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:

Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma

A Publisher Correction to this article was published on 05 October 2018

This article has been updated

Abstract

We describe results from IMmotion150, a randomized phase 2 study of atezolizumab (anti-PD-L1) alone or combined with bevacizumab (anti-VEGF) versus sunitinib in 305 patients with treatment-naive metastatic renal cell carcinoma. Co-primary endpoints were progression-free survival (PFS) in intent-to-treat and PD-L1+ populations. Intent-to-treat PFS hazard ratios for atezolizumab + bevacizumab or atezolizumab monotherapy versus sunitinib were 1.0 (95% confidence interval (CI), 0.69–1.45) and 1.19 (95% CI, 0.82–1.71), respectively; PD-L1+ PFS hazard ratios were 0.64 (95% CI, 0.38–1.08) and 1.03 (95% CI, 0.63–1.67), respectively. Exploratory biomarker analyses indicated that tumor mutation and neoantigen burden were not associated with PFS. Angiogenesis, T-effector/IFN-γ response, and myeloid inflammatory gene expression signatures were strongly and differentially associated with PFS within and across the treatments. These molecular profiles suggest that prediction of outcomes with anti-VEGF and immunotherapy may be possible and offer mechanistic insights into how blocking VEGF may overcome resistance to immune checkpoint blockade.

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: Positive independent review facility (IRF)-assessed efficacy associated with atezolizumab + bevacizumab in mRCC patients with PD-L1+ IC.
Fig. 2: Baseline tumor gene signature analyses.
Fig. 3: Association between tumor mutations and clinical outcome.

Similar content being viewed by others

Change history

  • 05 October 2018

    In the version of this article originally published, there was an error in Fig. 2n. The top line of the HR comparison chart originally was Atezo + bev vs sun. It should have been Atezo + bev vs atezo. The error has been corrected in the HTML and PDF versions of this article.

References

  1. Kaelin, W. G. Jr. The von Hippel-Lindau gene, kidney cancer, and oxygen sensing. J. Am. Soc. Nephrol. 14, 2703–2711 (2003).

    Article  PubMed  Google Scholar 

  2. George, D. J. & Kaelin, W. G. Jr. The von Hippel-Lindau protein, vascular endothelial growth factor, and kidney cancer. N. Engl. J. Med. 349, 419–421 (2003).

    Article  PubMed  Google Scholar 

  3. Motzer, R. J. et al. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a phase III trial. J. Clin. Oncol. 31, 3791–3799 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Clark, J. I. et al. Impact of sequencing targeted therapies with high-dose interleukin-2 immunotherapy: an analysis of outcome and survival of patients with metastatic renal cell carcinoma from an on-going observational Il-2 clinical trial: PROCLAIMSM. Clin. Genitourin. Cancer 15, 31–41.e4 (2017).

    Article  PubMed  Google Scholar 

  5. Herbst, R. S. et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515, 563–567 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Choueiri, T. K. et al. PD-L1 expression in nonclear-cell renal cell carcinoma. Ann. Oncol. 25, 2178–2184 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Thompson, R. H. et al. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin. Cancer Res. 13, 1757–1761 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Motzer, R. J. et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N. Engl. J. Med. 373, 1803–1813 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Thompson, R. H. et al. Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target. Proc. Natl. Acad. Sci. USA 101, 17174–17179 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Thompson, R. H. et al. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res. 66, 3381–3385 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Keir, M. E., Butte, M. J., Freeman, G. J. & Sharpe, A. H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704 (2008).

    Article  CAS  PubMed  Google Scholar 

  12. Latchman, Y. E. et al. PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. Proc. Natl. Acad. Sci. USA 101, 10691–10696 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yang, J. et al. The novel costimulatory programmed death ligand 1/B7.1 pathway is functional in inhibiting alloimmune responses in vivo. J. Immunol. 187, 1113–1119 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. McDermott, D. F. et al. Atezolizumab, an anti-programmed death-ligand 1 antibody, in metastatic renal cell carcinoma: long-term safety, clinical activity, and immune correlates from a phase Ia study. J. Clin. Oncol. 34, 833–842 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Elamin, Y. Y., Rafee, S., Toomey, S. & Hennessy, B. T. Immune effects of bevacizumab: killing two birds with one stone. Cancer Microenviron. 8, 15–21 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Kusmartsev, S. et al. Oxidative stress regulates expression of VEGFR1 in myeloid cells: link to tumor-induced immune suppression in renal cell carcinoma. J. Immunol. 181, 346–353 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Roland, C. L. et al. Inhibition of vascular endothelial growth factor reduces angiogenesis and modulates immune cell infiltration of orthotopic breast cancer xenografts. Mol. Cancer Ther. 8, 1761–1771 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Gabrilovich, D. I. & Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9, 162–174 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hodi, F. S. et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol. Res. 2, 632–642 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Roland, C. L. et al. Cytokine levels correlate with immune cell infiltration after anti-VEGF therapy in preclinical mouse models of breast cancer. PLoS One 4, e7669 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wallin, J. J. et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat. Commun. 7, 12624 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Osada, T. et al. The effect of anti-VEGF therapy on immature myeloid cell and dendritic cells in cancer patients. Cancer Immunol. Immunother. 57, 1115–1124 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Brauer, M. J. et al. Identification and analysis of in vivo VEGF downstream markers link VEGF pathway activity with efficacy of anti-VEGF therapies. Clin. Cancer Res. 19, 3681–3692 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. 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  PubMed  Google Scholar 

