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:

Mutant clones in normal epithelium outcompete and eliminate emerging tumours

Abstract

Human epithelial tissues accumulate cancer-driver mutations with age1,2,3,4,5,6,7,8,9, yet tumour formation remains rare. The positive selection of these mutations suggests that they alter the behaviour and fitness of proliferating cells10,11,12. Thus, normal adult tissues become a patchwork of mutant clones competing for space and survival, with the fittest clones expanding by eliminating their less competitive neighbours11,12,13,14. However, little is known about how such dynamic competition in normal epithelia influences early tumorigenesis. Here we show that the majority of newly formed oesophageal tumours are eliminated through competition with mutant clones in the adjacent normal epithelium. We followed the fate of nascent, microscopic, pre-malignant tumours in a mouse model of oesophageal carcinogenesis and found that most were rapidly lost with no indication of tumour cell death, decreased proliferation or an anti-tumour immune response. However, deep sequencing of ten-day-old and one-year-old tumours showed evidence of selection on the surviving neoplasms. Induction of highly competitive clones in transgenic mice increased early tumour removal, whereas pharmacological inhibition of clonal competition reduced tumour loss. These results support a model in which survival of early neoplasms depends on their competitive fitness relative to that of mutant clones in the surrounding normal tissue. Mutant clones in normal epithelium have an unexpected anti-tumorigenic role in purging early tumours through cell competition, thereby preserving tissue integrity.

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

Fig. 1: Most oesophageal tumours are lost over time.
Fig. 2: Sequencing of tumours and adjacent normal tissue.
Fig. 3: Elimination of early tumours by expansion of mutant clones in the adjacent normal epithelium.

Similar content being viewed by others

Data availability

Individual datasets are available in Supplementary Tables 111. The targeted DNA sequences from 10-day normal tissue and 10-day and 1-year tumours are available from the European Nucleotide Archive under accession ERP022921. Whole-exome sequences and whole-genome sequences of tumours are available from the European Nucleotide Archive under accessions ERP015469 and ERP122780, respectively. Source data are provided with this paper.

Code availability

Code is available at https://github.com/michaelhall28/Colom_lesions.

References

  1. Lee-Six, H. et al. The landscape of somatic mutation in normal colorectal epithelial cells. Nature 574, 532–537 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Martincorena, I. et al. Somatic mutant clones colonize the human esophagus with age. Science 362, 911–917 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Martincorena, I. et al. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348, 880–886 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Suda, K. et al. Clonal expansion and diversification of cancer-associated mutations in endometriosis and normal endometrium. Cell Rep. 24, 1777–1789 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Yokoyama, A. et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565, 312–317 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  6. Moore, L. et al. The mutational landscape of normal human endometrial epithelium. Nature 580, 640–646 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  7. Yoshida, K. et al. Tobacco smoking and somatic mutations in human bronchial epithelium. Nature 578, 266–272 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fowler, J. C. et al. Selection of oncogenic mutant clones in normal human skin varies with body site. Cancer Discov. 11, 340–361 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Lawson, A. R. J. et al. Extensive heterogeneity in somatic mutation and selection in the human bladder. Science 370, 75–82 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Vermeulen, L. et al. Defining stem cell dynamics in models of intestinal tumor initiation. Science 342, 995–998 (2013).

