A mechanism for increased sensitivity of acute myeloid leukemia to mitotoxic drugs

Svetlana B Panina, Natalia Baran, Fabio H Brasil da Costa, Marina Konopleva, Natalia V Kirienko, Svetlana B Panina, Natalia Baran, Fabio H Brasil da Costa, Marina Konopleva, Natalia V Kirienko

Abstract

Mitochondria play a central and multifunctional role in the progression of tumorigenesis. Although many recent studies have demonstrated correlations between mitochondrial function and genetic makeup or originating tissue, it remains unclear why some cancers are more susceptible to mitocans (anticancer drugs that target mitochondrial function to mediate part or all of their effect). Moreover, fundamental questions of efficacy and mechanism of action in various tumor types stubbornly remain. Here we demonstrate that cancer type is a significant predictor of tumor response to mitocan treatment, and that acute myeloid leukemias (AML) show an increased sensitivity to these drugs. We determined that AML cells display particular defects in mitochondrial metabolism that underlie their sensitivity to mitocan treatment. Furthermore, we demonstrated that combinatorial treatment with a mitocan (CCCP) and a glycolytic inhibitor (2-deoxyglucose) has substantial synergy in AML cells, including primary cells from patients with AML. Our results show that mitocans, either alone or in combination with a glycolytic inhibitor, display anti-leukemia effects in doses much lower than needed to induce toxicity against normal blood cells, indicating that mitochondria may be an effective and selective therapeutic target.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1. Cell lines derived from AML…
Fig. 1. Cell lines derived from AML are more susceptible to mitochondrial damage than cell lines derived from solid tumors.
aZ-scores of tumor cell lines from the NCI-60 panel. Sensitivity to non-mitocan drugs is shown on the x-axis, sensitivity to mitocans is shown on the y-axis. U251, a glioblastoma-derived cell line with high sensitivity to mitocans is shown in red, and ovarian cancer-derived cell lines, which show higher resistance, are shown in blue. b Density plot showing median rank for each possible permutation of six cell lines from the NCI-60 collection. The thick black line represents the median for leukemia cell lines (5.5) from the NCI-60 panel. c Sensitivity of AML (MV-4-11, THP-1, OCI-AML2, and MOLM-13), normal PBMCs, and solid tumor (U251, SKOV3, and OVCAR3) cell lines to mitocan treatment. Shown are LD50 values with 95% confidence intervals for MTX, DOX, CCCP, and ara-C based on results from 3–5 independent experiments. Comparisons of LD50 were done by the ratio test, the asterisk indicates significant difference compared with the next most sensitive cell line. d Fluorescence micrographs of MV-4-11 (top) or THP-1 (bottom) cells treated with either vehicle (left) or LD50 concentrations of CCCP. Cells were stained with acridine orange/propidium iodide. e The ratio of mitochondrial to genomic DNA was determined by quantitative PCR. Shown are mean values with SD. Statistical significance for comparison AML vs. healthy PBMCs was analyzed via the Student’s t-test. f Mitochondrial health in untreated MOLM-13 cells and normal PBMCs were compared, including mitochondrial mass (assessed via staining with MitoTracker Green), membrane potential (assessed via staining with JC-1), metabolic rate (assessed via Seahorse analysis of oxygen consumption rate and lactate production), steady-state ATP level, and protein level of the β subunit of ATP synthase. SKOV3, a mitocan-resistant cell line, is shown as a comparison. Shown is the mean of at least three independent experiments (in case of Seahorse data and ATP measurements dots represent all technical replicates), error bars are SD. Statistical analysis was performed using Student’s t-test with independent samples. ***p < 0.001; **p < 0.01; *p < 0.05
Fig. 2. AML-derived cells are more sensitive…
Fig. 2. AML-derived cells are more sensitive to mitocan treatment.
