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:

Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab

Abstract

Immune checkpoint blockade (ICB) provides clinical benefit to a subset of patients with cancer. However, existing biomarkers do not reliably predict treatment response across diverse cancer types. Limited data exist to show how serial circulating tumor DNA (ctDNA) testing may perform as a predictive biomarker in patients receiving ICB. We conducted a prospective phase II clinical trial to assess ctDNA in five distinct cohorts of patients with advanced solid tumors treated with pembrolizumab (NCT02644369). We applied bespoke ctDNA assays to 316 serial plasma samples obtained at baseline and every three cycles from 94 patients. Baseline ctDNA concentration correlated with progression-free survival, overall survival, clinical response and clinical benefit. This association became stronger when considering ctDNA kinetics during treatment. All 12 patients with ctDNA clearance during treatment were alive with median 25 months follow up. This study demonstrates the potential for broad clinical utility of ctDNA-based surveillance in patients treated with ICB.

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: Baseline ctDNA level is correlated with OS and PFS.
Fig. 2: The change in ctDNA levels from baseline to C3, clinical response and survival.
Fig. 3: The change in ctDNA levels from baseline to C3 is strongly correlated with OS and PFS.
Fig. 4: Risk groupings according to early clinical response and ctDNA kinetics.
Fig. 5: Objective clinical response to pembrolizumab is associated with ctDNA clearance.
Fig. 6: ctDNA clearance identifies patients with highly favorable survival.

Similar content being viewed by others

Data availability

Anonymized WES data of tumor and match normals are available with controlled access approval through the European Genome-Phenome Archive under accession number EGAS00001003280. Source data for Figs. 16 can be found in Source data Figs. 16 files, and Source data for Extended Data Fig. 28 can be found in Source data Extended Data Fig. 28 files. The supporting variant level data for all figures are available in Supplementary Table 4. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

Code necessary to reproduce the core analyses are available without restrictions at https://github.com/pughlab/inspire-ctdna.

References

  1. Bedognetti, D. et al. Toward a comprehensive view of cancer immune responsiveness: a synopsis from the SITC workshop. J. Immunother. Cancer 7, 131 (2019).

    Article  Google Scholar 

  2. Hause, R. J., Pritchard, C. C., Shendure, J. & Salipante, S. J. Classification and characterization of microsatellite instability across 18 cancer types. Nat. Med. 22, 1342–1350 (2016).

    Article  CAS  Google Scholar 

  3. Jenkins, R. W., Thummalapalli, R., Carter, J., Canadas, I. & Barbie, D. A. Molecular and genomic determinants of response to immune checkpoint inhibition in cancer. Annu. Rev. Med. 69, 333–347 (2018).

    Article  CAS  Google Scholar 

  4. Rizvi, N. A. et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).

    Article  CAS  Google Scholar 

  5. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).

    Article  Google Scholar 

  6. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 515, 568–571 (2014).

    Article  CAS  Google Scholar 

  7. Gibney, G. T., Weiner, L. M. & Atkins, M. B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 17, e542–e551 (2016).

    Article  CAS  Google Scholar 

  8. Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).

    Article  CAS  Google Scholar 

  9. Wolchok, J. D. et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Cancer Res. 15, 7412–7420 (2009).

    Article  CAS  Google Scholar 

  10. Ribas, A., Chmielowski, B. & Glaspy, J. A. Do we need a different set of response assessment criteria for tumor immunotherapy? Clin. Cancer Res. 15, 7116–7118 (2009).

    Article  CAS  Google Scholar 

  11. Corcoran, R. B. & Chabner, B. A. Application of cell-free DNA analysis to cancer treatment. N. Engl. J. Med. 379, 1754–1765 (2018).

    Article  CAS  Google Scholar 

  12. Cabel, L. et al. Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy. Nat. Rev. Clin. Oncol. 15, 639–650 (2018).

    Article  CAS  Google Scholar 

  13. Anagnostou, V. et al. Dynamics of tumor and immune responses during immune checkpoint blockade in non-small cell lung cancer. Cancer Res. 79, 1214–1225 (2019).

    Article  CAS  Google Scholar 

  14. Cabel, L. et al. Circulating tumor DNA changes for early monitoring of anti-PD1 immunotherapy: a proof-of-concept study. Ann. Oncol. 28, 1996–2001 (2017).

    Article  CAS  Google Scholar 

  15. Giroux Leprieur, E. et al. Circulating tumor DNA evaluated by Next-Generation Sequencing is predictive of tumor response and prolonged clinical benefit with nivolumab in advanced non-small cell lung cancer. Oncoimmunology. 7, e1424675 (2018).

    Article  Google Scholar 

  16. 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  Google Scholar 

  17. Gray, E. S. et al. Circulating tumor DNA to monitor treatment response and detect acquired resistance in patients with metastatic melanoma. Oncotarget 6, 42008–42018 (2015).

    Article  Google Scholar 

  18. Lee, J. H. et al. Circulating tumour DNA predicts response to anti-PD1 antibodies in metastatic melanoma. Ann. Oncol. 28, 1130–1136 (2017).

    Article  CAS  Google Scholar 

  19. Lee, J. H. et al. Association between circulating tumor DNA and pseudoprogression in patients with metastatic melanoma treated with anti-programmed cell death 1 antibodies. JAMA Oncol. 4, 717–721 (2018).

    Article  Google Scholar 

  20. Raja, R. et al. Early reduction in ctDNA predicts survival in patients with lung and bladder cancer treated with durvalumab. Clin. Cancer Res. 24, 6212–6222 (2018).

    Article  CAS  Google Scholar 

  21. Moding, E. J. et al. Circulating tumor DNA dynamics predict benefit from consolidation immunotherapy in locally advanced non-small-cell lung cancer. Nat. Cancer 1, 176–183 (2020).

    Article  Google Scholar 

  22. Clouthier, D. L. et al. An interim report on the investigator-initiated phase 2 study of pembrolizumab immunological response evaluation (INSPIRE). J. Immunother. Cancer 7, 72 (2019).

    Article  Google Scholar 

  23. Coombes, R. C. et al. Personalized detection of circulating tumor DNA antedates breast cancer metastatic recurrence. Clin. Cancer Res. 25, 4255–4263 (2019).

    Article  CAS  Google Scholar 

  24. Magbanua, M. J. M. et al. Circulating tumor DNA in neoadjuvant treated breast cancer reflects response and survival. Preprint at medRxiv https://doi.org/10.1101/2020.02.03.20019760 (2020).

  25. Christensen, E. et al. Early detection of metastatic relapse and monitoring of therapeutic efficacy by ultra-deep sequencing of plasma cell-free DNA in patients with urothelial bladder carcinoma. J. Clin. Oncol. 37, 1547–1557 (2019).

    Article  CAS  Google Scholar 

  26. Reinert, T. et al. Analysis of of plasma cell-free DNA by ultradeep sequencing in patients with stages I to III colorectal cancer. JAMA Oncol. 5, 1124–1131 (2019).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  28. Koboldt, D. C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    Article  CAS  Google Scholar 

  29. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  31. Lai, Z. et al. VarDict: a novel and versatile variant caller for next-generation sequencing in cancer research. Nucleic Acids Res. 44, e108 (2016).

    Article  Google Scholar 

  32. Salipante, S. J., Scroggins, S. M., Hampel, H. L., Turner, E. H. & Pritchard, C. C. Microsatellite instability detection by next generation sequencing. Clin. Chem. 60, 1192–1199 (2014).

    Article  CAS  Google Scholar 

  33. McGranahan, N. et al. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl. Med. 7, 283ra254 (2015).

    Article  Google Scholar 

Download references

Acknowledgements

Major funding support for the project was made possible by the Princess Margaret Cancer Foundation, Ontario Institute for Cancer Research and Terry Fox Research Institute. We thank Merck for contributing the study drug for the clinical trial. S.V.B. and T.J.P. were supported by the Gattuso-Slaight Personalized Cancer Medicine Fund. S.Y.C.Y. was supported in part by the University of Toronto Medical Biophysics OSOTF Excellence Award. M.A.J.I. was supported in part by a fellowship through the BMO Chair in Precision Cancer Genomics. L.L.S. holds the BMO Chair in Precision Cancer Genomics. T.J.P. holds the Canada Research Chair in Translational Genomics and is supported by a Senior Investigator Award from the Ontario Institute for Cancer Research. We are grateful to R. Salari, H.-T. Wu and S. Dashner for their technical support and insightful discussions of the project. We gratefully acknowledge the individuals from the Princess Margaret Tumour Immunotherapy Program (https://www.pm-tumorimmunotherapyprogram.ca/), including the immune monitoring (M. Butler, D. Gray, V. Sotov and D. Liu) and tissue correlatives (V. Speers, A. Giesler and S. Hakgor) teams. We thank the staff of the Princess Margaret Genomics Centre (https://www.pmgenomics.ca), Bioinformatics and High-Performance Computing Core, and PM-OICR Translational Genomics Laboratory (https://labs.oicr.on.ca/translational-genomics-laboratory) for their expertise in generating the sequencing data used in this study. Additional infrastructure support from the Canada Foundation for Innovation, Leaders Opportunity Fund (32383); Ontario Ministry of Research and Innovation, Ontario Research Fund Small Infrastructure Program and Ontario Institute for Cancer Research (https://genomics.oicr.on.ca).

