Skip to main content
Top
Published in: Cardiovascular Toxicology 3/2024

Open Access 13-02-2024 | Phenobarbital | Research

Comparative In Vitro Study of the Cytotoxic Effects of Doxorubicin’s Main Metabolites on Cardiac AC16 Cells Versus the Parent Drug

Authors: Ana Reis-Mendes, Cláudia Vitorino-Oliveira, Mariana Ferreira, Félix Carvalho, Fernando Remião, Emília Sousa, Maria de Lourdes Bastos, Vera Marisa Costa

Published in: Cardiovascular Toxicology | Issue 3/2024

Login to get access

Abstract

Doxorubicin (DOX; also known as adriamycin) serves as a crucial antineoplastic agent in cancer treatment; however, its clinical utility is hampered by its’ intrinsic cardiotoxicity. Although most DOX biotransformation occurs in the liver, a comprehensive understanding of the impact of DOX biotransformation and its’ metabolites on its induced cardiotoxicity remains to be fully elucidated. This study aimed to explore the role of biotransformation and DOX's main metabolites in its induced cardiotoxicity in human differentiated cardiac AC16 cells. A key discovery from our study is that modulating metabolism had minimal effects on DOX-induced cytotoxicity: even so, metyrapone (a non-specific inhibitor of cytochrome P450) increased DOX-induced cytotoxicity at 2 µM, while diallyl sulphide (a CYP2E1 inhibitor) decreased the 1 µM DOX-triggered cytotoxicity. Then, the toxicity of the main DOX metabolites, doxorubicinol [(DOXol, 0.5 to 10 µM), doxorubicinone (DOXone, 1 to 10 µM), and 7-deoxydoxorubicinone (7-DeoxyDOX, 1 to 10 µM)] was compared to DOX (0.5 to 10 µM) following a 48-h exposure. All metabolites evaluated, DOXol, DOXone, and 7-DeoxyDOX caused mitochondrial dysfunction in differentiated AC16 cells, but only at 2 µM. In contrast, DOX elicited comparable cytotoxicity, but at half the concentration. Similarly, all metabolites, except 7-DeoxyDOX impacted on lysosomal ability to uptake neutral red. Therefore, the present study showed that the modulation of DOX metabolism demonstrated minimal impact on its cytotoxicity, with the main metabolites exhibiting lower toxicity to AC16 cardiac cells compared to DOX. In conclusion, our findings suggest that metabolism may not be a pivotal factor in mediating DOX's cardiotoxic effects.

Graphical Abstract

Literature
2.
go back to reference Yang, F., et al. (2014). Doxorubicin, DNA torsion, and chromatin dynamics. Biochimica et Biophysica Acta, 1845(1), 84–89.PubMed Yang, F., et al. (2014). Doxorubicin, DNA torsion, and chromatin dynamics. Biochimica et Biophysica Acta, 1845(1), 84–89.PubMed
3.
go back to reference Speth, P. A. J., van Hoesel, Q. G. C. M., & Haanen, C. (1988). Clinical pharmacokinetics of doxorubicin. Clinical Pharmacokinetics, 15(1), 15–31.PubMedCrossRef Speth, P. A. J., van Hoesel, Q. G. C. M., & Haanen, C. (1988). Clinical pharmacokinetics of doxorubicin. Clinical Pharmacokinetics, 15(1), 15–31.PubMedCrossRef
4.
go back to reference Stewart, D. J., et al. (1993). Concentrations of doxorubicin and its metabolites in human autopsy heart and other tissues. Anticancer Research, 13(6a), 1945–1952.PubMed Stewart, D. J., et al. (1993). Concentrations of doxorubicin and its metabolites in human autopsy heart and other tissues. Anticancer Research, 13(6a), 1945–1952.PubMed
5.
go back to reference Cummings, J., Merry, S., & Willmott, N. (1986). Disposition kinetics of adriamycin, adriamycinol and their 7-deoxyaglycones in AKR mice bearing a sub-cutaneously growing ridgway osteogenic sarcoma (ROS). European Journal of Cancer & Clinical Oncology, 22(4), 451–460.CrossRef Cummings, J., Merry, S., & Willmott, N. (1986). Disposition kinetics of adriamycin, adriamycinol and their 7-deoxyaglycones in AKR mice bearing a sub-cutaneously growing ridgway osteogenic sarcoma (ROS). European Journal of Cancer & Clinical Oncology, 22(4), 451–460.CrossRef
6.
go back to reference Takanashi, S., & Bachur, N. R. (1976). Adriamycin metabolism in man. Evidence from urinary metabolites. Drug Metabolism & Disposition, 4(1), 79–87. Takanashi, S., & Bachur, N. R. (1976). Adriamycin metabolism in man. Evidence from urinary metabolites. Drug Metabolism & Disposition, 4(1), 79–87.