    Article  ADS  CAS  PubMed  Google Scholar 

  11. Alcolea, M. P. et al. Differentiation imbalance in single oesophageal progenitor cells causes clonal immortalization and field change. Nat. Cell Biol. 16, 615–622 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Fernandez-Antoran, D. et al. Outcompeting p53-mutant cells in the normal esophagus by redox manipulation. Cell Stem Cell 25, 329–341 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Colom, B. et al. Spatial competition shapes the dynamic mutational landscape of normal esophageal epithelium. Nat. Genet. 52, 604–614 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Murai, K. et al. Epidermal tissue adapts to restrain progenitors carrying clonal p53 mutations. Cell Stem Cell 23, 687–699.e688 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Doupe, D. P. et al. A single progenitor population switches behavior to maintain and repair esophageal epithelium. Science 337, 1091–1093 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Piedrafita, G. et al. A single-progenitor model as the unifying paradigm of epidermal and esophageal epithelial maintenance in mice. Nat. Commun. 11, 1429 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Frede, J., Greulich, P., Nagy, T., Simons, B. D. & Jones, P. H. A single dividing cell population with imbalanced fate drives oesophageal tumour growth. Nat. Cell Biol. 18, 967–978 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nair, R. R. et al. A role for keratin 17 during DNA damage response and tumor initiation. Proc. Natl Acad. Sci. USA 118, e2020150118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hobbs, R. P. et al. Keratin-dependent regulation of Aire and gene expression in skin tumor keratinocytes. Nat. Genet. 47, 933–938 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Liu, Z. et al. Keratin 17 activates AKT signalling and induces epithelial-mesenchymal transition in oesophageal squamous cell carcinoma. J. Proteomics 211, 103557 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. Narushima, K. et al. Establishment of a DEN-induced mouse model of esophageal squamous cell carcinoma metastasis. Esophagus 14, 131–137 (2017).

    Article  Google Scholar 

  22. Rubio, C. A., Liu, F. S., Chejfec, G. & Sveander, M. The induction of esophageal tumors in mice: dose and time dependency. In Vivo 1, 35–38 (1987).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  24. Greaves, M. & Maley, C. C. Clonal evolution in cancer. Nature 481, 306–313 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hall, M. W. J., Jones, P. H. & Hall, B. A. Relating evolutionary selection and mutant clonal dynamics in normal epithelia. J. R. Soc. Interface 16, 20190230 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell 171, 1029–1041.e1021 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Martincorena, I., Jones, P. H. & Campbell, P. J. Constrained positive selection on cancer mutations in normal skin. Proc. Natl Acad. Sci. USA 113, E1128–E1129 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Prasad, V. et al. Haploinsufficiency of Atp2a2, encoding the sarco(endo)plasmic reticulum Ca2+-ATPase isoform 2 Ca2+ pump, predisposes mice to squamous cell tumors via a novel mode of cancer susceptibility. Cancer Res. 65, 8655–8661 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Liu, L. H., Boivin, G. P., Prasad, V., Periasamy, M. & Shull, G. E. Squamous cell tumors in mice heterozygous for a null allele of Atp2a2, encoding the sarco(endo)plasmic reticulum Ca2+-ATPase isoform 2 Ca2+pump. J. Biol. Chem. 276, 26737–26740 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Tumbar, T. et al. Defining the epithelial stem cell niche in skin. Science 303, 359–363 (2004).

    Article  ADS  CAS  PubMed  Google Scholar 

  31. Brown, S. et al. Correction of aberrant growth preserves tissue homeostasis. Nature 548, 334–337 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kon, S. et al. Cell competition with normal epithelial cells promotes apical extrusion of transformed cells through metabolic changes. Nat. Cell Biol. 19, 530–541 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Shultz, L. D. et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2Rγnull mice engrafted with mobilized human hemopoietic stem cells. J. Immunol. 174, 6477–6489 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Ellis, P. et al. Reliable detection of somatic mutations in solid tissues by laser-capture microdissection and low-input DNA sequencing. Nat. Protoc. 16, 841–871 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

  36. Gerstung, M., Papaemmanuil, E. & Campbell, P. J. Subclonal variant calling with multiple samples and prior knowledge. Bioinformatics 30, 1198–1204, (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rosenthal, R., McGranahan, N., Herrero, J., Taylor, B. S. & Swanton, C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 17, 31 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Jones, D. et al. cgpCaVEManWrapper: simple execution of CaVEMan in order to detect somatic single nucleotide variants in NGS data. Curr. Protoc. Bioinformatics 56, 15.10.11–15.10.18 (2016).

    Google Scholar 

  40. Menzies, A. et al. VAGrENT: variation annotation generator. Curr. Protoc. Bioinformatics 52, 15.18.11–15.18.11 (2015).

