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.

Single-cell analyses reveal increased intratumoral heterogeneity after the onset of therapy resistance in small-cell lung cancer

Abstract

The natural history of small-cell lung cancer (SCLC) includes rapid evolution from chemosensitivity to chemoresistance, although mechanisms underlying this evolution remain obscure due to the scarcity of post-relapse tissue samples. We generated circulating tumor cell (CTC)-derived xenografts from patients with SCLC to study intratumoral heterogeneity (ITH) via single-cell RNA sequencing of chemosensitive and chemoresistant CTC-derived xenografts and patient CTCs. We found globally increased ITH, including heterogeneous expression of therapeutic targets and potential resistance pathways, such as epithelial-to-mesenchymal transition, between cellular subpopulations following treatment resistance. Similarly, serial profiling of patient CTCs directly from blood confirmed increased ITH post-relapse. These findings suggest that treatment resistance in SCLC is characterized by coexisting subpopulations of cells with heterogeneous gene expression leading to multiple, concurrent resistance mechanisms. These findings emphasize the need for clinical efforts to focus on rational combination therapies for treatment-naïve SCLC tumors to maximize initial responses and counteract the emergence of ITH and diverse resistance mechanisms.

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: SCLC CDXs mimic patient disease at the single-cell transcriptional level and by platinum response.
Fig. 2: Platinum-resistant disease is associated with increased ITH.
Fig. 3: Platinum resistance is associated with heterogeneous expression of therapeutic targets or EMT-related genes within specific clusters.
Fig. 4: Serial single-cell RNA-Seq analysis of patient CTCs revealed similar transcriptional heterogeneity to a paired CDX.
Fig. 5: Increased ITH and emergence of cell populations with EMT signatures occur following cisplatin relapse.
Fig. 6: Resistance to DNA-damaging targeted therapies resulted in the emergence of new, therapy-specific clusters.

Similar content being viewed by others

Data availability

The single-cell and bulk RNA-Seq data have been deposited in the NCBI Gene Expression Omnibus database with accession number GSE138474. Source data for Figs. 16 and Extended Data Figs. 16 are provided with the paper. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

Code availability

The bioinformatics analyses were performed using open-source software, including BWA-MEM version 0.7.9a49, VARSCAN2 version 2.3.9 (ref. 50), TOPHAT2 version 2.0.13 (ref. 51), HTSEQ version 0.9.1 (ref. 52), EdgeR version 3.7 (ref. 53), GSEA version 3.0 (ref. 42), ANNOVAR version 2018Apr16 (ref. 54), Seurat version 2.3 (ref. 55) and Cell Ranger version 2.0, as well as in-house R script that is available upon request.

References

  1. Horn, L. et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N. Engl. J. Med. 379, 2220–2229 (2018).

    CAS  PubMed  Google Scholar 

  2. H.R.733: Leech Lake Band of Ojibwe Reservation Restoration Act (Senate and House of Representatives of the United States of America in Congress, 2012). https://www.congress.gov/bill/112th-congress/house-bill/733

  3. Hodgkinson, C. L. et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat. Med. 20, 897–903 (2014).

    CAS  PubMed  Google Scholar 

  4. Drapkin, B. J. et al. Genomic and functional fidelity of small cell lung cancer patient-derived xenografts. Cancer Discov. 8, 600–615 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Chalishazar, M. D. et al. MYC-driven small-cell lung cancer is metabolically distinct and vulnerable to arginine depletion. Clin. Cancer Res. 25, 5107–5121 (2019).

    PubMed  PubMed Central  Google Scholar 

  6. Aggarwal, C. et al. Circulating tumor cells as a predictive biomarker in patients with small cell lung cancer undergoing chemotherapy. Lung Cancer 112, 118–125 (2017).

    PubMed  Google Scholar 

  7. Farago, A. F. et al. Combination olaparib and temozolomide in relapsed small-cell lung cancer. Cancer Discov. 9, 1372–1387 (2019).

