Skip to main content
Top
Published in: BMC Cancer 1/2016

Open Access 01-12-2016 | Research article

Tyrosine kinase inhibitor SU11274 increased tumorigenicity and enriched for melanoma-initiating cells by bioenergetic modulation

Authors: Lucia Kucerova, Lucia Demkova, Svetlana Skolekova, Roman Bohovic, Miroslava Matuskova

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

Small molecule inhibitor of tyrosine kinase activity, compound SU11274, was reported to have antitumorigenic and antimetastatic effect in melanoma. In this study, we evaluated, whether similar effect could be achieved also in other melanoma cells including highly tumorigenic and hypermetastatic variant.

Methods

The effect of SU11274 was evaluated in adherent and non-adherent melanosphere cultures of human melanoma cells M14, M4Beu, A375 and EGFP-A375/Rel3. Tumorigenicity of SU11274-treated cells was tested by limiting dilution assay in xenograft model in vivo.

Results

Here we show that SU11274 enriched for melanoma-initiating cells in vivo. SU11274 substantially decreased number of cells in adherent and spheroid cultures, but increased their tumorigenic potential as determined by higher frequency of tumor-initiating cells in vivo. SU11274 treatment was not associated with any significant alteration in the expression of stem cell markers, but the inhibitor stimulated higher level of pluripotent markers. SU11274-treated melanoma cells exhibited higher ATP content and lactate release indicative of increased glycolysis. Our data suggest that the SU11274 altered bioenergetic state of the cells. Indeed, pharmacological intervention with a glycolytic inhibitor dichloroacetate significantly reduced SU11274-promoted increase in melanoma-initiating cells and decreased their tumorigenicity.

