Skip to main content
Top
Published in: International Journal of Hematology 1/2015

01-07-2015 | Original Article

The mechanism of synergistic effects of arsenic trioxide and rapamycin in acute myeloid leukemia cell lines lacking typical t(15;17) translocation

Authors: Vilma Dembitz, Hrvoje Lalic, Alen Ostojic, Radovan Vrhovac, Hrvoje Banfic, Dora Visnjic

Published in: International Journal of Hematology | Issue 1/2015

Login to get access

Abstract

Arsenic trioxide (ATO) has potent clinical activity in the treatment of patients with acute promyelocytic leukemia (APL), but is much less efficacious in acute myeloid leukemia (AML) lacking t(15;17) translocation. Recent studies have indicated that the addition of mammalian target of rapamycin (mTOR) inhibitors may increase the sensitivity of malignant cells to ATO. The aim of the present study was to test for possible synergistic effects of ATO and rapamycin at therapeutically achievable doses in non-APL AML cells. In HL-60 and U937 cell lines, the inhibitory effects of low concentrations of ATO and rapamycin were synergistic and more pronounced in U937 cells. The combination of drugs increased apoptosis in HL-60 cells and increased the percentage of cells in G0/G1 phase in both cell lines. In U937 cells, rapamycin alone increased the activity of mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) and the addition of ATO decreased the level of phosphorylated ERK, Ser473 phosphorylated Akt and anti-apoptotic Mcl-1 protein. Primary AML cells show high sensitivity to growth-inhibitory effects of rapamycin alone or in combination with ATO. The results of the present study reveal the mechanism of the synergistic effects of two drugs at therapeutically achievable doses in non-APL AML cells.
Literature
1.
go back to reference Chen SJ, Zhou GB, Zhang XW, Mao JH, de Thé H, Chen Z. From an old remedy to a magic bullet: molecular mechanisms underlying the therapeutic effects of arsenic in fighting leukemia. Blood. 2011;117:6425–37.PubMedCentralPubMedCrossRef Chen SJ, Zhou GB, Zhang XW, Mao JH, de Thé H, Chen Z. From an old remedy to a magic bullet: molecular mechanisms underlying the therapeutic effects of arsenic in fighting leukemia. Blood. 2011;117:6425–37.PubMedCentralPubMedCrossRef
2.
go back to reference Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando SM, Iacobelli S, et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med. 2013;369:111–21.PubMedCrossRef Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando SM, Iacobelli S, et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med. 2013;369:111–21.PubMedCrossRef
3.
go back to reference de Thé H, Le Bras M, Lallemand-Breitenbach V. The cell biology of disease: acute promyelocytic leukemia, arsenic, and PML bodies. J Cell Biol. 2012;198:11–21.PubMedCentralPubMedCrossRef de Thé H, Le Bras M, Lallemand-Breitenbach V. The cell biology of disease: acute promyelocytic leukemia, arsenic, and PML bodies. J Cell Biol. 2012;198:11–21.PubMedCentralPubMedCrossRef
4.
go back to reference Nasr R, Guillemin MC, Ferhi O, Soilihi H, Peres L, Berthier C, et al. Eradication of acute promyelocytic leukemia-initiating cells through PML-RARA degradation. Nat Med. 2008;14:1333–42.PubMedCrossRef Nasr R, Guillemin MC, Ferhi O, Soilihi H, Peres L, Berthier C, et al. Eradication of acute promyelocytic leukemia-initiating cells through PML-RARA degradation. Nat Med. 2008;14:1333–42.PubMedCrossRef
5.
go back to reference Dai J, Weinberg RS, Waxman S, Jing Y. Malignant cells can be sensitized to undergo growth inhibition and apoptosis by arsenic trioxide through modulation of the glutathione redox system. Blood. 1999;93:268–77.PubMed Dai J, Weinberg RS, Waxman S, Jing Y. Malignant cells can be sensitized to undergo growth inhibition and apoptosis by arsenic trioxide through modulation of the glutathione redox system. Blood. 1999;93:268–77.PubMed
6.
go back to reference McCollum G, Keng PC, States JC, McCabe MJ Jr. Arsenite delays progression through each cell cycle phase and induces apoptosis following G2/M arrest in U937 myeloid leukemia cells. J Pharmacol Exp Ther. 2005;313:877–87.PubMedCrossRef McCollum G, Keng PC, States JC, McCabe MJ Jr. Arsenite delays progression through each cell cycle phase and induces apoptosis following G2/M arrest in U937 myeloid leukemia cells. J Pharmacol Exp Ther. 2005;313:877–87.PubMedCrossRef
7.
