Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2017

Open Access 01-12-2017 | Research

Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma

Authors: Jin Wang, Xiaoyang Liu, Yongzhi Hong, Songtao Wang, Pin Chen, Aihua Gu, Xiaoyuan Guo, Peng Zhao

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2017

Login to get access

Abstract

Background

Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Ibrutinib, a Bruton’s tyrosine kinase (BTK) inhibitor, is a novel anticancer drug used for treating several types of cancers. In this study, we aimed to determine the role of ibrutinib on GBM.

Methods

Cell proliferation was determined by using cell viability, colony formation, and 5-ethynyl-2′-deoxyuridine (EdU) assays. Cell cycle and cell apoptosis were analyzed by flow cytometry. Cell migratory ability was evaluated by wound healing assays and trans-well migration assays. ATG7 expression was knocked-down by transfection with Atg7-specific small interfering RNA. Overexpression of active Akt protein was achieved by transfecting the cells with a plasmid expressing constitutively active Akt (CA-Akt). Transmission electron microscopy was performed to examine the formation of autophagosomes in cells. Immunofluorescence and western blot analyses were used to analyze protein expression. Tumor xenografts in nude mice and immunohistochemistry were performed to evaluate the effect of ibrutinib on tumor growth in vivo.

Results

Ibrutinib inhibited cellular proliferation and migration, and induced apoptosis and autophagy in LN229 and U87 cells. Overexpression of the active Akt protein decreased ibrutinib-induced autophagy, while inhibiting Akt by LY294002 treatment enhanced ibrutinib-induced autophagy. Specific inhibition of autophagy by 3-methyladenine (3MA) or Atg7 targeting with small interfering RNA (si-Atg7) enhanced the anti-GBM effect of ibrutinib in vitro and in vivo.

