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:

Overall survival with circulating tumor DNA-guided therapy in advanced non-small-cell lung cancer

Abstract

Circulating tumor DNA (ctDNA) sequencing guides therapy decisions but has been studied mostly in small cohorts without sufficient follow-up to determine its influence on overall survival. We prospectively followed an international cohort of 1,127 patients with non-small-cell lung cancer and ctDNA-guided therapy. ctDNA detection was associated with shorter survival (hazard ratio (HR), 2.05; 95% confidence interval (CI), 1.74–2.42; P < 0.001) independently of clinicopathologic features and metabolic tumor volume. Among the 722 (64%) patients with detectable ctDNA, 255 (23%) matched to targeted therapy by ctDNA sequencing had longer survival than those not treated with targeted therapy (HR, 0.63; 95% CI, 0.52–0.76; P < 0.001). Genomic alterations in ctDNA not detected by time-matched tissue sequencing were found in 25% of the patients. These ctDNA-only alterations disproportionately featured subclonal drivers of resistance, including RICTOR and PIK3CA alterations, and were associated with short survival. Minimally invasive ctDNA profiling can identify heterogeneous drivers not captured in tissue sequencing and expand community access to life-prolonging therapy.

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
Fig. 2: ctDNA alterations and OS.
Fig. 3: Clinical utility of ctDNA and tissue sequencing.
Fig. 4: ctDNA mutations not detected on time-matched tissue sequencing (MSK-IMPACT).
Fig. 5: Correlates of plasma–tissue divergence.

Similar content being viewed by others

Data availability

Genomic and clinical data are available on cBioPortal (https://www.cbioportal.org/study/summary?id=nsclc_ctdx_msk_2022). The raw sequencing data for MSK-IMPACT and MSK-ACCESS are protected and are not broadly available due to privacy laws. Researchers at MSK with appropriate IRB permission may request the data from the Center for Molecular Oncology (skicmopm@mskcc.org).

Code availability

No software was used for data collection.

References

  1. Kris, M. G. et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 311, 1998–2006 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Howlader, N. et al. The effect of advances in lung-cancer treatment on population mortality. N. Engl. J. Med. 383, 640–649 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pasche, B. & Grant, S. C. Non-small cell lung cancer and precision medicine a model for the incorporation of genomic features into clinical trial design. JAMA 311, 1975 (2014).

    Article  PubMed  Google Scholar 

  4. Bruno, D. S. et al. Racial disparities in biomarker testing and clinical trial enrollment in non-small cell lung cancer (NSCLC). J. Clin. Oncol. 39, 9005 (2021).

    Article  Google Scholar 

  5. Robert, N. J. et al. Biomarker tissue journey among patients (pts) with untreated metastatic non-small cell lung cancer (mNSCLC) in the U.S. Oncology Network community practices. J. Clin. Oncol. 39, 9004 (2021).

    Article  Google Scholar 

  6. Zugazagoitia, J. et al. Clinical utility of plasma-based digital next-generation sequencing in patients with advance-stage lung adenocarcinomas with insufficient tumor samples for tissue genotyping. Ann. Oncol. 30, 290–296 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Sheridan, C. Investors keep the faith in cancer liquid biopsies. Nat. Biotechnol. 37, 972–974 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Rolfo, C. et al. Liquid biopsy for advanced non-small cell lung cancer: a consensus statement from the International Association for the Study of Lung Cancer (IASLC). J. Thorac. Oncol. 16, 1647–1662 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Heitzer, E., Haque, I. S., Roberts, C. E. S. & Speicher, M. R. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat. Rev. Genet. 20, 71–88 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Sabari, J. K. et al. A prosspective study of circulating tumor DNA to guide matched targeted therapy in lung cancers. J. Natl Cancer Inst. 111, 575–583 (2019).

    Article  PubMed  Google Scholar 

  11. Oxnard, G. R. et al. Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non-small-cell lung cancer. J. Clin. Oncol. 34, 3375–3382 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Aggarwal, C. et al. Clinical implications of plasma-based genotyping with the delivery of personalized therapy in metastatic non-small cell lung cancer. JAMA Oncol. 5, 173 (2019).