go back to reference Stępnik M, Ferlińska M, Smok-Pieniążek A, Gradecka-Meesters D, Arkusz J, Stańczyk M. Assessment of the involvement of oxidative stress and mitogen-activated protein kinase signaling pathways in the cytotoxic effects of arsenic trioxide and its combination with sulindac or its metabolites: sulindac sulfide and sulindac sulfone on human leukemic cell lines. Med Oncol. 2012;29:1161–72.PubMedCrossRef Stępnik M, Ferlińska M, Smok-Pieniążek A, Gradecka-Meesters D, Arkusz J, Stańczyk M. Assessment of the involvement of oxidative stress and mitogen-activated protein kinase signaling pathways in the cytotoxic effects of arsenic trioxide and its combination with sulindac or its metabolites: sulindac sulfide and sulindac sulfone on human leukemic cell lines. Med Oncol. 2012;29:1161–72.PubMedCrossRef
8.
go back to reference Ramos AM, Fernández C, Amrán D, Sancho P, de Blas E, Aller P. Pharmacologic inhibitors of PI3K/Akt potentiate the apoptotic action of the antileukemic drug arsenic trioxide via glutathione depletion and increased peroxide accumulation in myeloid leukemia cells. Blood. 2005;105:4013–20.PubMedCrossRef Ramos AM, Fernández C, Amrán D, Sancho P, de Blas E, Aller P. Pharmacologic inhibitors of PI3K/Akt potentiate the apoptotic action of the antileukemic drug arsenic trioxide via glutathione depletion and increased peroxide accumulation in myeloid leukemia cells. Blood. 2005;105:4013–20.PubMedCrossRef
9.
go back to reference Wetzler M, Andrews C, Ford LA, Tighe S, Barcos M, Sait SN, et al. Phase 1 study of arsenic trioxide, high-dose cytarabine, and idarubicin to down-regulate constitutive signal transducer and activator of transcription 3 activity in patients aged <60 years with acute myeloid leukemia. Cancer. 2011;117:4861–8.PubMedCrossRef Wetzler M, Andrews C, Ford LA, Tighe S, Barcos M, Sait SN, et al. Phase 1 study of arsenic trioxide, high-dose cytarabine, and idarubicin to down-regulate constitutive signal transducer and activator of transcription 3 activity in patients aged <60 years with acute myeloid leukemia. Cancer. 2011;117:4861–8.PubMedCrossRef
10.
go back to reference Wang R, Xia L, Gabrilove J, Waxman S, Jing Y. Downregulation of Mcl-1 through GSK-3β activation contributes to arsenic trioxide-induced apoptosis in acute myeloid leukemia cells. Leukemia. 2013;27:315–24.PubMedCentralPubMedCrossRef Wang R, Xia L, Gabrilove J, Waxman S, Jing Y. Downregulation of Mcl-1 through GSK-3β activation contributes to arsenic trioxide-induced apoptosis in acute myeloid leukemia cells. Leukemia. 2013;27:315–24.PubMedCentralPubMedCrossRef
12.
go back to reference Récher C, Beyne-Rauzy O, Demur C, Chicanne G, Dos Santos C, Mas VM, et al. Antileukemic activity of rapamycin in acute myeloid leukemia. Blood. 2005;105:2527–34.PubMedCrossRef Récher C, Beyne-Rauzy O, Demur C, Chicanne G, Dos Santos C, Mas VM, et al. Antileukemic activity of rapamycin in acute myeloid leukemia. Blood. 2005;105:2527–34.PubMedCrossRef
13.
go back to reference Wall M, Poortinga G, Hannan KM, Pearson RB, Hannan RD, McArthur GA. Translational control of c-MYC by rapamycin promotes terminal myeloid differentiation. Blood. 2008;112:2305–17.PubMedCrossRef Wall M, Poortinga G, Hannan KM, Pearson RB, Hannan RD, McArthur GA. Translational control of c-MYC by rapamycin promotes terminal myeloid differentiation. Blood. 2008;112:2305–17.PubMedCrossRef
14.
go back to reference Iwanami A, Gini B, Zanca C, Matsutani T, Assuncao A, Nael A, et al. PML mediates glioblastoma resistance to mammalian target of rapamycin (mTOR)-targeted therapies. Proc Natl Acad Sci USA. 2013;110:4339–44.PubMedCentralPubMedCrossRef Iwanami A, Gini B, Zanca C, Matsutani T, Assuncao A, Nael A, et al. PML mediates glioblastoma resistance to mammalian target of rapamycin (mTOR)-targeted therapies. Proc Natl Acad Sci USA. 2013;110:4339–44.PubMedCentralPubMedCrossRef
15.