7.
go back to reference Kassner, N., et al. (2008). Carbonyl reductase 1 is a predominant doxorubicin reductase in the human liver. Drug Metabolism and Disposition, 36(10), 2113–2120.PubMedCrossRef Kassner, N., et al. (2008). Carbonyl reductase 1 is a predominant doxorubicin reductase in the human liver. Drug Metabolism and Disposition, 36(10), 2113–2120.PubMedCrossRef
8.
go back to reference Schaupp, C. M., et al. (2015). Metabolism of doxorubicin to the cardiotoxic metabolite doxorubicinol is increased in a mouse model of chronic glutathione deficiency: A potential role for carbonyl reductase 3. Chemico-Biological Interactions, 234, 154–161.PubMedCrossRef Schaupp, C. M., et al. (2015). Metabolism of doxorubicin to the cardiotoxic metabolite doxorubicinol is increased in a mouse model of chronic glutathione deficiency: A potential role for carbonyl reductase 3. Chemico-Biological Interactions, 234, 154–161.PubMedCrossRef
9.
go back to reference Licata, S., et al. (2000). Doxorubicin metabolism and toxicity in human myocardium: Role of cytoplasmic deglycosidation and carbonyl reduction. Chemical Research in Toxicology, 13(5), 414–420.PubMedCrossRef Licata, S., et al. (2000). Doxorubicin metabolism and toxicity in human myocardium: Role of cytoplasmic deglycosidation and carbonyl reduction. Chemical Research in Toxicology, 13(5), 414–420.PubMedCrossRef
10.
go back to reference Mordente, A., et al. (2003). Anthracycline secondary alcohol metabolite formation in human or rabbit heart: Biochemical aspects and pharmacologic implications. Biochemical Pharmacology, 66(6), 989–998.PubMedCrossRef Mordente, A., et al. (2003). Anthracycline secondary alcohol metabolite formation in human or rabbit heart: Biochemical aspects and pharmacologic implications. Biochemical Pharmacology, 66(6), 989–998.PubMedCrossRef
12.
go back to reference Del Tacca, M., et al. (1985). Might adriamycinol contribute to adriamycin-induced cardiotoxicity? Pharmacological Research Communications, 17(11), 1073–1084.PubMedCrossRef Del Tacca, M., et al. (1985). Might adriamycinol contribute to adriamycin-induced cardiotoxicity? Pharmacological Research Communications, 17(11), 1073–1084.PubMedCrossRef
13.
go back to reference Reis-Mendes, A., et al. (2019). The main metabolites of fluorouracil + adriamycin + cyclophosphamide (FAC) are not major contributors to FAC toxicity in H9c2 cardiac differentiated cells. Biomolecules, 9(3), 98.PubMedPubMedCentralCrossRefADS Reis-Mendes, A., et al. (2019). The main metabolites of fluorouracil + adriamycin + cyclophosphamide (FAC) are not major contributors to FAC toxicity in H9c2 cardiac differentiated cells. Biomolecules, 9(3), 98.PubMedPubMedCentralCrossRefADS
14.
go back to reference Serrano, J., et al. (1999). Cardioselective and cumulative oxidation of mitochondrial DNA following subchronic doxorubicin administration. Biochimica et Biophysica Acta, 1411(1), 201–205.PubMedCrossRef Serrano, J., et al. (1999). Cardioselective and cumulative oxidation of mitochondrial DNA following subchronic doxorubicin administration. Biochimica et Biophysica Acta, 1411(1), 201–205.PubMedCrossRef
15.
go back to reference Doroshow, J. H., & Davies, K. J. (1986). Redox cycling of anthracyclines by cardiac mitochondria II Formation of superoxide anion, hydrogen peroxide, and hydroxyl radical. Journal of Biological Chemistry, 261(7), 3068–3074.PubMedCrossRef Doroshow, J. H., & Davies, K. J. (1986). Redox cycling of anthracyclines by cardiac mitochondria II Formation of superoxide anion, hydrogen peroxide, and hydroxyl radical. Journal of Biological Chemistry, 261(7), 3068–3074.PubMedCrossRef
16.
