Anticancer Properties of the Antipsychotic Drug Chlorpromazine and Its Synergism With Temozolomide in Restraining Human Glioblastoma Proliferation In Vitro

Silvia Matteoni, Paola Matarrese, Barbara Ascione, Mariachiara Buccarelli, Lucia Ricci-Vitiani, Roberto Pallini, Veronica Villani, Andrea Pace, Marco G Paggi, Claudia Abbruzzese, Silvia Matteoni, Paola Matarrese, Barbara Ascione, Mariachiara Buccarelli, Lucia Ricci-Vitiani, Roberto Pallini, Veronica Villani, Andrea Pace, Marco G Paggi, Claudia Abbruzzese

Abstract

The extremely poor prognosis of patients affected by glioblastoma (GBM, grade IV glioma) prompts the search for new and more effective therapies. In this regard, drug repurposing or repositioning can represent a safe, swift, and inexpensive way to bring novel pharmacological approaches from bench to bedside. Chlorpromazine, a medication used since six decades for the therapy of psychiatric disorders, shows in vitro several features that make it eligible for repositioning in cancer therapy. Using six GBM cell lines, three of which growing as patient-derived neurospheres and displaying stem-like properties, we found that chlorpromazine was able to inhibit viability in an apoptosis-independent way, induce hyperdiploidy, reduce cloning efficiency as well as neurosphere formation and downregulate the expression of stemness genes in all these cell lines. Notably, chlorpromazine synergized with temozolomide, the first-line therapeutic in GBM patients, in hindering GBM cell viability, and both drugs strongly cooperated in reducing cloning efficiency and inducing cell death in vitro for all the GBM cell lines assayed. These results prompted us to start a Phase II clinical trial on GBM patients (EudraCT # 2019-001988-75; ClinicalTrials.gov Identifier: NCT04224441) by adding chlorpromazine to temozolomide in the adjuvant phase of the standard first-line therapeutic protocol.

Keywords: antipsychotic drugs (APDs); cancer stem cells (CSC); clinical trials; drug repurposing and repositioning; drug synergism; glioblastoma; neurospheres.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Matteoni, Matarrese, Ascione, Buccarelli, Ricci-Vitiani, Pallini, Villani, Pace, Paggi and Abbruzzese.