    Article  PubMed  Google Scholar 

  13. Sacher, A. G. et al. Prospective validation of rapid plasma genotyping for the detection of EGFR and KRAS mutations in advanced lung cancer. JAMA Oncol. 2, 1014–1022 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  Google Scholar 

  15. Leighl, N. B. et al. Clinical utility of comprehensive cell-free DNA analysis to identify genomic biomarkers in patients with newly diagnosed metastatic non-small cell lung cancer. Clin. Cancer Res. 25, 4691–4700 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Remon, J. et al. Osimertinib benefit in EGFR-mutant NSCLC patients with T790M-mutation detected by circulating tumour DNA. Ann. Oncol. 28, 784–790 (2017).

    Article  CAS  PubMed  Google Scholar 

  17. Tran, H. T. et al. Clinical outcomes in non-small cell lung cancer patients treated with EGFR-tyrosine kinase inhibitors and other targeted therapies based on tumor versus plasma genomic profiling. JCO Precis. Oncol. 5, 1241–1249 (2021).

    Article  Google Scholar 

  18. Yang, M. et al. Incorporating blood-based liquid biopsy information into cancer staging: time for a TNMB system? Ann. Oncol. 29, 311–323 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Chaudhuri, A. A. et al. Early detection of molecular residual disease in localized lung cancer by circulating tumor DNA profiling. Cancer Discov. 7, 1394–1403 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nabet, B. Y. et al. Noninvasive early identification of therapeutic benefit from immune checkpoint inhibition. Cell 183, 363–376 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Goldberg, S. B. et al. Early assessment of lung cancer immunotherapy response via circulating tumor DNA. Clin. Cancer Res. 24, 1872–1880 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gandara, D. R. et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small cell lung cancer patients treated with atezolizumab. Nat. Med. 24, 1441–1448 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Karachaliou, N. et al. Association of EGFR L858R mutation in circulating free DNA with survival in the EURTAC trial. JAMA Oncol. 1, 149–157 (2015).

    Article  PubMed  Google Scholar 

  24. Mack, P. C. et al. Spectrum of driver mutations and clinical impact of circulating tumor DNA analysis in non-small cell lung cancer: analysis of over 8000 cases. Cancer 126, 3219–3228 (2020).

    Article  CAS  PubMed  Google Scholar 

  25. Newman, A. M. et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat. Biotechnol. 34, 547–555 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Razavi, P. et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat. Med. 25, 1928–1937 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Abbosh, C. et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 545, 446–451 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Merker, J. D. et al. Circulating tumor DNA analysis in patients with cancer: American Society of Clinical Oncology and College of American Pathologists joint review. J. Clin. Oncol. 36, 1631–1641 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Alix-Panabières, C. & Pantel, K. Liquid biopsy: from discovery to clinical application. Cancer Discov. 11, 858–873 (2021).

    Article  PubMed  Google Scholar 

  30. Paweletz, C. P. et al. Bias-corrected targeted next-generation sequencing for rapid, multiplexed detection of actionable alterations in cell-free DNA from advanced lung cancer patients. Clin. Cancer Res. 22, 915–922 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Supplee, J. G. et al. Sensitivity of next-generation sequencing assays detecting oncogenic fusions in plasma cell-free DNA. Lung Cancer 134, 96–99 (2019).

    Article  PubMed  Google Scholar 

  32. Chakravarty, D. et al. OncoKB: a precision oncology knowledge base. JCO Precis. Oncol. 2017, 1–16 (2017).

    Google Scholar 

  33. Mandelker, D. et al. Mutation detection in patients with advanced cancer by universal sequencing of cancer-related genes in tumor and normal DNA vs guideline-based germline testing. JAMA 318, 825–835 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Zehir, A. et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 23, 703–713 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Brannon, A. R. et al. Enhanced specificity of high sensitivity somatic variant profiling in cell-free DNA via paired normal sequencing: design, validation, and clinical experience of the MSK-ACCESS liquid biopsy assay. Nat. Comm. 12, 3770 (2021).

