Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A body map of somatic mutagenesis in morphologically normal human tissues

Abstract

Somatic mutations that accumulate in normal tissues are associated with ageing and disease1,2. Here we performed a comprehensive genomic analysis of 1,737 morphologically normal tissue biopsies of 9 organs from 5 donors. We found that somatic mutation accumulations and clonal expansions were widespread, although to variable extents, in morphologically normal human tissues. Somatic copy number alterations were rarely detected, except for in tissues from the oesophagus and cardia. Endogenous mutational processes with the SBS1 and SBS5 mutational signatures are ubiquitous among normal tissues, although they exhibit different relative activities. Exogenous mutational processes operate in multiple tissues from the same donor. We reconstructed the spatial somatic clonal architecture with sub-millimetre resolution. In the oesophagus and cardia, macroscopic somatic clones that expanded to hundreds of micrometres were frequently seen, whereas in tissues such as the colon, rectum and duodenum, somatic clones were microscopic in size and evolved independently, possibly restricted by local tissue microstructures. Our study depicts a body map of somatic mutations and clonal expansions from the same individual.

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

Access options

Buy this article

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

Fig. 1: Research strategy and summary of genomic alterations detected in normal tissues from five donors.
Fig. 2: Mutational signatures in normal tissues from five donors.
Fig. 3: Mutational landscape of driver genes across organs.
Fig. 4: Estimation of somatic mutant clonal sizes and construction of spatial clonal expansion maps.

Similar content being viewed by others

Data availability

The raw WES and WGS data generated in this study have been deposited in the European Genome-phenome Archive (EGA) (https://ega-archive.org) with accession number EGAD00001007859 and the Genome Sequence Archive (GSA) of the Beijing Institute of Genomics with accession number HRA000356 (https://ngdc.cncb.ac.cn/gsa-human). To gain access to the raw sequencing data, please submit requests to the Pan-body Mutagenesis Data Access Committee (EGA accession number EGAC00001002218) or through the GSA online page of this study (https://ngdc.cncb.ac.cn/gsa-human/browse/HRA000356). All somatic mutations detected from WES with functional annotations and allele count information can be found in Supplementary Table 3. RefSeq database: https://www.ncbi.nlm.nih.gov/refseq. NHLBI Exome Sequencing Project: http://evs.gs.washington.edu/EVS. dbSNP database: https://www.ncbi.nlm.nih.gov/snp. COSMIC database: https://cancer.sanger.ac.uk/cosmic. The GTEx project: https://gtexportal.org/home.

Code availability

Mutational signature analysis was performed using the HDP R package v.0.1.5 (https://github.com/nicolaroberts/hdp). Code for mutational signature analysis was adapted from https://github.com/HLee-Six/colon_microbiopsies. Code for the Bayesian Dirichlet process clustering of MCFs was adapted from https://github.com/sfbrunner/liver-pub-repo. Adapted code is available at Zenodo (https://doi.org/10.5281/zenodo.5012918). Driver gene analysis was performed using the dNdScv v0.01 (https://github.com/im3sanger/dndscv).

References

  1. Martincorena, I. & Campbell, P. J. Somatic mutation in cancer and normal cells. Science 349, 1483–1489 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Risques, R. A. & Kennedy, S. R. Aging and the rise of somatic cancer-associated mutations in normal tissues. PLoS Genet. 14, e1007108 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Martincorena, I. et al. Tumor evolution. 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. Tang, J. et al. The genomic landscapes of individual melanocytes from human skin. Nature 586, 600–605 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. 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 

  7. 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 

  8. Brunner, S. F. et al. Somatic mutations and clonal dynamics in healthy and cirrhotic human liver. Nature 574, 538–542 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  10. 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 

  11. Lodato, M. A. et al. Aging and neurodegeneration are associated with increased mutations in single human neurons. Science 359, 555–559 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  12. Bae, T. et al. Different mutational rates and mechanisms in human cells at pregastrulation and neurogenesis. Science 359, 550–555 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Ju, Y. S. et al. Somatic mutations reveal asymmetric cellular dynamics in the early human embryo. Nature 543, 714–718 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. Li, R. et al. Macroscopic somatic clonal expansion in morphologically normal human urothelium. Science 370, 82–89 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  16. Lee-Six, H. et al. Population dynamics of normal human blood inferred from somatic mutations. Nature 561, 473–478 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Watson, C. J. et al. The evolutionary dynamics and fitness landscape of clonal hematopoiesis. Science 367, 1449–1454 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Yizhak, K. et al. RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues. Science 364, eaaw0726 (2019).  