Figures

Figure 1
Figure 1
CPZ reduces cell viability in GBM cells. (A). Representative dose–response curves of all cell lines treated with CPZ for 48 h are shown. GBM anchorage-dependent cell lines are represented by black lines, while RPE-1 by a green line and neurospheres by red ones. (B). Table showing μM CPZ concentrations corresponding to the IC30 and IC50 calculated for each cell line. Values are expressed as mean ± SE.
Figure 2
Figure 2
CPZ induces alterations of the cell cycle and hyperdiploidy in GBM cells. Anchorage-dependent GBM cell lines [panels (A) and (B)] or neurospheres [panels (C) and (D)] underwent treatment with CPZ at the calculated IC30 for 48 h. Flow cytometry analysis of the cell cycle. Left panels (A), (C). Histograms obtained in a representative experiment. Numbers represent the percentage of cells in the different phases of the cell cycle or hyperdiploid cells. Right panels (A), (C). Bar graphs showing mean ± SE of data obtained from three independent experiments. Left panels (B), (D). Representative micrographs of the analysis of nuclear morphology performed after cell staining with Hoechst 33258. Right panels (B), (D). Bar graphs showing the quantification of hyperdiploid cells performed by counting at least 50 cells from 10 different fields observed with a 40× objective. Data are reported as mean ± SE of data obtained from three independent experiments. Statistical significance is referred toward the Control (*** p < 0.001).
Figure 3
Figure 3
CPZ reduces cloning efficiency in GBM cells. (A). Colony-forming assay. Anchorage-dependent cells were treated with increasing doses of CPZ, as indicated. At the end of the experiment, cells were stained and counted. The results are reported in the histogram on the far right of the panel along with statistical significance referred toward the Control (** p < 0.01; *** p < 0.001). (B). Neurosphere formation assay. Cells were treated with increasing doses of CPZ, as indicated. Of note, TS#83 neurospheres displayed a partial anchorage-dependent growing capability. At the end of the experiment, neurospheres were photographed under an inverted microscope using a 4× objective.
Figure 4
Figure 4
CPZ downregulates stemness gene expression in GBM cells. (A). Expression of stemness genes in the three stem-like cell lines (neurospheres). (B). Expression of stemness genes in the three anchorage-dependent cell lines. In all cases, determinations were performed via qRT-PCR after 24 h of exposure to CPZ. Histogram values represent the fold-changes referred to the respective value for untreated cells (Control), arbitrarily reported as 1.0 (gray columns on the left of each graph). Statistical significance is referred toward the Control (* p < 0.05; ** p < 0.01; *** p < 0.001). In TS#83 cells and U-251 MG cells, in panel A and in panel B respectively, the amount of OLIG2 mRNA was undetectable in Control as well in CPZ-treated cells.
Figure 5
Figure 5
CPZ synergizes with TMZ in reducing GBM cell viability. Anchorage-dependent GBM cell lines (A) and neurospheres (B). Dose–response effects of CPZ (blue), TMZ (green) and TMZ plus a constant (k) CPZ concentration, indicated for each cell line (red) on percent cell viability (left panels). Histograms show cell viability at selected drug concentrations, as indicated, to highlight the effect of the association of the two drugs (right panels). The effects of TMZ and CPZ combination were considered synergistic when the CI was <0.8 (in red). CPZ k values were 6.0, 6.0, 4.0, 10.0, 8.0 and 5.0 μM for T98G, U-87 MG, U-251 MG, TS#1, TS#83 and TS163 GBM cells, respectively.
Figure 6
Figure 6
CPZ cooperates with TMZ in inducing cell death. Anchorage-dependent GBM cell lines (A) and neurospheres (B) were analyzed after treatment with TMZ (96 h) and CPZ (48 h), or their combination, at the lowest concentrations considered synergistic on the basis of the viability analysis. Left panels. FACS analysis after staining with Calcein-AM (which is retained in the cytoplasm of live cells). Numbers represent the percentage of Calcein-negative cells (dead cells). One representative experiment is shown. Bar graphs below show the results obtained from four independent experiments, reported as means ± SE. Right panels. FACS analysis after double staining with Annexin V/PI. Dot plots from a representative experiment are shown. Numbers represent the percentages of Annexin V-positive cells (bottom right quadrant), Annexin V/PI double positive cells (upper right quadrant), or PI-positive cells (upper left quadrant). Note the high percentage of cells positive for PI in CPZ treated cells. Bar graphs below show results obtained from four independent experiments, reported as means ± SE. ** p < 0.01 and *** p < 0.001 indicate significant differences vs single drug treatments (TMZ or CPZ).
Figure 7
Figure 7
CPZ cooperates with TMZ in reducing GBM cell cloning efficiency. (A). Anchorage-dependent cells T98G, U-87 MG and U-251 MG were exposed to solvent(s) (Control), TMZ, CPZ or their association at the doses indicated, then rinsed and allowed to grow in the absence of drugs for the subsequent 12 d. Cell colonies, after staining with crystal violet (left), were counted, and the values reported as percent colony number in the histogram (right). In these panels, variance among groups was assessed via the Bartlett’s test for equal variances. Statistical analysis among groups was done using the One-way ANOVA test followed by the Tukey’s Multiple Comparison Test (### significance <0.001 vs. Control; ^^^ significance <0.001 vs. CPZ; §§§ significance <0.001 vs. TMZ; § significance <0.05 vs TMZ). (B). Neurospheres TS#1, TS#83, and TS#163 were treated as described and allowed to grow and form spheres for the subsequent 20 d. For each cell line, the four left images illustrate the effect of TMZ, CPZ or both drugs on neurosphere number and size, observed using a 4× objective. The two framed images on the right are enlarged pictures of the respective neurospheres indicated by the red arrows; these neurospheres were randomly chosen due to their dimensions, in order to appreciate their decrease in volume due to the treatment with the drug combo. This suggests a reduced sphere-forming ability in TMZ + CPZ-treated cells. In this panel, no histograms with values and statistical significance are reported, due to the intrinsic difficulty of objectively counting floating neurospheres.