go back to reference Liu N, Tai S, Ding B, Thor RK, Bhuta S, Sun Y, et al. Arsenic trioxide synergizes with everolimus (Rad001) to induce cytotoxicity of ovarian cancer cells through increased autophagy and apoptosis. Endocr Relat Cancer. 2012;19:711–23.PubMedCrossRef Liu N, Tai S, Ding B, Thor RK, Bhuta S, Sun Y, et al. Arsenic trioxide synergizes with everolimus (Rad001) to induce cytotoxicity of ovarian cancer cells through increased autophagy and apoptosis. Endocr Relat Cancer. 2012;19:711–23.PubMedCrossRef
16.
go back to reference Guilbert C, Annis MG, Dong Z, Siegel PM, Miller WH Jr, Mann KK. Arsenic trioxide overcomes rapamycin-induced feedback activation of AKT and ERK signaling to enhance the anti-tumor effects in breast cancer. PLoS One. 2013;8(12):e85995.PubMedCentralPubMedCrossRef Guilbert C, Annis MG, Dong Z, Siegel PM, Miller WH Jr, Mann KK. Arsenic trioxide overcomes rapamycin-induced feedback activation of AKT and ERK signaling to enhance the anti-tumor effects in breast cancer. PLoS One. 2013;8(12):e85995.PubMedCentralPubMedCrossRef
17.
go back to reference Altman JK, Yoon P, Katsoulidis E, Kroczynska B, Sassano A, Redig AJ, et al. Regulatory effects of mammalian target of rapamycin-mediated signals in the generation of arsenic trioxide responses. J Biol Chem. 2008;283:1992–2001.PubMedCrossRef Altman JK, Yoon P, Katsoulidis E, Kroczynska B, Sassano A, Redig AJ, et al. Regulatory effects of mammalian target of rapamycin-mediated signals in the generation of arsenic trioxide responses. J Biol Chem. 2008;283:1992–2001.PubMedCrossRef
18.
go back to reference Matkovic K, Brugnoli F, Bertagnolo V, Banfic H, Visnjic D. The role of the nuclear Akt activation and Akt inhibitors in all-trans-retinoic acid-differentiated HL-60 cells. Leukemia. 2006;20:941–51.PubMedCrossRef Matkovic K, Brugnoli F, Bertagnolo V, Banfic H, Visnjic D. The role of the nuclear Akt activation and Akt inhibitors in all-trans-retinoic acid-differentiated HL-60 cells. Leukemia. 2006;20:941–51.PubMedCrossRef
19.
go back to reference Mise J, Dembitz V, Banfic H, Visnjic D. Combined inhibition of PI3K and mTOR exerts synergistic antiproliferative effect, but diminishes differentiative properties of rapamycin in acute myeloid leukemia cells. Pathol Oncol Res. 2011;17:645–56.PubMedCrossRef Mise J, Dembitz V, Banfic H, Visnjic D. Combined inhibition of PI3K and mTOR exerts synergistic antiproliferative effect, but diminishes differentiative properties of rapamycin in acute myeloid leukemia cells. Pathol Oncol Res. 2011;17:645–56.PubMedCrossRef
20.
go back to reference Lalic H, Lukinovic-Skudar V, Banfic H, Visnjic D. Rapamycin enhances dimethyl sulfoxide-mediated growth arrest in human myelogenous leukemia cells. Leuk Lymph. 2012;53:2253–61.CrossRef Lalic H, Lukinovic-Skudar V, Banfic H, Visnjic D. Rapamycin enhances dimethyl sulfoxide-mediated growth arrest in human myelogenous leukemia cells. Leuk Lymph. 2012;53:2253–61.CrossRef
21.
go back to reference Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70:440–6.PubMedCrossRef Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70:440–6.PubMedCrossRef
22.
go back to reference Dalton WT Jr, Ahearn MJ, McCredie KB, Freireich EJ, Stass SA, Trujillo JM. Hl-60 cell line was derived from a patient with FAB-M2 and not FAB-M3. Blood. 1988;71:242–7.PubMed Dalton WT Jr, Ahearn MJ, McCredie KB, Freireich EJ, Stass SA, Trujillo JM. Hl-60 cell line was derived from a patient with FAB-M2 and not FAB-M3. Blood. 1988;71:242–7.PubMed
23.
