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:

Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab

Abstract

Although programmed death-ligand 1–programmed death 1 (PD-L1–PD-1) inhibitors are broadly efficacious, improved outcomes have been observed in patients with high PD-L1 expression or high tumor mutational burden (TMB). PD-L1 testing is required for checkpoint inhibitor monotherapy in front-line non-small-cell lung cancer (NSCLC). However, obtaining adequate tumor tissue for molecular testing in patients with advanced disease can be challenging. Thus, an unmet medical need exists for diagnostic approaches that do not require tissue to identify patients who may benefit from immunotherapy. Here, we describe a novel, technically robust, blood-based assay to measure TMB in plasma (bTMB) that is distinct from tissue-based approaches. Using a retrospective analysis of two large randomized trials as test and validation studies, we show that bTMB reproducibly identifies patients who derive clinically significant improvements in progression-free survival from atezolizumab (an anti-PD-L1) in second-line and higher NSCLC. Collectively, our data show that high bTMB is a clinically actionable biomarker for atezolizumab in NSCLC.

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: Development of a bTMB assay and correlation with tTMB.
Fig. 2: Forest plots of HRs for PFS and OS in the POPLAR study.
Fig. 3: Association of clinical outcome and bTMB in the OAK study.
Fig. 4: Forest plots showing the association between the clinical benefit of atezolizumab and bTMB in the OAK study.
Fig. 5: Comparison of PD-L1 expression and bTMB counts in the OAK study.

Similar content being viewed by others

References

  1. Rittmeyer, A. et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet 389, 255–265 (2017).

    Article  Google Scholar 

  2. Borghaei, H. et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015).

    Article  CAS  Google Scholar 

  3. Brahmer, J. et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N. Engl. J. Med. 373, 123–135 (2015).

    Article  CAS  Google Scholar 

  4. Reck, M. et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N. Engl. J. Med. 375, 1823–1833 (2016).

    Article  CAS  Google Scholar 

  5. Fehrenbacher, L. et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet 387, 1837–1846 (2016).

    Article  CAS  Google Scholar 

  6. Carbone, D. P. et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N. Engl. J. Med. 376, 2415–2426 (2017).

    Article  CAS  Google Scholar 

  7. Garon, E. B. Cancer immunotherapy trials not immune from imprecise selection of patients. N. Engl. J. Med. 376, 2483–2485 (2017).

    Article  Google Scholar 

  8. Hui, R. et al. Pembrolizumab as first-line therapy for patients with PD-L1-positive advanced non-small cell lung cancer: a phase 1 trial. Ann. Oncol. 28, 874–881 (2017).

    Article  CAS  Google Scholar 

  9. Tischer, B., Kim, E., Peters, M. & Hirsh, V. Physician patterns of care in patients with EGFR mutation+ NSCLC: an international survey into testing and treatment choice. J. Thorac. Oncol. 12, S1199–S1200 (2017).

    Article  Google Scholar 

  10. Lim, C. et al. Biomarker testing and time to treatment decision in patients with advanced nonsmall-cell lung cancer. Ann. Oncol. 26, 1415–1421 (2015).

    Article  CAS  Google Scholar 

  11. Green, M. R. et al. Molecular testing prior to first-line therapy in patients with stage IV nonsquamous non-small cell lung cancer (NSCLC): a survey of U.S. medical oncologists. J. Clin. Oncol. 26, abstr. 8097 (2014).

  12. 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 

  13. Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160, 48–61 (2015).

    Article  CAS  Google Scholar 

  14. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    Article  CAS  Google Scholar 

  15. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    Article  CAS  Google Scholar 

  16. Kowanetz, M. et al. Tumor mutation burden (TMB) is associated with improved efficacy of atezolizumab in 1L and 2L+ NSCLC patients. J. Thorac. Oncol. 12, S321–S322 (2017).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  18. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).

    Article  CAS  Google Scholar 

  19. Johnson, D. B. et al. Targeted next generation sequencing identifies markers of response to PD-1 blockade. Cancer Immunol. Res. 4, 959–967 (2016).

    Article  CAS  Google Scholar 

  20. Chalmers, Z. R. et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 9, 34 (2017).

    Article  Google Scholar 

  21. Campesato, L. F. et al. Comprehensive cancer-gene panels can be used to estimate mutational load and predict clinical benefit to PD-1 blockade in clinical practice. Oncotarget 6, 34221–34227 (2015).

    Article  Google Scholar 

  22. Rosenberg, J. E. et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 387, 1909–1920 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  25. Wan, J. C. et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat. Rev. Cancer 17, 223–238 (2017).

    Article  CAS  Google Scholar 

  26. Siravegna, G., Marsoni, S., Siena, S. & Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 14, 531–548 (2017).

    Article  CAS  Google Scholar 

  27. 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  Google Scholar 

  28. Frampton, G. M. et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat. Biotechnol. 31, 1023–1031 (2013).

    Article  CAS  Google Scholar 

  29. Fabrizio, D. et al. Analytic validation of a next generation sequencing assay to identify tumor mutational burden from blood (bTMB) to support investigation of an anti-PD-L1 agent, atezolizumab, in a first line non-small cell lung cancer trial (BFAST). Ann. Oncol. 28, mdx363.018 (2017).

    Article  Google Scholar 

  30. He, J. et al. Integrated genomic DNA/RNA profiling of hematologic malignancies in the clinical setting. Blood 127, 3004–3014 (2016).

    Article  CAS  Google Scholar 

  31. Friedman, J. H. & Roosen, C. B. An introduction to multivariate adaptive regression splines. Stat. Methods Med. Res. 4, 197–217 (1995).

