Skip to main content
Log in

Identification of the immune gene expression signature associated with recurrence of high-grade gliomas

  • Original Article
  • Published:
Journal of Molecular Medicine Aims and scope Submit manuscript

Abstract

High-grade gliomas (HGGs), the most common and aggressive primary brain tumors in adults, inevitably recur due to incomplete surgery or resistance to therapy. Intratumoral genomic and cellular heterogeneity of HGGs contributes to therapeutic resistance, recurrence, and poor clinical outcomes. Transcriptomic profiles of HGGs at recurrence have not been investigated in detail. Using targeted sequencing of cancer-related genes and transcriptomics, we identified single nucleotide variations, small insertions and deletions, copy number aberrations (CNAs), as well as gene expression changes and pathway deregulation in 16 pairs of primary and recurrent HGGs. Most of the somatic mutations identified in primary HGGs were not detected after relapse, suggesting a subclone substitution during the tumor progression. We found a novel frameshift insertion in the ZNF384 gene which may contribute to extracellular matrix remodeling. An inverse correlation of focal CNAs in EGFR and PTEN genes was detected. Transcriptomic analysis revealed downregulation of genes involved in messenger RNA splicing, cell cycle, and DNA repair, while genes related to interferon signaling and phosphatidylinositol (PI) metabolism are upregulated in secondary HGGs when compared to primary HGGs. In silico analysis of the tumor microenvironment identified M2 macrophages and immature dendritic cells as enriched in recurrent HGGs, suggesting a prominent immunosuppressive signature. Accumulation of those cells in recurrent HGGs was validated by immunostaining. Our findings point to a substantial transcriptomic deregulation and a pronounced infiltration of immature dendritic cells in recurrent HGG, which may impact the effectiveness of frontline immunotherapies in the GBM management.

Key messages

  • Most of the somatic mutations identified in primary HGGs were not detected after relapse.

  • Focal CNAs in EGFR and PTEN genes are inversely correlated in primary and recurrent HGGs.

  • Transcriptomic changes and distinct immune-related signatures characterize HGG recurrence.

  • Recurrent HGGs are characterized by a prominent infiltration of immature dendritic and M2 macrophages.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Data availability

The data that support the findings of this study are openly available at the European Genome-phenome Archive (EGA), reference number EGAS00001004606.

References

  1. Touat M, Idbaih A, Sanson M, Ligon KL (2017) Glioblastoma targeted therapy: updated approaches from recent biological insights. Ann Oncol 28:1457–1472

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Verhaak RGW, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP et al (2010) Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17:98–110

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Brennan CW, Verhaak RGW, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SH et al (2013) The somatic genomic landscape of glioblastoma. Cell 155:462–477

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Sturm D, Bender S, Jones DTW, Lichter P, Grill J, Becher O, Hawkins C, Majewski J, Jones C, Costello JF et al (2014) Paediatric and adult glioblastoma: multiform (epi)genomic culprits emerge. Nat Rev Cancer 14:92–107

    CAS  PubMed  PubMed Central  Google Scholar 

  5. An Z, Aksoy O, Zheng T, Fan Q-W, Weiss WA (2018) Epidermal growth factor receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies. Oncogene 37:1561–1575

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kim H, Zheng S, Amini SS, Virk SM, Mikkelsen T, Brat DJ, Grimsby J, Sougnez C, Muller F, Hu J et al (2015) Whole-genome and multisector exome sequencing of primary and post-treatment glioblastoma reveals patterns of tumor evolution. Genome Res 25:316–327

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, Misra A, Nigro JM, Colman H, Soroceanu L et al (2006) Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9:157–173

    CAS  PubMed  Google Scholar 

  8. Ceccarelli M, Barthel FP, Malta TM, Sabedot TS, Salama SR, Murray BA, Morozova O, Newton Y, Radenbaugh A, Pagnotta SM et al (2016) Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164:550–563

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Perng P, Lim M (2015) Immunosuppressive mechanisms of malignant gliomas: parallels at non-CNS sites. Front Oncol 6:5

    Google Scholar 

  10. Ellert-Miklaszewska A, Dabrowski M, Lipko M, Sliwa M, Maleszewska M, Kaminska B (2013) Molecular definition of the pro-tumorigenic phenotype of glioma-activated microglia. Glia. 61:1178–1190