    Google Scholar 

  41. Scheinin, I. et al. DNA copy number analysis of fresh and formalin-fixed specimens by shallow whole-genome sequencing with identification and exclusion of problematic regions in the genome assembly. Genome Res. 24, 2022–2032 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kuilman, T. et al. CopywriteR: DNA copy number detection from off-target sequence data. Genome Biol. 16, 49 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Nik-Zainal, S. et al. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Toni, T., Welch, D., Strelkowa, N., Ipsen, A. & Stumpf, M. P. Approximate Bayesian computation scheme for parameter inference and model selection in dynamical systems. J. R. Soc. Interface 6, 187–202 (2009).

    Article  PubMed  Google Scholar 

  45. Klinger, E., Rickert, D. & Hasenauer, J. pyABC: distributed, likelihood-free inference. Bioinformatics 34, 3591–3593 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank E. Choolun, T. Metcalf and staff at the MRC ARES and Sanger RSF animal facilities for technical support. This work was supported by grants from the Wellcome Trust to the Wellcome Sanger Institute (098051 and 296194) and Cancer Research UK Programme Grants to P.H.J. (C609/A17257 and C609/A27326). B.A.H. and M.W.J.H. are supported by the Medical Research Council (Grant-in-Aid to the MRC Cancer unit grant number MC_UU_12022/9 and NIRG to B.A.H., grant number MR/S000216/1). A.H. benefited from the award of an EMBO long term fellowship. M.W.J.H. acknowledges support from the Harrison Watson Fund at Clare College, Cambridge. B.A.H. acknowledges support from the Royal Society (grant no. UF130039). S.C.D. benefited from the award of an ESPOD fellowship, 2018-21, from the Wellcome Sanger Institute and the European Bioinformatics Institute EMBL-EBI. K.T.M. benefited from the support of the Chan Zuckerberg Initiative. We thank B. Mahler-Araujo and the MRC Metabolic Diseases Unit (MC_UU_00014/5) for the histological analysis of tumour samples. We are grateful to the Cambridge Biorepository for Translational Medicine for access to human tissue. The authors acknowledge the use and/or adaption of diagrams from Servier Medical Art (https://smart.servier.com).

Author information

Authors and Affiliations

Authors

Contributions

B.C. and M.P.A. designed experiments. B.C. performed the majority of experiments and data analysis. A.H. and M.P.A. contributed to mouse sample collection and performed preliminary experiments. M.W.J.H. conducted permutation analysis of leukocyte location. G.P. performed initial analysis of immune cell location in tumours and normal areas. D.F.-A. analysed human oesophageal samples. B.C., M.W.J.H., C.K., R.K.S. and S.H.O. analysed sequencing data. S.C.D. analysed chromosomal copy number alterations under the supervision of M.G. M.W.J.H. developed the mathematical modelling under the supervision of B.A.H. K.T.M., K.S.-P. and J.C.F. collected human samples. B.C., M.W.J.H. and P.H.J. wrote the paper. P.H.J. supervised the research.

Corresponding author

Correspondence to P. H. Jones.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Francesca Ciccarelli, James DeGregori and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data figures and tables

Extended Data Fig. 1 Architecture and dynamics of murine esophageal epithelium.

(a) H&E images and schematic of the mouse esophageal epithelium architecture and dynamics; b = basal cell layer (delineated by white dots), sb = suprabasal layers, str = stroma. Scale-bars: 500µm (left image), 50µm (inset). Dividing (progenitor) cells are confined to the basal layer. Differentiating cells exit the cell cycle, migrate out of the basal layer, through the suprabasal layers, and are finally shed into the lumen. (b–c) The single progenitor model (see Supplementary Note). All progenitor cells in the basal layer are functionally equivalent and following division produce either: two progenitors that will persist in the tissue, two differentiating cells that will cease division, stratify and be lost, or one cell of each type. In homeostasis (b), the likelihood of the two progenitor and two differentiating cell outcomes is equal. Mutations (c) may tip the balance towards a non-neutral behavior, resulting in clonal growth if they favor proliferation of daughter cells. (d) Expansion of mutant clones is defined by their relative competitive fitness to adjacent clones. Initially, a fit “winner” mutant progenitor (colored) shows a fate bias towards proliferation and outcompetes its less fit “loser” surrounding cells, resulting in clonal expansion. Eventually, mutant clones begin to collide with each other until surrounded by similarly competitive mutants, at which point their cell fate reverts towards balance and their expansion slows (see Supplementary Note).