  19. Garcia-Nieto, P. E., Morrison, A. J. & Fraser, H. B. The somatic mutation landscape of the human body. Genome Biol. 20, 298 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Alexandrov, L. B. et al. Clock-like mutational processes in human somatic cells. Nat. Genet. 47, 1402–1407 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Poon, S. L. et al. Mutation signatures implicate aristolochic acid in bladder cancer development. Genome Med. 7, 38 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Ng, A. W. T. et al. Aristolochic acids and their derivatives are widely implicated in liver cancers in Taiwan and throughout Asia. Sci. Transl. Med. 9, eaan6446 (2017).

  26. Du, Y. et al. Mutagenic factors and complex clonal relationship of multifocal urothelial cell carcinoma. Eur. Urol. 71, 841–843 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Chen, C. H. et al. Aristolochic acid-induced upper tract urothelial carcinoma in Taiwan: clinical characteristics and outcomes. Int. J. Cancer 133, 14–20 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513, 202–209 (2014).

    Article  ADS  CAS  Google Scholar 

  30. Bailey, M. H. et al. Comprehensive characterization of cancer driver genes and mutations. Cell 174, 1034–1035 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Moore, L. et al. The mutational landscape of human somatic and germline cells. Nature, https://doi.org/10.1038/s41586-021-03822-7 (2021).

  32. Blokzijl, F. et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538, 260–264 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Abascal, F. et al. Somatic mutation landscapes at single-molecule resolution. Nature 593, 405–410 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  34. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J. 17, 10–12 (2011).

    Article  Google Scholar 

  35. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cingolani, P. et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff. Fly 6, 80–92 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Coorens, T. H. H. et al. Inherent mosaicism and extensive mutation of human placentas. Nature 592, 80–85 (2021).

  42. Olafsson, S. et al. Somatic evolution in non-neoplastic IBD-affected colon. Cell 182, 672–684 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. The GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 1318–1330 (2020).

    Article  PubMed Central  CAS  Google Scholar 

  44. Roberts, N. D. Patterns of Somatic Genome Rearrangement In Human Cancer. PhD thesis, Univ. Cambridge (2018).

  45. 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 

  46. O’Leary, N. A. et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res. 44, D733–745 (2016).

    Article  PubMed  CAS  Google Scholar 

  47. Cancer Genome Atlas Research, N. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014).

    Article  ADS  CAS  Google Scholar 

  48. The Cancer Genome Atlas Research Network. Integrated genomic characterization of oesophageal carcinoma. Nature 541, 169–175 (2017).

    Article  PubMed Central  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  50. The Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 169, 1327–1341 (2017).

    Article  PubMed Central  CAS  Google Scholar 

  51. Wu, G., Feng, X. & Stein, L. A human functional protein interaction network and its application to cancer data analysis. Genome Biol. 11, R53 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Papastamoulis, P. label. switching: an R package for dealing with the label switching problem in MCMC outputs. J. Stat. Softw. 69, Code Snippet 1 (2016).

    Article  Google Scholar 

  53. Kumar, S., Stecher, G., Li, M., Knyaz, C. & Tamura, K. MEGA X: molecular evolutionary genetics analysis across computing platforms. Mol. Biol. Evol. 35, 1547–1549 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all the donors and their families for their consent and participation in this study. This project was jointly supported by the National Natural Science Foundation of China (81725015 to C.W., 81988101 to D.L. and C.W., 22050004 to J.W. and 22050002 to Y.H.); Beijing Outstanding Young Scientist Program (BJJWZYJH01201910023027 to C.W.); the Medical and Health Technology Innovation Project of the Chinese Academy of Medical Sciences (2019-I2M-2-001 to D.L. and C.W.); the National Key R&D Program of China (2019YFC1315702, 2018ZX10302205 to F.B. and 2018YFA0108100 to Y.H.); Guangdong Province Key Research and Development Program (2019B020226002 to F.B.); Beijing Municipal Science and Technology Commission (Z201100005320016 to Y.H.); Beijing Advanced Innovation Center for Genomics; and Shenzhen Bay Laboratory.