    Article  CAS  Google Scholar 

  32. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    Article  CAS  Google Scholar 

  33. Chen, L., Liu, P., Evans, T. C. Jr & Ettwiller, L. M. DNA damage is a pervasive cause of sequencing errors, directly confounding variant identification. Science 355, 752–756 (2017).

    Article  CAS  Google Scholar 

  34. Sun, J. X. et al. A computational method for somatic versus germline variant status determination from targeted next-generation sequencing of clinical cancer specimens without a matched normal control. Cancer Res. 74, 1893 (2014).

    Google Scholar 

  35. Sun, J. X. et al. A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal. PLoS Comput. Biol. 14, e1005965 (2018).

    Article  Google Scholar 

  36. 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  Google Scholar 

  37. 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  Google Scholar 

  38. Mok, T. et al. Detection and dynamic changes of EGFR mutations from circulating tumor DNA as a predictor of survival outcomes in NSCLC patients treated with first-line intercalated erlotinib and chemotherapy. Clin. Cancer Res. 21, 3196–3203 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the patients, their families and the clinical study-site investigators and staff for their contributions to the study. In addition, we acknowledge E. White, M. Coyne, T. Brennan, M. Kennedy, J. He, S. Zhong, G. Young, J. Ma, M. Zhao and G. Frampton for their contributions. This study was funded by F. Hoffmann-La Roche. Editorial support for this manuscript was provided by M. Balbas of Health Interactions and funded by F. Hoffmann-La Roche.

Author information

Authors and Affiliations

Authors

Contributions

D.R.G. interpreted the experiments and wrote the paper. S.M.P. designed and interpreted the experiments and wrote the paper. M.K. interpreted the experiments and wrote the paper. E.S. designed and interpreted the experiments and wrote the paper. W.Z. analyzed and interpreted the experiments and wrote the paper. Y.L. analyzed and interpreted the experiments and wrote the paper. A.R. designed and interpreted the experiments and wrote the paper. L.F. designed and interpreted the experiments and wrote the paper. G.O. designed, performed, analyzed and interpreted the experiments. C.M. analyzed the experiments. D.S.L. and D.L. performed and analyzed the experiments. J.S. performed and analyzed the experiments. L.A. interpreted the experiments and wrote the paper. T.R. interpreted the experiments and wrote the paper. C.A.C. designed, analyzed and interpreted the experiments and wrote the paper. P.S.H. interpreted the experiments and wrote the paper. A.S. designed and interpreted the experiments. M.B. designed and interpreted the experiments and wrote the paper. D.F. designed, performed, analyzed and interpreted the experiments and wrote the paper. T.M. interpreted the experiments and wrote the paper. D.S.S. designed, analyzed and interpreted the experiments and wrote the paper.

Corresponding authors

Correspondence to David R. Gandara, Tony Mok or David S. Shames.

Ethics declarations

Competing interests

The authors declare the following competing interests: D.R.G. has received a clinical trial grant and consultant fees from Genentech. Y.L., L.A., T.R., A.S. and M.B. are Genentech employees and holders of Roche stock. M.K., E.S., D.S.S., S.M.P., Y.L. and C.A.C. are Genentech employees and holders of Roche stock and have a patent pending for therapeutic and diagnostic methods for cancer. W.Z. is a Genentech employee and has received research funding from Roche. A.R. has received grants from AstraZeneca, Roche/Genentech, Bristol-Myers Squibb, Boehringer Ingelheim, Eli Lilly and Pfizer. L.F. declares no competing interests. D.F. is a Foundation Medicine employee and stockholder and has a patent pending for therapeutic and diagnostic methods for cancer. G.O., C.M., D.S.L., D.L. and J.S. are Foundation Medicine employees and stockholders. P.S.H. is a Genentech employee. T.M. has received grants/research support from AstraZeneca, Boehringer Ingelheim, Pfizer, Novartis, SFJ Pharmaceuticals, Roche, Merck Sharp & Dohme, Clovis Oncology, Bristol-Myers Squibb, Eisai and Taiho; speaker’s fees from AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Taiho and Ariad/Takeda; honoraria/honorarium from AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, Boehringer Ingelheim, Merck Serono, Merck Sharp & Dohme, Novartis, SFJ Pharmaceuticals, ACEA Biosciences, Vertex Pharmaceuticals, Bristol-Myers Squibb, OncoGenex Pharmaceuticals, Celgene and Ignyta; is a major stockholder in Sanomics and Cirina; participated in advisory boards for AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, Boehringer Ingelheim, Clovis Oncology, Merck Serono, Merck Sharp & Dohme, Novartis, SFJ Pharmaceuticals, ACEA Biosciences, Vertex Pharmaceuticals, Bristol-Myers Squibb, geneDecode, OncoGenex Technologies, Celgene, Ignyta and Cirina; and is on the board of directors for the International Association for the Study of Lung Cancer (IASLC), the Chinese Lung Cancer Research Foundation, the Chinese Society of Clinical Oncology (CSCO) and the Hong Kong Cancer Therapy Society (HKCTS).

Additional information

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

Supplementary Information

Supplementary Text and Figures

Supplementary Figures 1–11 and Supplementary Tables 1–7 and 9

Reporting Summary

Supplementary Table 8

Patient-level clinical variables, clinical outcome data and variant calls for bTMB in OAK and POPLAR

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gandara, D.R., Paul, S.M., Kowanetz, M. 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). https://doi.org/10.1038/s41591-018-0134-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0134-3

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