    Article  Google Scholar 

  36. Li, B. T. et al. Ultra-deep next-generation sequencing of plasma cell-free DNA in patients with advanced lung cancers: results from the Actionable Genome Consortium. Ann. Oncol. J. Eur. Soc. Med. Oncol. 30, 597–603 (2019).

    Article  CAS  Google Scholar 

  37. Nakamura, Y. et al. Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and GOZILA studies. Nat. Med. 26, 1859–1864 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Pairawan, S. et al. Cell-free circulating tumor DNA variant allele frequency associates with survival in metastatic cancer. Clin. Cancer Res. 26, 1924–1931 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Ramalingam, S. S. et al. Abstract CT078: tumor mutational burden (TMB) as a biomarker for clinical benefit from dual immune checkpoint blockade with nivolumab (nivo) + ipilimumab (ipi) in first-line (1L) non-small cell lung cancer (NSCLC): identification of TMB cutoff from CheckMate 568. Cancer Research 78, CT078 (2018).

  40. Chabon, J. J. et al. Integrating genomic features for non-invasive early lung cancer detection. Nature 580, 245–251 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Morbelli, S. et al. Circulating tumor DNA reflects tumor metabolism rather than tumor burden in chemotherapy-naive patients with advanced non-small cell lung cancer (NSCLC): 18F-FDG PET/CT study. J. Nucl. Med. 58, 1764–1769 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Piotrowska, Z. et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov. 8, 1529–1539 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Offin, M. et al. Acquired ALK and RET gene fusions as mechanisms of resistance to osimertinib in EGFR-mutant lung cancers. JCO Precis. Oncol. 2, PO.18.00126.

  44. Piper-Vallillo, A. J., Sequist, L. V. & Piotrowska, Z. Emerging treatment paradigms for EGFR-mutant lung cancers progressing on osimertinib: a review. J. Clin. Oncol. 38, 2926–2936 (2020).

    Article  CAS  Google Scholar 

  45. Tan, A. C. Targeting the PI3K/Akt/mTOR pathway in non-small cell lung cancer (NSCLC). Thorac. Cancer 11, 511–518 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Nicholas, M. et al. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl. Med. 7, 283ra54 (2015).

    Google Scholar 

  47. Shen, R. et al. Harnessing clinical sequencing data for survival stratification of patients with metastatic lung adenocarcinomas. JCO Precis. Oncol. https://doi.org/10.1200/po.18.00307 (2019).

  48. Bolton, K. L. et al. Cancer therapy shapes the fitness landscape of clonal hematopoiesis. Nat. Genet. 52, 1219–1226 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Coombs, C. C. et al. Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes. Cell Stem Cell 21, 374–382 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bolton, K. L. et al. Clonal hematopoiesis is associated with risk of severe Covid-19. Nat. Commun. 12, 5975 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ignatiadis, M., Sledge, G. W. & Jeffrey, S. S. Liquid biopsy enters the clinic – implementation issues and future challenges. Nat. Rev. Clin. Oncol. 18, 297–312 (2021).

    Article  PubMed  Google Scholar 

  52. Lisberg, A. et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor (TKI) naive patients with advanced NSCLC. J. Thorac. Oncol. 13, 1138–1145 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cristiano, S. et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 570, 385–389 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Parikh, A. R. et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat. Med. 25, 1415–1421 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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 

  56. Mateo, J. et al. A framework to rank genomic alterations as targets for cancer precision medicine: the ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT). Ann. Oncol. J. Eur. Soc. Med. Oncol. 29, 1895–1902 (2018).

