Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

01-12-2021 | Ovarian Cancer | Research

Choline kinase alpha impairment overcomes TRAIL resistance in ovarian cancer cells

Authors: Andrea Rizzo, Alessandro Satta, Giulia Garrone, Adalberto Cavalleri, Alessandra Napoli, Francesco Raspagliesi, Mariangela Figini, Loris De Cecco, Egidio Iorio, Antonella Tomassetti, Delia Mezzanzanica, Marina Bagnoli

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Choline kinase-α (ChoKα/CHKA) overexpression and hyper-activation sustain altered choline metabolism conferring the cholinic phenotype to epithelial ovarian cancer (OC), the most lethal gynecological tumor. We previously proved that CHKA down-modulation reduced OC cell aggressiveness and increased sensitivity to in vitro chemotherapeutics’ treatment also affecting intracellular content of one-carbon metabolites. In tumor types other than ovary, methionine decrease was shown to increase sensitivity to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-receptor 2 triggering. These effects were suggestive of a potential role for ChoKα in regulating susceptibility to TRAIL cytokine.

Methods

The relationship between ChoKα/CHKA and TRAIL-receptor 2 (TRAIL-R2) expression was investigated in silico in OC patients’ GEO datasets and in vitro in a panel of OC cell lines upon transient CHKA silencing (siCHKA). The effect of siCHKA on metabolites content was assessed by LC-MS. The triggered apoptotic signalling was studied following soluble-TRAIL or anti-TRAIL-R2 agonist antibody treatment. Lipid rafts were isolated by Triton X-100 fractionation. Preclinical ex vivo studies were performed in OC cells derived from patients’ ascites using autologous PBLs as effectors and a bispecific anti-TRAIL-R2/anti-CD3 antibody as triggering agent.

Results

Here we demonstrate that siCHKA specifically overcomes resistance to TRAIL-mediated apoptosis in OC cells. Upon siCHKA we detected: a significant sensitization to caspase-dependent apoptosis triggered by both soluble TRAIL and anti-TRAIL-R2 agonist antibody, a specific increase of TRAIL-R2 expression and TRAIL-R2 relocation into lipid rafts. In siCHKA-OC cells the acquired TRAIL sensitivity was completely reverted upon recovery of ChoKα expression but, at variance of other tumor cell types, TRAIL sensitivity was not efficiently phenocopied by methionine deprivation. Of note, we were also able to show that siCHKA sensitized tumor cells derived ex vivo from OC patients’ ascites to the cytotoxic activity of autologous lymphocytes redirected by a bispecific anti-TRAIL-R2/anti-CD3 antibody.