a Relative mtDNA content before and after 24 h exposure to doxorubicin treatment in AML cell lines (left) or in solid tumor cell lines (right). b Correlation between basal mtDNA content and LD50 of mitocans. Normalization of LD50 values was performed using min-max approach, linear functions were adjusted for the data using lm() method in R. Basal ATP level (c) and ATP-linked respiration (d) before and after exposure to doxorubicin for 24 h (c) or 4 h (d). e ATP synthase β subunit protein level before and after 24 h exposure to doxorubicin. β-actin was used as a loading control. f Changes in mitochondrial mass after 24 h doxorubicin treatment. Comparison of MOLM-13 vs. normal PBMCs is shown. g Mitochondrial membrane potential before and after 4 h doxorubicin treatment, as defined by JC-1 staining. Representative plots for red/green fluorescence (PE vs. FITC) after standard compensation for MOLM-13, healthy PBMCs, and SKOV3 cells are shown below for the untreated (blue) and doxorubicin-treated (red) cells. h Expression ratios of mitochondrial fusion/fission genes before and after 8 h doxorubicin treatment. The results of 3–6 independent experiments are presented as mean ± SD (in a, c, f, g, h) or mean ± SEM (in d, e). All technical replicates are shown in c. For statistical significance, either Student’s t-test with independent samples (a, c, eh) or ANOVA with subsequent Dunn’s test (d) was used. ***p < 0.001; **p < 0.01; *p < 0.05; ns: p > 0.05
Fig. 3. Combination therapy exhibits synergy in…
Fig. 3. Combination therapy exhibits synergy in AML cells.
FACI (combination index as a function of fraction affected) plots for combination treatment with CCCP and 2-DG in OCI-AML2, healthy PBMCs, and SKOV3 cells (a) or primary AML cells for one (upper) or multiple (n = 21, lower) patients (b). Non-linear regression was used to generate a function for combined FACI data for primary AML samples. c Drug combination landscapes for OCI-AML2 cells and normal PBMCs built using the Bioconductor package ‘synergyfinder’. d Viability of OCI-AML2 cells and normal PBMCs after treatment with CCCP/2-DG at LD25 concentrations corresponding to OCI-AML2 cell line. e ATP level before and after treatment with CCCP and 2-DG in AML cell lines, healthy PBMCs, and solid tumor cell lines (upper) or primary AML samples (lower). f Respiration (upper) and coupling efficiency (lower) before and after treatment with CCCP, 2-DG, or both. g Viability of OCI-AML2 and normal PBMC cells after treatment with ABT-199/2-DG. All graphs show results from at least three independent experiments (except b, where primary AML cells were limited by patient samples). FACI plots reflecting combination indices with 95%-confidence intervals were built using the Bioconductor package ‘drc’, * significant difference from combination index = 1 (additivity). Elsewhere, results are presented as mean ± SD (d, e, g) or mean ± SEM (f). Statistical significance was tested using Student’s t-test with independent samples (d, e, g) or by ANOVA with subsequent Dunn’s test (f). ***p < 0.001; **p < 0.01; *p < 0.05; ns: p > 0.05
Fig. 4. Bioenergetic profiling of AML cell…
Fig. 4. Bioenergetic profiling of AML cell lines, healthy PBMCs, and solid tumor cells.
a Oxygen consumption rate (OCR) measured in untreated cells using a Seahorse flux analyzer. Oligomycin (1), FCCP (2), and antimycin A (3) were added at the times indicated. b OCR measured in cells either untreated (blue) or treated with doxorubicin (red), CCCP (black), 2-DG (purple), or CCCP and 2-DG (orange). c ECAR in untreated AML, PBMCs, and SKOV3 cells. d Basal OCR, ECAR, ATP-linked OCR, proton leak, and coupling efficiency for AML cell lines, PBMCs, and a mitocan-resistant solid tumor cell line. (red—AML vs. PBMCs, blue—AML vs. SKOV3). e Intracellular ROS measured by dihydroethidium (DHE, 2.5 μg/ml) after 24 h treatment with CCCP. Mean fluorescence intensities (MFI) of DHE and Hoechst 33342 were quantified using ZEN software and presented as MFI ratios DHE/Hoechst 33342. All graphs show results as mean ± SEM from 3–6 independent experiments. Statistical testing was performed by Student's t-test with independent samples (e) or ANOVA with subsequent Dunn’s or Fisher’s LSD test (d). ***p < 0.001; **p < 0.01; *p < 0.05; ns: p > 0.05
Fig. 5. Mitocan treatment activates multiple cell…
Fig. 5. Mitocan treatment activates multiple cell death pathways.
