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.

  • Letter
  • Published:

Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers

An Author Correction to this article was published on 19 November 2019

This article has been updated

Abstract

During cancer therapy, tumor heterogeneity can drive the evolution of multiple tumor subclones harboring unique resistance mechanisms in an individual patient1,2,3. Previous case reports and small case series have suggested that liquid biopsy (specifically, cell-free DNA (cfDNA)) may better capture the heterogeneity of acquired resistance4,5,6,7,8. However, the effectiveness of cfDNA versus standard single-lesion tumor biopsies has not been directly compared in larger-scale prospective cohorts of patients following progression on targeted therapy. Here, in a prospective cohort of 42 patients with molecularly defined gastrointestinal cancers and acquired resistance to targeted therapy, direct comparison of postprogression cfDNA versus tumor biopsy revealed that cfDNA more frequently identified clinically relevant resistance alterations and multiple resistance mechanisms, detecting resistance alterations not found in the matched tumor biopsy in 78% of cases. Whole-exome sequencing of serial cfDNA, tumor biopsies and rapid autopsy specimens elucidated substantial geographic and evolutionary differences across lesions. Our data suggest that acquired resistance is frequently characterized by profound tumor heterogeneity, and that the emergence of multiple resistance alterations in an individual patient may represent the ‘rule’ rather than the ‘exception’. These findings have profound therapeutic implications and highlight the potential advantages of cfDNA over tissue biopsy in the setting of acquired resistance.

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: Identification of acquired resistance mechanisms in liquid versus tumor biopsy.
Fig. 2: Comparison of multiple tumor biopsies versus liquid biopsy in a BRAF-mutant colorectal cancer patient.
Fig. 3: Serial liquid biopsy and autopsy in a patient with FGFR2 fusion-positive gastric cancer (TPS177).

Similar content being viewed by others

Data availability

Sequencing data from WES and RNA-Seq are available in the database of Genotypes and Phenotypes with the accession number phs001853.v1.p1.

Change history

  • 19 November 2019

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Burrell, R. A. & Swanton, C. Tumour heterogeneity and the evolution of polyclonal drug resistance. Mol. Oncol. 8, 1095–1111 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gerlinger, M. et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 46, 225–233 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. McGranahan, N. & Swanton, C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell 27, 15–26 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Bettegowda, C. et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 6, 224ra24 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Goyal, L. et al. Polyclonal secondary mutations drive acquired resistance to FGFR inhibition in patients with FGFR2 fusion-positive cholangiocarcinoma. Cancer Discov. 7, 252–263 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Piotrowska, Z. et al. Heterogeneity underlies the emergence of EGFR T790 wild-type clones following treatment of T790M-positive cancers with a third-generation EGFR inhibitor. Cancer Discov. 5, 713–722 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Russo, M. et al. Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer. Cancer Discov. 6, 147–153 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Siravegna, G. et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat. Med. 21, 795–801 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Garraway, L. A. & Jänne, P. A. Circumventing cancer drug resistance in the era of personalized medicine. Cancer Discov. 2, 214–226 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jamal-Hanjani, M. et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 376, 2109–2121 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Misale, S., Di Nicolantonio, F., Sartore-Bianchi, A., Siena, S. & Bardelli, A. Resistance to anti-EGFR therapy in colorectal cancer: from heterogeneity to convergent evolution. Cancer Discov. 4, 1269–1280 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Misale, S. et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 486, 532–536 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Turajlic, S. et al. Deterministic evolutionary trajectories influence primary tumor growth: TRACERx Renal. Cell 173, 595–610.e11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hazar-Rethinam, M. et al. Convergent therapeutic strategies to overcome the heterogeneity of acquired resistance in BRAF V600E colorectal cancer. Cancer Discov. 8, 417–427 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Morelli, M. P. et al. Characterizing the patterns of clonal selection in circulating tumor DNA from patients with colorectal cancer refractory to anti-EGFR treatment. Ann. Oncol. 26, 731–736 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Piotrowska, Z. et al. Heterogeneity and coexistence of T790M and T790 wild-type resistant subclones drive mixed response to third-generation epidermal growth factor receptor inhibitors in lung cancer. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00263 (2018).