Author information

Authors and Affiliations

Authors

Contributions

S.V.B., L.L.S. and T.J.P. conceived the study. S.V.B., P.S.O., L.L.S. and T.J.P. secured funding. S.Y.C.Y. performed data analysis. M.A.J.I. reviewed the clinical data and performed data analysis. A.R.H., P.L.B., S.L., A.S., A.A.R. and L.L.S. accrued patients and supervised biospecimen sample collection. T.J.P., P.S.O., D.L.C. and D.T. supervised biospecimen sample processing by S.Y.C.Y., S.C.L., Y.H., I.C., K.M. and J.E. W.X. and Z.L. performed statistical analysis. S.S., M.L., B.Z., P.B., H.S. and A.A. conducted the ctDNA assays and provided technical expertise and scientific feedback. S.V.B., S.Y.C.Y., L.L.S., W.X., Z.L., D.T. and T.J.P. wrote the manuscript. All authors discussed the results and implications and commented on the manuscript at all stages.

Corresponding authors

Correspondence to Lillian L. Siu or Trevor J. Pugh.

Ethics declarations

Competing interests

S.V.B. provides consultation for Bristol-Myers Squibb, receives research support from Nektar Therapeutics and is a coinventor on a patent application relating to circulating tumor DNA mutation detection technology (PCT/US2014/02502, licensed to Roche Molecular Diagnostics) and on a patent application relating to ctDNA methylation analysis technology (PCT/CA2018/000203). L.L.S. provides consultation for Merck (compensated), Pfizer (compensated), Celgene (compensated), AstraZeneca (compensated), Morphosys (compensated), Roche (compensated), GeneSeeq (compensated), Loxo (compensated), Oncorus (compensated), Symphogen (compensated), Seattle Genetics (compensated), GlaxoSmithKline (compensated), Voronoi (compensated), Treadwell Therapeutics (compensated), Arvinas (compensated), Tessa (compensated), Navire (compensated); receives research support (institutional) from Novartis, Bristol-Myers Squibb, Pfizer, Boerhinger-Ingelheim, GlaxoSmithKline, Roche/Genentech, Karyopharm, AstraZeneca/Medimmune, Merck, Celgene, Astellas, Bayer, Abbvie, Amgen, Symphogen, Intensity Therapeutics, Mirati, Shattucks and Avid; and is a stockholder in Agios (spouse) and Treadwell Therapeutics (spouse). S.S., M.L., P.B., B.Z., H.S. and A.A. are employees of Natera, Inc. T.J.P. provides consultation for Merck, Chrysalis Biomedical Advisors and the Canadian Pension Plan Investment Board, and receives research support from Roche.

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 CONSORT diagram for the study.

The numbers of patients included in the study and reasons for exclusion are shown.

Extended Data Fig. 2 Factors affecting somatic mutation detection in tumor tissue and plasma cell-free DNA.

a, Somatic mutation burden per patient are grouped by INSPIRE cancer cohorts: Squamous cell cancer of head and neck (SCCHN, cohort A), triple negative breast cancer (TNBC, cohort B), high grade serous ovarian cancer (HGSOC, cohort C), malignant melanoma (MM, cohort D), and mixed solid tumors (MST, cohort E). The N (patients) for each group is shown in the top axis. Two-sided Kruskal-Wallis test p-value is shown. b, c, Comparison of 16 mutations vs 1 mutation from sequencing data for detecting ctDNA. To evaluate the impact of measuring 16 mutations on the likelihood of detecting ctDNA, we randomly selected one of the 16 mutations from each of the 94 baseline cell-free DNA samples and repeated this with 10,000 iterations. (b) An example of one of the 10,000 iteration shows that ctDNA levels (MTM/mL) are correlated (Spearman R = 0.87) among N = 94 baseline cell-free DNA samples. Two-sided Spearman correlation test p-value is shown. (c) Histogram of the detection likelihood (sensitivity for ctDNA) from 94 baseline cell-free DNA samples when using a randomly selected single mutation (N = 10,000 iterations). The mean sensitivity (red dashed line) is 84%. The sensitivity using all 16 mutations (black dashed line) is 98%. d, Influence of tumor tissue source used for bespoke assay design on detectability of ctDNA. The number of detected variants in baseline cell-free DNA samples are shown according to whether the bespoke assay design was conducted from FFPE vs fresh-frozen tumor exomes. Total number of samples in each group shown on the top axis. Two-sided Wilcoxon rank sum test p-value is shown. Box plots represent the median, upper and lower quartiles of each distribution with whiskers showing the limits of the distribution (1.5 times the interquartile range).