Author information

Authors and Affiliations

Authors

Contributions

F.B., C.W., Y.H., J.W., D.L. and R.L. conceived the study. R.L., L.D. and W.F. performed data analyses with assistance from L.L., D.X. and Y.W. W.F. performed sample collection, tissue sectioning and LCM with assistance from Y.L., W.G., W.L., L.C., Y.C., C.M., H.L. and Y.M. L.D. performed DNA extraction and library preparation. J.L. performed exome capture with assistance from Q.L. R.L., F.B., W.F., L.D. and J.L. wrote the manuscript with input from D.L., Y.H., J.W. and C.W. D.L., Y.H., J.W., F.B. and C.W. supervised all aspects of this study.

Corresponding authors

Correspondence to Dongxin Lin, Yanyi Huang, Jianbin Wang, Fan Bai or Chen Wu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Ziyue Gao and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Normal tissue histology.

Representative H&E-stained samples showing the histological features of normal tissues sampled from nine organs from the five donors. Blanks in the figure represent samples that are not available in corresponding organs and donors. Scale bars, 100 µm.

Extended Data Fig. 2 Detection of somatic mutations.

a, Bar plot showing the overlap of mutations detected from WES and WGS of 43 samples. b, Adjusted numbers of somatic mutations detected in the coding regions in tissue biopsies from the organs of five donors. Red vertical bars represent median mutation numbers and grey horizontal bars represent standard deviations. c, The mutation burdens (after the sensitivity correction) in samples with median VAFs between 0.08 and 0.14. Top, box plots showing the mutation burdens in organs from different donors. The lower edge, upper edge and centre of the box represent the 25th (Q1) percentile, 75th (Q3) percentile and the median, respectively. IQR = Q3 – Q1. Outliers are values beyond the whiskers (upper, Q3 + 1.5 × IQR; lower, Q1 − 1.5 × IQR). Detailed information about the box plots can be found in Supplementary Table 3. Bottom, dot plots showing the adjusted mutation burdens in different organs. Red bars represent the medians. d, Scatter plots showing the VAFs of somatic mutations detected in the normal tissues from the nine organs of the five donors. Dots are coloured by mutation type.

Extended Data Fig. 3 Correlations and interdependence between VAF distributions and mutation numbers.

In each tissue, we calculated the first quantile (Q1) and third quantile (Q3) of the VAF and mutation burden distribution. We defined IQR = Q3 – Q1 and considered samples with a median VAF or mutation burden greater than Q3 + 1.5 × IQR or less than Q1 – 1.5 × IQR as outliers. We excluded these outliers in this analysis. Corr., correlation. The error bands represent the 95% confidence intervals. P values are from two-sided correlation tests based on the Pearson correlation coefficient.

Extended Data Fig. 4 Mutation numbers and somatic CNAs.

a, Bar plots showing the number of mutations among the four intervals. Genes are divided into four intervals according to the tissue-specific gene expression levels. b, Heat maps showing somatic CNAs detected in the normal tissues from the nine organs of the five donors. Sex chromosomes were excluded.

Extended Data Fig. 5 Mutational spectra and signature analysis.

a, t-stochastic neighbour embedding (t-SNE) plots of the trinucleotide mutational spectra of biopsy samples from each donor, broken down by organ and donor. Only biopsy samples with more than 30 SNVs were included. b, Heat map showing the clustering of cosine similarities of the trinucleotide mutational context in different samples. Colour bars above indicate information of donors and tissue types. c, Trinucleotide mutational spectra for the unassigned signature and the seven signatures extracted using a Bayesian hierarchical Dirichlet process. The bars represent means (95% credible intervals) of the 96 trinucleotide contexts. d, Heat map depicting the cosine similarities between extracted mutational signatures and mutational signatures from COSMIC and PCAWG catalogues. Cosine similarities between the seven extracted mutational signatures and their most similar comparators are highlighted. e, Stacked bar plots showing the number of mutations that are caused by different mutational signatures.

Extended Data Fig. 6 Mutational signature analysis.

a, Transcriptional strand asymmetries across 96 mutation contexts for SBS4 and SBS22. Bar plots show the sum of assignment probabilities across trinucleotide contexts, split by whether the pyrimidine is on the template or coding strand. b, Transcriptional strand asymmetries across 96 mutation contexts for SBS4 and SBS22. Only mutations with an assignment probability greater than 0.5 are included. c, Trinucleotide mutational spectra of liver, oesophagus, duodenum and colon from donor PN1. Purple dots represent data points of the five tissue layers. Data are mean + s.d. Typical aristolochic-acid -associated mutational features are shaded in blue.