Conclusions

Our results indicate that ibrutinib exerts a profound antitumor effect and induces autophagy through Akt/mTOR signaling pathway in GBM cells. Autophagy inhibition promotes the antitumor activity of ibrutinib in GBM. Our findings provide important insights into the action of an anticancer agent combining with autophagy inhibitor for malignant glioma.
Appendix
Available only for authorised users
Literature
2.
3.
go back to reference Auffinger B, Spencer D, Pytel P, Ahmed AU, Lesniak MS. The role of glioma stem cells in chemotherapy resistance and glioblastoma multiforme recurrence. Expert Rev Neurother. 2015;15(7):741–52.CrossRefPubMedPubMedCentral Auffinger B, Spencer D, Pytel P, Ahmed AU, Lesniak MS. The role of glioma stem cells in chemotherapy resistance and glioblastoma multiforme recurrence. Expert Rev Neurother. 2015;15(7):741–52.CrossRefPubMedPubMedCentral
4.
go back to reference Eramo A, Ricci-Vitiani L, Zeuner A, Pallini R, Lotti F, Sette G, Pilozzi E, Larocca LM, Peschle C, De Maria R. Chemotherapy resistance of glioblastoma stem cells. Cell Death Differ. 2006;13(7):1238–41.CrossRefPubMed Eramo A, Ricci-Vitiani L, Zeuner A, Pallini R, Lotti F, Sette G, Pilozzi E, Larocca LM, Peschle C, De Maria R. Chemotherapy resistance of glioblastoma stem cells. Cell Death Differ. 2006;13(7):1238–41.CrossRefPubMed
5.
go back to reference Cohen MS, Zhang C, Shokat KM, Taunton J. Structural bioinformatics-based design of selective, irreversible kinase inhibitors. Science. 2005;308(5726):1318–21.CrossRefPubMedPubMedCentral Cohen MS, Zhang C, Shokat KM, Taunton J. Structural bioinformatics-based design of selective, irreversible kinase inhibitors. Science. 2005;308(5726):1318–21.CrossRefPubMedPubMedCentral
6.
go back to reference Leproult E, Barluenga S, Moras D, Wurtz JM, Winssinger N. Cysteine mapping in conformationally distinct kinase nucleotide binding sites: application to the design of selective covalent inhibitors. J Med Chem. 2011;54(5):1347–55.CrossRefPubMed Leproult E, Barluenga S, Moras D, Wurtz JM, Winssinger N. Cysteine mapping in conformationally distinct kinase nucleotide binding sites: application to the design of selective covalent inhibitors. J Med Chem. 2011;54(5):1347–55.CrossRefPubMed
8.
go back to reference Herman SE, Mustafa RZ, Gyamfi JA, Pittaluga S, Chang S, Chang B, Farooqui M, Wiestner A. Ibrutinib inhibits BCR and NF-kappaB signaling and reduces tumor proliferation in tissue-resident cells of patients with CLL. Blood. 2014;123(21):3286–95.CrossRefPubMedPubMedCentral Herman SE, Mustafa RZ, Gyamfi JA, Pittaluga S, Chang S, Chang B, Farooqui M, Wiestner A. Ibrutinib inhibits BCR and NF-kappaB signaling and reduces tumor proliferation in tissue-resident cells of patients with CLL. Blood. 2014;123(21):3286–95.CrossRefPubMedPubMedCentral
9.
go back to reference Cheng S, Ma J, Guo A, Lu P, Leonard JP, Coleman M, Liu M, Buggy JJ, Furman RR, Wang YL. BTK inhibition targets in vivo CLL proliferation through its effects on B-cell receptor signaling activity. Leukemia. 2014;28(3):649–57.CrossRefPubMed Cheng S, Ma J, Guo A, Lu P, Leonard JP, Coleman M, Liu M, Buggy JJ, Furman RR, Wang YL. BTK inhibition targets in vivo CLL proliferation through its effects on B-cell receptor signaling activity. Leukemia. 2014;28(3):649–57.CrossRefPubMed
10.
go back to reference Saleh LM, Wang W, Herman SE, Saba NS, Anastas V, Barber E, Corrigan-Cummins M, Farooqui M, Sun C, Sarasua SM, et al. Ibrutinib downregulates a subset of miRNA leading to upregulation of tumor suppressors and inhibition of cell proliferation in chronic lymphocytic leukemia. Leukemia. 2016;31(2):340–9.CrossRefPubMed Saleh LM, Wang W, Herman SE, Saba NS, Anastas V, Barber E, Corrigan-Cummins M, Farooqui M, Sun C, Sarasua SM, et al. Ibrutinib downregulates a subset of miRNA leading to upregulation of tumor suppressors and inhibition of cell proliferation in chronic lymphocytic leukemia. Leukemia. 2016;31(2):340–9.CrossRefPubMed
11.