    PubMed  Google Scholar 

  11. Barthel FP, Johnson KC, Varn FS, Moskalik AD, Tanner G, Kocakavuk E, Anderson KJ, Abiola O, Aldape K, Alfaro KD et al (2019) Longitudinal molecular trajectories of diffuse glioma in adults. Nature 576:112–120

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Gabrusiewicz K, Ellert-Miklaszewska A, Lipko M, Sielska M, Frankowska M, Kaminska B (2011) Characteristics of the alternative phenotype of microglia/macrophages and its modulation in experimental gliomas. PLoS One 6:e23902

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Bolger AM, Lohse M, Usadel B (2014) Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30:2114–2120

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Sedlazeck FJ, Rescheneder P, von Haeseler A (2013) NextGenMap: fast and accurate read mapping in highly polymorphic genomes. Bioinformatics 29:2790–2791

    CAS  PubMed  Google Scholar 

  15. (2019) Picard Tools - By Broad Institute. Github.Io. broadinstitute.github.io/picard/

  16. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, Miller CA, Mardis ER, Ding L, Wilson RK (2012) VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res 22:568–576

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang K, Li M, Hakonarson H (2010) ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res 38:e164–e164

    PubMed  PubMed Central  Google Scholar 

  18. Tamborero D, Gonzalez-Perez A, Lopez-Bigas N (2013) OncodriveCLUST: exploiting the positional clustering of somatic mutations to identify cancer genes. Bioinformatics 29:2238–2244

    CAS  PubMed  Google Scholar 

  19. Wang L, Wang S, Li W (2012) RSeQC: quality control of RNA-seq experiments. Bioinformatics 28:2184–2185

    CAS  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  21. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR (2012) STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29:15–21

    PubMed  PubMed Central  Google Scholar 

  22. Love MI, Huber W, Anders S (2014) Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biology 15:550

    PubMed  PubMed Central  Google Scholar 

  23. Aran D, Hu Z, Butte AJ (2017) xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol 18:220

  24. Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, Selves J, Laurent-Puig P, Sautès-Fridman C, Fridman WH, et al (2016) Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol 17:218

  25. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M, Alizadeh AA (2015) Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 12(5):453–457

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Finotello F, Mayer C, Plattner C, Laschober G, Rieder D, Hackl H, Krogsdam A, Loncova Z, Posch W, Wilflingseder D, et al (2019) Molecular and pharmacological modulators of the tumor immune contexture revealed by deconvolution of RNA-seq data. Genome Med 11:34

  27. Li B, Severson E, Pignon J-C, Zhao H, Li T, Novak J, Jiang P, Shen H, Aster JC, Rodig S, et al (2016) Comprehensive analyses of tumor immunity: implications for cancer immunotherapy. Genome Biol 17:174

  28. Körber V, Yang J, Barah P, Wu Y, Stichel D, Gu Z, Fletcher MNC, Jones D, Hentschel B, Lamszus K et al (2019) Evolutionary trajectories of IDHWT glioblastomas reveal a common path of early tumorigenesis instigated years ahead of initial diagnosis. Cancer Cell 35:692–704.e12

    PubMed  Google Scholar 

  29. Liu A, Hou C, Chen H, Zong X, Zong P (2016) Genetics and epigenetics of glioblastoma: applications and overall incidence of IDH1 mutation. Front Oncol 6:16

  30. Wang J, Cazzato E, Ladewig E, Frattini V, Rosenbloom DIS, Zairis S, Abate F, Liu Z, Elliott O, Shin Y-J et al (2016) Clonal evolution of glioblastoma under therapy. Nat Genet 48:768–776

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Gocho Y, Kiyokawa N, Ichikawa H, Nakabayashi K, Osumi T, Ishibashi T, Ueno H, Terada K, Oboki K, Sakamoto H et al (2015) A novel recurrent EP300–ZNF384 gene fusion in B-cell precursor acute lymphoblastic leukemia. Leukemia 29:2445–2448

    CAS  PubMed  Google Scholar 

  32. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SAJR, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Børresen-Dale A-L et al (2013) Signatures of mutational processes in human cancer. Nature 500:415–421

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Guo C, McDowell IC, Nodzenski M, Scholtens DM, Allen AS, Lowe WL, Reddy TE (2017) Transversions have larger regulatory effects than transitions. BMC Genomics 18:394