    Article  CAS  Google Scholar 

  57. Ramalingam, S. S. et al. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N. Engl. J. Med. 382, 41–50 (2020).

    Article  CAS  PubMed  Google Scholar 

  58. Newman, A. M. et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat. Med. 20, 548–554 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Presley, C. J. et al. Association of broad-based genomic sequencing with survival among patients with advanced non-small cell lung cancer in the community oncology setting. JAMA 320, 469–477 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. 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 

  61. Murthy, V. H., Krumholz, H. M. & Gross, C. P. Participation in cancer clinical trials: race-, sex-, and age-based disparities. JAMA 291, 2720–2726 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Scher, K. S. & Hurria, A. Under-representation of older adults in cancer registration trials: known problem, little progress. J. Clin. Oncol. 30, 2036–2038 (2012).

    Article  PubMed  Google Scholar 

  63. Sedrak, M. S. et al. Older adult participation in cancer clinical trials: a systematic review of barriers and interventions. CA Cancer J. Clin. 71, 78–92 (2021).

    Article  PubMed  Google Scholar 

  64. Rolfo, C., Russo, A. & de Miguel-Pérez, D. Speeding tumor genotyping during the SARS-CoV-2 outbreak through liquid biopsy. Cancer 126, 4089–4091 (2020).

    Article  CAS  PubMed  Google Scholar 

  65. Li, B. T. et al. Reimagining patient-centric cancer clinical trials: a multi-stakeholder international coalition. Nat. Med. 28, 620–626 (2022).

    Article  CAS  PubMed  Google Scholar 

  66. Syeda, M. M. et al. Circulating tumour DNA in patients with advanced melanoma treated with dabrafenib or dabrafenib plus trametinib: a clinical validation study. Lancet Oncol. 22, 370–380 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Turner, N. C. et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): a multicentre, multicohort, phase 2a, platform trial. Lancet Oncol. 21, 1296–1308 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. 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 

  69. Bardelli, A. & Pantel, K. Liquid biopsies, what we do not know (yet). Cancer Cell. 31, 172–179 (2017).

    Article  CAS  PubMed  Google Scholar 

  70. 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 

  71. Lennon, A. M. et al. Feasibility of blood testing combined with PET-CT to screen for cancer and guide intervention. Science 369, 6499 (2020).

    Article  Google Scholar 

  72. Karczewski, K. J. et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581, 434–443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. McNulty, S. N., Parikh, B. A., Duncavage, E. J., Heusel, J. W. & Pfeifer, J. D. Optimization of population frequency cutoffs for filtering common germline polymorphisms from tumor-only next-generation sequencing data. J. Mol. Diagn. 21, 903–912 (2019).

    Article  CAS  PubMed  Google Scholar 

  74. Shen, R. & Seshan, V. E. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 44, 16 (2016).

    Article  Google Scholar 

  75. Kanoun, S. et al. Influence of software tool and methodological aspects of total metabolic tumor volume calculation on baseline [18F]FDG PET to predict survival in Hodgkin lymphoma. PLoS ONE 10, e0140830 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Boellaard, R. et al. FDG PET and PET/CT: EANM procedure guidelines for tumour PET imaging: version 1.0. Eur. J. Nucl. Med. Mol. Imaging 37, 181 (2009).

    Article  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by a grant from the Antidote Health Foundation for Cure of Cancer (BTL), the National Institutes of Health (T32-CA009207 (J.J.), CTSA UL1TR00457 (M.R.M.-G.), P50 CA247749 01 and P30 CA008748), the Molecular Diagnostics Service in the Department of Pathology, and the Marie-Josee and Henry R. Kravis Center for Molecular Oncology.

