Skip to main content
Top
Published in: Cellular Oncology 2/2018

01-04-2018 | Report

Establishing cut-off points with clinical relevance for bcl-2, cyclin D1, p16, p21, p27, p53, Sox11 and WT1 expression in glioblastoma - a short report

Authors: Emma Camacho-Urkaray, Jorge Santos-Juanes, Francisco Borja Gutiérrez-Corres, Beatriz García, Luis M. Quirós, Isabel Guerra-Merino, José Javier Aguirre, Iván Fernández-Vega

Published in: Cellular Oncology | Issue 2/2018

Login to get access

Abstract

Purpose

Glioblastoma (GBM) ranks among the most challenging cancers to treat and there is an urgent need for clinically relevant prognostic and diagnostic biomarkers. Here, we set out to investigate the expression of eight proteins (bcl-2, cyclin D1, p16, p21, p27, p53, Sox11 and WT1) in GBM with the specific aim to establish immunohistochemistry cut-off points with clinical relevance.

Methods

Immunohistochemistry (IHC) was used to examine protein expression in 55 surgical GBM specimens using H-scores, and IHC cut-off points were established using the Cutoff Finder web platform. Protein co-expression and its correlation with histopathological features were assessed, and cases were classified according to IDH1 mutation status. Survival curves were determined using Kaplan-Meier analyses.

Results

Clinical and molecular parameters found to be correlated with overall survival (OS) were tumor size (r = −0.278; p = 0.048), p53 (r = −0.452; p = 0.001), p16 (r = 0.351; p = 0.012) and Sox11 (r = 0.324; p = 0.020). In addition, we found that tumor size correlated with cyclin D1 (r = −0.282; p = 0.037), p53 (r = 0.269; p = 0.041), Sox11 (r = −0.309; p = 0.022) and WT1 (r = −0.372; p = 0.003). Variables found to be significantly associated with IDH1 mutation status were OS (p < 0.01), age (p < 0.01), cyclin D1 (p = 0.046), p16 (p = 0.019) and Sox11 (p = 0.012). Variables found to be significantly associated with a poor survival were tumor size >5 cm (p < 0.001), bcl-2 score > 40 (p = 0.034), cyclin D1 score ≤ 70 (p = 0.004), p16 score ≤ 130 (p = 0.005), p53 score > 20 (p = 0.003), Sox11 score ≤ 40 (p < 0.001) and WT1 score ≤ 270 (p = 0.02).