Conclusions

Our data suggest critical role of glycolysis regulation in melanoma-initiating cells. Moreover, these data unravel substantial plasticity of melanoma cells and their adoptive mechanisms, which result in ambivalent response to therapeutic targeting.
Literature
1.
go back to reference Sattler M, Pride YB, Ma P, Gramlich JL, Chu SC, Quinnan LA, Shirazian S, Liang C, Podar K, Christensen JG, et al. A novel small molecule met inhibitor induces apoptosis in cells transformed by the oncogenic TPR-MET tyrosine kinase. Cancer Res. 2003;63(17):5462–9.PubMed Sattler M, Pride YB, Ma P, Gramlich JL, Chu SC, Quinnan LA, Shirazian S, Liang C, Podar K, Christensen JG, et al. A novel small molecule met inhibitor induces apoptosis in cells transformed by the oncogenic TPR-MET tyrosine kinase. Cancer Res. 2003;63(17):5462–9.PubMed
2.
4.
go back to reference Boccaccio C, Comoglio PM. MET, a driver of invasive growth and cancer clonal evolution under therapeutic pressure. Curr Opin Cell Biol. 2014;31:98–105.CrossRefPubMed Boccaccio C, Comoglio PM. MET, a driver of invasive growth and cancer clonal evolution under therapeutic pressure. Curr Opin Cell Biol. 2014;31:98–105.CrossRefPubMed
5.
go back to reference Knudsen BS, Vande Woude G. Showering c-MET-dependent cancers with drugs. Curr Opin Genet Dev. 2008;18(1):87–96.CrossRefPubMed Knudsen BS, Vande Woude G. Showering c-MET-dependent cancers with drugs. Curr Opin Genet Dev. 2008;18(1):87–96.CrossRefPubMed
6.
go back to reference Kucerova L, Skolekova S, Demkova L, Bohovic R, Matuskova M. Long-term efficiency of mesenchymal stromal cell-mediated CD-MSC/5FC therapy in human melanoma xenograft model. Gene Ther. 2014;21(10):874–87.CrossRefPubMed Kucerova L, Skolekova S, Demkova L, Bohovic R, Matuskova M. Long-term efficiency of mesenchymal stromal cell-mediated CD-MSC/5FC therapy in human melanoma xenograft model. Gene Ther. 2014;21(10):874–87.CrossRefPubMed
7.
go back to reference Kenessey I, Keszthelyi M, Kramer Z, Berta J, Adam A, Dobos J, Mildner M, Flachner B, Cseh S, Barna G, et al. Inhibition of c-Met with the specific small molecule tyrosine kinase inhibitor SU11274 decreases growth and metastasis formation of experimental human melanoma. Curr Cancer Drug Targets. 2010;10(3):332–42.CrossRefPubMed Kenessey I, Keszthelyi M, Kramer Z, Berta J, Adam A, Dobos J, Mildner M, Flachner B, Cseh S, Barna G, et al. Inhibition of c-Met with the specific small molecule tyrosine kinase inhibitor SU11274 decreases growth and metastasis formation of experimental human melanoma. Curr Cancer Drug Targets. 2010;10(3):332–42.CrossRefPubMed
8.
go back to reference Etnyre D, Stone AL, Fong JT, Jacobs RJ, Uppada SB, Botting GM, Rajanna S, Moravec DN, Shambannagari MR, Crees Z, et al. Targeting c-Met in melanoma: mechanism of resistance and efficacy of novel combinatorial inhibitor therapy. Cancer Biol Ther. 2014;15(9):1129–41.CrossRefPubMedPubMedCentral Etnyre D, Stone AL, Fong JT, Jacobs RJ, Uppada SB, Botting GM, Rajanna S, Moravec DN, Shambannagari MR, Crees Z, et al. Targeting c-Met in melanoma: mechanism of resistance and efficacy of novel combinatorial inhibitor therapy. Cancer Biol Ther. 2014;15(9):1129–41.CrossRefPubMedPubMedCentral
10.
go back to reference Girouard SD, Murphy GF. Melanoma stem cells: not rare, but well done. Lab Invest. 2011;91(5):647–64.CrossRefPubMed Girouard SD, Murphy GF. Melanoma stem cells: not rare, but well done. Lab Invest. 2011;91(5):647–64.CrossRefPubMed
11.
go back to reference Murphy GF, Wilson BJ, Girouard SD, Frank NY, Frank MH. Stem cells and targeted approaches to melanoma cure. Mol Aspects Med. 2014;39:33–49.CrossRefPubMed Murphy GF, Wilson BJ, Girouard SD, Frank NY, Frank MH. Stem cells and targeted approaches to melanoma cure. Mol Aspects Med. 2014;39:33–49.CrossRefPubMed