Source data

Extended Data Fig. 3 Rationale for calculation of absolute ctDNA levels from multiple mutation targets.

Analytical validation of the ctDNA quantification approach was conducted using Seraseq ctDNA Mutation Mix v2 (SeraCare, Milford, MA, USA) at various concentrations. For a given sample containing 16 target mutations, the mean tumor molecules per mL of plasma (MTM/mL) was calculated using two approaches (y-axis) and compared to known concentrations (x-axis). The blue data points represent the per-sample measurements of mean MTM/mL calculated over detected targets (slope=0.86), while the green data points represent the per-sample measurements of mean MTM/mL calculated over all 16 tested targets (slope=0.98). The red (X=Y) dashed line illustrates the ideal (expected) scenario in which the calculated experimental ctDNA levels are equal to the expected MTM/mL. The numbers of technical replicates at each concentration are shown (n = 2-18). In addition, all 10 negative control samples had no detected mutations (not shown). Pearson correlation coefficients (R2) are shown. This analytical comparison indicates that mean MTM/mL calculated over all tested targets provides a more accurate representation of the true amount of ctDNA, particularly at low levels of ctDNA.

Source data

Extended Data Fig. 4 ctDNA levels in the INSPIRE sub-cohorts and correlation with RECIST tumor measurements.

ctDNA levels (MTM/mL) at baseline a, and at cycle 3 b, as well as the fold-change in ctDNA levels from baseline to cycle 3 (∆ctDNAC3) c, are shown for each of the sub-cohorts: Squamous cell cancer of head and neck (SCCHN, cohort A), triple negative breast cancer (TNBC, cohort B), high grade serous ovarian cancer (HGSOC, cohort C), malignant melanoma (MM, cohort D), and mixed solid tumors (MST, cohort E). Box plots represent the median, upper and lower quartiles of each distribution with whiskers showing the limits of the distribution (1.5 times the interquartile range). The N (patients) for each group is shown in the top axis. Two-sided Kruskal-Wallis test p-values are shown for (ac). d, Relationship between ctDNAB and RECIST 1.1 target lesion sum of tumor measurements (N = 94 patients). Scatter plot with Spearman correlation coefficient (R) and two-sided p-value are shown. The poor correlation in this diverse cohort of advanced cancers suggests that ctDNA may provide complementary information to RECIST tumor measurements. Of note, RECIST target lesion sum of tumor measurements does not incorporate measurements from non-target lesions.

Source data

Extended Data Fig. 5 ∆ctDNAC3 for the INSPIRE sub-cohorts.

Clinical response is indicated by line color. The inset table summarizes ∆ctDNAC3 values among patients with distinct clinical responses in each sub-cohort. Closed circles indicate ctDNA positive samples and open circles indicate ctDNA negative samples; SD, stable disease; CR, complete response; PR, partial response; PD, progressive disease; B, baseline; C3, cycle 3; ND, not detected (below detection threshold).

Source data

Extended Data Fig. 6 Relationship between ctDNA, biomarkers, and prognosis.

a, Relationship between ctDNA metrics (ctDNAB or ∆ctDNAC3) and other candidate biomarkers (PD-L1 or TMB). Scatter plots with Spearman correlation coefficients (R) and p-values are shown. Top row: N = 94 patients with both ctDNAB and TMB levels; N = 93 patients with both ctDNAB and PD-L1 levels. Bottom row: N = 72 patients with ∆ctDNAC3, TMB, and PD-L1 levels. b, Performance of multivariable Cox models incorporating ctDNA metrics (ctDNAB and/or ∆ctDNAC3). Multivariable Cox models for OS (left) or PFS (right) were generated. The baseline model incorporates cohort, PD-L1, and TMB. C-statistic is plotted for each model. Statistically significant differences (two-sided likelihood ratio test) are indicated.

Source data

Extended Data Fig. 7 Definitions of ctDNA dynamic profiles.

Serial ctDNA measurements in patients grouped according ctDNA dynamics (clearance at any time point, no clearance with all on-treatment values below baseline, no clearance with any on-treatment value above baseline). Triangles indicate values off the displayed range of the y-axis.

Source data

Extended Data Fig. 8 Landmark analyses of ctDNA clearance.

Kaplan-Meier curves of OS among patients classified as one of 3 groups (Clearance; Decrease from baseline; Increase from baseline) based on ctDNA at each indicated landmark (that is, C3, C6, C9, or C12).

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–8.

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. 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.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bratman, S.V., Yang, S.Y.C., Iafolla, M.A.J. et al. Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nat Cancer 1, 873–881 (2020). https://doi.org/10.1038/s43018-020-0096-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-020-0096-5

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