Conclusions

Our findings suggest that ChoKα/CHKA impairment, by restoring drug-induced or receptor-mediated cell death, could be a suitable therapeutic strategy to be used in combination with chemotherapeutics or immunomodulators to improve OC patients’ outcome.
Appendix
Available only for authorised users
Literature
1.
go back to reference Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA A Cancer J Clin. 2020;70:7–30.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA A Cancer J Clin. 2020;70:7–30.CrossRef
2.
3.
go back to reference Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M, Kroemer G. Molecular mechanisms of cisplatin resistance. Oncogene. 2012;31:1869–83.PubMedCrossRef Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M, Kroemer G. Molecular mechanisms of cisplatin resistance. Oncogene. 2012;31:1869–83.PubMedCrossRef
4.
go back to reference Mezzanzanica D, Balladore E, Turatti F, et al. CD95-mediated apoptosis is impaired at receptor level by cellular FLICE-inhibitory protein (long form) in wild-type p53 human ovarian carcinoma. Clin Cancer Res. 2004;10:5202–14.PubMedCrossRef Mezzanzanica D, Balladore E, Turatti F, et al. CD95-mediated apoptosis is impaired at receptor level by cellular FLICE-inhibitory protein (long form) in wild-type p53 human ovarian carcinoma. Clin Cancer Res. 2004;10:5202–14.PubMedCrossRef
5.
go back to reference Bagnoli M, Balladore E, Luison E, Alberti P, Raspagliesi F, Marcomini B, Canevari S, Mezzanzanica D. Sensitization of p53-mutated epithelial ovarian cancer to CD95-mediated apoptosis is synergistically induced by cisplatin pretreatment. Mol Cancer Ther. 2007;6:762–72.PubMedCrossRef Bagnoli M, Balladore E, Luison E, Alberti P, Raspagliesi F, Marcomini B, Canevari S, Mezzanzanica D. Sensitization of p53-mutated epithelial ovarian cancer to CD95-mediated apoptosis is synergistically induced by cisplatin pretreatment. Mol Cancer Ther. 2007;6:762–72.PubMedCrossRef
6.
go back to reference Bagnoli M, Ambrogi F, Pilotti S, Alberti P, Ditto A, Barbareschi M, Galligioni E, Biganzoli E, Canevari S, Mezzanzanica D. C-FLIPL expression defines two ovarian cancer patient subsets and is a prognostic factor of adverse outcome. Endocr Relat Cancer. 2009;16:443–53.PubMedCrossRef Bagnoli M, Ambrogi F, Pilotti S, Alberti P, Ditto A, Barbareschi M, Galligioni E, Biganzoli E, Canevari S, Mezzanzanica D. C-FLIPL expression defines two ovarian cancer patient subsets and is a prognostic factor of adverse outcome. Endocr Relat Cancer. 2009;16:443–53.PubMedCrossRef
7.
go back to reference Almasan A, Ashkenazi A. Apo2L/TRAIL: apoptosis signaling, biology, and potential for cancer therapy. Cytokine Growth Factor Rev. 2003;14:337–48.PubMedCrossRef Almasan A, Ashkenazi A. Apo2L/TRAIL: apoptosis signaling, biology, and potential for cancer therapy. Cytokine Growth Factor Rev. 2003;14:337–48.PubMedCrossRef
9.
go back to reference Bevis KS, Buchsbaum DJ, Straughn JM Jr. Overcoming TRAIL resistance in ovarian carcinoma. Gynecol Oncol. 2010;119:157–63.PubMedCrossRef Bevis KS, Buchsbaum DJ, Straughn JM Jr. Overcoming TRAIL resistance in ovarian carcinoma. Gynecol Oncol. 2010;119:157–63.PubMedCrossRef
10.
11.
12.
go back to reference Strekalova E, Malin D, Good DM, Cryns VL. Methionine deprivation induces a targetable vulnerability in triple-negative breast cancer cells by enhancing TRAIL receptor-2 expression. Clin Cancer Res. 2015;21:2780–91.PubMedPubMedCentralCrossRef Strekalova E, Malin D, Good DM, Cryns VL. Methionine deprivation induces a targetable vulnerability in triple-negative breast cancer cells by enhancing TRAIL receptor-2 expression. Clin Cancer Res. 2015;21:2780–91.PubMedPubMedCentralCrossRef
13.