Author information

Authors and Affiliations

Authors

Contributions

Conception: J.J. and B.T.L. Radiomic analysis: J.J., E.S.L., R.Y. and J.P.D. Patient accrual: J.J., A.N., P.K.P., J.E.C., Y.R.M.-G., B.D., H.A.Y., M.O., M.D.H., K.C.A., M.G.Z., M.G.K., K.K.N., J.E., I.P., W.V.L., J.J.F., A.I., D.M., G.R., B.J.P., D.L.C., C.I.D., M.I., S.C., N.P., A.L., N.R., J.C., W.D.T., G.J.R., V.W.R., A.R., D.G., A.D., D.R.J., C.M.R., J.M.I. and B.T.L. Genomic data collection and analysis: J.J., G.J., A.R.B., R.B., A.Z., M.D., N.S., D.C., R.K., R.M., S.P.S., M.F.B., M.E.A., M.L., R.L., L.P.L. and M.L. Clinical data collection and analysis: J.J., E.S.L., N.S., Y.R.M.-G., H.-Y.T., C.-R.X., C.T.-L. and M.D.S. Administration: A.M., J.G., D.B.S., A.D., H.I.S., P.L., L.P.L., M.F.B., M.E.A., M.L., P.R., J.S.R.-F., D.R.J., C.M.R., J.M.I. and B.T.L. Statistical plan: J.J., M.G., R.S. and S.P.S. Writing: J.J., C.W., P.R., J.S.R.-F. and B.T.L. All co-authors reviewed and approved the final draft of the manuscript.

Corresponding author

Correspondence to Bob T. Li.