go back to reference Pawłowska, J., et al. (2003). Differential ability of cytostatics from anthraquinone group to generate free radicals in three enzymatic systems: NADH dehydrogenase, NADPH cytochrome P450 reductase, and xanthine oxidase. Oncology Research, 13(5), 245–252.PubMedCrossRef Pawłowska, J., et al. (2003). Differential ability of cytostatics from anthraquinone group to generate free radicals in three enzymatic systems: NADH dehydrogenase, NADPH cytochrome P450 reductase, and xanthine oxidase. Oncology Research, 13(5), 245–252.PubMedCrossRef
17.
go back to reference Doroshow, J. H. (1983). Anthracycline antibiotic-stimulated superoxide, hydrogen peroxide, and hydroxyl radical production by NADH dehydrogenase. Cancer Research, 43(10), 4543–4551.PubMed Doroshow, J. H. (1983). Anthracycline antibiotic-stimulated superoxide, hydrogen peroxide, and hydroxyl radical production by NADH dehydrogenase. Cancer Research, 43(10), 4543–4551.PubMed
18.
go back to reference Dresdale, A. R., et al. (1982). Prospective randomized study of the role of N-acetyl cysteine in reversing doxorubicin-induced cardiomyopathy. American Journal of Clinical Oncology, 5(6), 657–663.PubMedCrossRef Dresdale, A. R., et al. (1982). Prospective randomized study of the role of N-acetyl cysteine in reversing doxorubicin-induced cardiomyopathy. American Journal of Clinical Oncology, 5(6), 657–663.PubMedCrossRef
19.
go back to reference Myers, C., et al. (1983). A randomized controlled trial assessing the prevention of doxorubicin cardiomyopathy by N-acetylcysteine. Seminars in Oncology, 10(1 Suppl 1), 53–55.PubMed Myers, C., et al. (1983). A randomized controlled trial assessing the prevention of doxorubicin cardiomyopathy by N-acetylcysteine. Seminars in Oncology, 10(1 Suppl 1), 53–55.PubMed
20.
go back to reference Minotti, G., et al. (1998). The secondary alcohol metabolite of doxorubicin irreversibly inactivates aconitase/iron regulatory protein-1 in cytosolic fractions from human myocardium. The FASEB Journal, 12(7), 541–552.PubMedCrossRef Minotti, G., et al. (1998). The secondary alcohol metabolite of doxorubicin irreversibly inactivates aconitase/iron regulatory protein-1 in cytosolic fractions from human myocardium. The FASEB Journal, 12(7), 541–552.PubMedCrossRef
21.
go back to reference Olson, L. E., et al. (2003). Protection from doxorubicin-induced cardiac toxicity in mice with a null allele of carbonyl reductase 1. Cancer Research, 63(20), 6602–6606.PubMed Olson, L. E., et al. (2003). Protection from doxorubicin-induced cardiac toxicity in mice with a null allele of carbonyl reductase 1. Cancer Research, 63(20), 6602–6606.PubMed
22.
go back to reference Davidson, M. M., et al. (2005). Novel cell lines derived from adult human ventricular cardiomyocytes. Journal of Molecular and Cellular Cardiology, 39(1), 133–147.PubMedCrossRef Davidson, M. M., et al. (2005). Novel cell lines derived from adult human ventricular cardiomyocytes. Journal of Molecular and Cellular Cardiology, 39(1), 133–147.PubMedCrossRef
23.
go back to reference Dionísio, F., et al. (2022). Cardiotoxicity of cyclophosphamide’s metabolites: An in vitro metabolomics approach in AC16 human cardiomyocytes. Archives of Toxicology, 96(2), 653–671.PubMedCrossRef Dionísio, F., et al. (2022). Cardiotoxicity of cyclophosphamide’s metabolites: An in vitro metabolomics approach in AC16 human cardiomyocytes. Archives of Toxicology, 96(2), 653–671.PubMedCrossRef
24.