12.
go back to reference Quintana E, Shackleton M, Foster HR, Fullen DR, Sabel MS, Johnson TM, Morrison SJ. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell. 2010;18(5):510–23.CrossRefPubMedPubMedCentral Quintana E, Shackleton M, Foster HR, Fullen DR, Sabel MS, Johnson TM, Morrison SJ. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell. 2010;18(5):510–23.CrossRefPubMedPubMedCentral
13.
go back to reference Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA, Vultur A, Basu D, Gimotty P, Vogt T, Herlyn M. A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell. 2010;141(4):583–94.CrossRefPubMedPubMedCentral Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA, Vultur A, Basu D, Gimotty P, Vogt T, Herlyn M. A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell. 2010;141(4):583–94.CrossRefPubMedPubMedCentral
14.
go back to reference Pinner S, Jordan P, Sharrock K, Bazley L, Collinson L, Marais R, Bonvin E, Goding C, Sahai E. Intravital imaging reveals transient changes in pigment production and Brn2 expression during metastatic melanoma dissemination. Cancer Res. 2009;69(20):7969–77.CrossRefPubMedPubMedCentral Pinner S, Jordan P, Sharrock K, Bazley L, Collinson L, Marais R, Bonvin E, Goding C, Sahai E. Intravital imaging reveals transient changes in pigment production and Brn2 expression during metastatic melanoma dissemination. Cancer Res. 2009;69(20):7969–77.CrossRefPubMedPubMedCentral
15.
go back to reference Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S, Van Belle PA, Xu X, Elder DE, Herlyn M. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 2005;65(20):9328–37.CrossRefPubMed Fang D, Nguyen TK, Leishear K, Finko R, Kulp AN, Hotz S, Van Belle PA, Xu X, Elder DE, Herlyn M. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 2005;65(20):9328–37.CrossRefPubMed
16.
17.
go back to reference Perego M, Tortoreto M, Tragni G, Mariani L, Deho P, Carbone A, Santinami M, Patuzzo R, Mina PD, Villa, et al. Heterogeneous phenotype of human melanoma cells with in vitro and in vivo features of tumor-initiating cells. J Invest Dermatol. 2010;130(7):1877–86. Perego M, Tortoreto M, Tragni G, Mariani L, Deho P, Carbone A, Santinami M, Patuzzo R, Mina PD, Villa, et al. Heterogeneous phenotype of human melanoma cells with in vitro and in vivo features of tumor-initiating cells. J Invest Dermatol. 2010;130(7):1877–86.
18.
go back to reference Li C, Wu JJ, Hynes M, Dosch J, Sarkar B, Welling TH, Pasca di Magliano M, Simeone DM. c-Met is a marker of pancreatic cancer stem cells and therapeutic target. Gastroenterology. 2011;141(6):2218–27. e2215.CrossRefPubMed Li C, Wu JJ, Hynes M, Dosch J, Sarkar B, Welling TH, Pasca di Magliano M, Simeone DM. c-Met is a marker of pancreatic cancer stem cells and therapeutic target. Gastroenterology. 2011;141(6):2218–27. e2215.CrossRefPubMed
19.
go back to reference Isayev O, Rausch V, Bauer N, Liu L, Fan P, Zhang Y, Gladkich J, Nwaeburu CC, Mattern J, Mollenhauer M, et al. Inhibition of glucose turnover by 3-bromopyruvate counteracts pancreatic cancer stem cell features and sensitizes cells to gemcitabine. Oncotarget. 2014;5(13):5177–89.CrossRefPubMedPubMedCentral Isayev O, Rausch V, Bauer N, Liu L, Fan P, Zhang Y, Gladkich J, Nwaeburu CC, Mattern J, Mollenhauer M, et al. Inhibition of glucose turnover by 3-bromopyruvate counteracts pancreatic cancer stem cell features and sensitizes cells to gemcitabine. Oncotarget. 2014;5(13):5177–89.CrossRefPubMedPubMedCentral
20.
go back to reference Yuan P, Ito K, Perez-Lorenzo R, Del Guzzo C, Lee JH, Shen CH, Bosenberg MW, McMahon M, Cantley LC, Zheng B. Phenformin enhances the therapeutic benefit of BRAF (V600E) inhibition in melanoma. Proc Natl Acad Sci U S A. 2013;110(45):18226–31.CrossRefPubMedPubMedCentral Yuan P, Ito K, Perez-Lorenzo R, Del Guzzo C, Lee JH, Shen CH, Bosenberg MW, McMahon M, Cantley LC, Zheng B. Phenformin enhances the therapeutic benefit of BRAF (V600E) inhibition in melanoma. Proc Natl Acad Sci U S A. 2013;110(45):18226–31.CrossRefPubMedPubMedCentral
