Skip to main content
Top
Published in: Cellular Oncology 3/2017

01-06-2017 | Original Paper

Dual treatment with shikonin and temozolomide reduces glioblastoma tumor growth, migration and glial-to-mesenchymal transition

Authors: Diana Matias, Joana Balça-Silva, Luiz Gustavo Dubois, Bruno Pontes, Valéria Pereira Ferrer, Luciane Rosário, Anália do Carmo, Juliana Echevarria-Lima, Ana Bela Sarmento-Ribeiro, Maria Celeste Lopes, Vivaldo Moura-Neto

Published in: Cellular Oncology | Issue 3/2017

Login to get access

Abstract

Purpose

Glioblastomas (GBM) comprise 17% of all primary brain tumors. These tumors are extremely aggressive due to their infiltrative capacity and chemoresistance, with glial-to-mesenchymal transition (GMT) proteins playing a prominent role in tumor invasion. One compound that has recently been used to reduce the expression of these proteins is shikonin (SHK), a naphthoquinone with anti-tumor properties. Temozolomide (TMZ), the most commonly used chemotherapeutic agent in GBM treatment, has so far not been studied in combination with SHK. Here, we investigated the combined effects of these two drugs on the proliferation and motility of GBM-derived cells.

Methods

The cytotoxic and proliferative effects of SHK and TMZ on human GBM-derived cells were tested using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), Ki67 staining and BrdU incorporation assays. The migration capacities of these cells were evaluated using a scratch wound assay. The expression levels of β3 integrin, metalloproteinases (MMPs) and GMT-associated proteins were determined by Western blotting and immunocytochemistry.

Results

We found that GBM-derived cells treated with a combination of SHK and TMZ showed decreases in their proliferation and migration capacities. These decreases were followed by the suppression of GMT through a reduction of β3 integrin, MMP-2, MMP-9, Slug and vimentin expression via inactivation of PI3K/AKT signaling.