Extended Data Fig. 2 Characterization of Diethyl Nitrosamine (DEN) generated tumors.

(a, b) Confocal images of esophagus collected 10 days post-DEN treatment and stained with Dapi and KRT6. Images show top-down (a, b) and lateral (b) projections and illustrate typical morphological features and expression of KRT6 in tumors (arrowheads) and normal surrounding epithelium. (c) Quantification of the mean fluorescence intensity of KRT6 and Dapi in 10 days post-DEN tumors and the adjacent normal epithelium, measured from the images as shown. Two-sided Mann-Whitney test, n = 48 tumors from 3 mice. (d) Percentage of tumors positive for KRT6 staining. The number of tumors analyzed for each time point is shown between brackets. (e) Confocal images of 10-days and 12-months post-DEN tumors showing increased expression of KRT17 (green). Images are representative of 31 and 29 tumors, from 3 and 7 mice, respectively. (f) H&E images of esophageal tumors (bottom images) and normal epithelium (top images) from tissues collected at the indicated time points post-DEN treatment. (g) Percentage of angiogenic tumors at the indicated time points post-DEN treatment. The number of tumors analyzed at each time point is shown in brackets. (h, i) Projected top-down and lateral confocal images of 10 day (h) and 9 months (i) post-DEN tumors immuno-stained for the endothelial cell marker CD31 and KRT6. Images are representative of 18 and 6 tumors, respectively. Scale-bars for panels (a–c), (e–f) and (h–i) are 50µm.

Source data

Extended Data Fig. 3 Collection of 10-day and 1-year esophageal tumors.

(a–b) Mouse esophagus was collected 10-days (a) or 1-year (b) post-DEN treatment. The esophagus was cut open longitudinally and the epithelium separated from the underlying muscle and stroma. The epithelium was flattened, fixed, stained with KRT6 (grey), mounted and 3D-imaged on a confocal microscope. Tumors were identified from the processed images and manually dissected under a fluorescent microscope and sequenced (see Methods).

Extended Data Fig. 4 Targeted sequencing of 10 days and 1 year tumors.

(a) Protocol: wild-type mice were treated with DEN for two months and the esophageal tumors collected 10-days or 1-year later. Tumors were then sequenced with a targeted approach (192 gene panel). (b) Area of 10-day and 1-year post-DEN tumors (n = 89 and 64 tumors from 2 and 9 mice, respectively). Lines show mean ± s.e.m. (c) Mutational spectrum of 10-day and 1-year tumors. The bar plots illustrate the number of mutations in each of the 96 possible trinucleotides. The mutational spectrum of individual tumors is shown in (d). (e–f) Percentage of silent, missense, nonsense and splice mutations and indels identified in 10-day or 1-year tumors. Graphs show the values for individual tumors (e, each column is a tumor), or the average for all tumors at each time point (f). (g) Maximum VAF for mutations identified in each tumor at 10 days or 1 year post-DEN (n = 80 and 63 tumors, respectively). Colored shadow illustrates VAF > 0.35, as an estimation for clonality. (h) Number of positively selected mutant genes in tumors at 10 days or 1 year post-DEN treatment (n = 80 and 63 tumors, respectively). Lines show mean±s.e.m (two-tailed Mann-Whitney test). (i) Number and type of mutations in the positively selected genes identified by dN/dS analysis from 10-day or 1-year tumors. (j) Proportion of 10-day and 1-year tumors carrying nonsynonymous mutations in the indicated genes.