Conclusions

Correlations between protein biomarkers and main clinical GBM variables were identified. The establishment of distinct biomarker cut-off points may enable clinicians and pathologists to better weigh their prognostic value.
Literature
4.
go back to reference D. Matias, J. Balca-Silva, L.G. Dubois, B. Pontes, V.P. Ferrer, L. Rosario, A. do Carmo, J. Echevarria-Lima, A.B. Sarmento-Ribeiro, M.C. Lopes, V. Moura-Neto, Dual treatment with shikonin and temozolomide reduces glioblastoma tumor growth, migration and glial-to-mesenchymal transition. Cell Oncol 40, 247–261 (2017). https://doi.org/10.1007/s13402-017-0320-1 CrossRef D. Matias, J. Balca-Silva, L.G. Dubois, B. Pontes, V.P. Ferrer, L. Rosario, A. do Carmo, J. Echevarria-Lima, A.B. Sarmento-Ribeiro, M.C. Lopes, V. Moura-Neto, Dual treatment with shikonin and temozolomide reduces glioblastoma tumor growth, migration and glial-to-mesenchymal transition. Cell Oncol 40, 247–261 (2017). https://​doi.​org/​10.​1007/​s13402-017-0320-1 CrossRef
8.
go back to reference K.E. Tagscherer, A. Fassl, B. Campos, M. Farhadi, A. Kraemer, B.C. Bock, S. Macher-Goeppinger, B. Radlwimmer, O.D. Wiestler, C. Herold-Mende, W. Roth, Apoptosis-based treatment of glioblastomas with ABT-737, a novel small molecule inhibitor of Bcl-2 family proteins. Oncogene 27, 6646–6656 (2008). https://doi.org/10.1038/onc.2008.259 CrossRefPubMed K.E. Tagscherer, A. Fassl, B. Campos, M. Farhadi, A. Kraemer, B.C. Bock, S. Macher-Goeppinger, B. Radlwimmer, O.D. Wiestler, C. Herold-Mende, W. Roth, Apoptosis-based treatment of glioblastomas with ABT-737, a novel small molecule inhibitor of Bcl-2 family proteins. Oncogene 27, 6646–6656 (2008). https://​doi.​org/​10.​1038/​onc.​2008.​259 CrossRefPubMed
10.
go back to reference J. Wang, Q. Wang, Y. Cui, Z.Y. Liu, W. Zhao, C.L. Wang, Y. Dong, L. Hou, G. Hu, C. Luo, J. Chen, Y. Lu, Knockdown of cyclin D1 inhibits proliferation, induces apoptosis, and attenuates the invasive capacity of human glioblastoma cells. J Neuro-Oncol 106, 473–484 (2012). https://doi.org/10.1007/s11060-011-0692-4 CrossRef J. Wang, Q. Wang, Y. Cui, Z.Y. Liu, W. Zhao, C.L. Wang, Y. Dong, L. Hou, G. Hu, C. Luo, J. Chen, Y. Lu, Knockdown of cyclin D1 inhibits proliferation, induces apoptosis, and attenuates the invasive capacity of human glioblastoma cells. J Neuro-Oncol 106, 473–484 (2012). https://​doi.​org/​10.​1007/​s11060-011-0692-4 CrossRef
13.
go back to reference B. Weigle, R. Ebner, A. Temme, S. Schwind, M. Schmitz, A. Kiessling, M.A. Rieger, G. Schackert, H.K. Schackert, E.P. Rieber, Highly specific overexpression of the transcription factor SOX11 in human malignant gliomas. Oncol Rep 13, 139–144 (2005)PubMed B. Weigle, R. Ebner, A. Temme, S. Schwind, M. Schmitz, A. Kiessling, M.A. Rieger, G. Schackert, H.K. Schackert, E.P. Rieber, Highly specific overexpression of the transcription factor SOX11 in human malignant gliomas. Oncol Rep 13, 139–144 (2005)PubMed
15.
go back to reference P. Korkolopoulou, G. Levidou, E.A. El-Habr, C. Adamopoulos, P. Fragkou, E. Boviatsis, M.S. Themistocleous, K. Petraki, G. Vrettakos, M. Sakalidou, V. Samaras, A. Zisakis, A. Saetta, I. Chatziandreou, E. Patsouris, C. Piperi, Sox11 expression in astrocytic gliomas: correlation with nestin/c-Met/IDH1-R132H expression phenotypes, p-Stat-3 and survival. Br J Cancer 108, 2142–2152 (2013). https://doi.org/10.1038/bjc.2013.176 CrossRefPubMedPubMedCentral P. Korkolopoulou, G. Levidou, E.A. El-Habr, C. Adamopoulos, P. Fragkou, E. Boviatsis, M.S. Themistocleous, K. Petraki, G. Vrettakos, M. Sakalidou, V. Samaras, A. Zisakis, A. Saetta, I. Chatziandreou, E. Patsouris, C. Piperi, Sox11 expression in astrocytic gliomas: correlation with nestin/c-Met/IDH1-R132H expression phenotypes, p-Stat-3 and survival. Br J Cancer 108, 2142–2152 (2013). https://​doi.​org/​10.​1038/​bjc.​2013.​176 CrossRefPubMedPubMedCentral