Conclusion

From our results we conclude that dual treatment with SHK and TMZ may constitute a powerful new tool for GBM treatment by reducing therapy resistance and tumor recurrence.
Appendix
Available only for authorised users
Literature
1.
go back to reference F.B. Furnari, T. Fenton, R.M. Bachoo, A. Mukasa, J.M. Stommel, A. Stegh, W.C. Hahn, K.L. Ligon, D.N. Louis, C. Brennan, L. Chin, R.A. DePinho, W.K. Cavenee, Malignant astrocytic glioma: Genetics, biology, and paths to treatment. Genes. Dev. 21, 2683–2710 (2007). doi:10.1101/gad.1596707 CrossRefPubMed F.B. Furnari, T. Fenton, R.M. Bachoo, A. Mukasa, J.M. Stommel, A. Stegh, W.C. Hahn, K.L. Ligon, D.N. Louis, C. Brennan, L. Chin, R.A. DePinho, W.K. Cavenee, Malignant astrocytic glioma: Genetics, biology, and paths to treatment. Genes. Dev. 21, 2683–2710 (2007). doi:10.​1101/​gad.​1596707 CrossRefPubMed
2.
go back to reference F.R. Lima, S.A. Kahn, R.C. Soletti, D. Biasoli, T. Alves, A.C. da Fonseca, C. Garcia, L. Romao, J. Brito, R. Holanda-Afonso, J. Faria, H. Borges, V. Moura-Neto, Glioblastoma: Therapeutic challenges, what lies ahead. Biochim. Biophys. Acta. 1826, 338–349 (2012). doi:10.1016/j.bbcan.2012.05.004 PubMed F.R. Lima, S.A. Kahn, R.C. Soletti, D. Biasoli, T. Alves, A.C. da Fonseca, C. Garcia, L. Romao, J. Brito, R. Holanda-Afonso, J. Faria, H. Borges, V. Moura-Neto, Glioblastoma: Therapeutic challenges, what lies ahead. Biochim. Biophys. Acta. 1826, 338–349 (2012). doi:10.​1016/​j.​bbcan.​2012.​05.​004 PubMed
3.
go back to reference R. Stupp, M. E. Hegi, W. P. Mason, M. J. van den Bent, M. J. Taphoorn, R. C. Janzer, S. K. Ludwin, A. Allgeier, B. Fisher, K. Belanger, P. Hau, A. A. Brandes, J. Gijtenbeek, C. Marosi, C. J. Vecht, K. Mokhtari, P. Wesseling, S. Villa, E. Eisenhauer, T. Gorlia, M. Weller, D. Lacombe, J. G. Cairncross, R. O. Mirimanoff, European Organisation for Research, Treatment of Cancer Brain Tumour and Radiation Oncology Groups, National Cancer Institute of Canada Clinical Trials Group. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet. Oncol. 10, 459–466 (2009) doi: 10.1016/S1470–2045(09)70025–7 R. Stupp, M. E. Hegi, W. P. Mason, M. J. van den Bent, M. J. Taphoorn, R. C. Janzer, S. K. Ludwin, A. Allgeier, B. Fisher, K. Belanger, P. Hau, A. A. Brandes, J. Gijtenbeek, C. Marosi, C. J. Vecht, K. Mokhtari, P. Wesseling, S. Villa, E. Eisenhauer, T. Gorlia, M. Weller, D. Lacombe, J. G. Cairncross, R. O. Mirimanoff, European Organisation for Research, Treatment of Cancer Brain Tumour and Radiation Oncology Groups, National Cancer Institute of Canada Clinical Trials Group. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet. Oncol. 10, 459–466 (2009) doi: 10.1016/S1470–2045(09)70025–7
4.
go back to reference S.A. Kahn, D. Biasoli, C. Garcia, L.H. Geraldo, B. Pontes, M. Sobrinho, A.C. Frauches, L. Romao, R.C. Soletti, S. Assuncao Fdos, F. Tovar-Moll, J.M. de Souza, F.R. Lima, G. Anderluh, V. Moura-Neto, Equinatoxin II potentiates temozolomide- and etoposide-induced glioblastoma cell death. Current. Top. Med. Chem. 12, 2082–2093 (2012)CrossRef S.A. Kahn, D. Biasoli, C. Garcia, L.H. Geraldo, B. Pontes, M. Sobrinho, A.C. Frauches, L. Romao, R.C. Soletti, S. Assuncao Fdos, F. Tovar-Moll, J.M. de Souza, F.R. Lima, G. Anderluh, V. Moura-Neto, Equinatoxin II potentiates temozolomide- and etoposide-induced glioblastoma cell death. Current. Top. Med. Chem. 12, 2082–2093 (2012)CrossRef