    PubMed  PubMed Central  Google Scholar 

  8. Zhang, J. et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science 346, 256–259 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Mollaoglu, G. et al. MYC drives progression of small cell lung cancer to a variant neuroendocrine subtype with vulnerability to aurora kinase inhibition. Cancer Cell 31, 270–285 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Huang, Y. H. et al. POU2F3 is a master regulator of a tuft cell-like variant of small cell lung cancer. Genes Dev. 32, 915–928 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Cardnell, R. J. et al. Protein expression of TTF1 and cMYC define distinct molecular subgroups of small cell lung cancer with unique vulnerabilities to aurora kinase inhibition, DLL3 targeting, and other targeted therapies. Oncotarget 8, 73419–73432 (2017).

    PubMed  PubMed Central  Google Scholar 

  12. Lim, J. S. et al. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature 545, 360–364 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Shue, Y. T., Lim, J. S. & Sage, J. Tumor heterogeneity in small cell lung cancer defined and investigated in pre-clinical mouse models. Transl. Lung Cancer Res. 7, 21–31 (2018).

    PubMed  PubMed Central  Google Scholar 

  14. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-Seq. Science 352, 189–196 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Skoulidis, F. et al. Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities. Cancer Discov. 5, 860–877 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Rudin, C. M. et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat. Rev. Cancer 19, 289–297 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Misch, D. et al. Value of thyroid transcription factor (TTF)-1 for diagnosis and prognosis of patients with locally advanced or metastatic small cell lung cancer. Diagn. Pathol. 10, 21 (2015).

    PubMed  PubMed Central  Google Scholar 

  18. Zhang, W. et al. Small cell lung cancer tumors and preclinical models display heterogeneity of neuroendocrine phenotypes. Transl. Lung Cancer Res. 7, 32–49 (2018).

    PubMed  PubMed Central  Google Scholar 

  19. George, J. et al. Comprehensive genomic profiles of small cell lung cancer. Nature 524, 47–53 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Jahchan, N. S. et al. Identification and targeting of long-term tumor-propagating cells in small cell lung cancer. Cell Rep. 16, 644–656 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Singhi, A. D. et al. MYC gene amplification is often acquired in lethal distant breast cancer metastases of unamplified primary tumors. Mod. Pathol. 25, 378–387 (2012).

    CAS  PubMed  Google Scholar 

  22. Lee, H. Y. et al. c-MYC drives breast cancer metastasis to the brain, but promotes synthetic lethality with TRAIL. Mol. Cancer Res. 17, 544–554 (2019).

    CAS  PubMed  Google Scholar 

  23. Stewart, C. A. et al. Dynamic variations in epithelial-to-mesenchymal transition (EMT), ATM, and SLFN11 govern response to PARP inhibitors and cisplatin in small cell lung cancer. Oncotarget 8, 28575–28587 (2017).

    PubMed Central  Google Scholar 

  24. Gardner, E. E. et al. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2–SLFN11 axis. Cancer Cell 31, 286–299 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Wagner, A. H. et al. Recurrent WNT pathway alterations are frequent in relapsed small cell lung cancer. Nat. Commun. 9, 3787 (2018).

    PubMed  PubMed Central  Google Scholar 

  26. Byers, L. A. et al. Proteomic profiling identifies dysregulated pathways in small cell lung cancer and novel therapeutic targets including PARP1. Cancer Discov. 2, 798–811 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Dammert, M. A. et al. MYC paralog-dependent apoptotic priming orchestrates a spectrum of vulnerabilities in small cell lung cancer. Nat. Commun. 10, 3485 (2019).

    PubMed  PubMed Central  Google Scholar 

  28. Semenova, E. A. et al. Transcription factor NFIB is a driver of small cell lung cancer progression in mice and marks metastatic disease in patients. Cell Rep. 16, 631–643 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Bottger, F. et al. Tumor heterogeneity underlies differential cisplatin sensitivity in mouse models of small-cell lung cancer. Cell Rep. 27, 3345–3358.e4 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Wu, N. et al. NFIB overexpression cooperates with Rb/p53 deletion to promote small cell lung cancer. Oncotarget 7, 57514–57524 (2016).