  18. Blakely, C. M. et al. Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat. Genet. 49, 1693–1704 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Haber, D. A. & Velculescu, V. E. Blood-based analyses of cancer: circulating tumor cells and circulating tumor DNA. Cancer Discov. 4, 650–661 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chabon, J. J. et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat. Commun. 7, 11815 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Diaz, L. A. Jr. et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486, 537–540 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Strickler, J. H. et al. Genomic landscape of cell-free DNA in patients with colorectal cancer. Cancer Discov. 8, 164–173 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Thierry, A. R. et al. Circulating DNA demonstrates convergent evolution and common resistance mechanisms during treatment of colorectal cancer. Clin. Cancer Res. 23, 4578–4591 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ahronian, L. G. et al. Clinical acquired resistance to RAF inhibitor combinations in BRAF-mutant colorectal cancer through MAPK pathway alterations. Cancer Discov. 5, 358–367 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Arena, S. et al. MM-151 overcomes acquired resistance to cetuximab and panitumumab in colorectal cancers harboring EGFR extracellular domain mutations. Sci. Transl. Med. 8, 324ra14 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Bahcall, M. et al. Acquired MET D1228V mutation and resistance to MET inhibition in lung cancer. Cancer Discov. 6, 1334–1341 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bertotti, A. et al. A molecularly annotated platform of patient-derived xenografts (“xenopatients”) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 1, 508–523 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Bertotti, A. et al. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature 526, 263–267 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Heist, R. S. et al. Acquired resistance to crizotinib in NSCLC with MET exon 14 skipping. J. Thorac. Oncol. 11, 1242–1245 (2016).

    Article  PubMed  Google Scholar 

  30. Kwak, E. L. et al. Molecular heterogeneity and receptor coamplification drive resistance to targeted therapy in MET-amplified esophagogastric cancer. Cancer Discov. 5, 1271–1281 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Montagut, C. et al. Identification of a mutation in the extracellular domain of the epidermal growth factor receptor conferring cetuximab resistance in colorectal cancer. Nat. Med. 18, 221–223 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Oddo, D. et al. Molecular landscape of acquired resistance to targeted therapy combinations in BRAF-mutant colorectal cancer. Cancer Res. 76, 4504–4515 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Siravegna, G. et al. Radiologic and genomic evolution of individual metastases during HER2 blockade in colorectal cancer. Cancer Cell 34, 148–162.e7 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Tan, L. et al. Development of covalent inhibitors that can overcome resistance to first-generation FGFR kinase inhibitors. Proc. Natl Acad. Sci. USA 111, E4869–E4877 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yaeger, R. et al. Mechanisms of acquired resistance to BRAF V600E inhibition in colon cancers converge on RAF dimerization and are sensitive to its inhibition. Cancer Res. 77, 6513–6523 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Leshchiner, I., et al. Comprehensive analysis of tumour initiation, spatial and temporal progression under multiple lines of treatment. Preprint at bioRxiv https://www.biorxiv.org/content/10.1101/508127v1(2018).

  37. Janjigian, Y. Y. et al. Genetic predictors of response to systemic therapy in esophagogastric cancer. Cancer Discov. 8, 49–58 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Kim, S. T. et al. Impact of genomic alterations on lapatinib treatment outcome and cell-free genomic landscape during HER2 therapy in HER2+ gastric cancer patients. Ann. Oncol. 29, 1037–1048 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Pectasides, E. et al. Genomic heterogeneity as a barrier to precision medicine in gastroesophageal adenocarcinoma. Cancer Discov. 8, 37–48 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Rothwell, D. G. et al. Utility of ctDNA to support patient selection for early phase clinical trials: the TARGET study. Nat. Med. 25, 738–743 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Lanman, R. B. et al. Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA. PLoS ONE 10, e0140712 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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 

  43. Kim, S. et al. Strelka2: fast and accurate calling of germline and somatic variants. Nat. Methods 15, 591–594 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Taylor-Weiner, A. et al. DeTiN: overcoming tumor-in-normal contamination. Nat. Methods 15, 531–534 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Cibulskis, K. et al. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Bioinformatics 27, 2601–2602 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Carter, S. L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413–421 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Prelić, A. et al. A systematic comparison and evaluation of biclustering methods for gene expression data. Bioinformatics 22, 1122–1129 (2006).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Grant Support: This work is partially supported by the NIH/NCI Gastrointestinal Cancer Specialized Program of Research Excellence P50 (CA127003, R01CA208437, K08CA166510 and U54CA224068), a Damon Runyon Clinical Investigator Award and a Stand Up To Cancer Colorectal Dream Team Translational Research Grant (grant number SU2C-AACR-DT22-17 to R.B.C.). Research grants are administered by the American Association of Cancer Research, the scientific partners of Stand Up To Cancer. The work is partially supported by the Broad/IBM Cancer Resistance Research Project (G.G. and L.P.) and the Susan Eid Tumor Heterogeneity Initiative (D.J.). The research leading to these results has received funding from FONDAZIONE AIRC under 5 per Mille 2018 ID. 21091 program – P. I. Bardelli Alberto. The research leading to these results has received funding from FONDAZIONE AIRC: AIRC Investigator Grant 2015 – ID. 16788; AIRC Investigator Grant 2018 – ID. 21923; AIRC–CRUK–FC AECC Accelerator Award – contract 22795. This work was also supported by the European Community’s Seventh Framework Programme (grant agreement number 602901 MErCuRIC), H2020 (grant agreement number 635342-2 MoTriColor), IMI (contract number 115749 CANCER-ID), Ministero della Salute (project NET-2011-02352137) and Fondazione Piemontese per la Ricerca sul Cancro–ONLUS 5 per Mille 2014 and 2015 Ministero della Salute. G.S. was funded by a Roche per la Ricerca grant (2017) and AIRC three-year fellowship (2017).