References

    1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. . Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med (2005) 352(10):987–96. 10.1056/NEJMoa043330
    1. Korber V, Yang J, Barah P, Wu Y, Stichel D, Gu Z, et al. . Evolutionary Trajectories of IDH(WT) Glioblastomas Reveal a Common Path of Early Tumorigenesis Instigated Years ahead of Initial Diagnosis. Cancer Cell (2019) 35(4):692–704.e12. 10.1016/j.ccell.2019.02.007
    1. Horn AS, Snyder SH. Chlorpromazine and dopamine: conformational similarities that correlate with the antischizophrenic activity of phenothiazine drugs. Proc Natl Acad Sci USA (1971) 68(10):2325–8. 10.1073/pnas.68.10.2325
    1. Boyd-Kimball D, Gonczy K, Lewis B, Mason T, Siliko N, Wolfe J, et al. . Classics in Chemical Neuroscience: Chlorpromazine. ACS Chem Neurosci (2019) 10(1):79–88. 10.1021/acschemneuro.8b00258
    1. Caragher SP, Shireman JM, Huang M, Miska J, Atashi F, Baisiwala S, et al. . Activation of Dopamine Receptor 2 Prompts Transcriptomic and Metabolic Plasticity in Glioblastoma. J Neurosci (2019) 39(11):1982–93. 10.1523/JNEUROSCI.1589-18.2018
    1. Motohashi N, Sakagami H, Kamata K, Yamamoto Y. Cytotoxicity and differentiation-inducing activity of phenothiazine and benzo[a]phenothiazine derivatives. Anticancer Res (1991) 11(5):1933–7.
    1. Nordenberg J, Fenig E, Landau M, Weizman R, Weizman A. Effects of psychotropic drugs on cell proliferation and differentiation. Biochem Pharmacol (1999) 58(8):1229–36. 10.1016/s0006-2952(99)00156-2
    1. Lee MS, Johansen L, Zhang Y, Wilson A, Keegan M, Avery W, et al. . The novel combination of chlorpromazine and pentamidine exerts synergistic antiproliferative effects through dual mitotic action. Cancer Res (2007) 67(23):11359–67. 10.1158/0008-5472.CAN-07-2235
    1. Shin SY, Kim CG, Kim SH, Kim YS, Lim Y, Lee YH. Chlorpromazine activates p21Waf1/Cip1 gene transcription via early growth response-1 (Egr-1) in C6 glioma cells. Exp Mol Med (2010) 42(5):395–405. 10.3858/emm.2010.42.5.041
    1. Shin SY, Lee KS, Choi YK, Lim HJ, Lee HG, Lim Y, et al. . The antipsychotic agent chlorpromazine induces autophagic cell death by inhibiting the Akt/mTOR pathway in human U-87MG glioma cells. Carcinogenesis (2013) 34(9):2080–9. 10.1093/carcin/bgt169
    1. Pinheiro T, Otrocka M, Seashore-Ludlow B, Rraklli V, Holmberg J, Forsberg-Nilsson K, et al. . A chemical screen identifies trifluoperazine as an inhibitor of glioblastoma growth. Biochem Biophys Res Commun (2017) 494(3-4):477–83. 10.1016/j.bbrc.2017.10.106
    1. Oliva CR, Zhang W, Langford C, Suto MJ, Griguer CE. Repositioning chlorpromazine for treating chemoresistant glioma through the inhibition of cytochrome c oxidase bearing the COX4-1 regulatory subunit. Oncotarget (2017) 8(23):37568–83. 10.18632/oncotarget.17247
    1. Wiklund ED, Catts VS, Catts SV, Ng TF, Whitaker NJ, Brown AJ, et al. . Cytotoxic effects of antipsychotic drugs implicate cholesterol homeostasis as a novel chemotherapeutic target. Int J Cancer (2010) 126(1):28–40. 10.1002/ijc.24813
    1. Yang CE, Lee WY, Cheng HW, Chung CH, Mi FL, Lin CW, et al. . The antipsychotic chlorpromazine suppresses YAP signaling, stemness properties, and drug resistance in breast cancer cells. Chem Biol Interact (2019) 302:28–35. 10.1016/j.cbi.2019.01.033
    1. Abbruzzese C, Matteoni S, Persico M, Villani V, Paggi MG. Repurposing chlorpromazine in the treatment of glioblastoma multiforme: analysis of literature and forthcoming steps. J Exp Clin Cancer Res (2020) 39(1):26. 10.1186/s13046-020-1534-z