go back to reference Kokkinakis DM, Liu X, Chada S, Ahmed MM, Shareef MM, Singha UK, Yang S, Luo J. Modulation of gene expression in human central nervous system tumors under methionine deprivation-induced stress. Cancer Res. 2004;64:7513–25.PubMedCrossRef Kokkinakis DM, Liu X, Chada S, Ahmed MM, Shareef MM, Singha UK, Yang S, Luo J. Modulation of gene expression in human central nervous system tumors under methionine deprivation-induced stress. Cancer Res. 2004;64:7513–25.PubMedCrossRef
14.
go back to reference Yamamoto J, Miyake K, Han Q, et al. Oral recombinant methioninase increases TRAIL receptor-2 expression to regress pancreatic cancer in combination with agonist tigatuzumab in an orthotopic mouse model. Cancer Lett. 2020. Yamamoto J, Miyake K, Han Q, et al. Oral recombinant methioninase increases TRAIL receptor-2 expression to regress pancreatic cancer in combination with agonist tigatuzumab in an orthotopic mouse model. Cancer Lett. 2020.
15.
go back to reference Sreedhar A, Zhao Y. Dysregulated metabolic enzymes and metabolic reprogramming in cancer cells. Biomed Rep. 2018;8:3–10.PubMed Sreedhar A, Zhao Y. Dysregulated metabolic enzymes and metabolic reprogramming in cancer cells. Biomed Rep. 2018;8:3–10.PubMed
18.
go back to reference Iorio E, Mezzanzanica D, Alberti P, et al. Alterations of choline phospholipid metabolism in ovarian tumor progression. Cancer Res. 2005;65:9369–76.PubMedCrossRef Iorio E, Mezzanzanica D, Alberti P, et al. Alterations of choline phospholipid metabolism in ovarian tumor progression. Cancer Res. 2005;65:9369–76.PubMedCrossRef
19.
go back to reference Iorio E, Ricci A, Bagnoli M, et al. Activation of phosphatidylcholine cycle enzymes in human epithelial ovarian cancer cells. Cancer Res. 2010;70:2126–35.PubMedPubMedCentralCrossRef Iorio E, Ricci A, Bagnoli M, et al. Activation of phosphatidylcholine cycle enzymes in human epithelial ovarian cancer cells. Cancer Res. 2010;70:2126–35.PubMedPubMedCentralCrossRef
20.
go back to reference Wu G, Aoyama C, Young SG, Vance DE. Early embryonic lethality caused by disruption of the gene for choline kinase alpha, the first enzyme in phosphatidylcholine biosynthesis. J Biol Chem. 2008;283:1456–62.PubMedCrossRef Wu G, Aoyama C, Young SG, Vance DE. Early embryonic lethality caused by disruption of the gene for choline kinase alpha, the first enzyme in phosphatidylcholine biosynthesis. J Biol Chem. 2008;283:1456–62.PubMedCrossRef
21.
go back to reference Mori N, Glunde K, Takagi T, Raman V, Bhujwalla ZM. Choline kinase down-regulation increases the effect of 5-fluorouracil in breast cancer cells. Cancer Res. 2007;67:11284–90.PubMedCrossRef Mori N, Glunde K, Takagi T, Raman V, Bhujwalla ZM. Choline kinase down-regulation increases the effect of 5-fluorouracil in breast cancer cells. Cancer Res. 2007;67:11284–90.PubMedCrossRef
22.
go back to reference Bagnoli M, Granata A, Nicoletti R, Krishnamachary B, Bhujwalla ZM, Canese R, Podo F, Canevari S, Iorio E, Mezzanzanica D. Choline metabolism alteration: a focus on ovarian cancer. Front Oncol. 2016;6:153.PubMedPubMedCentralCrossRef Bagnoli M, Granata A, Nicoletti R, Krishnamachary B, Bhujwalla ZM, Canese R, Podo F, Canevari S, Iorio E, Mezzanzanica D. Choline metabolism alteration: a focus on ovarian cancer. Front Oncol. 2016;6:153.PubMedPubMedCentralCrossRef
23.
go back to reference Granata A, Nicoletti R, Tinaglia V, et al. Choline kinase-alpha by regulating cell aggressiveness and drug sensitivity is a potential druggable target for ovarian cancer. Br J Cancer. 2014;110:330–40.PubMedCrossRef Granata A, Nicoletti R, Tinaglia V, et al. Choline kinase-alpha by regulating cell aggressiveness and drug sensitivity is a potential druggable target for ovarian cancer. Br J Cancer. 2014;110:330–40.PubMedCrossRef
24.
go back to reference Granata A, Nicoletti R, Perego P, et al. Global metabolic profile identifies choline kinase alpha as a key regulator of glutathione-dependent antioxidant cell defense in ovarian carcinoma. Oncotarget. 2015;6:11216–30.PubMedPubMedCentralCrossRef Granata A, Nicoletti R, Perego P, et al. Global metabolic profile identifies choline kinase alpha as a key regulator of glutathione-dependent antioxidant cell defense in ovarian carcinoma. Oncotarget. 2015;6:11216–30.PubMedPubMedCentralCrossRef