go back to reference Hays, S. J., et al. (1984). Structure-activity relationship study of the inhibition of adrenal cortical 11 beta-hydroxylase by new metyrapone analogues. Journal of Medicinal Chemistry, 27(1), 15–19.PubMedCrossRef Hays, S. J., et al. (1984). Structure-activity relationship study of the inhibition of adrenal cortical 11 beta-hydroxylase by new metyrapone analogues. Journal of Medicinal Chemistry, 27(1), 15–19.PubMedCrossRef
25.
go back to reference Park, H., Lee, S., & Suh, J. (2005). Structural and dynamical basis of broad substrate specificity, catalytic mechanism, and inhibition of cytochrome P450 3A4. Journal of the American Chemical Society, 127(39), 13634–13642.PubMedCrossRef Park, H., Lee, S., & Suh, J. (2005). Structural and dynamical basis of broad substrate specificity, catalytic mechanism, and inhibition of cytochrome P450 3A4. Journal of the American Chemical Society, 127(39), 13634–13642.PubMedCrossRef
26.
go back to reference Asakura, T., & Shichi, H. (1992). Cytochrome P450-mediated prostaglandin omega/omega-1 hydroxylase activities in porcine ciliary body epithelial cells. Experimental Eye Research, 55(2), 377–384.PubMedCrossRef Asakura, T., & Shichi, H. (1992). Cytochrome P450-mediated prostaglandin omega/omega-1 hydroxylase activities in porcine ciliary body epithelial cells. Experimental Eye Research, 55(2), 377–384.PubMedCrossRef
27.
go back to reference Linder, C. D., Renaud, N. A., & Hutzler, J. M. (2009). Is 1-aminobenzotriazole an appropriate in vitro tool as a nonspecific cytochrome P450 inactivator? Drug Metabolism and Disposition, 37(1), 10–13.PubMedCrossRef Linder, C. D., Renaud, N. A., & Hutzler, J. M. (2009). Is 1-aminobenzotriazole an appropriate in vitro tool as a nonspecific cytochrome P450 inactivator? Drug Metabolism and Disposition, 37(1), 10–13.PubMedCrossRef
28.
go back to reference Jin, M., et al. (2013). Regulation of cytochrome P450 2e1 expression by ethanol: Role of oxidative stress-mediated pkc/jnk/sp1 pathway. Cell Death & Disease, 4(3), e554.CrossRef Jin, M., et al. (2013). Regulation of cytochrome P450 2e1 expression by ethanol: Role of oxidative stress-mediated pkc/jnk/sp1 pathway. Cell Death & Disease, 4(3), e554.CrossRef
29.
go back to reference Behnia, K., & Boroujerdi, M. (1999). Inhibition of aldo-keto reductases by phenobarbital alters metabolism, pharmacokinetics and toxicity of doxorubicin in rats. Journal of Pharmacy and Pharmacology, 51(11), 1275–1282.PubMedCrossRef Behnia, K., & Boroujerdi, M. (1999). Inhibition of aldo-keto reductases by phenobarbital alters metabolism, pharmacokinetics and toxicity of doxorubicin in rats. Journal of Pharmacy and Pharmacology, 51(11), 1275–1282.PubMedCrossRef
30.
go back to reference Deng, Y., et al. (2013). In vitro inhibition and induction of human liver cytochrome P450 enzymes by gentiopicroside: Potent effect on CYP2A6. Drug Metabolism and Pharmacokinetics, 28(4), 339–344.PubMedCrossRef Deng, Y., et al. (2013). In vitro inhibition and induction of human liver cytochrome P450 enzymes by gentiopicroside: Potent effect on CYP2A6. Drug Metabolism and Pharmacokinetics, 28(4), 339–344.PubMedCrossRef
31.
go back to reference Lin, J. H. (2006). CYP induction-mediated drug interactions: In vitro assessment and clinical implications. Pharmaceutical Research, 23(6), 1089–1116.PubMedCrossRef Lin, J. H. (2006). CYP induction-mediated drug interactions: In vitro assessment and clinical implications. Pharmaceutical Research, 23(6), 1089–1116.PubMedCrossRef
32.
go back to reference Soares, A. S., et al. (2014). Combination of Cl-IB-MECA with paclitaxel is a highly effective cytotoxic therapy causing mTOR-dependent autophagy and mitotic catastrophe on human melanoma cells. Journal of Cancer Research and Clinical Oncology, 140(6), 921–935.PubMedCrossRef Soares, A. S., et al. (2014). Combination of Cl-IB-MECA with paclitaxel is a highly effective cytotoxic therapy causing mTOR-dependent autophagy and mitotic catastrophe on human melanoma cells. Journal of Cancer Research and Clinical Oncology, 140(6), 921–935.PubMedCrossRef
33.