    1. Csatary LK. Chlorpromazines and cancer. Lancet (1972) 2(7772):338–9. 10.1016/s0140-6736(72)92955-8
    1. Huang J, Zhao D, Liu Z, Liu F. Repurposing psychiatric drugs as anti-cancer agents. Cancer Lett (2018) 419:257–65. 10.1016/j.canlet.2018.01.058
    1. Faraz S, Pannullo S, Rosenblum M, Smith A, Wernicke AG, et al. . Long-term survival in a patient with glioblastoma on antipsychotic therapy for schizophrenia: a case report and literature review. Ther Adv Med Oncol (2016) 8(6):421–8. 10.1177/1758834016659791
    1. D’Alessandris QG, Biffoni M, Martini M, Runci D, et al. . The clinical value of patient-derived glioblastoma tumorspheres in predicting treatment response. Neuro Oncol (2017) 19(8):1097–108. 10.1093/neuonc/now304
    1. McKinley KL, Cheeseman IM. Large-Scale Analysis of CRISPR/Cas9 Cell-Cycle Knockouts Reveals the Diversity of p53-Dependent Responses to Cell-Cycle Defects. Dev Cell (2017) 40(4):405–20.e2. 10.1016/j.devcel.2017.01.012
    1. Abbruzzese C, Catalogna G, Gallo E, di Martino S, Mileo AM, Carosi M, et al. . The small molecule SI113 synergizes with mitotic spindle poisons in arresting the growth of human glioblastoma multiforme. Oncotarget (2017) 8(67):110743–55. 10.18632/oncotarget.22500
    1. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. . The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol (2016) 131(6):803–20. 10.1007/s00401-016-1545-1
    1. Pallini R, Ricci-Vitiani L, Banna GL, Signore M, Lombardi D, Todaro M, et al. . Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin Cancer Res (2008) 14(24):8205–12. 10.1158/1078-0432.CCR-08-0644
    1. Lulli V, Buccarelli M, Ilari R, Castellani G, De Dominicis C, Di Giamberardino A, et al. . Mir-370-3p Impairs Glioblastoma Stem-Like Cell Malignancy Regulating a Complex Interplay between HMGA2/HIF1A and the Oncogenic Long Non-Coding RNA (lncRNA) NEAT1. Int J Mol Sci (2020) 21(10):3610. 10.3390/ijms21103610
    1. Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, et al. . Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res (2004) 64(19):7011–21. 10.1158/0008-5472.CAN-04-1364
    1. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. . Identification of human brain tumour initiating cells. Nature (2004) 432(7015):396–401. 10.1038/nature03128
    1. Matteoni S, Abbruzzese C, Matarrese P, De Luca G, Mileo AM, Miccadei S, et al. . The kinase inhibitor SI113 induces autophagy and synergizes with quinacrine in hindering the growth of human glioblastoma multiforme cells. J Exp Clin Cancer Res (2019) 38(1):202. 10.1186/s13046-019-1212-1
    1. Fransson A, Glaessgen D, Alfredsson J, Wiman KG, Bajalica-Lagercrantz S, Mohell N, et al. . Strong synergy with APR-246 and DNA-damaging drugs in primary cancer cells from patients with TP53 mutant High-Grade Serous ovarian cancer. J Ovarian Res (2016) 9(1):27. 10.1186/s13048-016-0239-6
    1. Wu J, Song R, Song W, Li Y, Zhang Q, Chen Y, et al. . Chlorpromazine protects against apoptosis induced by exogenous stimuli in the developing rat brain. PLoS One (2011) 6(7):e21966. 10.1371/journal.pone.0021966
    1. Wu J, Li A, Li Y, Li X, Zhang Q, Song W, et al. . Chlorpromazine inhibits mitochondrial apoptotic pathway via increasing expression of tissue factor. Int J Biochem Cell Biol (2016) 70:82–91. 10.1016/j.biocel.2015.11.008
    1. Stein GH. T98G: an anchorage-independent human tumor cell line that exhibits stationary phase G1 arrest in vitro. J Cell Physiol (1979) 99(1):43–54. 10.1002/jcp.1040990107
    1. Cheng HW, Liang YH, Kuo YL, Chuu CP, Lin CY, Lee MH, et al. . Identification of thioridazine, an antipsychotic drug, as an antiglioblastoma and anticancer stem cell agent using public gene expression data. Cell Death Dis (2015) 6:e1753. 10.1038/cddis.2015.77