5.
go back to reference V. Moura-Neto, L. Campanati, D. Matias, C.M. Pereira, C. Freitas, J. M. Coelho-Aguiar, T.C.L.S. Spohr, A.L. Tavares-Gomes, D. Pinheiro-Aguiar, S.A. Kahn, J. Balça-Silva, B. Pontes, I. Porto-Carreiro, J. Faria, R.A.P. Martins, S. Lima-Costa, M. F. Dias-Costa, M.C. Lopes, F.R.S. Lima. Glioblastoma and the special role of adhesion molecules in their invasion, ed. by A. Sedo, R. Mentlein (Springer-Verlag Wien, 2014), p. 293. doi:10.1007/978–3–7091-1431-5 V. Moura-Neto, L. Campanati, D. Matias, C.M. Pereira, C. Freitas, J. M. Coelho-Aguiar, T.C.L.S. Spohr, A.L. Tavares-Gomes, D. Pinheiro-Aguiar, S.A. Kahn, J. Balça-Silva, B. Pontes, I. Porto-Carreiro, J. Faria, R.A.P. Martins, S. Lima-Costa, M. F. Dias-Costa, M.C. Lopes, F.R.S. Lima. Glioblastoma and the special role of adhesion molecules in their invasion, ed. by A. Sedo, R. Mentlein (Springer-Verlag Wien, 2014), p. 293. doi:10.1007/978–3–7091-1431-5
6.
go back to reference H.J. Anderson, D.S. Galileo, Small-molecule inhibitors of FGFR, integrins and FAK selectively decrease L1CAM-stimulated glioblastoma cell motility and proliferation. Cell. Oncol. 39, 229–242 (2016). doi:10.1007/s13402-016-0267-7 H.J. Anderson, D.S. Galileo, Small-molecule inhibitors of FGFR, integrins and FAK selectively decrease L1CAM-stimulated glioblastoma cell motility and proliferation. Cell. Oncol. 39, 229–242 (2016). doi:10.​1007/​s13402-016-0267-7
8.
go back to reference J.S. Desgrosellier, D.A. Cheresh, Integrins in cancer: Biological implications and therapeutic opportunities. Nat. Rev. Cancer 10, 9–22 (2010). doi:10.1038/nrc2748 J.S. Desgrosellier, D.A. Cheresh, Integrins in cancer: Biological implications and therapeutic opportunities. Nat. Rev. Cancer 10, 9–22 (2010). doi:10.​1038/​nrc2748
10.
go back to reference C.Y. Liu, H.H. Lin, M.J. Tang, Y.K. Wang. Vimentin contributes to epithelial-mesenchymal transition cancer cell mechanics by mediating cytoskeletal organization and focal adhesion maturation. Oncotarget 6, 15966–15983 (2015) doi:10.18632/oncotarget.3862 C.Y. Liu, H.H. Lin, M.J. Tang, Y.K. Wang. Vimentin contributes to epithelial-mesenchymal transition cancer cell mechanics by mediating cytoskeletal organization and focal adhesion maturation. Oncotarget 6, 15966–15983 (2015) doi:10.​18632/​oncotarget.​3862
12.
go back to reference E. Godefroy, A. Moreau-Aubry, E. Diez, B. Dreno, F. Jotereau, Y. Guilloux. alpha v beta3-dependent cross-presentation of matrix metalloproteinase-2 by melanoma cells gives rise to a new tumor antigen. J. Exp. Med. 202, 61–72 (2005) doi: 10.1084/jem.20042138 E. Godefroy, A. Moreau-Aubry, E. Diez, B. Dreno, F. Jotereau, Y. Guilloux. alpha v beta3-dependent cross-presentation of matrix metalloproteinase-2 by melanoma cells gives rise to a new tumor antigen. J. Exp. Med. 202, 61–72 (2005) doi: 10.1084/jem.20042138
13.