17.
go back to reference Y. Chiba, N. Hashimoto, A. Tsuboi, C. Rabo, Y. Oka, M. Kinoshita, N. Kagawa, Y. Oji, H. Sugiyama, T. Yoshimine, Prognostic value of WT1 protein expression level and MIB-1 staining index as predictor of response to WT1 immunotherapy in glioblastoma patients. Brain Tumor Pathol 27(1), 29–34 (2010). https://doi.org/10.1007/s10014-010-0265-9 CrossRefPubMed Y. Chiba, N. Hashimoto, A. Tsuboi, C. Rabo, Y. Oka, M. Kinoshita, N. Kagawa, Y. Oji, H. Sugiyama, T. Yoshimine, Prognostic value of WT1 protein expression level and MIB-1 staining index as predictor of response to WT1 immunotherapy in glioblastoma patients. Brain Tumor Pathol 27(1), 29–34 (2010). https://​doi.​org/​10.​1007/​s10014-010-0265-9 CrossRefPubMed
18.
20.
go back to reference F.R. Hirsch, M. Varella-Garcia, P.A. Bunn Jr., M.V. Di Maria, R. Veve, R.M. Bremmes, A.E. Baron, C. Zeng, W.A. Franklin, Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol 21, 3798–3807 (2003). https://doi.org/10.1200/JCO.2003.11.069 CrossRefPubMed F.R. Hirsch, M. Varella-Garcia, P.A. Bunn Jr., M.V. Di Maria, R. Veve, R.M. Bremmes, A.E. Baron, C. Zeng, W.A. Franklin, Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol 21, 3798–3807 (2003). https://​doi.​org/​10.​1200/​JCO.​2003.​11.​069 CrossRefPubMed
24.
25.
go back to reference R.G. Verhaak, K.A. Hoadley, E. Purdom, V. Wang, Y. Qi, M.D. Wilkerson, C.R. Miller, L. Ding, T. Golub, J.P. Mesirov, G. Alexe, M. Lawrence, M. O'Kelly, P. Tamayo, B.A. Weir, S. Gabriel, W. Winckler, S. Gupta, L. Jakkula, H.S. Feiler, J.G. Hodgson, C.D. James, J.N. Sarkaria, C. Brennan, A. Kahn, P.T. Spellman, R.K. Wilson, T.P. Speed, J.W. Gray, M. Meyerson, G. Getz, C.M. Perou, D.N. Hayes, N. Cancer Genome Atlas Research, Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17(1), 98–110 (2010). https://doi.org/10.1016/j.ccr.2009.12.020 CrossRefPubMedPubMedCentral R.G. Verhaak, K.A. Hoadley, E. Purdom, V. Wang, Y. Qi, M.D. Wilkerson, C.R. Miller, L. Ding, T. Golub, J.P. Mesirov, G. Alexe, M. Lawrence, M. O'Kelly, P. Tamayo, B.A. Weir, S. Gabriel, W. Winckler, S. Gupta, L. Jakkula, H.S. Feiler, J.G. Hodgson, C.D. James, J.N. Sarkaria, C. Brennan, A. Kahn, P.T. Spellman, R.K. Wilson, T.P. Speed, J.W. Gray, M. Meyerson, G. Getz, C.M. Perou, D.N. Hayes, N. Cancer Genome Atlas Research, Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17(1), 98–110 (2010). https://​doi.​org/​10.​1016/​j.​ccr.​2009.​12.​020 CrossRefPubMedPubMedCentral
28.
go back to reference R.J. Molenaar, D. Verbaan, S. Lamba, C. Zanon, J.W.M. Jeuken, S.H.E. Boots-Sprenger, P. Wesseling, T.J.M. Hulsebos, D. Troost, A.A. van Tilborg, S. Leenstra, W.P. Vandertop, A. Bardelli, C.J.F. van Noorden, F.E. Bleeker, The combination of IDH1 mutations and MGMT methylation status predicts survival in glioblastoma better than either IDH1 or MGMT alone. Neuro-Oncology 16, 1263–1273 (2014). https://doi.org/10.1093/neuonc/nou005 CrossRefPubMedPubMedCentral R.J. Molenaar, D. Verbaan, S. Lamba, C. Zanon, J.W.M. Jeuken, S.H.E. Boots-Sprenger, P. Wesseling, T.J.M. Hulsebos, D. Troost, A.A. van Tilborg, S. Leenstra, W.P. Vandertop, A. Bardelli, C.J.F. van Noorden, F.E. Bleeker, The combination of IDH1 mutations and MGMT methylation status predicts survival in glioblastoma better than either IDH1 or MGMT alone. Neuro-Oncology 16, 1263–1273 (2014). https://​doi.​org/​10.​1093/​neuonc/​nou005 CrossRefPubMedPubMedCentral