    PubMed  PubMed Central  Google Scholar 

  31. Klameth, L. et al. Small cell lung cancer: model of circulating tumor cell tumorospheres in chemoresistance. Sci. Rep. 7, 5337 (2017).

    PubMed  PubMed Central  Google Scholar 

  32. Hamilton, G., Hochmair, M., Rath, B., Klameth, L. & Zeillinger, R. Small cell lung cancer: circulating tumor cells of extended stage patients express a mesenchymal–epithelial transition phenotype. Cell Adh. Migr. 10, 360–367 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Yu, N., Zhou, J., Cui, F. & Tang, X. Circulating tumor cells in lung cancer: detection methods and clinical applications. Lung 193, 157–171 (2015).

    CAS  PubMed  Google Scholar 

  34. Tanaka, F. et al. Circulating tumor cell as a diagnostic marker in primary lung cancer. Clin. Cancer Res. 15, 6980–6986 (2009).

    CAS  PubMed  Google Scholar 

  35. Rudin, C. M. et al. Rovalpituzumab tesirine, a DLL3-targeted antibody–drug conjugate, in recurrent small-cell lung cancer: a first-in-human, first-in-class, open-label, phase 1 study. Lancet Oncol. 18, 42–51 (2017).

    CAS  PubMed  Google Scholar 

  36. Giffin, M. et al. Targeting DLL3 with AMG 757, a BiTE® antibody construct, and AMG 119, a CAR-T, for the treatment of SCLC. J. Thorac. Oncol. 13, abstr. P3.12-03 (2018).

    Google Scholar 

  37. Carbone, D. P. et al. Efficacy and safety of rovalpituzumab tesirine in patients with DLL3-expressing, ≥ 3rd line small cell lung cancer: results from the phase 2 TRINITY study. J. Clin. Oncol. 36(Suppl.), 8507 (2018).

    Google Scholar 

  38. Paz-Ares, L. et al. Overall survival with durvalumab plus etoposide–platinum in first-line extensive-stage SCLC: results from the CASPIAN study. J. Thorac. Oncol. 14, S7–S8 (2019).

    Google Scholar 

  39. Nugent, J. L. et al. CNS metastases in small cell bronchogenic carcinoma: increasing frequency and changing pattern with lengthening survival. Cancer 44, 1885–1893 (1979).

    CAS  PubMed  Google Scholar 

  40. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Jamieson, A. R. et al. Exploring nonlinear feature space dimension reduction and data representation in breast CADx with laplacian eigenmaps and t-SNE. Med. Phys. 37, 339–351 (2010).

    PubMed  Google Scholar 

  42. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  45. Tong, P., Chen, Y., Su, X. & Coombes, K. R. SIBER: systematic identification of bimodally expressed genes using RNAseq data. Bioinformatics 29, 605–613 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Wang, J., Wen, S., Symmans, W. F., Pusztai, L. & Coombes, K. R. The bimodality index: a criterion for discovering and ranking bimodal signatures from cancer gene expression profiling data. Cancer Inform. 7, 199–216 (2009).

    PubMed  PubMed Central  Google Scholar 

  47. Byers, L. A. et al. An epithelial–mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin. Cancer Res. 19, 279–290 (2013).

    CAS  PubMed  Google Scholar 

  48. R Core Development Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2015).

  49. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).

    PubMed  PubMed Central  Google Scholar 

  50. Koboldt, D. C. et al. VarScan: variant detection in massively parallel sequencing of individual and pooled samples. Bioinformatics 25, 2283–2285 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  52. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  53. Dai, Z. et al. edgeR: a versatile tool for the analysis of shRNA-Seq and CRISPR–Cas9 genetic screens. F1000Res 3, 95 (2014).

    PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  55. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the patients who participated in this study, as well as their families. We also thank M. Vasquez for obtaining consent from the patients, E. Roarty for scientific input and editing, and K. Ramkumar for general laboratory assistance. This work was supported by NIH/NCI Cancer Center Support Grant P30-CA016672 (to the Bioinformatics Shared Resource), NIH/NCI T32 Award CA009666 (to C.M.G.), The University of Texas Southwestern Medical Center and MD Anderson Cancer Center Special Program of Research Excellence (5 P50 CA070907), NIH/NCI award R01-CA207295 (to L.A.B.), NIH/NCI award U01-CA213273 (to J.V.H. and L.A.B.), NIH/NCI award U01 CA231844 (to T.G.O.), award P30CA042014 (to the Huntsman Cancer Institute), the Department of Defense award LC170171 (to L.A.B.), the ASCO Young Investigator Award (to C.M.G.), generous philanthropic contributions to The University of Texas MD Anderson Cancer Center Moon Shots Program (to J.V.H., J.W. and L.A.B.), The University of Texas MD Anderson Cancer Center Small Cell Lung Cancer Working Group, Abell Hangar Foundation Distinguished Professor Endowment (to L.A.B. and B.G.), The University of Texas MD Anderson Cancer Center Physician Scientist Award (to L.A.B.), The Hope Foundation SWOG/ITSC Pilot Program (to P.R. and L.A.B.), an Andrew Sabin Family Fellowship (to L.A.B.) and Rexanna’s Foundation for Fighting Lung Cancer (to J.V.H. and L.A.B.).

Author information

Authors and Affiliations

Authors

Contributions

C.A.S., C.M.G. Y.X. and L.A.B. conceived of the project, analyzed and interpreted the data and wrote the manuscript. S.S., V.S., V.B., P.R., J.Z., B.G., J.d.G., S.G.S., J.A.R., M.D.C., T.G.O. and J.V.H. contributed to acquiring the data. J.F., C.M., N.K., J.S. and I.W. performed the pathology review and analysis. M.B. and J.W. contributed to analysis and interpretation of the data. P.M.H. collected liquid biopsies from the patients. H.T. coordinated the patient protocols. P.R., J.Z., B.G., J.d.G., S.G.S., J.A.R., M.D.C., T.G.O. and J.V.H. provided administrative and/or material support. All authors contributed to the writing, reviewing and/or revising of the manuscript.

Corresponding author

Correspondence to Lauren Averett Byers.

Ethics declarations

Competing interests

L.A.B. serves on advisory committees for AstraZeneca, AbbVie, Genmab, BerGenBio, Pharma Mar SA, Sierra Oncology, Merck, Bristol-Myers Squibb, Genentech and Pfizer and has research support from AbbVie, AstraZeneca, Genmab, Sierra Oncology and Tolero Pharmaceuticals. J.V.H. serves on advisory committees for AstraZeneca, Boehringer Ingelheim, Exelixis, Genentech, GlaxoSmithKline, Guardant Health, Hengrui, Lilly, Novartis, Spectrum, EMD Serono and Synta, and has research support from AstraZeneca, Bayer, GlaxoSmithKline and Spectrum and royalties and licensing fees from Spectrum. Otherwise, there are no pertinent financial or non-financial conflicts of interest to report.

Additional information

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

Extended data

Extended Data Fig. 1 CDXs exhibit common SCLC markers and mutations that are maintained over multiple generations.

a, Histological analysis of CDX tumors are consistent with SCLC. Scale bar = 100 µM. b, Patient expression of NCAM and TTF1 by staff pathologist review of diagnostic sample matches CDXs. c, Presence of parenchymal brain metastasis, confirmed by staff neuroradiologist and treating physician review, in the cerebellum (indicated by dashed circle) of the patient from which MDA-SC39 was derived. d, Genomic alterations in CDXs. Top panel: mutation load; middle panel: somatic mutations and genomic gain/loss status; lower panel: type of base-pair substitution. e, Mutational status of common SCLC genes and others unique to each CDX are maintained over multiple CDX passages in three separate models. f, Expression heatmap for ASCL1- and NEUROD1-associated genes. b, CDX and PDX models derived from patient SC49 exhibit similar patterns of expression for common SCLC markers, including loss of TTF1 expression. These experiments were repeated in three independent tumors from each model. Scale bar = 100 µM.

