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:

Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma

A Publisher Correction to this article was published on 25 October 2018

An Author Correction to this article was published on 25 October 2018

This article has been updated

Abstract

Preclinical studies suggest that treatment with neoadjuvant immune checkpoint blockade is associated with enhanced survival and antigen-specific T cell responses compared with adjuvant treatment1; however, optimal regimens have not been defined. Here we report results from a randomized phase 2 study of neoadjuvant nivolumab versus combined ipilimumab with nivolumab in 23 patients with high-risk resectable melanoma (NCT02519322). RECIST overall response rates (ORR), pathologic complete response rates (pCR), treatment-related adverse events (trAEs) and immune correlates of response were assessed. Treatment with combined ipilimumab and nivolumab yielded high response rates (RECIST ORR 73%, pCR 45%) but substantial toxicity (73% grade 3 trAEs), whereas treatment with nivolumab monotherapy yielded modest responses (ORR 25%, pCR 25%) and low toxicity (8% grade 3 trAEs). Immune correlates of response were identified, demonstrating higher lymphoid infiltrates in responders to both therapies and a more clonal and diverse T cell infiltrate in responders to nivolumab monotherapy. These results describe the feasibility of neoadjuvant immune checkpoint blockade in melanoma and emphasize the need for additional studies to optimize treatment regimens and to validate putative biomarkers.

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: Trial schema.
Fig. 2: Radiographic and pathologic responses to neoadjuvant nivolumab and the combination of ipilimumab and nivolumab.
Fig. 3: Kaplan–Meier estimates of survival.
Fig. 4: Immune correlates of response to ICB.

Similar content being viewed by others

Data availability

The authors declare that the data supporting the findings of this study are available within the paper and its Supplementary Information. Whole-exome sequencing, T cell receptor sequencing and Nanostring digital spatial profiling data are available from the European Genome-phenome Archive under accession number EGAS00001003178.

Change history

  • 25 October 2018

    In the version of this article originally published, there was an error in Fig. 1. In the neoadjuvant phase column, the n values for arms A and B were both reported to be 20. The n values for arms A and B were actually 12 and 11, respectively. Also, the URL underlying the accession code in the data availability section was incorrect. The URL was originally https://www.ebi.ac.uk/ega/studies/EGAS00001002698. It should have been https://www.ebi.ac.uk/ega/studies/EGAS00001003178. The errors have been corrected in the print, HTML and PDF versions of this article.

  • 25 October 2018

    In the version of this article originally published, there was an error in Fig. 2b. RECIST ORR and pCR were both listed as 25%. RECIST ORR was actually 73%, and pCR was 45%. Also, an author’s name was incorrect in the author list. Danny K. Wells should have been listed as Daniel K. Wells. The errors have been corrected in the print, HTML and PDF versions of this article.

References

  1. Liu, J. et al. Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discov. 6, 1382–1399 (2016).

    Article  CAS  Google Scholar 

  2. Gershenwald, J. E. et al. Melanoma staging: evidence-based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J. Clin. 67, 472–492 (2017).

    Article  Google Scholar 

  3. Eggermont, A. M. M., Suciu, S. & Testori, A. Ipilimumab adjuvant therapy in melanoma. N. Engl. J. Med. 376, 398––399 (2017).

    Article  Google Scholar 

  4. Weber, J. et al. Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N. Engl. J. Med. 377, 1824–1835 (2017).

    Article  CAS  Google Scholar 

  5. Eggermont, A. M. M. et al. Adjuvant pembrolizumab versus placebo in resected stage III melanoma. N. Engl. J. Med. 378, 1789–1801 (2018).

    Article  CAS  Google Scholar 

  6. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    Article  Google Scholar 

  7. Wolchok, J. D. et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 377, 1345–1356 (2017).

    Article  CAS  Google Scholar 

  8. Amaria, R. N. et al. Neoadjuvant plus adjuvant dabrafenib and trametinib versus standard of care in patients with high-risk, surgically resectable melanoma: a single-centre, open-label, randomised, phase 2 trial. Lancet Oncol. 19, 181–193 (2018).

    Article  CAS  Google Scholar 

  9. Forde, P. M. et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N. Engl. J. Med. 378, 1976–1986 (2018).

    Article  CAS  Google Scholar 

  10. Edge, S. B. AJCC Cancer Staging Manual 7th edn (Springer, New York, 2010).

  11. Chen, P. L. et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 6, 827–837 (2016).

    Article  Google Scholar 

  12. Roh, W. et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. 9, eaah3560 (2017).

    Article  Google Scholar 

  13. Eggermont, A. M. et al. Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy. N. Engl. J. Med. 375, 1845–1855 (2016).

    Article  CAS  Google Scholar 

  14. Long, G. V. et al. Adjuvant dabrafenib plus trametinib in stage III BRAF-mutated melanoma. N. Engl. J. Med. 377, 1813–1823 (2017).

    Article  CAS  Google Scholar 

  15. Huang A. C. et al. Safety, activity, and biomarkers for neoadjuvant anti-PD-1 therapy in melanoma. Cancer. Res. 78, abstr. CT181 (2018).

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

    Article  CAS  Google Scholar 

  17. Inoue, H. et al. Intratumoral expression levels of PD-L1, GZMA, and HLA-A along with oligoclonal T cell expansion associate with response to nivolumab in metastatic melanoma. OncoImmunology 5, e1204507 (2016).