Author information

Authors and Affiliations

Authors

Contributions

A.R.P., I.L., L.G., D.J., G.G. and R.B.C. conceived of the study; A.R.P., I.L., L.E., L.G., C.L., G.S., D.L., K.R., E.E.M., E.E.V.S., F.U., F.F.d.l.C., I.J.F., B.N., H.A.S., F.A., I.D.-G., M.H.-R., D.D.-S., D.T.T., A.J.I., A.B., L.P., D.J., G.G. and R.B.C. performed data analysis and interpretation; I.L., L.E., C.L., D.L., K.R., E.E.M., F.U., L.P. and G.G. performed analysis of whole exome sequencing and subclonal reconstruction; A.R.P., I.L., L.G., G.S., D.D.-S., D.T.T., A.J.I., A.B., L.P., D.J., G.G. and R.B.C. supervised the data analysis; A.R.P., I.L., L.E., C.L., K.R., E.E.V.S., F.U., K.S., B.P.D., L.P., G.G. and R.B.C. participated in writing of the manuscript and generated figures; M.H., B.P.D., T.R.G., and V.A.A. suggested manuscript edits; A.R.P., L.G., E.E.V.S., C.J.P., J.N.A., L.S.B., J.W.C., B.G., J.E.M., R.D.N., E.R., D.P.R., C.D.W., E.L.K., J.E.F., D.D.-S., D.T.T., A.X.Z., J.Y.W., T.S.H., A.J.I., D.J., R.B.C., M.H., A.W., V.A. performed sample and data collection; funding was obtained by T.R.G. and G.G.; and A.R.P., I.L., E.E.V.S., C.J.P., M.H., K.S., A.W., B.P.D., and G.G. and R.B.C. participated in project administration.

Corresponding authors

Correspondence to Gad Getz or Ryan B. Corcoran.

Ethics declarations

Competing interests

A.R.P. is a consultant/advisory board member for PureTech, Driver, Foundation Medicine and Eisai, and has institutional research funding from Array, Plexxikon, Guardant, BMS, MacroGenics, Genentech, Novartis, OncoMed and Tolero. L.G. is a consultant/advisory board member for Debiopharm, H3 Biomedicine and Pieris Pharmaceuticals, and a steering committee member for Agios Pharmaceuticals, Taiho Pharmaceuticals and Debiopharm. E.R. is an advisory board member/consultant for Helsinn, Heron, BASF, American Imaging Management, Napo, Immuneering and Vector Oncology. D.P.R. serves on advisory boards for MPM Capital, Gritstone Oncology, Oncorus and TCR2, has equity in MPM Capital and Acworth Pharmaceuticals, and serves as an author for Johns Hopkins University Press, UpToDate and McGraw Hill. C.D.W. is a consultant for Celgene. J.E.F. is an employee at Novartis, has served as an advisory board member/consultant for Merrimack and N-of-One, and received research funding from Novartis, Roche/Genentech, Agios, Takeda, Sanofi, Celgene and Exelixis, as well as travel support from Roche/Genentech. E.L.K. is an employee of Novartis. M.H.-R. is an employee of Forma Therapeutics. D.T.T. is a consultant/advisory board member for Merrimack, Roche Ventana and EMD Millipore–Sigma, is a founder and has equity in PanTher Therapeutics, and receives research funding from ACD-Biotechne. A.X.Z. is a consultant/advisor for AstraZeneca, Bayer, Bristol-Myers Squibb, Eisai, Eli Lilly, Exelixis, Merck, Novartis and Roche/Genentech, and received research funding from Bayer, Bristol-Myers Squibb, Eli Lilly, Merck and Novartis. T.S.H. is a consultant/advisory board member for Merck and EMD Serono, and received research support from Taiho, AstraZeneca, Bristol-Myers Squibb, Mobetron and Ipsen. A.J.I. is a consultant for DebioPharm, Chugai and Roche, received research support from Sanofi and has equity in ArcherDX. T.R.G. is or has been a consultant/advisor for Foundation Medicine, GlaxoSmithKline, Sherlock Biosciences and Forma Therapeutics, and holds equity in Sherlock Biosciences and Forma Therapeutics. R.B.C. is a consultant/advisory board member for Amgen, Array Biopharma, Astex Pharmaceuticals, Avidity Biosciences, BMS, C4 Therapeutics, Chugai, Elicio, Fog Pharma, Fount Therapeutics, Genentech, LOXO, Merrimack, N-of-One, Novartis, nRichDX, Revolution Medicines, Roche, Roivant, Shionogi, Shire, Spectrum Pharmaceuticals, Symphogen, Taiho and Warp Drive Bio, holds equity in Avidity Biosciences, C4 Therapeutics, Fount Therapeutics, nRichDX and Revolution Medicines, and has received research funding from Asana, AstraZeneca and Sanofi. D.J. is an advisor/consultant for Novartis, Genentech, Eisai, Ipsen and EMD Serono, and receives research support from Novartis, Genentech, Eisai, EMD Serono, Takeda, Celgene and Placon. G.G. receives research funds from IBM and Pharmacyclics, and is an inventor on patent applications related to MuTect, ABSOLUTE, and POLYSOLVER. K.R., F.U., C.L. and L.P. are listed as co-inventors on a patent application currently pending review at the USPTO. I.L. L.E., D.L., E.E.V.S., E.E.M., M.H., K.S., C.J.P., A.W., B.P.D., F.F.d.l.C., I.J.F., B.N., H.A.S., J.N.A., L.S.B., J.W.C., B.G., J.E.M., R.D.N., F.A., I.D.-G., D.D.-S. and V.A.A. declare no competing interests.