go back to reference R.C. Soletti, G.P. de Faria, J. Vernal, H. Terenzi, G. Anderluh, H.L. Borges, V. Moura-Neto, N.H. Gabilan, Potentiation of anticancer-drug cytotoxicity by sea anemone pore-forming proteins in human glioblastoma cells. Anti-Cancer Drugs 19, 517–525 (2008). doi:10.1097/CAD.0b013e3282faa704 R.C. Soletti, G.P. de Faria, J. Vernal, H. Terenzi, G. Anderluh, H.L. Borges, V. Moura-Neto, N.H. Gabilan, Potentiation of anticancer-drug cytotoxicity by sea anemone pore-forming proteins in human glioblastoma cells. Anti-Cancer Drugs 19, 517–525 (2008). doi:10.​1097/​CAD.​0b013e3282faa704​
14.
go back to reference B.L. Santos, M.N. Oliveira, P.L. Coelho, B.P. Pitanga, A.B. da Silva, T. Adelita, V.D. Silva, F. Costa Mde. R.S. El-Bacha, M. Tardy, H. Chneiweiss, M.P. Junier, V. Moura-Neto, S.L. Costa. Flavonoids suppress human glioblastoma cell growth by inhibiting cell metabolism, migration, and by regulating extracellular matrix proteins and metalloproteinases expression. Chem. Biol. Interact. 242, 123–138 (2015) doi: 10.1016/j.cbi.2015.07.014 B.L. Santos, M.N. Oliveira, P.L. Coelho, B.P. Pitanga, A.B. da Silva, T. Adelita, V.D. Silva, F. Costa Mde. R.S. El-Bacha, M. Tardy, H. Chneiweiss, M.P. Junier, V. Moura-Neto, S.L. Costa. Flavonoids suppress human glioblastoma cell growth by inhibiting cell metabolism, migration, and by regulating extracellular matrix proteins and metalloproteinases expression. Chem. Biol. Interact. 242, 123–138 (2015) doi: 10.1016/j.cbi.2015.07.014
15.
go back to reference J. Balca-Silva, D. Matias, A. do Carmo, H. Girao, V. Moura-Neto, A.B. Sarmento-Ribeiro, M.C. Lopes. Tamoxifen in combination with temozolomide induce a synergistic inhibition of PKC-pan in GBM cell lines. Biochim. Biophys. Acta 1850, 722–732 (2015) doi:10.1016/j.bbagen.2014.12.022 J. Balca-Silva, D. Matias, A. do Carmo, H. Girao, V. Moura-Neto, A.B. Sarmento-Ribeiro, M.C. Lopes. Tamoxifen in combination with temozolomide induce a synergistic inhibition of PKC-pan in GBM cell lines. Biochim. Biophys. Acta 1850, 722–732 (2015) doi:10.​1016/​j.​bbagen.​2014.​12.​022
17.
go back to reference Y. Chen, L. Zheng, J. Liu, Z. Zhou, X. Cao, X. Lv, F. Chen, Shikonin inhibits prostate cancer cells metastasis by reducing matrix metalloproteinase-2/−9 expression via AKT/mTOR and ROS/ERK1/2 pathways. Int. Immunopharmacol. 21, 447–455 (2014). doi:10.1016/j.intimp.2014.05.026 CrossRefPubMed Y. Chen, L. Zheng, J. Liu, Z. Zhou, X. Cao, X. Lv, F. Chen, Shikonin inhibits prostate cancer cells metastasis by reducing matrix metalloproteinase-2/−9 expression via AKT/mTOR and ROS/ERK1/2 pathways. Int. Immunopharmacol. 21, 447–455 (2014). doi:10.​1016/​j.​intimp.​2014.​05.​026 CrossRefPubMed
18.
go back to reference H. Wang, C. Wu, S. Wan, H. Zhang, S. Zhou, G. Liu, Shikonin attenuates lung cancer cell adhesion to extracellular matrix and metastasis by inhibiting integrin β1 expression and the ERK1/2 signaling pathway. Toxicology 308, 104–112 (2013). doi:10.1016/j.tox.2013.03.015 H. Wang, C. Wu, S. Wan, H. Zhang, S. Zhou, G. Liu, Shikonin attenuates lung cancer cell adhesion to extracellular matrix and metastasis by inhibiting integrin β1 expression and the ERK1/2 signaling pathway. Toxicology 308, 104–112 (2013). doi:10.​1016/​j.​tox.​2013.​03.​015
19.