go back to reference Greene, R. F., et al. (1983). Plasma pharmacokinetics of adriamycin and adriamycinol: Implications for the design of in vitro experiments and treatment protocols. Cancer Research, 43(7), 3417–3421.PubMed Greene, R. F., et al. (1983). Plasma pharmacokinetics of adriamycin and adriamycinol: Implications for the design of in vitro experiments and treatment protocols. Cancer Research, 43(7), 3417–3421.PubMed
34.
go back to reference Reis-Mendes, A., et al. (2023). Autophagy (but not metabolism) is a key event in mitoxantrone-induced cytotoxicity in differentiated AC16 cardiac cells. Archives of Toxicology, 97(1), 201–216.PubMedCrossRef Reis-Mendes, A., et al. (2023). Autophagy (but not metabolism) is a key event in mitoxantrone-induced cytotoxicity in differentiated AC16 cardiac cells. Archives of Toxicology, 97(1), 201–216.PubMedCrossRef
35.
go back to reference Repetto, G., del Peso, A., & Zurita, J. L. (2008). Neutral red uptake assay for the estimation of cell viability/cytotoxicity. Nature Protocols, 3(7), 1125–1131.PubMedCrossRef Repetto, G., del Peso, A., & Zurita, J. L. (2008). Neutral red uptake assay for the estimation of cell viability/cytotoxicity. Nature Protocols, 3(7), 1125–1131.PubMedCrossRef
36.
go back to reference Reers, M., Smith, T. W., & Chen, L. B. (1991). J-aggregate formation of a carbocyanine as a quantitative fluorescent indicator of membrane potential. Biochemistry, 30(18), 4480–4486.PubMedCrossRef Reers, M., Smith, T. W., & Chen, L. B. (1991). J-aggregate formation of a carbocyanine as a quantitative fluorescent indicator of membrane potential. Biochemistry, 30(18), 4480–4486.PubMedCrossRef
37.
go back to reference Cossarizza, A., et al. (1993). A new method for the cytofluorimetric analysis of mitochondrial membrane potential using the J-aggregate forming lipophilic cation 5,5’,6,6’-tetrachloro-1,1’,3,3’-tetraethylbenzimidazolcarbocyanine iodide (JC-1). Biochemical and Biophysical Research Communications, 197(1), 40–45.PubMedCrossRef Cossarizza, A., et al. (1993). A new method for the cytofluorimetric analysis of mitochondrial membrane potential using the J-aggregate forming lipophilic cation 5,5’,6,6’-tetrachloro-1,1’,3,3’-tetraethylbenzimidazolcarbocyanine iodide (JC-1). Biochemical and Biophysical Research Communications, 197(1), 40–45.PubMedCrossRef
38.
go back to reference Sampath-Kumar, R., et al. (1997). Metyrapone is a competitive inhibitor of 11beta-hydroxysteroid dehydrogenase type 1 reductase. Journal of Steroid Biochemistry and Molecular Biology, 62(2–3), 195–199.PubMedCrossRef Sampath-Kumar, R., et al. (1997). Metyrapone is a competitive inhibitor of 11beta-hydroxysteroid dehydrogenase type 1 reductase. Journal of Steroid Biochemistry and Molecular Biology, 62(2–3), 195–199.PubMedCrossRef
39.
go back to reference Joerger, M., et al. (2005). Pharmacokinetics of low-dose doxorubicin and metabolites in patients with AIDS-related Kaposi sarcoma. Cancer Chemotherapy and Pharmacology, 55(5), 488–496.PubMedCrossRef Joerger, M., et al. (2005). Pharmacokinetics of low-dose doxorubicin and metabolites in patients with AIDS-related Kaposi sarcoma. Cancer Chemotherapy and Pharmacology, 55(5), 488–496.PubMedCrossRef
40.
go back to reference Leow, J. W. H., et al. (2023). Investigating the relevance of CYP2J2 inhibition for drugs known to cause intermediate to high risk torsades de pointes. European Journal of Pharmaceutical Sciences, 1(187), 106475CrossRef Leow, J. W. H., et al. (2023). Investigating the relevance of CYP2J2 inhibition for drugs known to cause intermediate to high risk torsades de pointes. European Journal of Pharmaceutical Sciences, 1(187), 106475CrossRef
41.