  25. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rizvi, N. A. et al. Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. Lancet Oncol. 16, 257–265 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Balar, A. V. et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet 389, 67–76 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Scheller, J., Chalaris, A., Schmidt-Arras, D. & Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta 1813, 878–888 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Russo, R. C., Garcia, C. C., Teixeira, M. M. & Amaral, F. A. The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases. Expert Rev. Clin. Immunol. 10, 593–619 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Ha, H., Debnath, B. & Neamati, N. Role of the CXCL8-CXCR1/2 axis in cancer and inflammatory diseases. Theranostics 7, 1543–1588 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zelenay, S. et al. Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell 162, 1257–1270 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Powles, T. et al. Immune biomarkers associated with clinical benefit from atezolizumab (MPDL3280a; anti-PD-L1) in advanced urothelial bladder cancer (UBC). J. Immunother. Cancer 3, 83 (2015).

    Article  Google Scholar 

  34. 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  PubMed  PubMed Central  Google Scholar 

  35. 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  PubMed  PubMed Central  Google Scholar 

  36. Turajlic, S. et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol. 18, 1009–1021 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Motzer, R. J. et al. IMmotion151: a randomized phase III study of atezolizumab plus bevacizumab vs. sunitinib in untreated metastatic renal cell carcinoma (mRCC). J. Clin. Oncol. 36, abstr, 578 (2018).

    Article  Google Scholar 

  38. Choueiri, T. K. et al. First-line avelumab + axitinib therapy in patients (pts) with advanced renal cell carcinoma (aRCC): Results from a phase Ib trial. J. Clin. Oncol. 35, 4504 (2017).

    Article  Google Scholar 

  39. Motzer, R. Nivolumab plus ipilimumab versus aunitinib in advanced renal-cell carcinoma. N. Engl. J. Med. 378, 1277–1290 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Voss, M. H. et al. Integrated biomarker analysis for 412 renal cell cancer (RCC) patients (pts) treated on the phase 3 COMPARZ trial: correlating common mutation events in PBRM1 and BAP1 with angiogenesis expression signatures and outcomes on tyrosine kinase inhibitor (TKI) therapy. J. Clin. Oncol. 35, 4523 (2017).

    Article  Google Scholar 

  41. Prima, V., Kaliberova, L. N., Kaliberov, S., Curiel, D. T. & Kusmartsev, S. COX2/mPGES1/PGE2 pathway regulates PD-L1 expression in tumor-associated macrophages and myeloid-derived suppressor cells. Proc. Natl. Acad. Sci. USA 114, 1117–1122 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860–867 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu, Q. et al. The CXCL8-CXCR1/2 pathways in cancer. Cytokine Growth Factor Rev. 31, 61–71 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Yuan, M. et al. Tumor-derived CXCL1 promotes lung cancer growth via recruitment of tumor-associated neutrophils. J. Immunol. Res. https://doi.org/10.1155/2016/6530410 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Sumida, K. et al. Anti-IL-6 receptor mAb eliminates myeloid-derived suppressor cells and inhibits tumor growth by enhancing T-cell responses. Eur. J. Immunol. 42, 2060–2072 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Najjar, Y. G. et al. Myeloid-derived suppressor cell subset accumulation in renal cell carcinoma parenchyma is associated with intratumoral expression of IL1β, IL8, CXCL5, and Mip-1α. Clin. Cancer Res. 23, 2346–2355 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Draghiciu, O., Nijman, H. W., Hoogeboom, B. N., Meijerhof, T. & Daemen, T. Sunitinib depletes myeloid-derived suppressor cells and synergizes with a cancer vaccine to enhance antigen-specific immune responses and tumor eradication. OncoImmunology 4, e989764 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Reck, M. et al. Primary PFS and safety analyses of a randomized phase III study of carboplatin + paclitaxel +/− bevacizumab, with or without atezolizumab in 1 L non-squamous metastatic NSCLC (IMpower150). Ann. Oncol. https://doi.org/10.1093/annonc/mdx760.002 (2017).