go back to reference Q. Zhao, N. Kretschmer, R. Bauer, T. Efferth, Shikonin and its derivatives inhibit the epidermal growth factor receptor signaling and synergistically kill glioblastoma cells in combination with erlotinib. Int. J. Cancer 137, 1446–1456 (2015). doi:10.1002/ijc.29483 Q. Zhao, N. Kretschmer, R. Bauer, T. Efferth, Shikonin and its derivatives inhibit the epidermal growth factor receptor signaling and synergistically kill glioblastoma cells in combination with erlotinib. Int. J. Cancer 137, 1446–1456 (2015). doi:10.​1002/​ijc.​29483
20.
go back to reference F.L. Zhang, P. Wang, Y.H. Liu, L.B. Liu, X.B. Liu, Z. Li, Y.X. Xue, Topoisomerase I inhibitors, shikonin and topotecan, inhibit growth and induce apoptosis of glioma cells and glioma stem cells. PLoS One 8, e81815 (2013). doi:10.1371/journal.pone.0081815 F.L. Zhang, P. Wang, Y.H. Liu, L.B. Liu, X.B. Liu, Z. Li, Y.X. Xue, Topoisomerase I inhibitors, shikonin and topotecan, inhibit growth and induce apoptosis of glioma cells and glioma stem cells. PLoS One 8, e81815 (2013). doi:10.​1371/​journal.​pone.​0081815
21.
go back to reference D. Hong, S.Y. Jang, E.H. Jang, B. Jung, I.H. Cho, M.J. Park, S.Y. Jeong, J.H. Kim, Shikonin as an inhibitor of the LPS-induced epithelial-to-mesenchymal transition in human breast cancer cells. Int. J. Mol. Med. 36, 1601–1606 (2015). doi:10.3892/ijmm.2015.2373 PubMed D. Hong, S.Y. Jang, E.H. Jang, B. Jung, I.H. Cho, M.J. Park, S.Y. Jeong, J.H. Kim, Shikonin as an inhibitor of the LPS-induced epithelial-to-mesenchymal transition in human breast cancer cells. Int. J. Mol. Med. 36, 1601–1606 (2015). doi:10.​3892/​ijmm.​2015.​2373 PubMed
22.
go back to reference R. Mahabir, M. Tanino, A. Elmansuri, L. Wang, T. Kimura, T. Itoh, Y. Ohba, H. Nishihara, H. Shirato, M. Tsuda, S. Tanaka, Sustained elevation of snail promotes glial-mesenchymal transition after irradiation in malignant glioma. Neuro-Oncology 16, 671–685 (2014). doi:10.1093/neuonc/not239 R. Mahabir, M. Tanino, A. Elmansuri, L. Wang, T. Kimura, T. Itoh, Y. Ohba, H. Nishihara, H. Shirato, M. Tsuda, S. Tanaka, Sustained elevation of snail promotes glial-mesenchymal transition after irradiation in malignant glioma. Neuro-Oncology 16, 671–685 (2014). doi:10.​1093/​neuonc/​not239
23.
go back to reference J. Faria, L. Romao, S. Martins, T. Alves, F.A. Mendes, G.P. de Faria, R. Hollanda, C. Takiya, L. Chimelli, V. Morandi, J.M. de Souza, J.G. Abreu, V. Moura Neto, Interactive properties of human glioblastoma cells with brain neurons in culture and neuronal modulation of glial laminin organization. Differentiation 74, 562–572 (2006). doi:10.1111/j.1432-0436.2006.00090.x J. Faria, L. Romao, S. Martins, T. Alves, F.A. Mendes, G.P. de Faria, R. Hollanda, C. Takiya, L. Chimelli, V. Morandi, J.M. de Souza, J.G. Abreu, V. Moura Neto, Interactive properties of human glioblastoma cells with brain neurons in culture and neuronal modulation of glial laminin organization. Differentiation 74, 562–572 (2006). doi:10.​1111/​j.​1432-0436.​2006.​00090.​x
24.
go back to reference L.F. Romao, F.A. Mendes, N.M. Feitosa, J.C. Faria, J.M. Coelho-Aguiar, J.M. de Souza, V. Moura-Neto, J.G. Abreu, Connective tissue growth factor (CTGF/CCN2) is negatively regulated during neuron-glioblastoma interaction. PLoS One 8, e55605 (2013). doi:10.1371/journal.pone.0055605 L.F. Romao, F.A. Mendes, N.M. Feitosa, J.C. Faria, J.M. Coelho-Aguiar, J.M. de Souza, V. Moura-Neto, J.G. Abreu, Connective tissue growth factor (CTGF/CCN2) is negatively regulated during neuron-glioblastoma interaction. PLoS One 8, e55605 (2013). doi:10.​1371/​journal.​pone.​0055605