go back to reference Zhou, B., et al. (2017). Mitochondrial activity and oxidative stress functions are influenced by the activation of AhR-induced CYP1A1 overexpression in cardiomyocytes. Molecular Medicine Reports, 16(1), 174–180PubMedPubMedCentralCrossRef Zhou, B., et al. (2017). Mitochondrial activity and oxidative stress functions are influenced by the activation of AhR-induced CYP1A1 overexpression in cardiomyocytes. Molecular Medicine Reports, 16(1), 174–180PubMedPubMedCentralCrossRef
42.
go back to reference Lopes, M. A., et al. (2008). Doxorubicin induces biphasic neurotoxicity to rat cortical neurons. Neurotoxicology, 29(2), 286–293.PubMedCrossRef Lopes, M. A., et al. (2008). Doxorubicin induces biphasic neurotoxicity to rat cortical neurons. Neurotoxicology, 29(2), 286–293.PubMedCrossRef
43.
go back to reference Branco, A. F., et al. (2012). Differentiation-dependent doxorubicin toxicity on H9c2 cardiomyoblasts. Cardiovascular Toxicology, 12(4), 326–340.PubMedCrossRef Branco, A. F., et al. (2012). Differentiation-dependent doxorubicin toxicity on H9c2 cardiomyoblasts. Cardiovascular Toxicology, 12(4), 326–340.PubMedCrossRef
44.
go back to reference Xiao, B., et al. (2019). The true colors of autophagy in doxorubicin-induced cardiotoxicity. Oncology Letters, 18(3), 2165–2172.PubMedPubMedCentral Xiao, B., et al. (2019). The true colors of autophagy in doxorubicin-induced cardiotoxicity. Oncology Letters, 18(3), 2165–2172.PubMedPubMedCentral
45.
go back to reference Brandão, S. R., et al. (2023). The metabolic fingerprint of doxorubicin-induced cardiotoxicity in male CD-1 mice fades away with time while autophagy increases. Pharmaceuticals, 16(11), 1613.PubMedPubMedCentralCrossRef Brandão, S. R., et al. (2023). The metabolic fingerprint of doxorubicin-induced cardiotoxicity in male CD-1 mice fades away with time while autophagy increases. Pharmaceuticals, 16(11), 1613.PubMedPubMedCentralCrossRef
46.
go back to reference Montalvo, R. N., et al. (2020). Protection against doxorubicin-induced cardiac dysfunction is not maintained following prolonged autophagy inhibition. International Journal of Molecular Sciences, 21(21), 8105.PubMedPubMedCentralCrossRef Montalvo, R. N., et al. (2020). Protection against doxorubicin-induced cardiac dysfunction is not maintained following prolonged autophagy inhibition. International Journal of Molecular Sciences, 21(21), 8105.PubMedPubMedCentralCrossRef
47.
go back to reference Parrish, K. E., et al. (2016). In vitro and in vivo characterization of CYP inhibition by 1-aminobenzotriazole in rats. Biopharmaceutics & Drug Disposition, 37(4), 200–211.CrossRef Parrish, K. E., et al. (2016). In vitro and in vivo characterization of CYP inhibition by 1-aminobenzotriazole in rats. Biopharmaceutics & Drug Disposition, 37(4), 200–211.CrossRef
48.
go back to reference Emoto, C., et al. (2003). In vitro inhibitory effect of 1-aminobenzotriazole on drug oxidations catalyzed by human cytochrome P450 enzymes: A comparison with SKF-525A and ketoconazole. Drug Metabolism and Pharmacokinetics, 18(5), 287–295.PubMedCrossRef Emoto, C., et al. (2003). In vitro inhibitory effect of 1-aminobenzotriazole on drug oxidations catalyzed by human cytochrome P450 enzymes: A comparison with SKF-525A and ketoconazole. Drug Metabolism and Pharmacokinetics, 18(5), 287–295.PubMedCrossRef
49.
go back to reference Creemers, S. G., et al. (2019). Osilodrostat is a potential novel steroidogenesis inhibitor for the treatment of cushing syndrome: an in vitro study. Journal of Clinical Endocrinology and Metabolism, 104(8), 3437–3449.PubMedCrossRef Creemers, S. G., et al. (2019). Osilodrostat is a potential novel steroidogenesis inhibitor for the treatment of cushing syndrome: an in vitro study. Journal of Clinical Endocrinology and Metabolism, 104(8), 3437–3449.PubMedCrossRef
50.