Survival rates of AML cells and healthy PBMCs treated with mitoxantrone (a), doxorubicin (b), CCCP (c), or 2-DG (d) at LD50 doses. For each drug, cells were also tested with or without the following cell death pathway inhibitors: z-VAD-fmk (40 μM, pan-caspase inhibitor), 3-methyladenine (5 mM, autophagy inhibitor), or VX-765 (10 μM, caspase-1 inhibitor). Results show the mean ± SD from at least three replicates. Statistical testing was performed by Student’s t-test with independent samples. ***p < 0.001; **p < 0.01; *p < 0.05; ns: p > 0.05

References

    1. Pelicano H, Martin DS, Xu RH, Huang P. Glycolysis inhibition for anticancer treatment. Oncogene. 2006;25:4633–4646. doi: 10.1038/sj.onc.1209597.
    1. Sánchez-Mendoza SE, Rego EM. Targeting the mitochondria in acute myeloid leukemia. Appl. Cancer Res. 2017;37:22. doi: 10.1186/s41241-017-0022-z.
    1. Lee HC, Wei YH. Mitochondrial DNA instability and metabolic shift in human cancers. Int J. Mol. Sci. 2009;10:674–701. doi: 10.3390/ijms10020674.
    1. Zorova LD, et al. Mitochondrial membrane potential. Anal. Biochem. 2018;552:50–59. doi: 10.1016/j.ab.2017.07.009.
    1. Cui Q, Wen S, Huang P. Targeting cancer cell mitochondria as a therapeutic approach: recent updates. Future Med. Chem. 2017;9:929–949. doi: 10.4155/fmc-2017-0011.
    1. Neuzil J, Dong LF, Rohlena J, Truksa J, Ralph SJ. Classification of mitocans, anti-cancer drugs acting on mitochondria. Mitochondrion. 2013;13:199–208. doi: 10.1016/j.mito.2012.07.112.
    1. Basak NP, Banerjee S. Mitochondrial dependency in progression of acute myeloid leukemia. Mitochondrion. 2015;21:41–48. doi: 10.1016/j.mito.2015.01.006.
    1. Hahn T, et al. Use of anti-cancer drugs, mitocans, to enhance the immune responses against tumors. Curr. Pharm. Biotechnol. 2013;14:357–376. doi: 10.2174/1389201011314030010.
    1. Vyas S, Zaganjor E, Haigis MC. Mitochondria and cancer. Cell. 2016;166:555–566. doi: 10.1016/j.cell.2016.07.002.
    1. Shoemaker RH. The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer. 2006;6:813–823. doi: 10.1038/nrc1951.
    1. Shankavaram UT, et al. CellMiner: a relational database and query tool for the NCI-60 cancer cell lines. BMC Genom. 2009;10:277. doi: 10.1186/1471-2164-10-277.
    1. Wang L, et al. Oxidative degradation of polyamines by serum supplement causes cytotoxicity on cultured cells. Sci. Rep. 2018;8:10384. doi: 10.1038/s41598-018-28648-8.
    1. Mahbub AA, Le Maitre CL, Haywood-Small SL, Cross NA, Jordan-Mahy N. Polyphenols act synergistically with doxorubicin and etoposide in leukaemia cell lines. Cell Death Discov. 2015;1:15043. doi: 10.1038/cddiscovery.2015.43.
    1. Ritz, C., Baty, F., Streibig, J. C. & Gerhard, D. Dose-response analysis using R. PLoS ONE10, e0146021 (2015).
    1. Wheeler MW, Park RM, Bailer AJ. Comparing median lethal concentration values using confidence interval overlap or ratio tests. Environ. Toxicol. Chem. 2006;25:1441–1444. doi: 10.1897/05-320R.1.