25.
go back to reference C.C. Liang, A.Y. Park, J.L. Guan, In vitro scratch assay: A convenient and inexpensive method for analysis of cell migration in vitro. Nat. Protoc. 2, 329–333 (2007). doi:10.1038/nprot.2007.30 C.C. Liang, A.Y. Park, J.L. Guan, In vitro scratch assay: A convenient and inexpensive method for analysis of cell migration in vitro. Nat. Protoc. 2, 329–333 (2007). doi:10.​1038/​nprot.​2007.​30
26.
go back to reference A. Boudaoud, A. Burian, D. Borowska-Wykret, M. Uyttewaal, R. Wrzalik, D. Kwiatkowska, O. Hamant, FibrilTool, an ImageJ plug-in to quantify fibrillar structures in raw microscopy images. Nat. Protoc. 9, 457–463 (2014). doi:10.1038/nprot.2014.024 CrossRefPubMed A. Boudaoud, A. Burian, D. Borowska-Wykret, M. Uyttewaal, R. Wrzalik, D. Kwiatkowska, O. Hamant, FibrilTool, an ImageJ plug-in to quantify fibrillar structures in raw microscopy images. Nat. Protoc. 9, 457–463 (2014). doi:10.​1038/​nprot.​2014.​024 CrossRefPubMed
27.
go back to reference H. Towbin, T. Staehelin, J. Gordon, Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applications. Biotechnology 24, 145–149 (1979) H. Towbin, T. Staehelin, J. Gordon, Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applications. Biotechnology 24, 145–149 (1979)
28.
go back to reference K.A. McDowell, G.J. Riggins, G.L. Gallia, Targeting the AKT pathway in glioblastoma. Curr. Pharm. Des. 17, 2411–2420 (2011)CrossRefPubMed K.A. McDowell, G.J. Riggins, G.L. Gallia, Targeting the AKT pathway in glioblastoma. Curr. Pharm. Des. 17, 2411–2420 (2011)CrossRefPubMed
29.
go back to reference N. Fenouille, M. Tichet, M. Dufies, A. Pottier, A. Mogha, J.K. Soo, S. Rocchi, A. Mallavialle, M.D. Galibert, A. Khammari, J.P. Lacour, R. Ballotti, M. Deckert, S. Tartare-Deckert, The epithelial-mesenchymal transition (EMT) regulatory factor SLUG (SNAI2) is a downstream target of SPARC and AKT in promoting melanoma cell invasion. PLoS One 7, e40378 (2012). doi:10.1371/journal.pone.0040378 N. Fenouille, M. Tichet, M. Dufies, A. Pottier, A. Mogha, J.K. Soo, S. Rocchi, A. Mallavialle, M.D. Galibert, A. Khammari, J.P. Lacour, R. Ballotti, M. Deckert, S. Tartare-Deckert, The epithelial-mesenchymal transition (EMT) regulatory factor SLUG (SNAI2) is a downstream target of SPARC and AKT in promoting melanoma cell invasion. PLoS One 7, e40378 (2012). doi:10.​1371/​journal.​pone.​0040378
31.
go back to reference W.Y. Cheng, J.J. Kandel, D.J. Yamashiro, P. Canoll, D. Anastassiou, A multi-cancer mesenchymal transition gene expression signature is associated with prolonged time to recurrence in glioblastoma. PLoS One 7, e34705 (2012). doi:10.1371/journal.pone.0034705 W.Y. Cheng, J.J. Kandel, D.J. Yamashiro, P. Canoll, D. Anastassiou, A multi-cancer mesenchymal transition gene expression signature is associated with prolonged time to recurrence in glioblastoma. PLoS One 7, e34705 (2012). doi:10.​1371/​journal.​pone.​0034705
32.