go back to reference Grabinski N, Ewald F. Ibrutinib (ImbruvicaTM) potently inhibits ErbB receptor phosphorylation and cell viability of ErbB2-positive breast cancer cells. Invest New Drugs. 2014;32(6):1096–104.CrossRefPubMed Grabinski N, Ewald F. Ibrutinib (ImbruvicaTM) potently inhibits ErbB receptor phosphorylation and cell viability of ErbB2-positive breast cancer cells. Invest New Drugs. 2014;32(6):1096–104.CrossRefPubMed
12.
go back to reference Zucha MA, Wu AT, Lee WH, Wang LS, Lin WW, Yuan CC, Yeh CT. Bruton’s tyrosine kinase (Btk) inhibitor ibrutinib suppresses stem-like traits in ovarian cancer. OncOtarget. 2015;6(15):13255–68.CrossRefPubMedPubMedCentral Zucha MA, Wu AT, Lee WH, Wang LS, Lin WW, Yuan CC, Yeh CT. Bruton’s tyrosine kinase (Btk) inhibitor ibrutinib suppresses stem-like traits in ovarian cancer. OncOtarget. 2015;6(15):13255–68.CrossRefPubMedPubMedCentral
13.
go back to reference Wang JD, Chen XY, Ji KW, Tao F. Targeting Btk with ibrutinib inhibit gastric carcinoma cells growth. AM J Transl Res. 2016;8(7):3003–12.PubMedPubMedCentral Wang JD, Chen XY, Ji KW, Tao F. Targeting Btk with ibrutinib inhibit gastric carcinoma cells growth. AM J Transl Res. 2016;8(7):3003–12.PubMedPubMedCentral
14.
go back to reference Wei L, Su YK, Lin CM, Chao TY, Huang SP, Huynh TT, Jan HJ, Whang-Peng J, Chiou JF, Wu AT, et al. Preclinical investigation of ibrutinib, a Bruton’s kinase tyrosine (Btk) inhibitor, in suppressing glioma tumorigenesis and stem cell phenotypes. Oncotarget. 2016;7(43):69961–75.PubMedPubMedCentral Wei L, Su YK, Lin CM, Chao TY, Huang SP, Huynh TT, Jan HJ, Whang-Peng J, Chiou JF, Wu AT, et al. Preclinical investigation of ibrutinib, a Bruton’s kinase tyrosine (Btk) inhibitor, in suppressing glioma tumorigenesis and stem cell phenotypes. Oncotarget. 2016;7(43):69961–75.PubMedPubMedCentral
15.
17.
19.
20.
go back to reference Pap M, Cooper GM. Role of glycogen synthase kinase-3 in the phosphatidylinositol 3-Kinase/Akt cell survival pathway. J Biol Chem. 1998;273(32):19929–32.CrossRefPubMed Pap M, Cooper GM. Role of glycogen synthase kinase-3 in the phosphatidylinositol 3-Kinase/Akt cell survival pathway. J Biol Chem. 1998;273(32):19929–32.CrossRefPubMed
21.
go back to reference Xie Z, Klionsky DJ. Autophagosome formation: core machinery and adaptations. Nat Cell Biol. 2007;9(10):1102–9.CrossRefPubMed Xie Z, Klionsky DJ. Autophagosome formation: core machinery and adaptations. Nat Cell Biol. 2007;9(10):1102–9.CrossRefPubMed
22.
go back to reference Shi J, Wang H, Guan H, Shi S, Li Y, Wu X, Li N, Yang C, Bai X, Cai W, et al. IL10 inhibits starvation-induced autophagy in hypertrophic scar fibroblasts via cross talk between the IL10-IL10R-STAT3 and IL10-AKT-mTOR pathways. Cell Death Dis. 2016;7:e2133.CrossRefPubMedPubMedCentral Shi J, Wang H, Guan H, Shi S, Li Y, Wu X, Li N, Yang C, Bai X, Cai W, et al. IL10 inhibits starvation-induced autophagy in hypertrophic scar fibroblasts via cross talk between the IL10-IL10R-STAT3 and IL10-AKT-mTOR pathways. Cell Death Dis. 2016;7:e2133.CrossRefPubMedPubMedCentral
23.
go back to reference Livesey KM, Tang D, Zeh HJ, Lotze MT. Autophagy inhibition in combination cancer treatment. Curr Opin Investig Drugs. 2009;10(12):1269–79.PubMed Livesey KM, Tang D, Zeh HJ, Lotze MT. Autophagy inhibition in combination cancer treatment. Curr Opin Investig Drugs. 2009;10(12):1269–79.PubMed
24.
go back to reference Vazquez-Martin A, Oliveras-Ferraros C, Menendez JA. Autophagy facilitates the development of breast cancer resistance to the anti-HER2 monoclonal antibody trastuzumab. Plos One. 2009;4(7):e6251.CrossRefPubMedPubMedCentral Vazquez-Martin A, Oliveras-Ferraros C, Menendez JA. Autophagy facilitates the development of breast cancer resistance to the anti-HER2 monoclonal antibody trastuzumab. Plos One. 2009;4(7):e6251.CrossRefPubMedPubMedCentral
25.
go back to reference Dalby KN, Tekedereli I, Lopez-Berestein G, Ozpolat B. Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer. Autophagy. 2010;6(3):322–9.CrossRefPubMedPubMedCentral Dalby KN, Tekedereli I, Lopez-Berestein G, Ozpolat B. Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer. Autophagy. 2010;6(3):322–9.CrossRefPubMedPubMedCentral