21.
go back to reference Abildgaard C, Dahl C, Basse AL, Ma T, Guldberg P. Bioenergetic modulation with dichloroacetate reduces the growth of melanoma cells and potentiates their response to BRAFV600E inhibition. J Transl Med. 2014;12:247.CrossRefPubMedPubMedCentral Abildgaard C, Dahl C, Basse AL, Ma T, Guldberg P. Bioenergetic modulation with dichloroacetate reduces the growth of melanoma cells and potentiates their response to BRAFV600E inhibition. J Transl Med. 2014;12:247.CrossRefPubMedPubMedCentral
23.
go back to reference Kankotia S, Stacpoole PW. Dichloroacetate and cancer: new home for an orphan drug? Biochim Biophys Acta. 2014;1846(2):617–29.PubMed Kankotia S, Stacpoole PW. Dichloroacetate and cancer: new home for an orphan drug? Biochim Biophys Acta. 2014;1846(2):617–29.PubMed
24.
25.
go back to reference Martin MJ, Hayward R, Viros A, Marais R. Metformin accelerates the growth of BRAF V600E-driven melanoma by upregulating VEGF-A. Cancer Discov. 2012;2(4):344–55.CrossRefPubMedPubMedCentral Martin MJ, Hayward R, Viros A, Marais R. Metformin accelerates the growth of BRAF V600E-driven melanoma by upregulating VEGF-A. Cancer Discov. 2012;2(4):344–55.CrossRefPubMedPubMedCentral
26.
go back to reference Cerezo M, Tichet M, Abbe P, Ohanna M, Lehraiki A, Rouaud F, Allegra M, Giacchero D, Bahadoran P, Bertolotto C, et al. Metformin blocks melanoma invasion and metastasis development in AMPK/p53-dependent manner. Mol Cancer Ther. 2013;12(8):1605–15.CrossRefPubMed Cerezo M, Tichet M, Abbe P, Ohanna M, Lehraiki A, Rouaud F, Allegra M, Giacchero D, Bahadoran P, Bertolotto C, et al. Metformin blocks melanoma invasion and metastasis development in AMPK/p53-dependent manner. Mol Cancer Ther. 2013;12(8):1605–15.CrossRefPubMed
27.
go back to reference Eleveld-Trancikova D, Kudela P, Majerciak V, Regendova M, Zelnik V, Pastorek J, Pastorekova S, Bizik J. Suppression subtractive hybridisation to isolate differentially expressed genes involved in invasiveness of melanoma cell line cultured under different conditions. Int J Oncol. 2002;20(3):501–8.PubMed Eleveld-Trancikova D, Kudela P, Majerciak V, Regendova M, Zelnik V, Pastorek J, Pastorekova S, Bizik J. Suppression subtractive hybridisation to isolate differentially expressed genes involved in invasiveness of melanoma cell line cultured under different conditions. Int J Oncol. 2002;20(3):501–8.PubMed
28.
go back to reference Kucerova L, Matuskova M, Pastorakova A, Tyciakova S, Jakubikova J, Bohovic R, Altanerova V, Altaner C. Cytosine deaminase expressing human mesenchymal stem cells mediated tumour regression in melanoma bearing mice. J Gene Med. 2008;10(10):1071–82.CrossRefPubMed Kucerova L, Matuskova M, Pastorakova A, Tyciakova S, Jakubikova J, Bohovic R, Altanerova V, Altaner C. Cytosine deaminase expressing human mesenchymal stem cells mediated tumour regression in melanoma bearing mice. J Gene Med. 2008;10(10):1071–82.CrossRefPubMed
29.
go back to reference Kucerova L, Skolekova S, Matuskova M, Bohac M, Kozovska Z. Altered features and increased chemosensitivity of human breast cancer cells mediated by adipose tissue-derived mesenchymal stromal cells. BMC Cancer. 2013;13(1):535.CrossRefPubMedPubMedCentral Kucerova L, Skolekova S, Matuskova M, Bohac M, Kozovska Z. Altered features and increased chemosensitivity of human breast cancer cells mediated by adipose tissue-derived mesenchymal stromal cells. BMC Cancer. 2013;13(1):535.CrossRefPubMedPubMedCentral
30.
go back to reference Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347(1–2):70–8.PubMed Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347(1–2):70–8.PubMed