go back to reference Carracedo A, Ma L, Teruya-Feldstein J, Rojo F, Salmena L, Alimonti A, et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest. 2008;118:3065–74.PubMedCentralPubMed Carracedo A, Ma L, Teruya-Feldstein J, Rojo F, Salmena L, Alimonti A, et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest. 2008;118:3065–74.PubMedCentralPubMed
24.
go back to reference Tamburini J, Chapuis N, Bardet V, Park S, Sujobert P, Willems L, et al. Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways. Blood. 2008;111:379–82.PubMedCrossRef Tamburini J, Chapuis N, Bardet V, Park S, Sujobert P, Willems L, et al. Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways. Blood. 2008;111:379–82.PubMedCrossRef
25.
go back to reference Mills JR, Hippo Y, Robert F, Chen SM, Malina A, Lin CJ, Trojahn U, Wendel HG, Charest A, Bronson RT, Kogan SC, Nadon R, Housman DE, Lowe SW, Pelletier J. mTORC1 promotes survival through translational control of Mcl-1. Proc Natl Acad Sci USA. 2008;105:10853–8.PubMedCentralPubMedCrossRef Mills JR, Hippo Y, Robert F, Chen SM, Malina A, Lin CJ, Trojahn U, Wendel HG, Charest A, Bronson RT, Kogan SC, Nadon R, Housman DE, Lowe SW, Pelletier J. mTORC1 promotes survival through translational control of Mcl-1. Proc Natl Acad Sci USA. 2008;105:10853–8.PubMedCentralPubMedCrossRef
26.
go back to reference Chen GQ, Shi XG, Tang W, Xiong SM, Zhu J, Cai X, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): I. As2O3 exerts dose-dependent dual effects on APL cells. Blood. 1997;89:3345–53.PubMed Chen GQ, Shi XG, Tang W, Xiong SM, Zhu J, Cai X, et al. Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): I. As2O3 exerts dose-dependent dual effects on APL cells. Blood. 1997;89:3345–53.PubMed
27.
go back to reference Nishioka C, Ikezoe T, Yang J, Nishioka C, Ikezoe T, Yang J, et al. Inhibition of mammalian target of rapamycin signaling potentiates the effects of all-trans retinoic acid to induce growth arrest and differentiation of human acute myelogenous leukemia cells. Int J Cancer. 2009;125:1710–20.PubMedCrossRef Nishioka C, Ikezoe T, Yang J, Nishioka C, Ikezoe T, Yang J, et al. Inhibition of mammalian target of rapamycin signaling potentiates the effects of all-trans retinoic acid to induce growth arrest and differentiation of human acute myelogenous leukemia cells. Int J Cancer. 2009;125:1710–20.PubMedCrossRef
28.
go back to reference Lalic H, Dembitz V, Lukinovic-Skudar V, Banfic H, Visnjic D. 5-Aminoimidazole-4-carboxamide ribonucleoside induces differentiation of acute myeloid leukemia cells. Leuk Lymph. 2014;55:2375–83.CrossRef Lalic H, Dembitz V, Lukinovic-Skudar V, Banfic H, Visnjic D. 5-Aminoimidazole-4-carboxamide ribonucleoside induces differentiation of acute myeloid leukemia cells. Leuk Lymph. 2014;55:2375–83.CrossRef
29.
go back to reference Verges B, Walter T, Cariou B. Endocrine side effects of anti-cancer drugs: effects of anti-cancer targeted therapies on lipid and glucose metabolism. Eur J Endocrinol. 2014;170:R43–55.PubMedCrossRef Verges B, Walter T, Cariou B. Endocrine side effects of anti-cancer drugs: effects of anti-cancer targeted therapies on lipid and glucose metabolism. Eur J Endocrinol. 2014;170:R43–55.PubMedCrossRef
30.
go back to reference Suganuma K, Miwa H, Imai N, Shikami M, Gotou M, Goto M, et al. Energy metabolism of leukemia cells: glycolysis versus oxidative phosphorylation. Leuk Lymph. 2010;51:2112–9.CrossRef Suganuma K, Miwa H, Imai N, Shikami M, Gotou M, Goto M, et al. Energy metabolism of leukemia cells: glycolysis versus oxidative phosphorylation. Leuk Lymph. 2010;51:2112–9.CrossRef
31.