Additional information

Peer review information Javier Carmona was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended Data

Extended Data Fig. 1 Phylogenetic trees for paired tumor biopsies.

Phylogenetic trees for patients with WES for pre- and post-treatment samples. The numbers of somatic alterations assigned to each cluster and detected events in known cancer genes appear on the branches. Two-dimensional plots show the CCF distributions of subclones in the pre- and post-treatment samples. Events in known cancers are shown next to their subclonal cluster.

Extended Data Fig. 2 Patients with multiple postprogression tumor biopsies.

Three of five patients with multiple postprogression tumor biopsies are shown. The other two patients (TPS037 and TPS177) are shown in Figs. 2 and 3, respectively. For each patient, the location of each tumor biopsy, as well as the resistance alterations (blue) and truncal alterations (orange) detected in cfDNA and in each tumor specimen, are shown. TPS001 is a patient with RAS wild type colorectal cancer who developed resistance to an anti-EGFR antibody8. Biopsy of one liver lesion revealed an activating MEK1 (MAP2K1) mutation, and biopsy of a second liver lesion identified a KRAS mutation. Both resistance alterations were detected in postprogression cfDNA. TPS007 is an FGFR2 fusion-positive cholangiocarcinoma patient who developed resistance to an FGFR inhibitor5. Five FGFR2 resistance mutations were identified in postprogression cfDNA, and three of these alterations were identified in distinct liver lesions harvested through a rapid autopsy program, with one lesion harboring two FGFR2 alterations. TPS011 is a patient with RAS wild type colorectal cancer who developed resistance to an anti-EGFR antibody. A recurrent colon tumor harbored a KRAS mutation, and an EGFR extracellular domain mutation known to interfere with antibody binding was identified in an ovarian metastasis, whereas both alterations were detected in cfDNA. Importantly, in all patients, individual resistance mechanisms emerging in distinct metastatic lesions were detectable in cfDNA.

Extended Data Fig. 3 Biclusters of patients based on similar changes (δ) in somatic alteration.

Biclustering of four δ matrices, reflecting changes in CCF of mutations or copy number in known cancer genes or genesets, yielded significant biclusters (all empirical P values < 0.0001; Methods). The biclusters from all four δ matrices included at least one bicluster with patient TPS130 (a patient with an unknown mechanism of resistance). Patient TPS130 consistently biclustered together with TPS021 and TPS037 (patients with known mechanisms of resistance) across all matrices, highlighting the possibility that additional genomic alterations contribute to resistance beyond the identified resistance alterations. a, The change in somatic alterations, δ, was calculated based on WES data of the samples closest to the start and end of therapy. We biclustered four δ matrices (\(\delta _{{\rm{cancer}}\;{\rm{gene}}}^{{\rm{copy}}\,{\rm{number}}}\), \(\delta _{{\rm{geneset}}}^{{\rm{mutation}}}\), \(\delta _{{\rm{geneset}}}^{{\rm{copy}}\,{\rm{number}}}\) and \(\delta _{{\rm{geneset}}}^{{\rm{copy}}\,{\rm{number}}\;{\rm{and}}\;{\rm{mutation}}}\)) and assessed their significance by comparing the size of biclusters against n = 10,000 permuted matrices with a two-sided t-test (Methods). be, Illustration of the biclustering results (using BiMax) of the four δ matrices (biclusters are listed in Supplementary Table 6). Outlined in red are biclusters containing TPS130 observed in all four δ matrices.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Parikh, A.R., Leshchiner, I., Elagina, L. et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat Med 25, 1415–1421 (2019). https://doi.org/10.1038/s41591-019-0561-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0561-9

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