Source data

Extended Data Fig. 2 ITH among SCLC molecular subtypes.

a, t-SNE visualization of NE gene expression status in all CDXs. b, t-SNE visualization of cell populations from biological replicates of MDA-SC39s and MDA-SC16r obtained from tumors grown in the same passage, but different mice. Note mixing of cell populations indicate that clustering is not due to variations in replicate. c, Heatmap analysis of NE gene expression indicating that all CDXs are considered high neuroendocrine subtypes. d, Expression of ASCL1 and NEUROD1 in all CDXs by both violin plot to indicate range in expression and feature plot to show abundance. e, Violin plots indicating expression of MYC family members in the CDXs. Each dot represents one cell and the violin curve represent the density of the cells at different expression levels. f, EMT score is elevated within MDA-SC39s and MDA-SC49r, which corresponds with increased expression of VIM and decreased EPCAM. In a, b, d, e, and f, n=2,000 cells each.

Source data

Extended Data Fig. 3 Validation of cluster calls and visualization.

a, Silhouette analysis to determine cluster number in each of the eight CDXs. b, UMAP visualization of the clusters in all CDXs. c, Barplot of variations of absolute normalized enrichment scores (NES) for hallmark pathways in GSEA analysis in sensitive clusters (blue) and resistant clusters (red). The variation of pathway enrichment is higher in resistant clusters than sensitive clusters by one-sided Wilcoxon rank sum test (P=2.9e-6; n=21 pathways).

Source data

Extended Data Fig. 4 CDX copy number and expression of DNA repair genes between clusters.

a,b, Inferred copy number between clusters in MDA-SC16r (a) and MDA-SC49r (b). c, Expression heatmap of genes associated with DNA repair in all CDX clusters. d, Violin plots indicating range of expression of several therapeutic targets within individual clusters. AURKA, AURKB and DLL3 were relatively unchanged between clusters. MDA-SC4s: n=978, 1,022 cells for clusters 1-2; MDA-SC39s: n=1172, 828 cells for clusters 1-2; MDA-SC68s: n=733, 704, 563 cells for clusters 1-3; HCI-008s: n=596, 1,404 cells for clusters 1-2; MDA-SC49r: n=683, 317, 652, 348 cells for clusters 1-4. Each dot represents one cell and the violin curve represent the density of the cells at different expression levels.

Source data

Extended Data Fig. 5 Validation of CTC identification within a patient liquid biopsy by positive expression of epithelial, NE and SCLC genes.

Percentage of cells expressing epithelial, NE genes (for example, UCHL1, NCAM1, SYP, and CHGA) or SCLC lineage-specific genes (for example, ASCL1, NEUROD1, etc.) in the CTC population and non-CTC populations.

Source data

Extended Data Fig. 6 Emergence of a mesenchymal cell cluster following cisplatin-treatment.

Violin plot of VIM (a) and EXPCAM (b) expression in the clusters of MDA-SC68s vehicle and cisplatin-treated CDXs. MDA-SC68 vehicle: n=733, 704, 563 cells for clusters 1-3; MDA-SC68 cisplatin: n=635, 489, 71, 467, 338 cells for clusters 1-5. Each dot represents one cell and the violin curve represent the density of the cells at different expression levels Source data.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Stewart, C.A., Gay, C.M., Xi, Y. et al. Single-cell analyses reveal increased intratumoral heterogeneity after the onset of therapy resistance in small-cell lung cancer. Nat Cancer 1, 423–436 (2020). https://doi.org/10.1038/s43018-019-0020-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-019-0020-z

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