go back to reference Sims JT, Plattner R. MTT assays cannot be utilized to study the effects of STI571/Gleevec on the viability of solid tumor cell lines. Cancer Chemother Pharmacol. 2009;64:629–33.PubMedCentralPubMedCrossRef Sims JT, Plattner R. MTT assays cannot be utilized to study the effects of STI571/Gleevec on the viability of solid tumor cell lines. Cancer Chemother Pharmacol. 2009;64:629–33.PubMedCentralPubMedCrossRef
32.
go back to reference Wang X, Yue P, Kim YA, Fu H, Khuri FR, Sun S. Enhancing mammalian target of rapamycin (mTOR)-targeted cancer therapy by preventing mTOR/Raptor inhibition-initiated, mTOR/Rictor-independent Akt activation. Cancer Res. 2008;68:7409–18.PubMedCentralPubMedCrossRef Wang X, Yue P, Kim YA, Fu H, Khuri FR, Sun S. Enhancing mammalian target of rapamycin (mTOR)-targeted cancer therapy by preventing mTOR/Raptor inhibition-initiated, mTOR/Rictor-independent Akt activation. Cancer Res. 2008;68:7409–18.PubMedCentralPubMedCrossRef
33.
go back to reference Zeng Z, Sarbassov DD, Samudio IJ, Yee KWL, Munsell MF, Jackson CE, et al. Rapamycin derivatives reduce mTORC2 signaling and inhibit Akt activation i AML. Blood. 2007;109:3509–12.PubMedCentralPubMedCrossRef Zeng Z, Sarbassov DD, Samudio IJ, Yee KWL, Munsell MF, Jackson CE, et al. Rapamycin derivatives reduce mTORC2 signaling and inhibit Akt activation i AML. Blood. 2007;109:3509–12.PubMedCentralPubMedCrossRef
34.
go back to reference Altman JK, Sassano A, Kaur S, Glaser H, Kroczynska B, Redig AJ, et al. Dual mTORC2/mTORC1 targeting results in potent suppressive effectson acute myeloid leukemia (AML) progenitors. Clin Cancer Res. 2011;17:4378–88.PubMedCentralPubMedCrossRef Altman JK, Sassano A, Kaur S, Glaser H, Kroczynska B, Redig AJ, et al. Dual mTORC2/mTORC1 targeting results in potent suppressive effectson acute myeloid leukemia (AML) progenitors. Clin Cancer Res. 2011;17:4378–88.PubMedCentralPubMedCrossRef
35.
go back to reference Sánchez Y, Simón GP, Calviño E, de Blas E, Aller P. Curcumin stimulates reactive oxygen species production and potentiates apoptosis induction by the antitumor drugs arsenic trioxide and lonidamine in human myeloid leukemia cell lines. J Pharmacol Exp Ther. 2010;335:114–23.PubMedCrossRef Sánchez Y, Simón GP, Calviño E, de Blas E, Aller P. Curcumin stimulates reactive oxygen species production and potentiates apoptosis induction by the antitumor drugs arsenic trioxide and lonidamine in human myeloid leukemia cell lines. J Pharmacol Exp Ther. 2010;335:114–23.PubMedCrossRef
36.
go back to reference Ramirez-Valle F, Badura ML, Braunstein S, Narasimhan M, Schneider RJ. Mitotic raptor promotes mTORC1 activity, G(2)/M cell cycle progression, and internal ribosome entry site-mediated mRNA translation. Mol Cell Biol. 2010;30:3151–64.PubMedCentralPubMedCrossRef Ramirez-Valle F, Badura ML, Braunstein S, Narasimhan M, Schneider RJ. Mitotic raptor promotes mTORC1 activity, G(2)/M cell cycle progression, and internal ribosome entry site-mediated mRNA translation. Mol Cell Biol. 2010;30:3151–64.PubMedCentralPubMedCrossRef
37.
go back to reference Johnson DE. Src family kinases and the MEK/ERK pathway in the regulation of myeloid differentiation and myeloid leukemogenesis. Adv Enzyme Regul. 2008;48:98–112.PubMedCentralPubMedCrossRef Johnson DE. Src family kinases and the MEK/ERK pathway in the regulation of myeloid differentiation and myeloid leukemogenesis. Adv Enzyme Regul. 2008;48:98–112.PubMedCentralPubMedCrossRef
38.
go back to reference Biggs JR, Ahn NG, Kraft AS. Activation of the mitogen-activated protein kinase pathway in U937 leukemic cells induces phosphorylation of the amino terminus of the TATA-binding protein. Cell Growth Differ. 1998;9:667–76.PubMed Biggs JR, Ahn NG, Kraft AS. Activation of the mitogen-activated protein kinase pathway in U937 leukemic cells induces phosphorylation of the amino terminus of the TATA-binding protein. Cell Growth Differ. 1998;9:667–76.PubMed
39.