Ethics declarations

Competing interests

J.J. has a patent licensed by MDSeq. A.N. reports serving as a one-time paid consultant for Bayer. P.K.P. receives compensation for consulting or advisory board participation from Bicara Therapeutics, Boehringer Ingelheim, EMD Serono, GlaxoSmithKline, Takeda Pharmaceuticals, WC Communications and Xencor and receives honoraria for participation in CME educational programs from PeerVoice, ACE Oncology Research to Practice, Clinical Care Options, Spring to Life and Touch Independent Medical Education. J.E.C. has served as a consultant for Astra Zeneca, Bristol-Myers Squib, Genentech, Merck, Flame Biosciences, Novartis, Regeneron-Sanofi, Guardant Health and Janssen and has received research funding to her institution from Astra Zeneca, Bristol-Myers Squib, Genentech and Merck. A.R.B. has stock ownership in Johnson & Johnson. R.B. reports a grant from Archer, honoraria for advisory board participation from Loxo oncology and speaking fees from Illumina. A.Z. has received speaking fees from Illumina. D.C. has consulted with/received honoraria from Pfizer, Loxo/Lilly Oncology, BridgeBio, FORE Therapeutics, Scorpion Therapeutics and Vividion Therapeutics. Y.R.M.-G. acknowledges receipt of training through an institutional K30 grant from the NIH (CTSA UL1TR00457). She has received funding from a Kristina M. Day Young Investigator Award from Conquer Cancer, the ASCO Foundation, funded by Charles M. Baum and Carol A. Baum. She reports travel, accommodation and expenses from AstraZeneca and honoraria from Virology Education. She acknowledges research funding to the institution from Loxo Oncology at Eli Lilly, Elucida Oncology, Taiho Oncology, Hengrui USA/Jiangsu Hengrui Pharmaceuticals and Endeavor Biomedicines. She acknowledges royalties from Rutgers University Press and Wolters Kluwer. H.-Y.T. has received academic travel support from Resolution Bioscience. C.X. has received honoraria from AstraZeneca, BeiGene, Boehringer Ingelheim, Bristol Myers Squibb, Lilly, Merck & Co., Novartis, Pfizer and Roche; has received research support from BeiGene and Hengrui Pharmaceutical; and has received reimbursement for travel and accommodation expenses from AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Lilly, Merck & Co., Novartis, Pfizer and Roche. B.D. reports equity interest in Eli Lilly and Company, Roche and CVS Health and family member involvement with Eli Lilly and Company and CVS Health. H.A.Y. has consulted for AstraZeneca, Blueprint Medicine, Janssen Oncology, Cullinan and Daiichi. Her institution has received research funding for clinical trials from AstraZeneca, Daiichi, Pfizer, Novartis, Cullinan Oncology and Lilly. M.O. reports personal fees from PharmaMar, personal fees from Novartis, personal fees from Targeted Oncology, personal fees from Bristol-Myers Squibb, personal fees from Merck Sharp & Dohme, personal fees from Jazz Pharmaceuticals and personal fees from Astro Pharmaceuticals, outside the submitted work. M.D.H. reports grants from BMS and personal fees from Achilles, Adagene, Adicet, Arcus, AstraZeneca, Blueprint, BMS, DaVolterra, Eli Lilly, Genentech/Roche, Genzyme/Sanofi, Janssen, Immunai, Instil Bio, Mana Therapeutics, Merck, Mirati, Natera, Pact Pharma, Shattuck Labs and Regeneron, as well as equity options from Factorial, Immunai, Shattuck Labs and Arcus. A patent filed by Memorial Sloan Kettering related to the use of TMB to predict response to immunotherapy (PCT/US2015/062208) is pending and licensed by PGDx. P.L. is listed as an inventor on patent applications filed by MSKCC that describe approaches to treat KRAS or BRAF-mutant tumors. K.C.A. reports personal fees from AstraZeneca and nonfinancial support from Takeda and Novartis outside the submitted work. In the last 3 years, M.G.Z. has received consulting fees from Takeda, GlaxoSmithKline, Expert Connect, Aldeyra Therapeutics, Novocure and Atara and honoraria from Research to Practice, Medical Learning Institute and OncLive. Memorial Sloan Kettering receives research funding from the Department of Defense, the National Institutes of Health, Precog, GlaxoSmithKline, Epizyme, Polaris, Sellas Life Sciences, Bristol Myers Squibb, Millenium/Takeda, Curis and Atara for research conducted by M.G.Z. M.G.Z. serves as chair of the board of directors of the Mesothelioma Applied Research Foundation, uncompensated. M.G.K. receives personal fees from Novartis, Sanofi-Genzyme, AstraZeneca, Pfizer, Janssen and Daiichi-Sankyo; received honoraria for participation in educational programs from WebMD, OncLive, Physicians Education Resources, Prime Oncology, Intellisphere, Creative Educational Concepts, Peerview, i3 Health, Paradigm Medical Communications, AXIS, Carvive Systems and AstraZeneca; received travel support from AstraZeneca, Pfizer and Genentech; and received editorial support from Hoffman La-Roche. Memorial Sloan Kettering has received research funding from the National Cancer Institute (USA), the Lung Cancer Research Foundation and Genentech Roche for research conducted by M.G.K. I.P. has consulted or served on the advisory boards of Pfizer, AstraZeneca, Blueprint Medicine, DavaOncology, Eli Lilly and Curio Science. W.V.L. receives institutional research funding from Daiichi Sankyo, Amgen and Abbvie and has been a compensated consultant for PharmaMar, G1 Therapeutics, AstraZeneca and Jazz Pharmaceuticals. D.M. is a consultant for AstraZeneca, Johnson & Johnson, Boston Scientific, Bristol Myers Squibb and Merck. G.R. receives royalties from Scanlan International. B.J.P. is a consultant for AstraZeneca. L.P.L. is an employee and shareholder of Agilent Technologies. M.L. is an employee and shareholder of Agilent Technologies. C.I.D. reports serving on the advisory board for Amgen and Ipsen, honoraria for Merck and academic travel support from Roche. M.I. reports serving on the advisory boards of Pfizer and Takeda; receiving honoraria from Roche, AstraZeneca, MSD, Bristol Myers Squibb, Pfizer, Takeda and Novartis; and travel support from Roche, AstraZeneca and MSD. S.C. reports advisory board fees from Roche and Astra Zeneca and travel support from Bristol Myers Squibb, outside the submitted work. N.P. has served on advisory boards or received personal honoraria from Boehringer Ingelheim, MSD, Merck, Bristol-Myers Squib, Astra Zeneca, Takeda, Pfizer, Roche, Novartis, Ipsen