26.
go back to reference Eisenberg-Lerner A, Bialik S, Simon HU, Kimchi A. Life and death partners: apoptosis, autophagy and the cross-talk between them. Cell Death Differ. 2009;16(7):966–75.CrossRefPubMed Eisenberg-Lerner A, Bialik S, Simon HU, Kimchi A. Life and death partners: apoptosis, autophagy and the cross-talk between them. Cell Death Differ. 2009;16(7):966–75.CrossRefPubMed
27.
go back to reference Denton D, Nicolson S, Kumar S. Cell death by autophagy: facts and apparent artefacts. Cell Death Differ. 2012;19(1):87–95.CrossRefPubMed Denton D, Nicolson S, Kumar S. Cell death by autophagy: facts and apparent artefacts. Cell Death Differ. 2012;19(1):87–95.CrossRefPubMed
29.
go back to reference Shimizu S, Kanaseki T, Mizushima N, Mizuta T, Arakawa-Kobayashi S, Thompson CB, Tsujimoto Y. Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes. Nat Cell Biol. 2004;6(12):1221–8.CrossRefPubMed Shimizu S, Kanaseki T, Mizushima N, Mizuta T, Arakawa-Kobayashi S, Thompson CB, Tsujimoto Y. Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes. Nat Cell Biol. 2004;6(12):1221–8.CrossRefPubMed
30.
go back to reference Yu L, Alva A, Su H, Dutt P, Freundt E, Welsh S, Baehrecke EH, Lenardo MJ. Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8. Science. 2004;304(5676):1500–2.CrossRefPubMed Yu L, Alva A, Su H, Dutt P, Freundt E, Welsh S, Baehrecke EH, Lenardo MJ. Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8. Science. 2004;304(5676):1500–2.CrossRefPubMed
31.
go back to reference Sharma V, Joseph C, Ghosh S, Agarwal A, Mishra MK, Sen E. Kaempferol induces apoptosis in glioblastoma cells through oxidative stress. Mol Cancer Ther. 2007;6(9):2544–53.CrossRefPubMed Sharma V, Joseph C, Ghosh S, Agarwal A, Mishra MK, Sen E. Kaempferol induces apoptosis in glioblastoma cells through oxidative stress. Mol Cancer Ther. 2007;6(9):2544–53.CrossRefPubMed
32.
go back to reference Sharma V, Tewari R, Sk UH, Joseph C, Sen E. Ebselen sensitizes glioblastoma cells to Tumor Necrosis Factor (TNFalpha)-induced apoptosis through two distinct pathways involving NF-kappaB downregulation and Fas-mediated formation of death inducing signaling complex. Int J Cancer. 2008;123(9):2204–12.CrossRefPubMed Sharma V, Tewari R, Sk UH, Joseph C, Sen E. Ebselen sensitizes glioblastoma cells to Tumor Necrosis Factor (TNFalpha)-induced apoptosis through two distinct pathways involving NF-kappaB downregulation and Fas-mediated formation of death inducing signaling complex. Int J Cancer. 2008;123(9):2204–12.CrossRefPubMed
34.
go back to reference Solca FF, Baum A, Langkopf E, Dahmann G, Heider KH, Himmelsbach F, van Meel JC. Inhibition of epidermal growth factor receptor activity by two pyrimidopyrimidine derivatives. J Pharmacol Exp Ther. 2004;311(2):502–9.CrossRefPubMed Solca FF, Baum A, Langkopf E, Dahmann G, Heider KH, Himmelsbach F, van Meel JC. Inhibition of epidermal growth factor receptor activity by two pyrimidopyrimidine derivatives. J Pharmacol Exp Ther. 2004;311(2):502–9.CrossRefPubMed
35.
go back to reference Aoki H, Kondo Y, Aldape K, Yamamoto A, Iwado E, Yokoyama T, Hollingsworth EF, Kobayashi R, Hess K, Shinojima N, et al. Monitoring autophagy in glioblastoma with antibody against isoform B of human microtubule-associated protein 1 light chain 3. Autophagy. 2008;4(4):467–75.CrossRefPubMed Aoki H, Kondo Y, Aldape K, Yamamoto A, Iwado E, Yokoyama T, Hollingsworth EF, Kobayashi R, Hess K, Shinojima N, et al. Monitoring autophagy in glioblastoma with antibody against isoform B of human microtubule-associated protein 1 light chain 3. Autophagy. 2008;4(4):467–75.CrossRefPubMed
36.
go back to reference Lippai M, Szatmar Z. Autophagy-from molecular mechanisms to clinical relevance. Cell Biol Toxicol. 2016;33(2):145–68.CrossRefPubMed Lippai M, Szatmar Z. Autophagy-from molecular mechanisms to clinical relevance. Cell Biol Toxicol. 2016;33(2):145–68.CrossRefPubMed