30.
go back to reference A.X. Ming Li, D. Floyd, I. Olmez, J. Lee, J. Godlewski, A. Bronisz, K.P.L. Bhat, E.P. Sulman, I. Nakano, B. Purow, CDK4/6 inhibition is more active against the glioblastoma proneural subtype. Oncotarget 8, 12 (2017) A.X. Ming Li, D. Floyd, I. Olmez, J. Lee, J. Godlewski, A. Bronisz, K.P.L. Bhat, E.P. Sulman, I. Nakano, B. Purow, CDK4/6 inhibition is more active against the glioblastoma proneural subtype. Oncotarget 8, 12 (2017)
33.
go back to reference H.S. Phillips, S. Kharbanda, R. Chen, W.F. Forrest, R.H. Soriano, T.D. Wu, A. Misra, J.M. Nigro, H. Colman, L. Soroceanu, P.M. Williams, Z. Modrusan, B.G. Feuerstein, K. Aldape, Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9, 157–173 H.S. Phillips, S. Kharbanda, R. Chen, W.F. Forrest, R.H. Soriano, T.D. Wu, A. Misra, J.M. Nigro, H. Colman, L. Soroceanu, P.M. Williams, Z. Modrusan, B.G. Feuerstein, K. Aldape, Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9, 157–173
35.
go back to reference S.S.C. Romagosa, L. López-Vicente, A. Mazo, M.E. Lleonart, J. Castellvi, S. Ramon y Cajal, p16 (Ink4) a overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors. Oncogene 30, 10 (2011)CrossRef S.S.C. Romagosa, L. López-Vicente, A. Mazo, M.E. Lleonart, J. Castellvi, S. Ramon y Cajal, p16 (Ink4) a overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors. Oncogene 30, 10 (2011)CrossRef
37.
go back to reference N. Kijima, N. Hosen, N. Kagawa, N. Hashimoto, M. Kinoshita, Y. Oji, H. Sugiyama, T. Yoshimine, Wilms' tumor 1 is involved in tumorigenicity of glioblastoma by regulating cell proliferation and apoptosis. Anticancer Res 34, 61–67 (2014)PubMed N. Kijima, N. Hosen, N. Kagawa, N. Hashimoto, M. Kinoshita, Y. Oji, H. Sugiyama, T. Yoshimine, Wilms' tumor 1 is involved in tumorigenicity of glioblastoma by regulating cell proliferation and apoptosis. Anticancer Res 34, 61–67 (2014)PubMed
38.
go back to reference S. Izumoto, A. Tsuboi, Y. Oka, T. Suzuki, T. Hashiba, N. Kagawa, N. Hashimoto, M. Maruno, O.A. Elisseeva, T. Shirakata, M. Kawakami, Y. Oji, S. Nishida, S. Ohno, I. Kawase, J. Hatazawa, S. Nakatsuka, K. Aozasa, S. Morita, J. Sakamoto, H. Sugiyama, T. Yoshimine, Phase II clinical trial of Wilms tumor 1 peptide vaccination for patients with recurrent glioblastoma multiforme, J Neurosurg 108(5), 963–971 (2008). https://doi.org/10.3171/JNS/2008/108/5/0963 CrossRefPubMed S. Izumoto, A. Tsuboi, Y. Oka, T. Suzuki, T. Hashiba, N. Kagawa, N. Hashimoto, M. Maruno, O.A. Elisseeva, T. Shirakata, M. Kawakami, Y. Oji, S. Nishida, S. Ohno, I. Kawase, J. Hatazawa, S. Nakatsuka, K. Aozasa, S. Morita, J. Sakamoto, H. Sugiyama, T. Yoshimine, Phase II clinical trial of Wilms tumor 1 peptide vaccination for patients with recurrent glioblastoma multiforme, J Neurosurg 108(5), 963–971 (2008). https://​doi.​org/​10.​3171/​JNS/​2008/​108/​5/​0963 CrossRefPubMed
Metadata
Title
Establishing cut-off points with clinical relevance for bcl-2, cyclin D1, p16, p21, p27, p53, Sox11 and WT1 expression in glioblastoma - a short report
Authors
Emma Camacho-Urkaray
Jorge Santos-Juanes
Francisco Borja Gutiérrez-Corres
Beatriz García
Luis M. Quirós
Isabel Guerra-Merino
José Javier Aguirre
Iván Fernández-Vega
Publication date
01-04-2018
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 2/2018
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-017-0362-4

Other articles of this Issue 2/2018

Cellular Oncology 2/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine