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:

ERK1/2 phosphorylation predicts survival following anti-PD-1 immunotherapy in recurrent glioblastoma

A Publisher Correction to this article was published on 11 February 2022

This article has been updated

Abstract

Only a subset of recurrent glioblastoma (rGBM) responds to anti-PD-1 immunotherapy. Previously, we reported enrichment of BRAF/PTPN11 mutations in 30% of rGBM that responded to PD-1 blockade. Given that BRAF and PTPN11 promote MAPK/ERK signaling, we investigated whether activation of this pathway is associated with response to PD-1 inhibitors in rGBM, including patients that do not harbor BRAF/PTPN11 mutations. Here we show that immunohistochemistry for ERK1/2 phosphorylation (p-ERK), a marker of MAPK/ERK pathway activation, is predictive of overall survival following adjuvant PD-1 blockade in two independent rGBM patient cohorts. Single-cell RNA-sequencing and multiplex immunofluorescence analyses revealed that p-ERK was mainly localized in tumor cells and that high-p-ERK GBMs contained tumor-infiltrating myeloid cells and microglia with elevated expression of MHC class II and associated genes. These findings indicate that ERK1/2 activation in rGBM is predictive of response to PD-1 blockade and is associated with a distinct myeloid cell phenotype.

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: ERK1/2 activation is a predictive biomarker of radiographic response to anti-PD-1 immunotherapy in patients with recurrent GBM.
Fig. 2: ERK1/2 phosphorylation evaluated by semiautomatic IHC quantification shows that is a predictive biomarker following PD-1 blockade in recurrent GBM.
Fig. 3: Validation of pretreatment p-ERK staining correlates with OS in an independent recurrent GBM cohort treated with adjuvant PD-1 blockade.
Fig. 4: Multiplex immunofluorescence of recurrent GBM samples shows p-ERK positivity in SOX2+ cells and associated myeloid cell infiltration.
Fig. 5: Spatial analysis of tumor cells expressing p-ERK and their associated myeloid cells.
Fig. 6: scRNA-seq in patients with GBM from high- and low-p-ERK groups.

Similar content being viewed by others

Data availability

Single-cell RNA-seq data supporting the findings of this study have been deposited in the Gene Expression Omnibus (GEO) under accession code nos. GSE103224 and GSE141383. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding authors on reasonable request.

Code availability

Code is available at https://github.com/RabadanLab/GBMsinglecell.

Change history

References

  1. Stupp, R. et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. JAMA 318, 2306–2316 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Neftel, C. et al. An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell 178, 835–849 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gong, J., Chehrazi-Raffle, A., Reddi, S. & Salgia, R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J. Immunother. Cancer 6, 8 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Reardon, D. A. et al. Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the CheckMate 143 Phase 3 randomized clinical trial. JAMA Oncol. 6, 1003–1010 (2020).

    Article  PubMed  Google Scholar 

  6. Nayak, L. et al. Randomized phase II and biomarker study of pembrolizumab plus bevacizumab versus pembrolizumab alone for recurrent glioblastoma patients. Clin. Cancer Res. 27, 1048–1057 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. de Groot, J. et al. Window-of-opportunity clinical trial of pembrolizumab in patients with recurrent glioblastoma reveals predominance of immune-suppressive macrophages. Neuro. Oncol. 22, 539–549 (2020).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  9. Chongsathidkiet, P. et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat. Med. 24, 1459–1468 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Jackson, C. M., Choi, J. & Lim, M. Mechanisms of immunotherapy resistance: lessons from glioblastoma. Nat. Immunol. 20, 1100–1109 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Arrieta, V. A. et al. Can patient selection and neoadjuvant administration resuscitate PD-1 inhibitors for glioblastoma? J. Neurosurg. 132, 1667–1672 (2019).