    1. Ritz C, Streibig JC. From additivity to synergism—a modelling perspective. Synergy. 2014;1:22–29. doi: 10.1016/j.synres.2014.07.010.
    1. Ianevski A, He L, Aittokallio T, Tang J. SynergyFinder: a web application for analyzing drug combination dose-response matrix data. Bioinformatics. 2017;33:2413–2415. doi: 10.1093/bioinformatics/btx162.
    1. Mei H, et al. Reduced mtDNA copy number increases the sensitivity of tumor cells to chemotherapeutic drugs. Cell Death Dis. 2015;6:e1710. doi: 10.1038/cddis.2015.78.
    1. Pardee TS, et al. A Phase I Study of CPI-613 in combination with high-dose cytarabine and mitoxantrone for relapsed or refractory acute myeloid leukemia. Clin. Cancer Res. 2018;24:2060–2073. doi: 10.1158/1078-0432.CCR-17-2282.
    1. Pollyea DA, et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat. Med. 2018;24:1859–1866. doi: 10.1038/s41591-018-0233-1.
    1. Cang S, Iragavarapu C, Savooji J, Song Y, Liu D. ABT-199 (venetoclax) and BCL-2 inhibitors in clinical development. J. Hematol. Oncol. 2015;8:129. doi: 10.1186/s13045-015-0224-3.
    1. Konopleva M, Letai A. BCL-2 inhibition in AML: an unexpected bonus? Blood. 2018;132:1007–1012. doi: 10.1182/blood-2018-03-828269.
    1. Jia L, Gribben JG. Dangerous power: mitochondria in CLL cells. Blood. 2014;123:2596–2597. doi: 10.1182/blood-2014-03-558965.
    1. Mailloux RJ, McBride SL, Harper ME. Unearthing the secrets of mitochondrial ROS and glutathione in bioenergetics. Trends Biochem. Sci. 2013;38:592–602. doi: 10.1016/j.tibs.2013.09.001.
    1. Wu YT, et al. Dual role of 3-methyladenine in modulation of autophagy via different temporal patterns of inhibition on class I and III phosphoinositide 3-kinase. J. Biol. Chem. 2010;285:10850–10861. doi: 10.1074/jbc.M109.080796.
    1. Antczak C, Takagi T, Ramirez CN, Radu C, Djaballah H. Live-cell imaging of caspase activation for high-content screening. J. Biomol. Screen. 2009;14:956–969. doi: 10.1177/1087057109343207.
    1. Stack JH, et al. IL-converting enzyme/caspase-1 inhibitor VX-765 blocks the hypersensitive response to an inflammatory stimulus in monocytes from familial cold autoinflammatory syndrome patients. J. Immunol. 2005;175:2630–2634. doi: 10.4049/jimmunol.175.4.2630.
    1. Lu SZ, Harrison-Findik DD. Autophagy and cancer. World J. Biol. Chem. 2013;4:64–70. doi: 10.4331/wjbc.v4.i3.64.
    1. Yan C, Li TS. Dual role of mitophagy in cancer drug resistance. Anticancer Res. 2018;38:617–621. doi: 10.21873/anticanres.13034.
    1. Jitschin R, et al. Mitochondrial metabolism contributes to oxidative stress and reveals therapeutic targets in chronic lymphocytic leukemia. Blood. 2014;123:2663–2672. doi: 10.1182/blood-2013-10-532200.
    1. Sriskanthadevan S, et al. AML cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress. Blood. 2015;125:2120–2130. doi: 10.1182/blood-2014-08-594408.
    1. Vélez J, et al. Mitochondrial uncoupling and the reprograming of intermediary metabolism in leukemia cells. Front Oncol. 2013;3:67. doi: 10.3389/fonc.2013.00067.