Extended Data Fig. 7 Intra-donor comparisons of mutational signatures.

a, The 96 mutation context profiles in two oesophagus samples from donor PN7 (top) and two liver samples from donor PN9 (bottom) based on somatic mutations detected from WGS. b, Trinucleotide mutational spectra of two dissected duodenum layers from donor PN7. Typical aristolochic-acid -associated mutational features are shaded in blue. c, H&E stained liver tissue (PN9 layer 2 to 4) with superimposed donut charts showing the proportional contributions of mutational signatures, as estimated by deconstructSigs. Scale bars, 200 µm.

Extended Data Fig. 8 Landscape of driver mutations.

a, Mutational landscape of the 32 putative driver genes across different organs from the 5 donors. b, The functional interaction (network of the 32 driver genes. Driver genes are in blue nodes and linker genes (those not significantly mutated but highly connected to driver genes in the network) are in pink nodes. c, Significantly enriched pathways of the 32 driver genes. The vertical red line marks a false discovery rate (FDR) of 0.01. d, Bar plot showing the numbers of total mutations and cancer hotspot mutations in driver genes. The percentages of hotspot mutations are labelled on the top of the bar plot. e, Fraction of driver mutations that are private or shared by more than one biopsy sample. f, Heat maps showing the ratio of the numbers of observed to expected (O/E) driver mutations across different organs (left) and the P values for the enrichment (right). P values from one-sided hypergeometric tests. g, Bar plots comparing the number of mutations in gastric cancer top-10 most frequently mutated driver genes in TCGA with normal stomach and cardia samples in this study. Adjustment for multiple comparisons was performed. Adjusted P values (q-value) are labelled.

Extended Data Fig. 9 Relationships between mutational burdens and average mutant MCFs.

Bubble plots show the correlations between average MCFs and mutational burdens in biopsy samples across different organs in donors PN1, PN2, PN7, PN8 and PN9.

Extended Data Fig. 10 Mutant clonal expansion in oesophageal epithelium.

a, Phylogenetic tree depicting the clonal relationships of the biopsy samples of the oesophagus of donor PN9. b, Heat maps show mutation clustering, spatial clonal architecture and potential driver mutations or CNAs in samples from the oesophagus. Scale bars, 800 µm. c, Heat maps showing potential driver mutations and CNAs in oesophagus samples from donor PN9.

Extended Data Fig. 11 Representative examples of large scale mutant clonal expansion.

a, Heat map showing the mutation clustering in liver samples from donor PN9. b, Spatial clonal architecture of liver tissue from donor PN9. The numbers in each layer represent the positions of LCM biopsy samples. The overlaid colours correspond to a and indicate the ranges of clonal expansions. c, Heat maps show mutation clustering in samples from the representative organs. Each cluster contains mutations with similar MCFs.

Extended Data Fig. 12 Representative examples of independent clonal evolution.

a, Phylogenetic tree depicting the clonal relationships of colon biopsy samples from donor PN9. b, Heat maps showing clustered mutations in samples from representative organs. Each cluster contains mutations with similar MCFs.

Supplementary information

Supplementary Information

This file contains the Supplementary Discussion, including additional discussions about the sampling strategy, mutation burden, clonal expansion patterns, mutational signatures, and copy number alterations in the current study.

Reporting Summary

Supplementary Table 1

Clinical information for the five donors in the current study.

Supplementary Table 2

Whole-exome sequencing information. Nomenclature of sample IDs: for example, ‘PN1E-1-2’ represents the number 2 oesophagus sample dissected from tissue layer 1 from donor PN1.

Supplementary Table 3

Somatic mutations with functional annotations and allele count information detected from whole-exome sequencing. Detailed information for box plots in Fig. 1c and Extended Data Fig. 2c.

Supplementary Table 4

Low-depth and high-depth whole-genome sequencing information and copy number analysis.

Supplementary Table 5

Input matrix for mutational signature analysis using HDP. SNVs detected in both coding and non-coding regions from the whole-exome sequencing data are included. Mutations detected from normal biopsy samples from each dissected layer are merged.

Supplementary Table 6

Extracted mutational signatures and their cosine similarities to the known signatures.

Supplementary Table 7

Relative activates of extracted mutational signatures.

Supplementary Table 8

Driver gene candidates from previous studies (126 gene list).

Supplementary Table 9

Cancer hotspot mutations detected in normal tissues in the current study.

Supplementary Table 10

P values for driver gene enrichment in different tissues. P values were calculated from one-sided hypergeometric tests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, R., Di, L., Li, J. et al. A body map of somatic mutagenesis in morphologically normal human tissues. Nature 597, 398–403 (2021). https://doi.org/10.1038/s41586-021-03836-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03836-1

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