    Article  Google Scholar 

  49. 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  PubMed  PubMed Central  Google Scholar 

  50. Miao, D. et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 359, 801–806 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hsieh, J. J. et al. Genomic biomarkers of a randomized trial comparing first-line everolimus and sunitinib in patients with metastatic renal cell carcinoma. Eur. Urol. 71, 405–414 (2017).

    Article  CAS  PubMed  Google Scholar 

  52. Motzer, R. J. et al. Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J. Clin. Oncol. 17, 2530–2540 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Wu, T. D. & Nacu, S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics 26, 873–881 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wu, T. D., Reeder, J., Lawrence, M., Becker, G. & Brauer, M. J. GMAP and GSNAP for genomic sequence alignment: enhancements to speed, accuracy, and functionality. Methods Mol. Biol. 1418, 283–334 (2016).

    Article  PubMed  Google Scholar 

  55. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLOS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Ritchie, M. E. et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wilm, A. et al. LoFreq: a sequence-quality aware, ultra-sensitive variant caller for uncovering cell-population heterogeneity from high-throughput sequencing datasets. Nucleic Acids Res. 40, 11189–11201 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Saunders, C. T. et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 28, 1811–1817 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. McLaren, W. et al. The Ensembl variant effect predictor. Genome Biol. 17, 122 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lawrence, M., Degenhardt, J. & Gentleman, R. VariantTools: tools for working with genetic variants. version 1.12.0 Bioconductor https://bioconductor.org/packages/release/bioc/html/VariantTools.html (2018).

  62. Karosiene, E., Lundegaard, C., Lund, O. & Nielsen, M. NetMHCcons: a consensus method for the major histocompatibility complex class I predictions. Immunogenetics 64, 177–186 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Shukla, S. A. et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 33, 1152–1158 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Clopper, C. J. & Pearson, E. S. The use of confidence or fiducial limits illustrated in the case of the binomial. Biometrika 26, 404–413 (1934).

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank A. Bailey for her contributions to development of the protocol and Z. Boyd for his contributions to the development of the PD-L1 IHC assay and its implementation in this study. Support for third-party writing assistance for this manuscript—by P.S. Davies of Health Interactions, Inc.—was provided by F. Hoffmann-La Roche, AG. This study was sponsored by F. Hoffmann-La Roche, AG. Authors were funded by NCI grants P50 CA101942-13 to D.F.M, M.B.A., and T.K.C.; P30 CA008748 to R.J.M.; and P30 CA14599 to W.M.S.

Author information

Authors and Affiliations

Authors

Contributions

D.F.M., M.B.A., R.J.M., B.I.R., B.E., and T.P. contributed to the conception, trial design, and data acquisition, analysis, and interpretation; T.P. was the principal investigator of the study; M.A.H., S.J., and D.N. performed biomarker analyses and interpretation; J.Q. supervised the analysis of the clinical data; M.A.H. and P.S.H. supervised the analysis of biomarker data; L.F., R.W.J., S.K.P., J.A.R., M.S., J.H., W.K.R., W.M.S., T.H., M.E.G., A.R., S.B., C. Suárez, V.G., T.K.C., D.N., A.T., C. Schiff, E.P.-L., R.D., and G.D.F. made substantial contributions to the acquisition of data and data analysis and interpretation; P.S.H., M.A.H., and D.S.C. had overall biomarker oversight; C. Schiff, G.D.F., and D.S.C. had overall medical oversight.

Corresponding author

Correspondence to David F. McDermott.

Ethics declarations

Competing interests

D.F.M. reports a consulting/advisory role for Bristol-Myers Squibb, Merck, Roche/Genentech, Pfizer, Exelixis, Novartis, Eisai, X4 Pharmaceuticals, and Array BioPharma; and reports that his home institution receives research funding from Prometheus Laboratories. M.B.A. has been a paid consultant to Roche/Genentech, Bristol-Myers Squibb, Merck, Pfizer, Novartis, Exelixis, and Eisai. R.J.M. reports consulting fees from Roche/Genentech, Novartis, Pfizer, Eisai, and Exelixis, and research funds from Roche/Genentech, Bristol Myers Squibb, Pfizer, Novartis, Eisai, and Exelixis to the hospital for which he is employed. B.I.R. reports research funding to his institution from Roche/Genentech during the conduct of the study and grants/fees from Pfizer and Merck outside the submitted work. B.E. reports honoraria and research funding from Bristol-Myers Squibb, Novartis, Pfizer, and Ipsen; honoraria from Eusa Pharma, Roche, and Eisai; and research funding from Aveo. L.F. reports research funding to his institution from Roche/Genentech outside the submitted work. R.W.J. reports a consulting/advisory role with Bristol-Myers Squibb, Nektar, Genoptix, Eisai, Novartis, and Exilixis and research funding from Merck and Bristol-Myers Squibb; his home institution is in a consulting/advisory role with Merck and receives research funding from Roche/Genentech, X4 Pharmaceuticals, and Amgen. S.K.P. reports honoraria and a consulting/advisory role with Novartis, Astellas Pharma, Pfizer, Aveo, Myriad Pharmaceuticals, Roche/Genentech, Exelixis, Bristol-Myers Squibb, Ipsen, and Eisai and honoraria and research funding from Medivation. M.S. reports stock option interest in Amphivena Therapeutics, Intensity Therapeutics, and Adaptive Biotechnologies; a consulting role with Bristol-Myers Squibb, Roche/Genentech, AstraZeneca/MedImmune, Pfizer, Nektar, Lilly, Merck, Alexion Pharmaceuticals, Theravance, Baxalta/Shire, Seattle Genetics, Ignyta, Pierre Fabre, Incyte, Newlink Genetics, Celldex, Gritstone, and Innate Pharma; and an advisory role with Symphogen, Adaptimmune, Omniox, Lycera, and Molecular Partners. W.K.R. reports research funding to her home institution from Pfizer, Novartis, Tracon Pharmaceuticals, Bristol-Myers Squibb, Calithera Biosciences, and Peloton Therapeutics and research funding to an immediate family member from Incyte and Merck. W.M.S. reports honoraria and a consulting/advisory role with CVS Caremark, AstraZeneca, Bristol-Myers Squibb, Roche/Genentech, and Pfizer; research funding to his home institution from AstraZeneca, Bayer, Bristol-Myers Squibb, Boehringer Ingelheim, Exelixis, Novartis, Roche/Genentech, Pfizer, Merck, Janssen, and X4 Pharmaceuticals; and other relationships with UpToDate and American Cancer Society. M.E.G. acknowledges NHS funding to the NIHR Biomedical Research Centre at the Royal Marsden Hospital and Institute of Cancer Research, London UK. A.R. reports honoraria, accommodations, and a consulting/advisory role with Pfizer, Novartis, and Bristol-Myers Squibb; a consulting/advisory role with Ipsen and Roche; and research funding to his home institution from Pfizer and Novartis. S.B. reports personal fees and nonfinancial support for advisory boards from Pfizer, Astellas, Bristol-Myers Squibb, and Novartis; nonfinancial support for advisory boards from Bayer and Roche/Genentech; and nonfinancial support from Exelixis. V.G. reports grants from Bristol-Myers Squibb, Merck, Pfizer, and AstraZeneca, personal fees and nonfinancial support from Bristol-Myers Squibb, Merck, Roche, Novartis, Ipsen, Pfizer, AstraZeneca, Eisai, Eusa Pharma, and Cerulean outside the submitted work. T.K.C. reports consulting/advisory fees from AstraZeneca, Bayer, Bristol-Myers Squibb, Cerulean, Eisai, Foundation Medicine, Exelixis, Roche/Genentech, GlaxoSmithKline, Merck, Novartis, Peloton, Pfizer, Prometheus Laboratories, and Corvus; and research funding to his home institution from AstraZeneca, Bristol-Myers Squibb, Exelixis, Genentech, GlaxoSmithKline, Merck, Novartis, Peloton, Pfizer, Roche, Tracon, and Eisai. C. Schiff and P.S.H. report employment, including stock, with Genentech, Inc. G.D.F. reports employment, including stock, with Genentech, Inc. and stock with Foundation Medicine. T.P. reports honoraria and a consulting/advisory role with Roche/Genentech, Bristol-Myers Squibb, and Merck; a consulting/advisory role with AstraZeneca and Novartis; research funding from AstraZeneca/MedImmune and Roche/Genentech; and other relationships with Ipsen and Bristol-Myers Squibb (ASCO). M.A.H, D.N., S.J., E.P-L., J.Q., and D.S.C. are employees of Genentech, Inc. A.T. is an employee of Roche Products Ltd. J.A.R., J.H., T.H., C. Suárez, and R.D. have nothing to disclose.

Supplementary information

Supplementary Figures and Tables

Supplementary Figures 1–8 and Supplementary Tables 1–3

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

McDermott, D.F., Huseni, M.A., Atkins, M.B. et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med 24, 749–757 (2018). https://doi.org/10.1038/s41591-018-0053-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0053-3

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