    Article  PubMed  Google Scholar 

  12. Woroniecka, K. et al. T-cell exhaustion signatures vary with tumor type and are severe in glioblastoma. Clin. Cancer Res. 24, 4175–4186 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhao, J. et al. Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat. Med. 25, 462–469 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Cloughesy, T. F. et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 25, 477–486 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dhillon, A. S., Hagan, S., Rath, O. & Kolch, W. MAP kinase signalling pathways in cancer. Oncogene 26, 3279–3290 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Ruess, D. A. et al. Mutant KRAS-driven cancers depend on PTPN11/SHP2 phosphatase. Nat. Med. 24, 954–960 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Svensson, S. et al. ERK phosphorylation is linked to VEGFR2 expression and Ets-2 phosphorylation in breast cancer and is associated with tamoxifen treatment resistance and small tumours with good prognosis. Oncogene 24, 4370–4379 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Roth, P., Valavanis, A. & Weller, M. Long-term control and partial remission after initial pseudoprogression of glioblastoma by anti-PD-1 treatment with nivolumab. Neuro. Oncol. 19, 454–456 (2017).

    Article  PubMed  Google Scholar 

  19. Okada, H. et al. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol. 16, e534–e542 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Vassilakopoulou, M. et al. Preanalytical variables and phosphoepitope expression in FFPE tissue: quantitative epitope assessment after variable cold ischemic time. Lab. Invest. 95, 334–341 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Nagasawa-Masuda, A. & Terai, K. ERK activation in endothelial cells is a novel marker during neovasculogenesis. Genes Cells 21, 1164–1175 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Pombo Antunes, A. R. et al. Single-cell profiling of myeloid cells in glioblastoma across species and disease stage reveals macrophage competition and specialization. Nat. Neurosci. 24, 595–610 (2021).

    Article  CAS  PubMed  Google Scholar 

  23. Hara, T. et al. Interactions between cancer cells and immune cells drive transitions to mesenchymal-like states in glioblastoma. Cancer Cell 39, 779–792 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Bennett, M. L. et al. New tools for studying microglia in the mouse and human CNS. Proc. Natl Acad. Sci. USA 113, E1738–E1746 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sankowski, R. et al. Mapping microglia states in the human brain through the integration of high-dimensional techniques. Nat. Neurosci. 22, 2098–2110 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Friebel, E. et al. Single-cell mapping of human brain cancer reveals tumor-specific instruction of tissue-invading leukocytes. Cell 181, 1626–1642 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Klemm, F. et al. Interrogation of the microenvironmental landscape in brain tumors reveals disease-specific alterations of immune cells. Cell 181, 1643–1660 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Saha, D., Martuza, R. L. & Rabkin, S. D. Macrophage polarization contributes to glioblastoma eradication by combination immunovirotherapy and immune checkpoint blockade. Cancer Cell 32, 253–267 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Goswami, S. et al. Immune profiling of human tumors identifies CD73 as a combinatorial target in glioblastoma. Nat. Med. 26, 39–46 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Rao, G. et al. Anti-PD-1 induces M1 polarization in the glioma microenvironment and exerts therapeutic efficacy in the absence of CD8 cytotoxic T cells. Clin. Cancer Res. 26, 4699–4712 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Keren, L. et al. A structured tumor-immune microenvironment in triple negative breast cancer revealed by multiplexed ion beam imaging. Cell 174, 1373–1387 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Schalper, K. A. et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat. Med. 25, 470–476 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hellmann, M. D. et al. Nivolumab plus Ipilimumab in lung cancer with a high tumor mutational burden. N. Engl. J. Med. 378, 2093–2104 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Cristescu, R. et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 362, eaar3593 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Johanns, T. M. et al. Immunogenomics of hypermutated glioblastoma: a patient with germline POLE deficiency treated with checkpoint blockade immunotherapy. Cancer Discov. 6, 1230–1236 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Lukas, R. V. et al. Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma. J. Neurooncol. 140, 317–328 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Bouffet, E. et al. Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J. Clin. Oncol. 34, 2206–2211 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Ahmad, H., Fadul, C. E., Schiff, D. & Purow, B. Checkpoint inhibitor failure in hypermutated and mismatch repair-mutated recurrent high-grade gliomas. Neurooncol. Pract. 6, 424–427 (2019).

    PubMed  PubMed Central  Google Scholar 

  42. Samstein, R. M. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 51, 202–206 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Touat, M. et al. Mechanisms and therapeutic implications of hypermutation in gliomas. Nature 580, 517–523 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wick, W. et al. Lomustine and bevacizumab in progressive glioblastoma. N. Engl. J. Med. 377, 1954–1963 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Azoulay, M. et al. Benefit of re-operation and salvage therapies for recurrent glioblastoma multiforme: results from a single institution. J. Neurooncol. 132, 419–426 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Robert, K. J. et al. CD8+ T-cell-mediated immunoediting influences genomic evolution and immune evasion in murine gliomas. Clin. Cancer Res. 26, 4390–4401 (2020).

    Article  Google Scholar 

  48. Chen, P. et al. Symbiotic macrophage–glioma cell interactions reveal synthetic lethality in PTEN-null glioma. Cancer Cell 35, 868–884 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hutter, G. et al. Microglia are effector cells of CD47-SIRPα antiphagocytic axis disruption against glioblastoma. Proc. Natl Acad. Sci. USA 116, 997–1006 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Strauss, L. et al. Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci. Immunol. 5, eaay1863 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gromeier, M. et al. Very low mutation burden is a feature of inflamed recurrent glioblastomas responsive to cancer immunotherapy. Nat. Commun. 12, 352 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Liu, D. et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 25, 1916–1927 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Alspach, E. et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 574, 696–701 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mrdjen, D. et al. High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity 48, 380–395 (2018).

    Article  CAS  PubMed  Google Scholar 

  55. Mundt, S. et al. Conventional DCs sample and present myelin antigens in the healthy CNS and allow parenchymal T cell entry to initiate neuroinflammation. Sci. Immunol. 4, eaau8380 (2019).

    Article  CAS  PubMed  Google Scholar 

  56. Qian, J. et al. TLR2 promotes glioma immune evasion by downregulating MHC class II molecules in microglia. Cancer Immunol. Res. 6, 1220–1233 (2018).

    CAS  PubMed  Google Scholar 

  57. Schartner, J. M. et al. Impaired capacity for upregulation of MHC class II in tumor-associated microglia. Glia 51, 279–285 (2005).

    Article  PubMed  Google Scholar 

  58. Sarkar, S. et al. Therapeutic activation of macrophages and microglia to suppress brain tumor-initiating cells. Nat. Neurosci. 17, 46–55 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Sarkar, S. et al. Microglia induces Gas1 expression in human brain tumor-initiating cells to reduce tumorigenecity. Sci. Rep. 8, 15286 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Zhang, J. et al. Compartmental analysis of T-cell clonal dynamics as a function of pathologic response to neoadjuvant PD-1 blockade in resectable non-small cell lung cancer. Clin. Cancer Res. 26, 1327–1337 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. Robin, X. et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics 12, 77 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Hothorn, T., Hornik, K. & Zeileis, A. Unbiased recursive partitioning: a conditional inference framework. J. Comput. Graph. Stat. 15, 651–674 (2006).

    Article  Google Scholar 

  63. Hothorn, T. & Zeileis, A. partykit: A modular toolkit for recursive partytioning in R. https://www2.uibk.ac.at/downloads/c4041030/wpaper/2014-10.pdf (2015).

  64. McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: Uniform Manifold Approximation and Projection. J. Open Source Softw. 3, 861 (2018).

    Article  Google Scholar 

  65. Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grant nos. 1R01NS110703-01A1 and 5DP5OD021356-05 (both A.M.S.); no. P50CA221747 SPORE for Translational Approaches to Brain Cancer (principal investigator M. S. Lesniak, with support to A.M.S.); developmental funds from the Robert H. Lurie Cancer Center Support Grant (no. P30CA060553, A.M.S.); Vagelos Precision Medicine Award (F.M.I., R.R. and J.Z.), U54CA193313 (R.R.), U54CA209997 (R.R.), R35CA253126 (R.R.); Keep Punching (F.M.I.); The William Rhodes and Louise Tilzer-Rhodes Center for Glioblastoma at New York-Presbyterian Hospital (F.M.I.); NIAID nos. 1R01AI099195 (U.B.) and R01AI134988 (U.B.). V.A.A. is financially supported by the Mexican government through the Mexican National Council for Science and Technology and the Plan of Combined Studies in Medicine of the National Autonomous University of Mexico. A.X.C. is funded by the Medical Scientist Training Program (no. T32GM007367). We thank T. Sudhakar for sample collection at Columbia University; K. McCortney, R. Javier and J. Walshon from the Nervous System Tumor Bank supported by the P50CA221747 SPORE for Translational Approaches to Brain Cancer; and B. Shmaltsuyeva for immunohistochemistry at the Northwestern University Pathology Core Facility funded by Cancer Center Support Grant (no. NCI CA060553). We thank L. Kai for technical support on multiplex staining performed at the Immunotherapy Assessment Core at Northwestern University and in the Flow Cytometry & Cellular Imaging Core Facility, which is supported in part by NIH through MD Anderson´s Cancer Center Support Grant no. CA016672, the NCI´s Research Specialist 1 (no. R50 CA23707, J.K.B.) and NIH grant nos. R01-CA120813 and 1R01-CA237418 (both A.B.H.).

Author information

Authors and Affiliations

Authors

Contributions

V.A.A., A.X.C. and J.R.K. performed the majority of experiments and analyses. V.A.A., J.R.K., R.R., F.M.I. and A.M.S. conceptualized and designed the study. V.A.A. and L.A.D.C. quantified IHC images. V.A.A., A.X.C. and A.M.S. wrote the manuscript. R.S., P.U., J.Z., R.V.L., C.D., D.C., X.L., A.G., S.J.K., J.S., D.Z., J.N.B. and J.T.Y. compiled the clinical data for analysis. T.F.C. and R.P. provided tumor samples and clinical data for the validation cohort. C.A. and L.C. provided administrative support. V.A.A., A.X.C., K.B.B. and H.Z. performed survival and statistical analyses. P.C. and J.N.B. are responsible for the Tumor Bank at Columbia University. C.H. is responsible for the Tumor Bank at Northwestern University. P.U. acquired tumor specimens. C.H., M.M., D.J.B. and P.C. scored the tumor slides. J.Y., W.Z. and P.S. provided scRNA-seq data. A.X.C., J.Z. and R.R. performed scRNA-seq data analysis. D.J., B.Z., C.K., J.K.B., X.L. and A.B.H. performed Opal multiplex tissue staining. V.A.A., A.X.C. and C.L.-C. analyzed multiplex immunofluorescence images. V.A.A., C.L.-C., B.L., G.R. and U.B. acquired and analyzed flow cytometry data. V.A.A. and S.J.K. performed immunoblots and the peptide competition assay. A.M.S., R.R. and F.M.I. supervised the entire study.

Corresponding authors

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

Ethics declarations

Competing interests

F.M.I. is a consultant for Merck, Novocure, Regeneron, Abbvie, Alexion, Guidepoint and Tocagen. He has received clinical or research support (including equipment or material) from Bristol Myers Squibb and Merck. A.M.S., F.M.I. and R.R. are co-authors for the following patent filed by Columbia University: Systems and methods for predicting clinical responses to immunotherapies. A.M.S., V.A.A, F.M.I. and R.R. are co-authors of the following patent filed by Northwestern University: 5. Methods for treating glioblastoma. A.M.S. has received a consulting honorarium from Abbvie. R.R. is a member of the SAB of AimedBio and a founder of Genotwin. R.V.L. has received support from Roche-Genentech for meeting travel to present study results; honoraria for advisory boards for AstraZeneca, Abbvie and Ziopharm; and honoraria for medical editing for EBSCO publishing, Medlink Neurology and American Physician Institute. He has also received honoraria for consultation with Eisai and Abbvie, and honoraria for creating and presenting CME Board review material for the American Physician Institute. R.V.L. received drug support (but no additional support) from BMS for an investigator-initiated trial. A.B.H. is a consultant for Caris Life Sciences and WCG Oncology Advisory Board, receives royalties and milestone payments from Celldex Therapeutics and DNAtrix and receives clinical or research support (including equipment and materials) from Celularity, Codiak Biosciences, Moleculin and Carthera. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Cancer thanks Robert Manguso and Ignacio Melero for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Optimization of the staining technique and quantification of p-ERK.