Source data

Extended Data Fig. 5 Whole exome sequencing, whole genome sequencing, and chromosomal alteration of surviving tumors.

(a) Wild-type mice received DEN for two months and the esophagus was collected 9 or 18 months after treatment (n=49 tumors from 16 mice). Tissues were stained for Dapi (blue) and KRT6 (red) and confocal imaged to identify tumors. Scale-bars=2mm (main), 150µm (inset). Individual tumors were manually cut under a fluorescent microscope, digested and separated in triplicates. Each triplicate was whole genome amplified (WGA) and whole exome sequenced. To exclude artefactual SNVs generated during WGA, only mutations shared by all three amplified triplicates were considered for further analysis. A total of 32,736 mutations, including silent, missense, nonsense and splice mutations and indels were identified. (b) Cumulative sequencing coverage of the whole exome triplicate samples. (c) Number of synonymous and non-synonymous mutations per tumor, ranked by mutation burden. (d) Distribution of the variant allele fraction (VAF) for the mutations common within triplicates in each tumor. (e) Percentage of silent, missense, nonsense and splice mutations and indels for individual tumors and for all tumors combined. (f) Mutational spectrum of tumors. The bar plots illustrate the percentage of mutations in each of the 96 possible trinucleotides. (g) dN/dS ratios for missense and truncating (nonsense + essential splice site) substitutions indicating genes under significant positive selection in the tumors (q<0.05, calculated with R package dNdScv26). (h) Analysis of chromosomal copy number alterations (CNAs) by whole genome sequencing of 1-year post-DEN tumors (n=64 tumors from 9 mice). Only 2 tumors, MD5924e and MD5928e, exhibited CNAs. (i) Summary of chromosomal alterations found by whole exome sequencing data of 9 or 18 months post-DEN tumors (n = 49 tumors from 16 mice). Only alterations present in all 3 whole genome amplified triplicates (j) were considered valid calls. 8140nT2 (top) is a representative example of a tumor without chromosomal alterations. 2 tumors, n2T1 and n34_T1, showed small alterations.

Source data

Extended Data Fig. 6 Early tumors are not eliminated by tumor cell apoptosis, abnormal proliferation or the immune system.

(a) Representative confocal image of a 10-day post-DEN esophageal epithelium immuno-stained for activated Caspase 3+ (Yellow) and KRT6 (red). No apoptotic cells were detected in the tumors (arrowhead) (n = 23 tumors, 2 mice). Scale-bar: 20 µm. (b) Protocol: wild-type mice were treated with DEN for two months and the tissues collected ten days later. Mice received EdU 1h before tissue collection. (c) Representative confocal images showing EdU incorporation (1h pulse) in tumors (dotted line) and the surrounding normal epithelium (n = 22 tumors, 3 mice). Scale-bars: 20µm. (d) In vivo label-retaining assay using Rosa26M2rtTA/TetO-HGFP transgenic mice to measure the rate of progenitor cell division (see Methods). (e) Protocol: Rosa26M2rtTA/TetO-HGFP mice received DEN for one month followed by DEN+Doxy for another month. Tissues were collected at times 0, 10 and 30 days after Doxy/DEN withdrawal. (f–g) Representative confocal images (f) and quantification (g) of the histone- green fluorescent protein (H2BGFP) intensity in esophageal tumors (dotted lines) and the surrounding normal epithelium at the indicated time points post-Doxy withdrawal. Scale bars: 50µm. Graph shows median (central box line), 25th–75th percentiles (box) and 5th–95th percentiles (whiskers). Two-tailed Mann-Whitney test (n = 10 images per group). (h) Confocal images depicting CD45+ (immune) cells (green) within a 10-days post-DEN tumor (white dotted line) and its adjacent normal epithelium. (i–j) Correlation between the size of tumors and the number of CD45+ cells within them (i) and the number of CD45+ cells in the tumor and the normal epithelium (j). Lines show the two-tailed Pearson correlations: R2=0.5039 (i), and R2 = 0.1464 (j), with 95% confidence interval (dotted lines). (k) Number of CD45+ cells per area of tumor or normal esophageal epithelium at 10- or 30-days post-DEN treatment. Graph shows median (central box line), 25th–75th percentiles (box), 5th–95th percentiles (whiskers) and outliers (dots). Two tailed Wilcoxon matched-pairs test, n = 53 and 50 images, respectively. (l) Permutation analysis of leukocyte location within tumors, based on the experimental measurements obtained from (h). For each image, the location of CD45+ cells (black dots) was left intact while the location of the tumor (colored circles) was randomly shuffled (blue shows original location, red shows shuffled “phantom” tumors), and the number of immune cells in contact with the tumor was counted. This was repeated 1000 times to produce the expected distribution. (m) Calculated (within the original location) vs. experimentally observed number of CD45+ cells in 10- or 30-days post-DEN tumors. (n) Distribution of the average number of CD45+ cells in 10- or 30-days post-DEN tumors, obtained from the permutation analysis in (l). Red dotted line shows the experimentally observed average number of CD45+ cells within tumors at the indicated time-points. Statistics are two-tailed permutation tests (Methods). (o) Protocol: Wild-type and immunocompromised NOD Cg-Prkdcscid Il2rgtm1Wjl SzJ (Scid-NOD-IL2r) mice were treated with DEN for two months and the tissues collected 10 or 30 days post-DEN withdrawal. (p) Tumor density in mice collected 10-days (n = 4) or 30-days (n = 5 and 4, respectively) post-DEN treatment. Mean ± s.e.m (two-tailed Mann-Whitney test).