37.
go back to reference Galluzzi L, Pietrocola f, Bravo-San Pedro JM, Amaravadi RK, Baehrecke EH, Cecconi F, Codogno P, Debnath J, Gewirtz DA, Karantza V, et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015;34(7):856–80.CrossRefPubMedPubMedCentral Galluzzi L, Pietrocola f, Bravo-San Pedro JM, Amaravadi RK, Baehrecke EH, Cecconi F, Codogno P, Debnath J, Gewirtz DA, Karantza V, et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015;34(7):856–80.CrossRefPubMedPubMedCentral
38.
go back to reference Huang C, Lin MZ, Cheng D, Braet F, Pollock CA, Chen XM. KCa3.1 mediates dysfunction of tubular autophagy in diabetic kidneys via PI3k/Akt/mTOR signaling pathways. Sci Rep. 2016;6:23884.CrossRefPubMedPubMedCentral Huang C, Lin MZ, Cheng D, Braet F, Pollock CA, Chen XM. KCa3.1 mediates dysfunction of tubular autophagy in diabetic kidneys via PI3k/Akt/mTOR signaling pathways. Sci Rep. 2016;6:23884.CrossRefPubMedPubMedCentral
40.
go back to reference Gao W, Wang M, Wang L, Lu H, Wu S, Dai B, Ou Z, Zhang L, Heymach JV, Gold KA, et al. Selective antitumor activity of ibrutinib in EGFR-mutant non-small cell lung cancer cells. J Natl Cancer Inst. 2014;106(9):dju204.CrossRefPubMedPubMedCentral Gao W, Wang M, Wang L, Lu H, Wu S, Dai B, Ou Z, Zhang L, Heymach JV, Gold KA, et al. Selective antitumor activity of ibrutinib in EGFR-mutant non-small cell lung cancer cells. J Natl Cancer Inst. 2014;106(9):dju204.CrossRefPubMedPubMedCentral
41.
go back to reference Fan QW, Cheng C, Hackett C, Feldman M, Houseman BT, Nicolaides T, Haas-Kogan D, James CD, Oakes SA, Debnath J, et al. Akt and autophagy cooperate to promote survival of drug-resistant glioma. Sci Signal. 2010;3(147):ra81.CrossRefPubMedPubMedCentral Fan QW, Cheng C, Hackett C, Feldman M, Houseman BT, Nicolaides T, Haas-Kogan D, James CD, Oakes SA, Debnath J, et al. Akt and autophagy cooperate to promote survival of drug-resistant glioma. Sci Signal. 2010;3(147):ra81.CrossRefPubMedPubMedCentral
42.
go back to reference Degtyarev M, De Maziere A, Orr C, Lin J, Lee BB, Tien JY, Prior WW, van Dijk S, Wu H, Gray DC, et al. Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents. J Cell Biol. 2008;183(1):101–16.CrossRefPubMedPubMedCentral Degtyarev M, De Maziere A, Orr C, Lin J, Lee BB, Tien JY, Prior WW, van Dijk S, Wu H, Gray DC, et al. Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents. J Cell Biol. 2008;183(1):101–16.CrossRefPubMedPubMedCentral
43.
go back to reference Eimer S, Belaud-Rotureau MA, Airiau K, Jeanneteau M, Laharanne E, Veron N, Vital A, Loiseau H, Merlio JP, Belloc F. Autophagy inhibition cooperates with erlotinib to induce glioblastoma cell death. Cancer Biol Ther. 2011;11(12):1017–27.CrossRefPubMed Eimer S, Belaud-Rotureau MA, Airiau K, Jeanneteau M, Laharanne E, Veron N, Vital A, Loiseau H, Merlio JP, Belloc F. Autophagy inhibition cooperates with erlotinib to induce glioblastoma cell death. Cancer Biol Ther. 2011;11(12):1017–27.CrossRefPubMed
44.
go back to reference Shingu T, Fujiwara K, Bogler O, Akiyama Y, Moritake K, Shinojima N, Tamada Y, Yokoyama T, Kondo S. Inhibition of autophagy at a late stage enhances imatinib-induced cytotoxicity in human malignant glioma cells. Int J Cancer. 2009;124(5):1060–71.CrossRefPubMed Shingu T, Fujiwara K, Bogler O, Akiyama Y, Moritake K, Shinojima N, Tamada Y, Yokoyama T, Kondo S. Inhibition of autophagy at a late stage enhances imatinib-induced cytotoxicity in human malignant glioma cells. Int J Cancer. 2009;124(5):1060–71.CrossRefPubMed
Metadata
Title
Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma
Authors
Jin Wang
Xiaoyang Liu
Yongzhi Hong
Songtao Wang
Pin Chen
Aihua Gu
Xiaoyuan Guo
Peng Zhao
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2017
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-017-0549-6

Other articles of this Issue 1/2017

Journal of Experimental & Clinical Cancer Research 1/2017 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