a, Titration of the p-ERK antibody (clone: D13.14.4E) using different dilutions performed in GBM samples. We show the same region of a GBM sample stained with the indicated dilutions of the p-ERK antibody with a low and high magnification image for each dilution. b, (left) Microarray containing breast cancer tissues stained with p-ERK antibody (1:500 dilution) representing a positive control. (right) Magnification of one the breast cancer tissues showing specific staining in the endothelium (red rectangle). c, (left) Nontumoral brain tissue stained with p-ERK antibody (1:500 dilution) representing a negative control. (right) Magnification of the white matter showing p-ERK staining with minimal background. Dilution titration and staining of positive and negative controls were performed as a single experiment in one standardized run. d, Workflow used for the software-based quantification of p-ERK+ cells.

Extended Data Fig. 2 Quantification and cut point optimization of p-ERK+ cell density in tumoral regions.

a, Dot plot showing the distribution of p-ERK quantification of all GBM samples treated and nontreated with PD-1 blockade N = 62 tumors). b, From top to bottom, micrographs showing one high p-ERK tumor sample and two low p-ERK tumor samples with positive staining in the endothelial cells (red arrows). In the dot plot, the magenta dot represents CU100 patient, the green dot represents NU01688 patient, and the red dot represents CU110 patient. IHC images are representative of 62 independent GBM samples. c, Conditional inference trees analysis for cut-point optimization in the GBM cohort treated with PD-1 blockade reveals a cut-point value similar to the median of all tumor samples. d, Forest plot representing the univariable analysis using a Cox regression model evaluating the clinical and molecular prognostic factors that might confound the association between survival p-ERK and presented as Hazard ratio (95% CI). N = 29 GBM patients. P value by two-sided Wald test. e, Kaplan-Meier curve comparing OS of recurrent GBM patients scored as either high or low p-ERK by assessment of a neuropathologist counting from initiation of PD-1 blockade (anti-PD-1 therapy group, N = 29 GBM patients) and from surgery at recurrence (no-immunotherapy group, N = 33 GBM patients). p-ERK scores in tumor regions were designated as follows: 0-1 were considered as low, and 2–3 as high; P value by two-sided log-rank test.

Extended Data Fig. 3 Preservation of the p-ERK epitope and peptide competition assay neutralizing the p-ERK1/2 antibody tested in FFPE GBM samples.

a, Protein extraction from FFPE GBM tissues for assessment of selected phosphoproteins. b, Western blot targeting p-ERK, ERK1, ERK2, p-AKT, AKT, and β-actin in a subset of GBM samples used for survival analysis. Western blotting was done as a single experiment in 12 independent GBM samples and 2 GBM cell lines. c, Peptide competition assay in which p-ERK1/2 antibody was neutralized with a blocking peptide employing extracted proteins obtained from GBM samples. The peptide competition assay was assessed by western blot. One western blot was incubated with the neutralized p-ERK antibody and the other with the free p-ERK antibody. d, Peptide competition assay employing IHC using the same GBM samples used to perform western blot employing the neutralized and free p-ERK antibody to perform the staining. The experiments were done in 4 independent GBM samples and 2 GBM cell lines as a single experiment.