go back to reference E.M. Goellner, B. Grimme, A.R. Brown, Y.C. Lin, X.H. Wang, K.F. Sugrue, L. Mitchell, R.N. Trivedi, J.B. Tang, R.W. Sobol, Overcoming temozolomide resistance in glioblastoma via dual inhibition of NAD+ biosynthesis and base excision repair. Cancer Res. 71, 2308–2317 (2011). doi:10.1158/0008-5472.CAN-10-3213 E.M. Goellner, B. Grimme, A.R. Brown, Y.C. Lin, X.H. Wang, K.F. Sugrue, L. Mitchell, R.N. Trivedi, J.B. Tang, R.W. Sobol, Overcoming temozolomide resistance in glioblastoma via dual inhibition of NAD+ biosynthesis and base excision repair. Cancer Res. 71, 2308–2317 (2011). doi:10.​1158/​0008-5472.​CAN-10-3213
35.
go back to reference M.E. Hegi, A.C. Diserens, T. Gorlia, M.F. Hamou, N. de Tribolet, M. Weller, J.M. Kros, J.A. Hainfellner, W. Mason, L. Mariani, J.E. Bromberg, P. Hau, R.O. Mirimanoff, J.G. Cairncross, R.C. Janzer, R. Stupp, MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 352, 997–1003 (2005). doi:10.1056/NEJMoa043331 CrossRefPubMed M.E. Hegi, A.C. Diserens, T. Gorlia, M.F. Hamou, N. de Tribolet, M. Weller, J.M. Kros, J.A. Hainfellner, W. Mason, L. Mariani, J.E. Bromberg, P. Hau, R.O. Mirimanoff, J.G. Cairncross, R.C. Janzer, R. Stupp, MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 352, 997–1003 (2005). doi:10.​1056/​NEJMoa043331 CrossRefPubMed
36.
go back to reference C. Kubelt, K. Hattermann, S. Sebens, H.M. Mehdorn, J. Held-Feindt, Epithelial-to-mesenchymal transition in paired human primary and recurrent glioblastomas. Int. J. Oncol. 46, 2515–2525 (2015). doi:10.3892/ijo.2015.2944 PubMed C. Kubelt, K. Hattermann, S. Sebens, H.M. Mehdorn, J. Held-Feindt, Epithelial-to-mesenchymal transition in paired human primary and recurrent glioblastomas. Int. J. Oncol. 46, 2515–2525 (2015). doi:10.​3892/​ijo.​2015.​2944 PubMed
37.
go back to reference Y.W. Heng, H.H. Lim, T. Mina, P. Utomo, S. Zhong, C.T. Lim, C.G. Koh, TPPP acts downstream of RhoA-ROCK-LIMK2 to regulate astral microtubule organization and spindle orientation. J. Cell. Sci. 125, 1579–1590 (2012). doi:10.1242/jcs.096818 CrossRefPubMed Y.W. Heng, H.H. Lim, T. Mina, P. Utomo, S. Zhong, C.T. Lim, C.G. Koh, TPPP acts downstream of RhoA-ROCK-LIMK2 to regulate astral microtubule organization and spindle orientation. J. Cell. Sci. 125, 1579–1590 (2012). doi:10.​1242/​jcs.​096818 CrossRefPubMed
38.
39.
40.
go back to reference S.S. Lakka, C.S. Gondi, N. Yanamandra, W.C. Olivero, D.H. Dinh, M. Gujrati, J.S. Rao, Inhibition of cathepsin B and MMP-9 gene expression in glioblastoma cell line via RNA interference reduces tumor cell invasion, tumor growth and angiogenesis. Oncogene 23, 4681–4689 (2004). doi:10.1038/sj.onc.1207616 S.S. Lakka, C.S. Gondi, N. Yanamandra, W.C. Olivero, D.H. Dinh, M. Gujrati, J.S. Rao, Inhibition of cathepsin B and MMP-9 gene expression in glioblastoma cell line via RNA interference reduces tumor cell invasion, tumor growth and angiogenesis. Oncogene 23, 4681–4689 (2004). doi:10.​1038/​sj.​onc.​1207616
41.
go back to reference C. Chetty, S.S. Lakka, P. Bhoopathi, J.S. Rao, MMP-2 alters VEGF expression via alphaVbeta3 integrin-mediated PI3K/AKT signaling in A549 lung cancer cells. Int. J. Cancer 127, 1081–1095 (2010). doi:10.1002/ijc.25134 C. Chetty, S.S. Lakka, P. Bhoopathi, J.S. Rao, MMP-2 alters VEGF expression via alphaVbeta3 integrin-mediated PI3K/AKT signaling in A549 lung cancer cells. Int. J. Cancer 127, 1081–1095 (2010). doi:10.​1002/​ijc.​25134
43.