31.
go back to reference Jun HJ, Acquaviva J, Chi D, Lessard J, Zhu H, Woolfenden S, Bronson RT, Pfannl R, White F, Housman DE, et al. Acquired MET expression confers resistance to EGFR inhibition in a mouse model of glioblastoma multiforme. Oncogene. 2012;31(25):3039–50.CrossRefPubMedPubMedCentral Jun HJ, Acquaviva J, Chi D, Lessard J, Zhu H, Woolfenden S, Bronson RT, Pfannl R, White F, Housman DE, et al. Acquired MET expression confers resistance to EGFR inhibition in a mouse model of glioblastoma multiforme. Oncogene. 2012;31(25):3039–50.CrossRefPubMedPubMedCentral
32.
go back to reference Cao HH, Cheng CY, Su T, Fu XQ, Guo H, Li T, Tse AK, Kwan HY, Yu H, Yu ZL. Quercetin inhibits HGF/c-Met signaling and HGF-stimulated melanoma cell migration and invasion. Mol Cancer. 2015;14:103.CrossRefPubMedPubMedCentral Cao HH, Cheng CY, Su T, Fu XQ, Guo H, Li T, Tse AK, Kwan HY, Yu H, Yu ZL. Quercetin inhibits HGF/c-Met signaling and HGF-stimulated melanoma cell migration and invasion. Mol Cancer. 2015;14:103.CrossRefPubMedPubMedCentral
33.
go back to reference Wang X, Le P, Liang C, Chan J, Kiewlich D, Miller T, Harris D, Sun L, Rice A, Vasile S, et al. Potent and selective inhibitors of the Met [hepatocyte growth factor/scatter factor (HGF/SF) receptor] tyrosine kinase block HGF/SF-induced tumor cell growth and invasion. Mol Cancer Ther. 2003;2(11):1085–92.PubMed Wang X, Le P, Liang C, Chan J, Kiewlich D, Miller T, Harris D, Sun L, Rice A, Vasile S, et al. Potent and selective inhibitors of the Met [hepatocyte growth factor/scatter factor (HGF/SF) receptor] tyrosine kinase block HGF/SF-induced tumor cell growth and invasion. Mol Cancer Ther. 2003;2(11):1085–92.PubMed
34.
go back to reference Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, Koudriakova TB, Alton G, Cui JJ, Kung PP, et al. An orally available small-molecule inhibitor of c-Met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 2007;67(9):4408–17.CrossRefPubMed Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, Koudriakova TB, Alton G, Cui JJ, Kung PP, et al. An orally available small-molecule inhibitor of c-Met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 2007;67(9):4408–17.CrossRefPubMed
35.
go back to reference Ghislin S, Deshayes F, Lauriol J, Middendorp S, Martins I, Al-Daccak R, Alcaide-Loridan C. Plasticity of melanoma cells induced by neural cell crest conditions and three-dimensional growth. Melanoma Res. 2012;22(3):184–94.CrossRefPubMed Ghislin S, Deshayes F, Lauriol J, Middendorp S, Martins I, Al-Daccak R, Alcaide-Loridan C. Plasticity of melanoma cells induced by neural cell crest conditions and three-dimensional growth. Melanoma Res. 2012;22(3):184–94.CrossRefPubMed
36.
go back to reference Mo J, Sun B, Zhao X, Gu Q, Dong X, Liu Z, Ma Y, Zhao N, Liu Y, Chi J, et al. The in-vitro spheroid culture induces a more highly differentiated but tumorigenic population from melanoma cell lines. Melanoma Res. 2013;23(4):254–63.PubMed Mo J, Sun B, Zhao X, Gu Q, Dong X, Liu Z, Ma Y, Zhao N, Liu Y, Chi J, et al. The in-vitro spheroid culture induces a more highly differentiated but tumorigenic population from melanoma cell lines. Melanoma Res. 2013;23(4):254–63.PubMed
37.
go back to reference Davey RJ, Westhuizen A, Bowden NA. Metastatic melanoma treatment: Combining old and new therapies. Crit Rev Oncol Hematol. 2016;98:242–53.CrossRefPubMed Davey RJ, Westhuizen A, Bowden NA. Metastatic melanoma treatment: Combining old and new therapies. Crit Rev Oncol Hematol. 2016;98:242–53.CrossRefPubMed
38.