    PubMed  PubMed Central  Google Scholar 

  34. van Alphen RJ, Wiemer EAC, Burger H, Eskens FALM (2008) The spliceosome as target for anticancer treatment. Br J Cancer 100:228–232

    PubMed  PubMed Central  Google Scholar 

  35. Szulzewsky F, Pelz A, Feng X, Synowitz M, Markovic D, Langmann T, Holtman IR, Wang X, Eggen BJL, Boddeke HWGM et al (2015) Glioma-associated microglia/macrophages display an expression profile different from M1 and M2 polarization and highly express Gpnmb and Spp1. Harrison JK, editor. PLoS One 10:e0116644

    PubMed  PubMed Central  Google Scholar 

  36. Barthel FP, Johnson KC, Varn FS, Moskalik AD, Tanner G, Kocakavuk E, Anderson KJ, Abiola O, Aldape K, Alfaro KD et al (2019) Longitudinal molecular trajectories of diffuse glioma in adults. Nature 576(7785):112–120

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hirabayashi S, Ohki K, Nakabayashi K, Ichikawa H, Momozawa Y, Okamura K, Yaguchi A, Terada K, Saito Y, Yoshimi A et al (2016) ZNF384-related fusion genes define a subgroup of childhood B-cell precursor acute lymphoblastic leukemia with a characteristic immunotype. Haematologica 102:118–129

    PubMed  Google Scholar 

  38. Childress P, Stayrook KR, Alvarez MB, Wang Z, Shao Y, Hernandez-Buquer S, Mack JK, Grese ZR, He Y, Horan D et al (2015) Genome-wide mapping and interrogation of the Nmp4 antianabolic bone axis. Mol Endocrinol 29:1269–1285

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Young SK, Shao Y, Bidwell JP, Wek RC (2016) Nuclear matrix protein 4 is a novel regulator of ribosome biogenesis and controls the unfolded protein response via repression of Gadd34 expression. J Biol Chem 291:13780–13788

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Danan-Gotthold M, Golan-Gerstl R, Eisenberg E, Meir K, Karni R, Levanon EY (2015) Identification of recurrent regulated alternative splicing events across human solid tumors. Nucleic Acids Res 43:5130–5144

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Yang CH, Wang Y, Sims M, Cai C, He P, Häcker H, Yue J, Cheng J, Boop FA, Pfeffer LM (2017) MicroRNA203a suppresses glioma tumorigenesis through an ATM-dependent interferon response pathway. Oncotarget 8:112980–112991

    PubMed  PubMed Central  Google Scholar 

  42. Tanabe T, Kominsky SL, Subramaniam PS, Johnson HM (2000) Torres BA. J. Neuro-Oncol 48:225–232

    CAS  Google Scholar 

  43. Silginer M, Nagy S, Happold C, Schneider H, Weller M, Roth P (2017) Autocrine activation of the IFN signaling pathway may promote immune escape in glioblastoma. Neuro-Oncol 19:1338–1349

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Gardner A, Ruffell B (2016) Dendritic cells and cancer immunity. Trends Immunol 37:855–865

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kim R, Emi M, Tanabe K (2006) Functional roles of immature dendritic cells in impaired immunity of solid tumour and their targeted strategies for provoking tumour immunity. Clin Exp Immunol 146:189–196

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Ma Y, Shurin GV, Peiyuan Z, Shurin MR (2013) Dendritic cells in the cancer microenvironment. J Cancer 4:36–44

    CAS  PubMed  Google Scholar 

  47. Tran Janco JM, Lamichhane P, Karyampudi L, Knutson KL (2015) Tumor-infiltrating dendritic cells in cancer pathogenesis. J Immunol 194:2985–2991

    PubMed  Google Scholar 

  48. Aras S, Zaidi MR (2017) TAMeless traitors: macrophages in cancer progression and metastasis. Br J Cancer 117:1583–1591

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We would like to thank the physicians who performed the surgeries, the patients for their consent for the use of their biological material for this research, and Dr. Chinchu Jayaprakash for improving the language quality of this manuscript.

Code availability

Codes generated during the current study are available upon reasonable request.

Funding

Studies were supported by the Foundation for Polish Science TEAM-TECH Core Facility project “NGS platform for comprehensive diagnostics and personalized therapy in neuro-oncology.” The use of CePT infrastructure, financed by the European Union, The European Regional Development Fund within the Operational Programme “Innovative economy” for 2007–2013, is highly appreciated.