go back to reference U. Jadhav, S. Chigurupati, S.S. Lakka, S. Mohanam, Inhibition of matrix metalloproteinase-9 reduces in vitro invasion and angiogenesis in human microvascular endothelial cells. Int. J. Oncol. 25, 1407–1414 (2004)PubMed U. Jadhav, S. Chigurupati, S.S. Lakka, S. Mohanam, Inhibition of matrix metalloproteinase-9 reduces in vitro invasion and angiogenesis in human microvascular endothelial cells. Int. J. Oncol. 25, 1407–1414 (2004)PubMed
44.
go back to reference Y. Jiao, X. Feng, Y. Zhan, R. Wang, S. Zheng, W. Liu, X. Zeng, Matrix metalloproteinase-2 promotes αvβ3 integrin-mediated adhesion and migration of human melanoma cells by cleaving fibronectin. PLoS One 7, e41591 (2012). doi:10.1371/journal.pone.0041591 Y. Jiao, X. Feng, Y. Zhan, R. Wang, S. Zheng, W. Liu, X. Zeng, Matrix metalloproteinase-2 promotes αvβ3 integrin-mediated adhesion and migration of human melanoma cells by cleaving fibronectin. PLoS One 7, e41591 (2012). doi:10.​1371/​journal.​pone.​0041591
45.
go back to reference J.M. Yang, Z. Xu, H. Wu, H. Zhu, X. Wu, W.N. Hait, Overexpression of extracellular matrix metalloproteinase inducer in multidrug resistant cancer cells. Mol. Cancer Res. 1, 420–427 (2003) J.M. Yang, Z. Xu, H. Wu, H. Zhu, X. Wu, W.N. Hait, Overexpression of extracellular matrix metalloproteinase inducer in multidrug resistant cancer cells. Mol. Cancer Res. 1, 420–427 (2003)
46.
go back to reference F.Y. Zhang, Y. Hu, Z.Y. Que, P. Wang, Y.H. Liu, Z.H. Wang, Y.X. Xue, Shikonin inhibits the migration and invasion of human glioblastoma cells by targeting phosphorylated β-catenin and phosphorylated PI3K/Akt: A potential mechanism for the anti-glioma efficacy of a traditional Chinese herbal medicine. Int. J. Mol. Sci. 16, 23823–23848 (2015). doi:10.3390/ijms161023823 CrossRefPubMedPubMedCentral F.Y. Zhang, Y. Hu, Z.Y. Que, P. Wang, Y.H. Liu, Z.H. Wang, Y.X. Xue, Shikonin inhibits the migration and invasion of human glioblastoma cells by targeting phosphorylated β-catenin and phosphorylated PI3K/Akt: A potential mechanism for the anti-glioma efficacy of a traditional Chinese herbal medicine. Int. J. Mol. Sci. 16, 23823–23848 (2015). doi:10.​3390/​ijms161023823 CrossRefPubMedPubMedCentral
47.
go back to reference P.L. Wei, C.C. Tu, C.H. Chen, Y.S. Ho, C.T. Wu, H.Y. Su, W.Y. Chen, J.J. Liu, Y.J. Chang, Shikonin suppresses the migratory ability of hepatocellular carcinoma cells. J. Agric. Food Chem. 61, 8191–8197 (2013). doi:10.1021/jf4009586 P.L. Wei, C.C. Tu, C.H. Chen, Y.S. Ho, C.T. Wu, H.Y. Su, W.Y. Chen, J.J. Liu, Y.J. Chang, Shikonin suppresses the migratory ability of hepatocellular carcinoma cells. J. Agric. Food Chem. 61, 8191–8197 (2013). doi:10.​1021/​jf4009586
Metadata
Title
Dual treatment with shikonin and temozolomide reduces glioblastoma tumor growth, migration and glial-to-mesenchymal transition
Authors
Diana Matias
Joana Balça-Silva
Luiz Gustavo Dubois
Bruno Pontes
Valéria Pereira Ferrer
Luciane Rosário
Anália do Carmo
Juliana Echevarria-Lima
Ana Bela Sarmento-Ribeiro
Maria Celeste Lopes
Vivaldo Moura-Neto
Publication date
01-06-2017
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 3/2017
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-017-0320-1

Other articles of this Issue 3/2017

Cellular Oncology 3/2017 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