go back to reference Liu PP, Liao J, Tang ZJ, Wu WJ, Yang J, Zeng ZL, Hu Y, Wang P, Ju HQ, Xu RH, et al. Metabolic regulation of cancer cell side population by glucose through activation of the Akt pathway. Cell Death Differ. 2014;21(1):124–35.CrossRefPubMedPubMedCentral Liu PP, Liao J, Tang ZJ, Wu WJ, Yang J, Zeng ZL, Hu Y, Wang P, Ju HQ, Xu RH, et al. Metabolic regulation of cancer cell side population by glucose through activation of the Akt pathway. Cell Death Differ. 2014;21(1):124–35.CrossRefPubMedPubMedCentral
39.
go back to reference Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, Peng J, Lin E, Wang Y, Sosman J, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487(7408):505–9.CrossRefPubMedPubMedCentral Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, Peng J, Lin E, Wang Y, Sosman J, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487(7408):505–9.CrossRefPubMedPubMedCentral
40.
go back to reference Liu YR, Sun B, Zhao XL, Gu Q, Liu ZY, Dong XY, Che N, Mo J. Basal caspase-3 activity promotes migration, invasion, and vasculogenic mimicry formation of melanoma cells. Melanoma Res. 2013;23(4):243–53.PubMed Liu YR, Sun B, Zhao XL, Gu Q, Liu ZY, Dong XY, Che N, Mo J. Basal caspase-3 activity promotes migration, invasion, and vasculogenic mimicry formation of melanoma cells. Melanoma Res. 2013;23(4):243–53.PubMed
41.
go back to reference Furlan A, Roux B, Lamballe F, Conti F, Issaly N, Daian F, Guillemot JF, Richelme S, Contensin M, Bosch J, et al. Combined drug action of 2-phenylimidazo [2,1-b] benzothiazole derivatives on cancer cells according to their oncogenic molecular signatures. PLoS One. 2012;7(10):e46738.CrossRefPubMedPubMedCentral Furlan A, Roux B, Lamballe F, Conti F, Issaly N, Daian F, Guillemot JF, Richelme S, Contensin M, Bosch J, et al. Combined drug action of 2-phenylimidazo [2,1-b] benzothiazole derivatives on cancer cells according to their oncogenic molecular signatures. PLoS One. 2012;7(10):e46738.CrossRefPubMedPubMedCentral
43.
go back to reference Vlashi E, Pajonk F. The metabolic state of cancer stem cells-a valid target for cancer therapy? Free Radic Biol Med. 2015;79:264–8.CrossRefPubMed Vlashi E, Pajonk F. The metabolic state of cancer stem cells-a valid target for cancer therapy? Free Radic Biol Med. 2015;79:264–8.CrossRefPubMed
44.
go back to reference Shakhova O, Sommer L. Testing the cancer stem cell hypothesis in melanoma: the clinics will tell. Cancer Lett. 2013;338(1):74–81.CrossRefPubMed Shakhova O, Sommer L. Testing the cancer stem cell hypothesis in melanoma: the clinics will tell. Cancer Lett. 2013;338(1):74–81.CrossRefPubMed
45.
go back to reference Quan L, Shi J, Tian Y, Zhang Q, Zhang Y, Hui Q, Tao K. Identification of potential therapeutic targets for melanoma using gene expression analysis. Neoplasma. 2015;62(5):733–9.CrossRefPubMed Quan L, Shi J, Tian Y, Zhang Q, Zhang Y, Hui Q, Tao K. Identification of potential therapeutic targets for melanoma using gene expression analysis. Neoplasma. 2015;62(5):733–9.CrossRefPubMed
46.
go back to reference Bohovic R, Demkova L, Cihova M, Skolekova S, Durinikova E, Toro L, Tyciakova S, Kozovska Z, Matuskova M, Kucerova L. 3D multicellular models reflect the efficiency of MSC-directed enzyme/prodrug treatment. Neoplasma. 2015;62(4):521–30.CrossRefPubMed Bohovic R, Demkova L, Cihova M, Skolekova S, Durinikova E, Toro L, Tyciakova S, Kozovska Z, Matuskova M, Kucerova L. 3D multicellular models reflect the efficiency of MSC-directed enzyme/prodrug treatment. Neoplasma. 2015;62(4):521–30.CrossRefPubMed
Metadata
Title
Tyrosine kinase inhibitor SU11274 increased tumorigenicity and enriched for melanoma-initiating cells by bioenergetic modulation
Authors
Lucia Kucerova
Lucia Demkova
Svetlana Skolekova
Roman Bohovic
Miroslava Matuskova
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2016
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2341-y

Other articles of this Issue 1/2016

BMC Cancer 1/2016 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