Source data

Extended Data Fig. 7 Mutational landscape of normal esophageal epithelium at 10 days post-DEN treatment.

(a) Protocol: wild-type mice were treated for two months with DEN. 10 days after DEN withdrawal, normal epithelial samples matching the size of the tumors (Extended Data Fig. 4b) were collected and sequenced with a targeted approach (192 gene panel). (b) Number of mutations per sample, including essential splice, frameshift, missense, nonsense and silent mutations (n = 81 samples from 2 mice). (c) Percentage of mutation types identified in normal epithelium and tumors collected 10 days post-DEN treatment. (d) Mutational spectrum of 10-day post-DEN tumors and normal epithelium. The bar plots illustrate the number of mutations in each of the 96 possible trinucleotides. (e) Dots show the value of the mutation with maximum VAF identified in each sample (n = 81 samples). Lines are mean ± s.e.m. (f) dN/dS ratios for missense and truncating (nonsense + essential splice site) mutations of positively selected genes (dN/dS >1) (only significant genes are shown, q < 0.05 calculated with R package dNdScv26). (g) Number and type of mutations in the positively selected genes identified by dN/dS. (h) Estimated percentage of 10 days post-DEN normal epithelium carrying non-synonymous mutations in the positively selected genes. Range indicates upper and lower bound estimates.

Source data

Extended Data Fig. 8 Expansion of highly competitive clones in normal tissue eliminates early tumors.