    1. Gorini S, et al. Chemotherapeutic Drugs and Mitochondrial Dysfunction: Focus on Doxorubicin, Trastuzumab, and Sunitinib. Oxid. Med. Cell Longev. 2018;2018:7582730. doi: 10.1155/2018/7582730.
    1. Kluza J, et al. Mitochondrial proliferation during apoptosis induced by anticancer agents: effects of doxorubicin and mitoxantrone on cancer and cardiac cells. Oncogene. 2004;23:7018–7030. doi: 10.1038/sj.onc.1207936.
    1. Yan C, et al. Doxorubicin-induced mitophagy contributes to drug resistance in cancer stem cells from HCT8 human colorectal cancer cells. Cancer Lett. 2017;388:34–42. doi: 10.1016/j.canlet.2016.11.018.
    1. Shin HJ, et al. Doxorubicin-induced necrosis is mediated by poly-(ADP-ribose) polymerase 1 (PARP1) but is independent of p53. Sci. Rep. 2015;5:15798. doi: 10.1038/srep15798.
    1. Lee S, Zhang C, Liu X. Role of glucose metabolism and ATP in maintaining PINK1 levels during Parkin-mediated mitochondrial damage responses. J. Biol. Chem. 2015;290:904–917. doi: 10.1074/jbc.M114.606798.
    1. Brand MD, Nicholls DG. Assessing mitochondrial dysfunction in cells. Biochem. J. 2011;435:297–312. doi: 10.1042/BJ20110162.
    1. Roy Chowdhury S, Banerji V. Targeting Mitochondrial Bioenergetics as a Therapeutic Strategy for Chronic Lymphocytic Leukemia. Oxid. Med. Cell Longev. 2018;2018:2426712. doi: 10.1155/2018/2426712.
    1. Brookes PS. Mitochondrial H(+) leak and ROS generation: an odd couple. Free Radic. Biol. Med. 2005;38:12–23. doi: 10.1016/j.freeradbiomed.2004.10.016.
    1. Cheng J, et al. Mitochondrial proton leak plays a critical role in pathogenesis of cardiovascular diseases. Adv. Exp. Med. Biol. 2017;982:359–370. doi: 10.1007/978-3-319-55330-6_20.
    1. Dar S, et al. Bioenergetic adaptations in chemoresistant ovarian cancer cells. Sci. Rep. 2017;7:8760. doi: 10.1038/s41598-017-09206-0.
    1. Pfleger J, He M, Abdellatif M. Mitochondrial complex II is a source of the reserve respiratory capacity that is regulated by metabolic sensors and promotes cell survival. Cell Death Dis. 2015;6:e1835. doi: 10.1038/cddis.2015.202.
    1. Cheng G, et al. Profiling and targeting of cellular bioenergetics: inhibition of pancreatic cancer cell proliferation. Br. J. Cancer. 2014;111:85–93. doi: 10.1038/bjc.2014.272.
    1. Cheng G, et al. Mitochondria-targeted drugs synergize with 2-deoxyglucose to trigger breast cancer cell death. Cancer Res. 2012;72:2634–2644. doi: 10.1158/0008-5472.CAN-11-3928.
    1. Hao JH, Yu M, Liu FT, Newland AC, Jia L. Bcl-2 inhibitors sensitize tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by uncoupling of mitochondrial respiration in human leukemic CEM cells. Cancer Res. 2004;64:3607–3616. doi: 10.1158/0008-5472.CAN-03-3648.
    1. Roberts DJ, Miyamoto S. Hexokinase II integrates energy metabolism and cellular protection: Akting on mitochondria and TORCing to autophagy. Cell Death Differ. 2015;22:248–257. doi: 10.1038/cdd.2014.173.
    1. Chen Z, Zhang H, Lu W, Huang P. Role of mitochondria-associated hexokinase II in cancer cell death induced by 3-bromopyruvate. Biochim. Biophys. Acta. 2009;1787:553–560. doi: 10.1016/j.bbabio.2009.03.003.

Source: PubMed

3
Abonneren