26.
27.
go back to reference De Cecco L, Bossi P, Locati L, Canevari S, Licitra L. Comprehensive gene expression meta-analysis of head and neck squamous cell carcinoma microarray data defines a robust survival predictor. Ann Oncol. 2014;25:1628–35.PubMedCrossRef De Cecco L, Bossi P, Locati L, Canevari S, Licitra L. Comprehensive gene expression meta-analysis of head and neck squamous cell carcinoma microarray data defines a robust survival predictor. Ann Oncol. 2014;25:1628–35.PubMedCrossRef
28.
go back to reference Bagnoli M, Tomassetti A, Figini M, Flati S, Dolo V, Canevari S, Miotti S. Downmodulation of caveolin-1 expression in human ovarian carcinoma is directly related to alpha-folate receptor overexpression. Oncogene. 2000;19:4754–63.PubMedCrossRef Bagnoli M, Tomassetti A, Figini M, Flati S, Dolo V, Canevari S, Miotti S. Downmodulation of caveolin-1 expression in human ovarian carcinoma is directly related to alpha-folate receptor overexpression. Oncogene. 2000;19:4754–63.PubMedCrossRef
29.
go back to reference Satta A, Mezzanzanica D, Caroli F, et al. Design, selection and optimization of an anti-TRAIL-R2/anti-CD3 bispecific antibody able to educate T cells to recognize and destroy cancer cells. MAbs. 2018;10:1084–97.PubMedPubMedCentral Satta A, Mezzanzanica D, Caroli F, et al. Design, selection and optimization of an anti-TRAIL-R2/anti-CD3 bispecific antibody able to educate T cells to recognize and destroy cancer cells. MAbs. 2018;10:1084–97.PubMedPubMedCentral
30.
go back to reference Ducker GS, Rabinowitz JD. One-carbon metabolism in health and disease. Cell Metab. 2017;25:27–42.PubMedCrossRef Ducker GS, Rabinowitz JD. One-carbon metabolism in health and disease. Cell Metab. 2017;25:27–42.PubMedCrossRef
31.
go back to reference Ashkenazi A, Dixit VM. Apoptosis control by death and decoy receptors. Curr Opin Cell Biol. 1999;11:255–60.PubMedCrossRef Ashkenazi A, Dixit VM. Apoptosis control by death and decoy receptors. Curr Opin Cell Biol. 1999;11:255–60.PubMedCrossRef
32.
34.
go back to reference Deng D, Shah K. TRAIL of hope meeting resistance in cancer, trends Cancer; 2020. Deng D, Shah K. TRAIL of hope meeting resistance in cancer, trends Cancer; 2020.
35.
go back to reference Walczak H, Miller RE, Ariail K, et al. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med. 1999;5:157–63.PubMedCrossRef Walczak H, Miller RE, Ariail K, et al. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med. 1999;5:157–63.PubMedCrossRef
36.
go back to reference Yagita H, Takeda K, Hayakawa Y, Smyth MJ, Okumura K. TRAIL and its receptors as targets for cancer therapy. Cancer Sci. 2004;95:777–83.PubMedCrossRef Yagita H, Takeda K, Hayakawa Y, Smyth MJ, Okumura K. TRAIL and its receptors as targets for cancer therapy. Cancer Sci. 2004;95:777–83.PubMedCrossRef
37.
go back to reference Dyer MJ, MacFarlane M, Cohen GM. Barriers to effective TRAIL-targeted therapy of malignancy. J Clin Oncol. 2007;25:4505–6.PubMedCrossRef Dyer MJ, MacFarlane M, Cohen GM. Barriers to effective TRAIL-targeted therapy of malignancy. J Clin Oncol. 2007;25:4505–6.PubMedCrossRef
38.
go back to reference Zhang L, Fang B. Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 2005;12:228–37.PubMedCrossRef Zhang L, Fang B. Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 2005;12:228–37.PubMedCrossRef
39.
go back to reference von Karstedt S, Montinaro A, Walczak H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer. 2017;17:352–66.CrossRef von Karstedt S, Montinaro A, Walczak H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer. 2017;17:352–66.CrossRef
40.