and Bayer and has received research funding to his institution from Bayer, Pfizer and Roche. A.L. reports personal fees and travel funding from Mundipharma/Helsinn, personal fees from Bayer and personal fees from Eisai. G.J.R. reports grants from National Institutes of Health/National Cancer Institute, has been an uncompensated consultant to Daiichi, Pfizer and Mirati and has institutional research support from Mirati, Takeda, Merck, Roche, Pfizer and Novartis. In addition, G.J.R. has pending patents US20170273982A1 and WO2017164887A8. D.B.S. has served as a consultant for/received honoraria from Loxo Oncology, Lilly Oncology, Pfizer, QED Therapeutics, Vivideon Therapeutics and Illumina. M.L. has received honoraria for advisory board participation from Merck, Astra-Zeneca, Bristol Myers Squibb, Blueprint Medicines, Janssen Pharmaceuticals, Takeda Pharmaceuticals, Lilly Oncology, LOXO Oncology, Bayer, ADC Therapeutics, Riken Genesis and Paige AI and research support from LOXO Oncology, Merus, and Helsinn Therapeutics. Marc Ladanyi reports honoraria for ad-hoc advisory board participation from Merck, AstraZeneca, Bristol Myers Squibb, Takeda, Bayer, and Lilly Oncology; and research support from LOXO Oncology, Merus and Helsinn Therapeutics. V.W.R. reports grants from Genelux, grants from Genentech, other from DaVinci Surgery, nonfinancial support from Bristol Myers Squibb and personal fees from NIH/Coordinating Center for Clinical Trials, outside the submitted work. A.R. reports grants from Varian Medical Systems, grants and personal fees from AstraZeneca, grants and personal fees from Merck, grants and personal fees from Boehringer Ingelheim, grants from Pfizer, personal fees from Research to Practice, personal fees from Cybrexa, personal fees from More Health and nonfinancial support from Philips/Elekta, outside the submitted work. D.G. reports grants from Varian, AstraZeneca, Merck and Bristol Myers Squibb and personal fees from Varia, AstraZeneca, Merck, US Oncology, Bristol Myers Squibb, Relfexion, WebMD, Vindico and Medscape and has served on the advisory board for AstraZeneca. A.D. has received honoraria or worked on the advisory boards of Ignyta/Genentech/Roche, Loxo/Bayer/Lilly, Takeda/Ariad/Millenium, TP Therapeutics, AstraZeneca, Pfizer, Blueprint Medicines, Helsinn, Beigene, BergenBio, Hengrui Therapeutics, Exelixis, Tyra Biosciences, Verastem, MORE Health, Abbvie, 14ner/Elevation Oncology, Remedica, ArcherDX, Monopteros, Novartis, EMD Serono, Melendi, Liberum, Repare RX, Nuvalent, Merus, AXIS, Chugai Pharm and EPG Health; has received funding through his institution from Pfizer, Exelixis, GlaxoSmithKline, Teva, Taiho and PharmaMar; has received research support from Foundation Medicine; receives royalties from Wolters Kluwer; has received other support from Boehringer Ingelheim; has received food/beverage from Merck, Puma and Merus; and has received CME honoraria from Medscape, OncLive, PeerVoice, Physicians Education Resources, Targeted Oncology, Research to Practice, Axis, Peerview Institute, Paradigm Medical Communications, WebMD, MJH Life Sciences, Med Learning, Imedex, Answers in CME and Clinical Care Options. H.I.S. reports the following support: compensated consultant/advisor to Ambry Genetics, Konica Minolta, Bayer, Pfizer, Sun Pharmaceuticals and WCG Oncology; uncompensated consultant/advisory to Amgen, ESSA Pharma, Janssen Research & Development, Janssen Biotech and Sanofi Aventis; he has received research funding (to his institution) from Epic Sciences, Illumina, Janssen, Menarini Silicon Biosystems, Prostate Cancer Foundation and ThermoFisher; intellectual property rights from BioNTech, Elucida Oncology, MaBVAX and Y-mAbs Therapeutics; and nonfinancial support from Amgen, Asterias Biotherapeutics, Bayer, ESSA Pharma, Menarini Silicon Biosystems, Phosplatin, Pfizer, Prostate Cancer Foundation and WCG Oncology. S.P.S. is a shareholder and consultant of Canesia Health. M.F.B. reports a consulting/advisory role with PetDx and Eli Lilly; research support from Grail; and a patent pending related to cfDNA profiling. R.L. is on the supervisory board of Qiagen and is a scientific advisor to Imago, Mission Bio, Syndax. Zentalis, Ajax, Bakx, Auron, Prelude, C4 Therapeutics and Isoplexis for which he receives equity support. He receives research support from Ajax and Abbvie and has consulted for Incyte, Janssen, Morphosys and Novartis. He has received honoraria from Astra Zeneca and Kura for invited lectures and from Gilead for grant reviews. P.R. received institutional grant/funding from Grail, Illumina, Novartis, Epic Sciences and ArcherDx and consultation/ad board/honoraria from Novartis, Foundation Medicine, AstraZeneca, Epic Sciences, Inivata, Natera and Tempus. J.R.-F. is a paid consultant of Goldman Sachs, Paige.AI and REPARE Therapeutics, a member of the scientific advisory board of Goldman Sachs, Paige.AI and Volition RX, and an ad hoc member of the scientific advisory board of Roche, Genentech, Roche Tissue Diagnostics, Ventana, Novartis, InVicro and GRAIL. D.R.J. serves as a consultant for AstraZeneca and Merck. C.M.R. reports personal fees from AbbVie, Amgen, Ascentage, AstraZeneca, Bicycle, Celgene, Daiichi Sankyo, Genentech/Roche, Ipsen, Jansen, Jazz, Lilly/Loxo, Pfizer, PharmaMar, Syros, Vavotek, Bridge Medicines and Harpoon Therapeutics, outside the submitted work. J M.I. reports equity in LumaCyte and has served as an uncompensated member of a steering committee for Genentech. B.T.L. has served as an uncompensated advisor and consultant to Amgen, Genentech, Boehringer Ingelheim, Lilly, AstraZeneca and Daiichi Sankyo. He has received research grants to his institution from Amgen, Genentech, AstraZeneca, Daiichi Sankyo, Lilly, Illumina, GRAIL, Guardant Health, Hengrui Therapeutics, MORE Health and Bolt Biotherapeutics. He has received academic travel support from MORE Health and Jiangsu Hengrui Medicine. He is an inventor on two institutional patents at Memorial Sloan Kettering (US62/685,057, US62/514,661) and has intellectual property rights as a book author at Karger Publishers and Shanghai Jiao Tong University Press. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks Ana Vivancos, Benjamin Besse and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Joao Monteiro, in collaboration with the Nature Medicine team.