Author information

Authors and Affiliations

Authors

Contributions

Design of the study, data analysis, data interpretation, and manuscript preparation were performed by Adria-Jaume Roura. RNA/DNA isolation, sequencing, and clinical table preparation were performed by Bartlomiej Gielniewski and Paulina Pilanc. Immunohistochemistry was carried out by Paulina Pilanc. Experimental design and preparation of the materials for experiments were performed by Marta Maleszweska and Sylwia K. Krol. Preparation of the clinical samples and collection of clinical information were performed by Ryszard Czepko and Wojciech Kaspera. Study design, data interpretation, and manuscript preparation were performed by Bartosz Wojtas and Bozena Kaminska. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Bartosz Wojtas.

Ethics declarations

Conflict of interest

The authors declare that they have no conflicts of interest.

Ethical approval

This study was approved by the Bioethics Committees of St. Raphael Hospital, Andrzej Frycz Modrzewski Krakow University, Krakow, Poland (Nr. 73/KBL/OIL/2015); Medical University of Silesia, Sosnowiec, Poland; and Mazovian Brodno Hospital, Warsaw, Poland (Nr. KNW/0022/KB1/46/I/16), and have therefore been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki and its later amendments.

Consent to participate

All persons gave their informed consent prior to their inclusion in the study. Details that might disclose the identity of the subjects under study have been omitted.

Consent for publication

Not applicable.

Additional information

Publisher’s note

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

Supplementary Information

Supplementary Figure S1

High-throughput sequencing (HTS) pipeline, genetic alterations and ZNF384 survival analysis. (A) Representation of the HTS pipeline implemented in this study. (B) Oncoplot showing recurrent somatic mutations. Each column represents a sample and each row a different gene; genes are ordered by the frequency of occurrence. (C) Mutational hot-spot and its effect on the ZNF384 protein structure, where domain and aminoacidic change are indicated. (D) Kaplan-Meier survival analysis between low- and high- expressed ZNF384 in High-grade glioma (TCGA-GBM/LGG data). Dashed lines represent the survival median line for each of the groups and Log Rank Test was utilized (PDF 665 kb)

Supplementary Figure S2

Identification of cancer driver genes based on spatial clustering (A) Potential cancer driver genes in primary and (B) recurrent cohorts, respectively, based on oncodriveCLUST algorithm. Numbers enclosed in square brackets represent the number of clusters found in the gene, and dots in red correspond to statistically significant clusters (PDF 632 kb)

Supplementary Figure S3

Focal copy number changes in EGFR and PTEN genes. Somatic copy number changes from matched tumor-normal pairs showing relative changes in the tumor samples. Each dot corresponds to the adjusted copynumber median of EGFR and PTEN, which were calculated for each of the primary (A) and recurrent (B) HGGs (PDF 85 kb)

Supplementary Figure S4

Functional enrichment obtained using Reactome database. Visualization of the relationship among (A) up-regulated and down-regulated (B) enriched gene sets that are differentially expressed between recurrent and primary HGGs (PDF 305 kb)

Supplementary Figure S5

Functional enrichment obtained using KEGG database. Visualization of the relationship among (A) up-regulated and down-regulated (B) enriched gene sets among differentially expressed between recurrent and primary HGGs (PDF 158 kb)

Supplementary Figure S6

In-silico validation of xCell cell enrichment analysis obtained by xCell. Cell type enrichment analysis from normalized gene expression using (A) CIBERSORT, (B) QuantiSEQ, (C) mcp-COUNTER and (D) TIMER approaches. Scatter- and density plots are shown for each of the analysis (A-D). Top density plots show the cell enrichment score distribution of x-axis while right density plots show the distribution in the y-axis. Selected and significant signatures are presented based on P-values, which were calculated using Wilcoxon signed-rank test (PDF 133 kb)

ESM 7

(XLSX 234 kb)

ESM 8

(XLSX 26 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roura, AJ., Gielniewski, B., Pilanc, P. et al. Identification of the immune gene expression signature associated with recurrence of high-grade gliomas. J Mol Med 99, 241–255 (2021). https://doi.org/10.1007/s00109-020-02005-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00109-020-02005-7

Keywords

Navigation