go back to reference Maddocks OD, Berkers CR, Mason SM, Zheng L, Blyth K, Gottlieb E, Vousden KH. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature. 2013;493:542–6.PubMedCrossRef Maddocks OD, Berkers CR, Mason SM, Zheng L, Blyth K, Gottlieb E, Vousden KH. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature. 2013;493:542–6.PubMedCrossRef
41.
go back to reference Mecham JO, Rowitch D, Wallace CD, Stern PH, Hoffman RM. The metabolic defect of methionine dependence occurs frequently in human tumor cell lines. Biochem Biophys Res Commun. 1983;117:429–34.PubMedCrossRef Mecham JO, Rowitch D, Wallace CD, Stern PH, Hoffman RM. The metabolic defect of methionine dependence occurs frequently in human tumor cell lines. Biochem Biophys Res Commun. 1983;117:429–34.PubMedCrossRef
42.
go back to reference Poirson-Bichat F, Goncalves RA, Miccoli L, Dutrillaux B, Poupon MF. Methionine depletion enhances the antitumoral efficacy of cytotoxic agents in drug-resistant human tumor xenografts. Clin Cancer Res. 2000;6:643–53.PubMed Poirson-Bichat F, Goncalves RA, Miccoli L, Dutrillaux B, Poupon MF. Methionine depletion enhances the antitumoral efficacy of cytotoxic agents in drug-resistant human tumor xenografts. Clin Cancer Res. 2000;6:643–53.PubMed
43.
go back to reference Lu S, Hoestje SM, Choo E, Epner DE. Induction of caspase-dependent and -independent apoptosis in response to methionine restriction. Int J Oncol. 2003;22:415–20.PubMed Lu S, Hoestje SM, Choo E, Epner DE. Induction of caspase-dependent and -independent apoptosis in response to methionine restriction. Int J Oncol. 2003;22:415–20.PubMed
44.
go back to reference Mills GB, May C, Hill M, Campbell S, Shaw P, Marks A. Ascitic fluid from human ovarian cancer patients contains growth factors necessary for intraperitoneal growth of human ovarian adenocarcinoma cells. J Clin Invest. 1990;86:851–5.PubMedPubMedCentralCrossRef Mills GB, May C, Hill M, Campbell S, Shaw P, Marks A. Ascitic fluid from human ovarian cancer patients contains growth factors necessary for intraperitoneal growth of human ovarian adenocarcinoma cells. J Clin Invest. 1990;86:851–5.PubMedPubMedCentralCrossRef
45.
46.
go back to reference Lane D, Robert V, Grondin R, Rancourt C, Piche A. Malignant ascites protect against TRAIL-induced apoptosis by activating the PI3K/akt pathway in human ovarian carcinoma cells. Int J Cancer. 2007;121:1227–37.PubMedCrossRef Lane D, Robert V, Grondin R, Rancourt C, Piche A. Malignant ascites protect against TRAIL-induced apoptosis by activating the PI3K/akt pathway in human ovarian carcinoma cells. Int J Cancer. 2007;121:1227–37.PubMedCrossRef
47.
go back to reference Lane D, Goncharenko-Khaider N, Rancourt C, Piche A. Ovarian cancer ascites protects from TRAIL-induced cell death through alphavbeta5 integrin-mediated focal adhesion kinase and akt activation. Oncogene. 2010;29:3519–31.PubMedCrossRef Lane D, Goncharenko-Khaider N, Rancourt C, Piche A. Ovarian cancer ascites protects from TRAIL-induced cell death through alphavbeta5 integrin-mediated focal adhesion kinase and akt activation. Oncogene. 2010;29:3519–31.PubMedCrossRef
48.
go back to reference Lacal J, Andera L. Choline kinase inhibitors synergize with TRAIL in the treatment of colorectal tumors and overcomes TRAIL resistance. Cancer Transl Med. 2016;2:163–74.CrossRef Lacal J, Andera L. Choline kinase inhibitors synergize with TRAIL in the treatment of colorectal tumors and overcomes TRAIL resistance. Cancer Transl Med. 2016;2:163–74.CrossRef
Metadata
Title
Choline kinase alpha impairment overcomes TRAIL resistance in ovarian cancer cells
Authors
Andrea Rizzo
Alessandro Satta
Giulia Garrone
Adalberto Cavalleri
Alessandra Napoli
Francesco Raspagliesi
Mariangela Figini
Loris De Cecco
Egidio Iorio
Antonella Tomassetti
Delia Mezzanzanica
Marina Bagnoli
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01794-6

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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