live cell imaging of chromosome segregations in human intestinal stem cell organoids

Representative live cell imaging experiments of wild-type (1), APCKO (2), APCKO/P53KO (3), KRASG12D/APCKO/P53KO (4) and KRASG12D/APCKO/P53KO/SMAD4KO (5) organoids and P53KO in the second human intestinal organoid line (6). Organoids were subjected to confocal imaging for 16 – 20 hours. Shown are representative parts of this time window and organoids presented are depicted as green-lined dots in quantification plot Figure 4a. Upper right quadrant shows H2B-mNeon fluorescence after maximum-projection of 3D z-stacks; upper left represents same data, color-coded for depth (z), facilitating tracking of individual events; lower quadrants include transmitted light images with and without merged H2B-mNeon (green). Scale bars as indicated in video. (MP4 19297 kb)

Live cell imaging of chromosome segregations in human intestinal stem cell organoids

Representative live cell imaging experiments of wild-type (1), APCKO (2), APCKO/P53KO (3), KRASG12D/APCKO/P53KO (4) and KRASG12D/APCKO/P53KO/SMAD4KO (5) organoids and P53KO in the second human intestinal organoid line (6). Organoids were subjected to confocal imaging for 16 – 20 hours. Shown are representative parts of this time window and organoids presented are depicted as green-lined dots in quantification plot Figure 4a. Upper right quadrant shows H2B-mNeon fluorescence after maximum-projection of 3D z-stacks; upper left represents same data, color-coded for depth (z), facilitating tracking of individual events; lower quadrants include transmitted light images with and without merged H2B-mNeon (green). Scale bars as indicated in video. (MP4 18622 kb)

Live cell imaging of chromosome segregations in human intestinal stem cell organoids

Representative live cell imaging experiments of wild-type (1), APCKO (2), APCKO/P53KO (3), KRASG12D/APCKO/P53KO (4) and KRASG12D/APCKO/P53KO/SMAD4KO (5) organoids and P53KO in the second human intestinal organoid line (6). Organoids were subjected to confocal imaging for 16 – 20 hours. Shown are representative parts of this time window and organoids presented are depicted as green-lined dots in quantification plot Figure 4a. Upper right quadrant shows H2B-mNeon fluorescence after maximum-projection of 3D z-stacks; upper left represents same data, color-coded for depth (z), facilitating tracking of individual events; lower quadrants include transmitted light images with and without merged H2B-mNeon (green). Scale bars as indicated in video. (MP4 17866 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Drost, J., van Jaarsveld, R., Ponsioen, B. et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 521, 43–47 (2015). https://doi.org/10.1038/nature14415

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14415

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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