Source data

Extended Data Fig. 4 Evaluation of the ischemic time on p-ERK degradation by IHC and western blot, and comparison to the samples used in this study.

a, Representatives images of the analysis conducted to evaluate p-ERK degradation in endothelial cells of GBM samples at different periods of ischemic time. For this, 3 human tumor specimens were obtained during surgery, and immediately divided into similar size portions, which then were subjected to different ischemic times before processing. Specific endothelial cells subjected to analysis are labeled with colors assigned by the software. b, Blue bars represent p-ERK+ cells mm2 in tumor regions, and dots represent p-ERK intensity on individual endothelial cells within the same samples used to evaluate the effect of ischemic time on p-ERK degradation, and tumor samples used for survival analysis (PD-1 immunotherapy cohort and no immunotherapy cohort). Each dot represents one ROI analyzing one endothelial cell. Green dots (N = 24, 20, 13 endothelial cells from NU02608, NU02617, and NU02609, respectively) represent a statistically significant group compared to the group of 0 hrs. of ischemic time represented as gray dots (N = 18, 19, 14 endothelial cells from NU02608, NU02617, and NU02609, respectively). All samples were normalized to the average of values of the three 0 hrs. groups. P values by two-sided Kruskal Wallis test with post hoc Dunn’s multiple comparison test. c, Western blot showing p-ERK and other phosphoproteins in samples subjected to different ischemic times. Densitometry analysis for p-ERK western blot was performed using ERK1 and ERK2 staining. For this densitometry, every patient had density normalized by 0 minutes of ischemic time. N = 3 GBM samples. Error bars represent SEM. Western blot was done as a single experiment in 3 independent GBM samples.

Source data

Extended Data Fig. 5 Progression-free survival of the validation cohort from the Cloughesy T et al.14 clinical trial.

a, b, Kaplan-Meier showing progression-free survival following PD-1 blockade based on p-ERK high vs low for pre-study (a) and on-study (b) tumor samples. N = 13 GBM patients. P values by two-sided log rank test.

Extended Data Fig. 6 Multiplex immunofluorescence staining of recurrent GBM samples employing GFAP marker.

a, Bar plot showing the comparison of GFAP+ p-ERK+ cells and other cells expressing p-ERK+. N = 6 tumor samples. P value by two-sided Mann Whitney U test. Data is presented as mean ± s.d. b, Representative images of three different tumor samples derived form results in a. From top to bottom: a BRAFV600E GBM sample having high p-ERK staining, a wild-type BRAF/PTPN11 GBM having high p-ERK staining, and a wild-type BRAF/PTPN11 GBM displaying low p-ERK staining. For the three tumor samples: (left) H&E and p-ERK IHC images of the same tumor region. (middle), Multiplex immunofluorescence images showing the markers for GFAP, p-ERK, and DAPI. (right) Multiplex immunofluorescence images showing the markers for GFAP, CD163, and DAPI. Experiment was done using a tumor sample in one standardized run per patient.

Source data

Extended Data Fig. 7 Single-cell RNA seq of GBM patients with high and low p-ERK IHC staining.

UMAP representation of 28,194 individual cells from 10 GBM patients measured with scRNA-seq (left). UMAP graph showing the representation of 3,153 myeloid cells derived from the 10 GBM patients (right). Each dot represents an individual cell.

Supplementary information

Reporting Summary

Supplementary Tables

Table 1. Clinical characteristics of our GBM cohort treated with anti-PD-1 therapy. Table 2. Clinical characteristics of our GBM cohort that did not receive immunotherapy. Table 3. Clinical characteristics of the validation cohort treated with adjuvant PD-1 blockade. Table 4. GO term enrichment in myeloid cells from high-p-ERK GBMs versus myeloid cells from low-p-ERK GBMs.

Supplementary Data 1

Flow cytometry gating for GBM patients treated with PD-1 blockade.

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. 3

Unprocessed immunoblots.

Source Data Extended Data Fig. 4

Unprocessed immunoblots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Arrieta, V.A., Chen, A.X., Kane, J.R. et al. ERK1/2 phosphorylation predicts survival following anti-PD-1 immunotherapy in recurrent glioblastoma. Nat Cancer 2, 1372–1386 (2021). https://doi.org/10.1038/s43018-021-00260-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00260-2

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