    1. Toledo-Guzman ME, Hernandez MI, Gomez-Gallegos AA, Ortiz-Sanchez E. ALDH as a Stem Cell Marker in Solid Tumors. Curr Stem Cell Res Ther (2019) 14(5):375–88. 10.2174/1574888X13666180810120012
    1. Li G, Li Y, Liu X, Wang Z, Zhang C, Wu F, et al. . ALDH1A3 induces mesenchymal differentiation and serves as a predictor for survival in glioblastoma. Cell Death Dis (2018) 9(12):1190. 10.1038/s41419-018-1232-3
    1. Lee CAA, Banerjee P, Wilson BJ, Wu S, Guo Q, Berg G, et al. . Targeting the ABC transporter ABCB5 sensitizes glioblastoma to temozolomide-induced apoptosis through a cell-cycle checkpoint regulation mechanism. J Biol Chem (2020) 295(22):7774–88. 10.1074/jbc.RA120.013778
    1. D’Atri S, Tentori L, Lacal PM, Graziani G, Pagani E, Benincasa E, et al. . Involvement of the mismatch repair system in temozolomide-induced apoptosis. Mol Pharmacol (1998) 54(2):334–41. 10.1124/mol.54.2.334
    1. Buccarelli M, Marconi M, Pacioni S, De Pasqualis I, D'Alessandris QG, Martini M, et al. . Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis (2018) 9(8):841. 10.1038/s41419-018-0864-7
    1. Abbruzzese C, Matteoni S, Signore M, Cardone L, Nath K, Glickson JD, et al. . Drug repurposing for the treatment of glioblastoma multiforme. J Exp Clin Cancer Res (2017) 36(1):169. 10.1186/s13046-017-0642-x
    1. Schonberg DL, Lubelski D, Miller TE, Rich JN. Brain tumor stem cells: Molecular characteristics and their impact on therapy. Mol Asp Med (2014) 39:82–101. 10.1016/j.mam.2013.06.004
    1. Gimple RC, Bhargava S, Dixit D, Rich JN. Glioblastoma stem cells: lessons from the tumor hierarchy in a lethal cancer. Genes Dev (2019) 33(11-12):591–609. 10.1101/gad.324301.119
    1. Cheng L, Bao S, Rich JN. Potential therapeutic implications of cancer stem cells in glioblastoma. Biochem Pharmacol (2010) 80(5):654–65. 10.1016/j.bcp.2010.04.035
    1. Couturier CP, Ayyadhury S, Le PU, Nadaf J, Monlong J, Riva G, et al. . Single-cell RNA-seq reveals that glioblastoma recapitulates a normal neurodevelopmental hierarchy. Nat Commun (2020) 11(1):3406. 10.1038/s41467-020-17186-5
    1. Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev (2011) 63(1):182–217. 10.1124/pr.110.002642
    1. Barygin OI, Nagaeva EI, Tikhonov DB, Belinskaya DA, Vanchakova NP, Shestakova NN, et al. . Inhibition of the NMDA and AMPA receptor channels by antidepressants and antipsychotics. Brain Res (2017) 1660:58–66. 10.1016/j.brainres.2017.01.028
    1. Venkatesh HS, Morishita W, Geraghty AC, Silverbush D, Gillespie SM, Arzt M, et al. . Electrical and synaptic integration of glioma into neural circuits. Nature (2019) 573(7775):539–45. 10.1038/s41586-019-1563-y
    1. Venkataramani V, Tanev DI, Strahle C, Studier-Fischer A, Fankhauser L, Kessler T, et al. . Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature (2019) 573(7775):532–8. 10.1038/s41586-019-1564-x
    1. Meyer M, Reimand J, Lan X, Head R, Zhu X, Kushida M, et al. . Single cell-derived clonal analysis of human glioblastoma links functional and genomic heterogeneity. Proc Natl Acad Sci U S A (2015) 112(3):851–6. 10.1073/pnas.1320611111
    1. Lan X, Jorg DJ, Cavalli FMG, Richards LM, Nguyen LV, Vanner RJ, et al. . Fate mapping of human glioblastoma reveals an invariant stem cell hierarchy. Nature (2017) 549(7671):227–32. 10.1038/nature23666
    1. World Health Organization Model List of Essential Medicines, 21st List. Geneva: World Health Organization; (2019).

Source: PubMed

3
購読する