(a) Cartoon illustrating the elimination of tumors due to competition with the highly competitive DN-Maml1 clones. Following the generation of tumors by DEN treatment, DN-Maml1 clones are induced by BNF and TAM injections. As DN-Maml1 clones in the normal epithelium expand they eliminate less fit tumors. (b) Qualitative representation of tumor dynamics following DN-Maml1 induction as compared to non-induced controls (see Supplementary Note). (c) In vivo genetic lineage tracing using AhcreERT/R26DNM−GFP/wt reporter mice. Upon injection of the drugs tamoxifen (TAM) and ß-napthoflavone (BNF), Cre-mediated recombination results in the heritable expression of the highly competitive dominant negative allele of Maml-1 fused to GFP fluorescent protein (DN-Maml1), which will then be expressed in all the progeny of the single marked cells, generating clusters of labelled mutant clones. (d) Confocal image (representative of 9 mice) of a DEN-treated AhcreERT/R26DNM−GFP/wt mouse whole esophageal epithelium, depicting DN-Maml1 clones (green). Mice were induced ten days after DEN withdrawal and esophagus collected twenty days later (as in Fig. 3b). Scale-bar: 2 mm. (e) Percentage of epithelium covered by DN-Maml1 clones in induced and non-induced (control) mice (n = 9 and 7 mice, respectively). Error-bars are mean ± s.e.m (two-tailed Mann-Whitney test). (f) Number of tumors per mm2 of esophageal epithelium in control non-induced (n = 7) and induced (n=9) AhcreERT/R26DNM−GFP/wt mice. Data for the induced mice is show as the number of tumors per mm2 of whole tissue area and per mm2 of area not covered by DN-Maml1 clones (DN-Maml1- area). Error-bars are mean±s.e.m. (two-sided Mann-Whitney test). (g) Correlation between the area covered by DN-Mam1 clones and the number of tumors in induced and non-induced AhcreERT/R26DNM−GFP/wt mice. Line shows the Pearson correlation (two-tailed, R2 = 0.7526) with 95% confidence interval (dotted lines). Inset shows the correlation of the induced mice only (R2 = 0.4577).

Source data

Extended Data Fig. 9 Spatial interaction between mutant clones in mutagenized normal epithelium and tumors.

(a) Confocal images and cartoons of esophageal epithelium from AhcreERT/R26DNM−GFP/wt mice treated with DEN for 2 months, induced 10-days post-DEN withdrawal and tissues collected 20-days later (as in Fig. 3b). The images show different categories of DN-Maml1 clones (green) interacting with early tumors (red). (b) Percentage of tumors in each category. Number of tumors in each group is shown between brackets (n = 386 tumors from 9 mice). (c) Top-down confocal images depicting a DN-Maml1 clone surrounding a tumor (see also Fig. 3e). A smaller tumor surface area at the base is consistent with a process of extrusion by the expanding DN-Maml1 clone as illustrated in (g). (d) In vivo genetic lineage tracing using the multicolor confetti reporter allele in R26creERT2R26flConfetti mice. Injection of the drug tamoxifen (TAM), results in Cre-mediated inversion and excision recombination events in scattered single cells. This confers heritable expression of one of four fluorescent proteins (YFP, GFP, RFP or CFP) resulting in labelled clones. (e) Protocol: R26creERT2R26flConfetti mice were induced with TAM to label Confetti clones, followed by 2-months DEN treatment. Esophagi were collected 10 days or 30 days after DEN withdrawal. (f) Confocal images of Confetti clones surrounding early tumors (KRT6, shown in red) in the categories 2-4 as in (a), suggesting that tumor extrusion events may also take place in mutagenized epithelium in the absence of DN-Maml1 induced clones. Images are representative of 13 tumors from 8 mice. (g) Cartoon illustrating a potential mechanism of tumor extrusion by expanding mutant clones in the normal epithelium. Once the tumor footprint in the basal layer is displaced by expanding mutant clones in the adjacent normal tissue, the tumor will be shed. Scale-bars in panels (a), (c) and (f) are 25µm.

Source data

Extended Data Fig. 10 Increasing competitive fitness throughout the epithelium slows down tumor loss. Human esophageal lesions.