    Article  Google Scholar 

  18. 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 

  19. Provenzano, E. et al. Standardization of pathologic evaluation and reporting of postneoadjuvant specimens in clinical trials of breast cancer: recommendations from an international working group. Mod. Pathol. 28, 1185–1201 (2015).

    Article  Google Scholar 

  20. NCI. National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) version 4.0 (2009).

  21. Cha, E. et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci. Transl. Med. 6, 238ra270 (2014).

    Article  Google Scholar 

  22. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  Google Scholar 

  23. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    Article  CAS  Google Scholar 

  24. Rimmer, A. et al. Integrating mapping-, assembly- and haplotype-based approaches for calling variants in clinical sequencing applications. Nat. Genet. 46, 912–918 (2014).

    Article  CAS  Google Scholar 

  25. 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 

  26. Ye, K., Schulz, M. H., Long, Q., Apweiler, R. & Ning, Z. Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics 25, 2865–2871 (2009).

    Article  CAS  Google Scholar 

  27. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    Article  CAS  Google Scholar 

  28. Wolchok, J. D. et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 369, 122–133 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the patients and their families for participating in this clinical trial. The clinical aspects of this study were funded by Bristol-Myers Squibb. The correlative research was supported by the philanthropic contributions to The University of Texas MD Anderson Cancer Center Melanoma Moon Shot Program, The Parker Institute for Cancer Immunotherapy and US Department of Defense Grant CA150619.

Author information

Authors and Affiliations

Authors

Contributions

R.N.A. and J.A.W. designed the study. R.N.A., H.A.T., M.A.D., M.I.R., I.C.G., J.N.C., C.L., W.-J.H., E.H., A.D., M.K.W., R.R., N.G., R.W., S.Y.L., R.E., P.H., S.P.P., A.L., A.H., J.E.L., J.G., L.S. and J.A.W. recruited and/or treated patients. R.N.A., S.M.R., J.B., D.R.M., S.W., R.K., D.K.W., L.H., L.S., E.M.B., L.P., L.W., S.Z., A.J.L., C.W.H., V.G., A.R., M.C.A., C.N.S., V.P., P.S., J.A., M.T.T. and J.A.W. analyzed and interpreted data. D.R.M. developed the statistical analysis plan. All authors developed and approved the manuscript.

Corresponding author

Correspondence to Jennifer A. Wargo.

Ethics declarations

Competing Interests

R.N.A. received grants from Merck, Bristol-Myers Squibb and Array Biopharma, all outside the submitted work. S.M.R. received support from National Institutes of Health T32 Training Grant T32 CA 009666, outside the submitted work. H.A.T. received personal fees from Novartis, grants from Merck and Celgene, and grants and personal fees from BMS and Genentech, all outside of the submitted work. M.A.D. received personal fees from Novartis, BMS and Vaccinex, grants from AstraZeneca and Merck, and grants and personal fees from Roche/Genentech and Sanofi-Aventis, all outside the submitted work. W.-J.H. received research grants from Merck, Bristol-Myers Squibb, MedImmune, and GlaxoSmithKline and has served on an advisory board for Merck, all outside the submitted work. M.K.W. received personal fees from Merck and EMD Serono, outside the submitted work. J.G. has participated in the advisory board of Merck and Castle Biosciences. A.J.L. received personal fees from BMS, Novartis, Merck and Genentech/Roche, personal fees and nonfinancial support from ArcherDX and Beta-Cat, grants and nonfinancial support from Medimmune/AstraZeneca and Sanofi and grants, personal fees and nonfinancial support from Janssen, all outside the submitted work. V.G. reports a US patent (PCT/US17/53,717), consultant fees from Microbiome DX, and honoraria from CAP18, outside of the submitted work. A.R. reports a US patent (PCT/US17/53,717) and is supported by the Kimberley Clark Foundation Award for Scientific Achievement provided by MD Anderson’s Odyssey Fellowship Program. M.C.A. is supported by the National Health and Medical Research Council of Australia CJ Martin Early Career Fellowship (1148680), and reports advisory board participation, travel support and honoraria from Merck Sharpe and Dohme. C.N.S. reports a US patent (PCT/US17/53,717), outside of the submitted work. P.S. received consultant or advisor fees from Bristol-Myers Squibb, GlaxoSmithKline, AstraZeneca, Amgen, Jounce, Kite Pharma, Neon, Evelo, EMD Serono and Astellas, during the conduct of the study; has stocks from Jounce, Kite Pharma, Evelo, Constellation and Neon, outside the submitted work; and has a patent licensed to Jounce, outside the submitted work. M.T.T. reports personal fees from Myriad Genetics, Seattle Genetics and Novartis, all outside the submitted work. J.A.W. reports a US patent (PCT/US17/53,717), has received compensation for speaker’s bureau and honoraria from Dava Oncology, Bristol-Myers Squibb and Illumina and has served on advisory committees for GlaxoSmithKline, Roche/Genentech, Novartis and AstraZeneca. All other authors declare no competing interests.

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 1–10, Supplementary Tables 1–11 and Supplementary Note

Reporting Summary

Supplementary Dataset

Integrated correlative data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Amaria, R.N., Reddy, S.M., Tawbi, H.A. et al. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med 24, 1649–1654 (2018). https://doi.org/10.1038/s41591-018-0197-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0197-1

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