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 Turnaround time of plasma and tissue sequencing.

Boxplots showing median (center) +/−25%ile (boxes) and 95%ile (whiskers) of turnaround time for liquid biopsy (MSK-ACCESS and ctDx Lung, N independent samples = 2,162) and tissue (MSK-IMPACT, N independent samples = 612) sequencing from date of blood collection. Tissue start time is the date of white blood cell control collection. The turnaround time for plasma ctDNA sequencing was significantly faster than for tissue sequencing (*two-sided Mann-Whitney U, p < 0.001).

Extended Data Fig. 2 Correlates of ctDNA alteration levels.

Histograms showing the proportion of patients with either number of ctDNA alterations or maximum mutation variant allele frequency (VAF) per sample. P-values are from Mann-Whitney U tests or Kruskal-Wallis tests for histologic subgroups. Raw VAFs of zero are set to the minimum value of the log axis.

Extended Data Fig. 3 Survival of patients without tissue sequencing matched to targeted therapy by ctDNA.

Kaplan-Meier survival curves for patients without tissue sequencing matched or not matched to targeted therapy. Number at risk in each category is adjusted for left truncation and time-dependent nature of targeted therapy variables.

Supplementary information

Supplementary Information

Supplemental text and Supplementary Figs. S1–14.

Reporting Summary

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jee, J., Lebow, E.S., Yeh, R. et al. Overall survival with circulating tumor DNA-guided therapy in advanced non-small-cell lung cancer. Nat Med 28, 2353–2363 (2022). https://doi.org/10.1038/s41591-022-02047-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-022-02047-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing