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:

Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma

An Author Correction to this article was published on 17 April 2019

This article has been updated

Abstract

Immune checkpoint inhibitors have been successful across several tumor types; however, their efficacy has been uncommon and unpredictable in glioblastomas (GBM), where <10% of patients show long-term responses. To understand the molecular determinants of immunotherapeutic response in GBM, we longitudinally profiled 66 patients, including 17 long-term responders, during standard therapy and after treatment with PD-1 inhibitors (nivolumab or pembrolizumab). Genomic and transcriptomic analysis revealed a significant enrichment of PTEN mutations associated with immunosuppressive expression signatures in non-responders, and an enrichment of MAPK pathway alterations (PTPN11, BRAF) in responders. Responsive tumors were also associated with branched patterns of evolution from the elimination of neoepitopes as well as with differences in T cell clonal diversity and tumor microenvironment profiles. Our study shows that clinical response to anti-PD-1 immunotherapy in GBM is associated with specific molecular alterations, immune expression signatures, and immune infiltration that reflect the tumor’s clonal evolution during treatment.

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: Analysis pipeline and clinical characteristics of the cohort.
Fig. 2: Mutational landscape, genomic correlates of response, and tumor evolution under anti-PD-1 therapy.
Fig. 3: Transcriptomic signatures related to response to anti-PD-1 therapy.
Fig. 4: Tumor microenvironment profiling through qmIF.

Similar content being viewed by others

Code availability

All the custom code will be made available upon request.

Data availability

All the sequencing data have been deposited in SRA PRJNA482620. Processed data and basic association analyses will be made available upon request.

Change history

  • 17 April 2019

    In the version of this article originally published, the graph in Extended Data Fig. 2c was a duplication of Extended Data Fig. 2b. The correct version of Extended Data Fig. 2c is now available online.

References

  1. Gilbert, M. R. et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 370, 699–708 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. Garon, E. B. et al. Pembrolizumab for the treatment of non–small-cell lung cancer. N. Engl. J. Med. 372, 2018–2028 (2015).

    Article  Google Scholar 

  4. Ansell, S. M. et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 372, 311–319 (2015).

    Article  Google Scholar 

  5. Filley, A. C., Henriquez, M. & Dey, M. Recurrent glioma clinical trial, CheckMate-143: the game is not over yet. Oncotarget 8, 91779 (2017).

    Article  Google Scholar 

  6. Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  10. Wen, P. Y. et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J. Clin. Oncol. 28, 1963–1972 (2010).

    Article  Google Scholar 

  11. Wang, J. et al. Clonal evolution of glioblastoma under therapy. Nat. Genet. 48, 768 (2016).

    Article  CAS  Google Scholar 

  12. Hugo, W. et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016).

    Article  CAS  Google Scholar 

  13. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399 (2017).

    Article  Google Scholar 

  14. Chowell, D. et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 359, 582–587 (2018).

    Article  CAS  Google Scholar 

  15. Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).

    Article  CAS  Google Scholar 

  16. Peng, W. et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov. 6, 202–216 (2016).

    Article  CAS  Google Scholar 

  17. Parsa, A. T. et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat. Med. 13, 84 (2007).

    Article  CAS  Google Scholar 

  18. Dong, C., Davis, R. J. & Flavell, R. A. MAP kinases in the immune response. Annu. Rev. Immunol. 20, 55–72 (2002).

    Article  CAS  Google Scholar 

  19. Pan, D. et al. A major chromatin regulator determines resistance of tumor cells to T cell–mediated killing. Science 359, 770–775 (2018).

    Article  CAS  Google Scholar 

  20. Arrieta, V. A. et al. The possibility of cancer immune editing in gliomas. A critical review. Oncoimmunology 7, e1445458 (2018).

    Article  Google Scholar 

  21. Wang, Q. et al. Tumor evolution of glioma-intrinsic gene expression subtypes associates with immunological changes in the microenvironment. Cancer Cell 32, 42–56. e46 (2017).

    Article  Google Scholar 

  22. Yao, X. et al. Levels of peripheral CD4+FoxP3+ regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 119, 5688–5696 (2012).

    Article  CAS  Google Scholar 

  23. Lin, K.-Y. et al. Ectopic expression of vascular cell adhesion molecule-1 as a new mechanism for tumor immune evasion. Cancer Res. 67, 1832–1841 (2007).

    Article  CAS  Google Scholar 

  24. Yuan, J. et al. Single-cell transcriptome analysis of lineage diversity and microenvironment in high-grade glioma. Genome Med. 10, 57 (2018).

    Article  Google Scholar 

  25. Rizvi, A. H. et al. Single-cell topological RNA-seq analysis reveals insights into cellular differentiation and development. Nat. Biotechnol. 35, 551–560 (2017).

    Article  CAS  Google Scholar 

  26. Senbabaoglu, Y. et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol. 17, 231 (2016).

    Article  Google Scholar 

  27. Zhang, C. et al. Tumor purity as an underlying key factor in glioma. Clin. Cancer Res. 23, 6279–6291 (2017).

    Article  CAS  Google Scholar 

  28. Hussain, S. F. et al. The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neuro-oncology 8, 261–279 (2006).

    Article  CAS  Google Scholar 

  29. Gartrell, R. D. et al. Quantitative analysis of immune infiltrates in primary melanoma. Cancer Immunol. Res. 6, 481–493 (2018).

    Article  CAS  Google Scholar 

  30. Stack, E. C., Wang, C., Roman, K. A. & Hoyt, C. C. Multiplexed immunohistochemistry, imaging, and quantitation: a review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods 70, 46–58 (2014).

    Article  CAS  Google Scholar 

  31. Mooney, K. L. et al. The role of CD44 in glioblastoma multiforme. J. Clin. Neurosci. 34, 1–5 (2016).

    Article  CAS  Google Scholar 

  32. George, S. et al. Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma. Immunity 46, 197–204 (2017).

    Article  CAS  Google Scholar 

  33. Lastwika, K. J. et al. Control of PD-L1 expression by oncogenic activation of the AKT–mTOR pathway in non-small cell lung cancer. Cancer Res. 76, 227–238 (2016).

    Article  CAS  Google Scholar 

  34. Noh, K. H. et al. Activation of Akt as a mechanism for tumor immune evasion. Mol. Ther. 17, 439–447 (2009).

    Article  CAS  Google Scholar 

  35. Bedognetti, D., Roelands, J., Decock, J., Wang, E. & Hendrickx, W. The MAPK hypothesis: immune-regulatory effects of MAPK-pathway genetic dysregulations and implications for breast cancer immunotherapy. Emerg. Top. Life Sci. 1, 429–445 (2017).

    Article  Google Scholar 

  36. Ebert, P. J. et al. MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade. Immunity 44, 609–621 (2016).

    Article  CAS  Google Scholar 

  37. Deken, M. A. et al. Targeting the MAPK and PI3K pathways in combination with PD1 blockade in melanoma. Oncoimmunology 5, e1238557 (2016).

    Article  Google Scholar 

  38. Toso, A. et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 9, 75–89 (2014).

    Article  CAS  Google Scholar 

  39. Tran, E. et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 350, 1387–1390 (2015).

    Article  CAS  Google Scholar 

  40. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  Google Scholar 

  41. Stupp, R. et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 352, 987–996 (2005).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  43. Trifonov, V., Pasqualucci, L., Tiacci, E., Falini, B. & Rabadan, R. SAVI: a statistical algorithm for variant frequency identification. BMC Syst. Biol. 7, S2 (2013).

    Article  Google Scholar 

  44. Talevich, E., Shain, A. H., Botton, T. & Bastian, B. C. CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput. Biol. 12, e1004873 (2016).

    Article  Google Scholar 

  45. Carter, S. L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413 (2012).

    Article  CAS  Google Scholar 

  46. Yoshihara, K. et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat. Commun. 4, 2612 (2013).

    Article  Google Scholar 

  47. Iyer, M. K., Chinnaiyan, A. M. & Maher, C. A. ChimeraScan: a tool for identifying chimeric transcription in sequencing data. Bioinformatics 27, 2903–2904 (2011).

    Article  CAS  Google Scholar 

  48. Abate, F. et al. Pegasus: a comprehensive annotation and prediction tool for detection of driver gene fusions in cancer. BMC Syst. Biol. 8, 97 (2014).

    Article  Google Scholar 

  49. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Google Scholar 

  50. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2013).

    Article  Google Scholar 

  51. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  Google Scholar 

  52. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  54. Hundal, J. et al. pVAC-Seq: a genome-guided in silico approach to identifying tumor neoantigens. Genome Med. 8, 11 (2016).

    Article  Google Scholar 

  55. 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  Google Scholar 

  56. Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380 (2015).

    Article  CAS  Google Scholar 

  57. Baddeley, A, Rubak, E. & Turner, R. Spatial Point Patterns: Methodology and Applications with R (Chapman and Hall/CRC Press, Boca Raton, FL, USA, 2015).

    Book  Google Scholar 

Download references

Acknowledgements

This work has been funded by NIH grants R01 CA185486 (R.R.), R01 CA179044 (R.R.), U54 CA193313, U54 209997 (R.R.), R01 NS103473 (P.C., J.N.B., P.S.), NSF/SU2C/V-Foundation Ideas Lab Multidisciplinary Team (PHY-1545805) (R.R.), 2018 Stand Up To Cancer Phillip A. Sharp Innovation in Collaboration Awards (R.R.) and Keep Punching Foundation (F.M.I.). Funding support from Northwestern 5DP5OD021356-04 (A.M. Sonabend), P50CA221747 SPORE for Translational Approaches to Brain Cancer (A.M.Sonabend, R.V.L., C.H.). Developmental funds from The Robert H Lurie NCI Cancer Center Support Grant no. P30CA060553 (A.M. Sonabend). A.X.C. is funded by the Medical Scientist Training Program (T32GM007367). R.D.G. is funded by CUIMC CTSA as TL1 Precision Medicine Fellow (1TL1TR001875-01) and Swim Across America.

Author information

Authors and Affiliations

Authors

Contributions

J.Z. performed the majority of experiments and analyses. R.R., A.M. Sonabend, J.Z., and A.X.C wrote the manuscript. A.I.R., J.T.Y., M.C., R.V.L., and J.R. compiled the clinical data for analysis. J.N.B., J.S., A.M., P.C., and C.H. procured and reviewed the tumor specimens for sequencing. J.Y. and P.S. provided the single-cell transcriptomic data. R.D.G., A.M. Silverman, D.B., D.S., and Y.M.S. provided the single-cell immunofluorescence. L.A. performed single-cell transcriptomic data analysis. I.F. and R.O. performed HLA genotyping. M.G. performed survival analysis. T.C. conducted figure design. R.R., A.M. Sonabend, and F.M.I. designed and supervised the entire project.

Corresponding authors

Correspondence to Fabio M. Iwamoto, Adam M. Sonabend or Raul Rabadan.

Ethics declarations

Competing interests

The 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.

Extended data

Extended Data Fig. 1 Additional clinical characteristics of the cohort.

a, Venn diagram of the data modalities available across the 66-patient cohort. b,c, Kaplan–Meier curve comparing post-treatment survival (b) and overall survival from diagnosis (c) of patients who responded to anti-PD-1 therapy (n = 13) with those that did not respond (n = 29; P =4.5 × 10–5 (b), P = 0.0045 (c), two-sided log-rank test), assessed across the entire cohort. d, Univariate survival analysis reveals that response to anti-PD-1 therapy is still most correlated with post-treatment survival of the patients when assessed across the entire cohort (n = 42, 13 responders, 29 non-responders; P value, two-sided log-rank test).

Extended Data Fig. 2 Additional analysis of genomic correlates of response to anti-PD-1 immunotherapy.

a, Mutation burden by response group (n = 17 patients). b, Tumor purity, as estimated by ABSOLUTE, by response group. c, Ratio of subclonal to clonal mutations, as estimated by ABSOLUTE, by response group. d, Aneuploidy score analysis of non-responders versus responders. Boxplots show the median, interquartile range, and whiskers (1.5 times interquartile range).

Extended Data Fig. 3 Additional analysis of transcriptomic correlates of response to anti-PD-1 immunotherapy.

a, GSEA enrichment score of gene-set KIM_PTEN_TARGETS_UP for non-responders versus responders (n = 12 patients). The boxplot shows the median, interquartile range, and whiskers (1.5 times interquartile range). b, Boxplot of CD274 (encoding PD-L1) messenger RNA expression in responders versus non-responders (n = 12 patients). The boxplot shows the median, interquartile range, and whiskers (1.5 times interquartile range).

Extended Data Fig. 4 Clonal diversity of lymphocytes before and after immunotherapy.

Within seven patients with longitudinal information on TCR and immunoglobulin (Ig) RNA expression, MiXCR was used to group reads into T cell (a) and B cell clones (b). Each color on a bar represents the fractional presence of a different clone, with the total clonal read count, n, listed above.

Extended Data Figure 5 Non-responders demonstrate a greater increase in clonal diversity of B cells following immunotherapy.

B cell clonal diversity before and after immunotherapy was assessed by identifying immunoglobulin RNA sequences within the tumor. Non-responders had a greater increase in Shannon entropy among B cells compared with responders (P = 0.048, two-sided exact Mann–Whitney U test; n = 16 independent timepoints from seven patients). The boxplot shows the median, interquartile range, and whiskers (1.5 times interquartile range); the violin plot represents sample distributions via kernel density estimation.

Extended Data Figure 6 Tumor subtype.

Expression subtyping of tumors from nine patients (pre- and post-treatment) into proneural, mesenchymal, and classical subtypes.

Extended Data Figure 7 GSEA analysis.

GSEA enrichment plots (n = 12 patients; six responders versus six non-responders) of two Treg-cell-related gene sets; P = 0.004 (left), P = 0.013 (right), two-sided Kolmogorov–Smirnov test.

Extended Data Figure 8 Enrichment of Treg cell signature.

a, Cells associated with the Treg cell signature were enriched in a PTEN-mutated tumor. b, Tumors associated with the Treg cell signature were enriched in PTEN-mutated samples.

Extended Data Figure 9 Single-cell RNA-seq data analysis.

Topological data analysis of single-cell RNA-seq data (n = 4,000 cells) from a PTEN-mutated tumor, demonstrating clusters of cells with high expression of CD44 (A, in red) and of microglial signatures (B, in red).

Extended Data Figure 10 Tumor purity analysis.

PTEN-mutated GBM tumors have significantly lower tumor purity compared with PTEN wild-type tumors (n = 172, two-sided Wilcoxon rank-sum test). The boxplot shows the median, interquartile range, and whiskers (1.5 times interquartile range); the violin plot represents sample distributions via kernel density estimation.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, J., Chen, A.X., Gartrell, R.D. et al. Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med 25, 462–469 (2019). https://doi.org/10.1038/s41591-019-0349-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0349-y

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