go back to reference Kandilci A, Grosveld GC. SET-induced calcium signaling and MAPK/ERK pathway activation mediate dendritic cell-like differentiation of U937 cells. Leukemia. 2005;19:1439–45.PubMedCrossRef Kandilci A, Grosveld GC. SET-induced calcium signaling and MAPK/ERK pathway activation mediate dendritic cell-like differentiation of U937 cells. Leukemia. 2005;19:1439–45.PubMedCrossRef
40.
go back to reference Yan H, Peng ZG, Wu YL, Jiang Y, Yu Y, Huang Y, et al. Hypoxia-simulating agents and selective stimulation of arsenic trioxide-induced growth arrest and cell differentiation in acute promyelocytic leukemic cells. Haematologica. 2005;90:1607–16.PubMed Yan H, Peng ZG, Wu YL, Jiang Y, Yu Y, Huang Y, et al. Hypoxia-simulating agents and selective stimulation of arsenic trioxide-induced growth arrest and cell differentiation in acute promyelocytic leukemic cells. Haematologica. 2005;90:1607–16.PubMed
41.
go back to reference Yamamoto-Yamaguchi Y, Okabe-Kado J, Kasukabe T, Honma Y. Induction of differentiation of human myeloid leukemia cells by immunosuppressant macrolides (rapamycin and FK506) and calcium/calmodulin-dependent kinase inhibitors. Exp Hematol. 2001;29:582–8.PubMedCrossRef Yamamoto-Yamaguchi Y, Okabe-Kado J, Kasukabe T, Honma Y. Induction of differentiation of human myeloid leukemia cells by immunosuppressant macrolides (rapamycin and FK506) and calcium/calmodulin-dependent kinase inhibitors. Exp Hematol. 2001;29:582–8.PubMedCrossRef
42.
go back to reference Yang J, Ikezoe T, Nishioka C, Ni L, Koeffler HP, Yokoyama A. Inhibition of mTORC1 by RAD001 (everolimus) potentiates the effect of 1,25-dihydroxyvitamin D3 to induce growth arrest and differentiation of AML cells in vitro and in vivo. Exp Hematol. 2010;38:666–76.PubMedCrossRef Yang J, Ikezoe T, Nishioka C, Ni L, Koeffler HP, Yokoyama A. Inhibition of mTORC1 by RAD001 (everolimus) potentiates the effect of 1,25-dihydroxyvitamin D3 to induce growth arrest and differentiation of AML cells in vitro and in vivo. Exp Hematol. 2010;38:666–76.PubMedCrossRef
43.
go back to reference Mercalli A, Calavita I, Dugnani E, Citro A, Cantarelli E, Nano R, et al. Rapamycin unbalances the polarization of human macrophages to M1. Immunology. 2013;140:179–90.PubMedCentralPubMedCrossRef Mercalli A, Calavita I, Dugnani E, Citro A, Cantarelli E, Nano R, et al. Rapamycin unbalances the polarization of human macrophages to M1. Immunology. 2013;140:179–90.PubMedCentralPubMedCrossRef
44.
go back to reference Hackstein H, Taner T, Logar AJ, Thomson AW. Rapamycin inhibits macropinocytosis and mannose receptor-mediated endocytosis by bone marrow-derived dendritic cells. Blood. 2002;100:1084–7.PubMedCrossRef Hackstein H, Taner T, Logar AJ, Thomson AW. Rapamycin inhibits macropinocytosis and mannose receptor-mediated endocytosis by bone marrow-derived dendritic cells. Blood. 2002;100:1084–7.PubMedCrossRef
Metadata
Title
The mechanism of synergistic effects of arsenic trioxide and rapamycin in acute myeloid leukemia cell lines lacking typical t(15;17) translocation
Authors
Vilma Dembitz
Hrvoje Lalic
Alen Ostojic
Radovan Vrhovac
Hrvoje Banfic
Dora Visnjic
Publication date
01-07-2015
Publisher
Springer Japan
Published in
International Journal of Hematology / Issue 1/2015
Print ISSN: 0925-5710
Electronic ISSN: 1865-3774
DOI
https://doi.org/10.1007/s12185-015-1776-2

Other articles of this Issue 1/2015

International Journal of Hematology 1/2015 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