(a) A stochastic drift model (Supplementary Note) fit to the observed number (mean ± s.e.m) of tumors following DEN treatment. (b) Experimentally observed (black dots) and model predicted (dashed lines) % of tumors eliminated when highly fit mutant clones (such as DN-Maml1) are induced in esophageal epithelium following DEN-treatment. Dashed lines represent models where the induced mutant clones remove tumors they encounter (blue) or where tumor loss is independent of clones in the surrounding tissue (red) (Supplementary Note). (c) Cartoon illustrating the predicted effects of DBZ administration. When clones in the normal surrounding epithelium have a higher competitive fitness than the tumors, the probability that tumors will be eliminated from the tissue may be higher (left) than when administration of DBZ levels the competition between the clones and tumors (right). (d) Qualitative representation of tumor dynamics following DBZ vs. control treatment. (e) Protocol: wild-type mice were treated with DEN for two months. Ten days post-DEN withdrawal mice received DBZ or vehicle control. Tissues were harvested two weeks later. (f–g) Experimental (f) and simulated (g, Supplementary Note) tumors per mm2 of esophageal epithelium in DBZ and vehicle control treated mice (n = 5 mice/group). Lines in (f) are mean±s.e.m (two-tailed Mann-Whitney test). Black dots in (g) show experimental data. (h–i) Numerical example (see Supplementary Note) of the model showing increased proportion of tumors resistant to displacement by mutant clones (h) and the decrease in tumor density following DEN-treatment (i). Experimental data depicts mean ± s.e.m. (j) Images of human (top) and mouse (bottom) esophagus. Dotted lines delineate lesions. (k) Confocal images of human normal (top) and neoplastic (bottom) esophageal epithelium stained with KRT6 (red) and Topro3 (nuclei, blue). Scale-bars: 100 µm. Simulations in (a, g, h and i) show the mean and range between the minimum-maximum outputs of the model run with the accepted parameters from Approximate Bayesian Computation (Methods).

Source data

Supplementary information

Supplementary Note

This document sets out the theory and mathematical modelling of tumour dynamics in Sections 1–7. Section 1 discusses previous results on the growth and competition of mutant clones in normal oesophageal epithelium. Section 2 describes a previously proposed stochastic model of tumour dynamics. Section 3 describes the elimination of tumours by highly competitive mutant clones in the surrounding normal epithelium. Section 4 shows how reducing the competitive imbalance between tumours and highly fit mutant clones in the normal tissue affects tumour survival. Section 5 describes the selection pressure on tumours from competition with surrounding clones in the normal epithelium. In Section 6, we substitute simple mathematical equations into the model to numerically illustrate the principles described in the previous sections. Section 7 is a summary of our conclusions.

Reporting Summary

Peer Review File

Supplementary Tables

This file contains Supplementary Tables 1–11. Supplementary Table 1 lists the area (in µm2) and number of tumours measured at different time points after DEN treatment. Supplementary Table 2 displays the targeted sequencing data of isolated tumours from 10-day post-DEN-treated mouse oesophageal epithelium. Supplementary Table 3 displays the targeted sequencing data of isolated tumours from 1-year post-DEN-treated mouse oesophageal epithelium. Supplementary Table 4 shows the dN/dS results from targeted sequencing data of isolated tumours from 10-day post-DEN-treated mouse oesophageal epithelium. Supplementary Table 5 shows the dN/dS results from targeted sequencing data of isolated tumours from 1-year post-DEN-treated mouse oesophageal epithelium. Supplementary Table 6 shows the whole-exome sequencing results of 9- and 18-month post-DEN tumours. Supplementary Table 7 displays the dN/dS results for whole-exome sequencing of 9- and 18-month post-DEN tumours. Supplementary Table 8 displays the HGFP mean intensity in tumours and surrounding normal oesophageal epithelium. Supplementary Table 9 shows the targeted sequencing data of normal oesophageal epithelium from 10-day post-DEN-treated mice. Supplementary Table 10 displays the dN/dS results from targeted sequencing data of normal oesophageal epithelium from 10-day post-DEN-treated mice. Supplementary Table 11 lists the parameters for the mathematical model.

Supplementary Video 1

The video illustrates the mouse oesophageal epithelium at 1 month post-DEN treatment. The video initially shows the entire tissue before zooming in on a tumour.

Supplementary Video 2

The video shows small capillary circling a tumour in the mouse oesophageal epithelium collected 10 days post-DEN treatment, illustrating the early steps of angiogenesis.

Supplementary Video 3

The video shows a developed vasculature surrounding an established tumour in the mouse oesophageal epithelium, collected 9 months post-DEN treatment.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Colom, B., Herms, A., Hall, M.W.J. et al. Mutant clones in normal epithelium outcompete and eliminate emerging tumours. Nature 598, 510–514 (2021). https://doi.org/10.1038/s41586-021-03965-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03965-7

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