go back to reference Sevrioukova, I. (2019). Interaction of human drug-metabolizing CYP3A4 with small inhibitory molecules. Biochemistry, 58(7), 930–939.PubMedCrossRef Sevrioukova, I. (2019). Interaction of human drug-metabolizing CYP3A4 with small inhibitory molecules. Biochemistry, 58(7), 930–939.PubMedCrossRef
51.
go back to reference Aubrecht, J., et al. (1996). Differential induction of mRNA expression of cytochromes P450 (CYP2B1 and CYP1A1/2) by metyrapone in primary rat hepatocyte cultures. Research Communications in Molecular Pathology and Pharmacology, 94(1), 47–61.PubMed Aubrecht, J., et al. (1996). Differential induction of mRNA expression of cytochromes P450 (CYP2B1 and CYP1A1/2) by metyrapone in primary rat hepatocyte cultures. Research Communications in Molecular Pathology and Pharmacology, 94(1), 47–61.PubMed
52.
go back to reference Brady, J. F., et al. (1991). Inhibition of cytochrome P-450 2E1 by diallyl sulfide and its metabolites. Chemical Research in Toxicology, 4(6), 642–647.PubMedCrossRef Brady, J. F., et al. (1991). Inhibition of cytochrome P-450 2E1 by diallyl sulfide and its metabolites. Chemical Research in Toxicology, 4(6), 642–647.PubMedCrossRef
53.
go back to reference Rossato, L. G., et al. (2013). The metabolic profile of mitoxantrone and its relation with mitoxantrone-induced cardiotoxicity. Archives of Toxicology, 87(10), 1809–1820.PubMedCrossRef Rossato, L. G., et al. (2013). The metabolic profile of mitoxantrone and its relation with mitoxantrone-induced cardiotoxicity. Archives of Toxicology, 87(10), 1809–1820.PubMedCrossRef
54.
go back to reference Grant, M. K. O., et al. (2020). Sexual dimorphism in doxorubicin-induced systemic inflammation: implications for hepatic cytochrome P450 regulation. International Journal of Molecular Sciences, 21(4), 1279.PubMedPubMedCentralCrossRef Grant, M. K. O., et al. (2020). Sexual dimorphism in doxorubicin-induced systemic inflammation: implications for hepatic cytochrome P450 regulation. International Journal of Molecular Sciences, 21(4), 1279.PubMedPubMedCentralCrossRef
56.
go back to reference Cantiello, M., et al. (2022). Induction by phenobarbital of phase I and II xenobiotic-metabolizing enzymes in bovine liver: An overall catalytic and immunochemical characterization. International Journal of Molecular Sciences, 23(7), 3564.PubMedPubMedCentralCrossRef Cantiello, M., et al. (2022). Induction by phenobarbital of phase I and II xenobiotic-metabolizing enzymes in bovine liver: An overall catalytic and immunochemical characterization. International Journal of Molecular Sciences, 23(7), 3564.PubMedPubMedCentralCrossRef
57.
go back to reference Sturgill, M. G., August, D. A., & Brenner, D. E. (2000). Hepatic enzyme induction with phenobarbital and doxorubicin metabolism and myelotoxicity in the rabbit. Cancer Investigation, 18(3), 197–205.PubMedCrossRef Sturgill, M. G., August, D. A., & Brenner, D. E. (2000). Hepatic enzyme induction with phenobarbital and doxorubicin metabolism and myelotoxicity in the rabbit. Cancer Investigation, 18(3), 197–205.PubMedCrossRef
58.
go back to reference Forrest, G. L., et al. (2000). Human carbonyl reductase overexpression in the heart advances the development of doxorubicin-induced cardiotoxicity in transgenic mice1. Cancer Research, 60(18), 5158–5164.PubMed Forrest, G. L., et al. (2000). Human carbonyl reductase overexpression in the heart advances the development of doxorubicin-induced cardiotoxicity in transgenic mice1. Cancer Research, 60(18), 5158–5164.PubMed
59.
go back to reference Revis, N. W., & Marusić, N. (1979). Effects of doxorubicin and its aglycone metabolite on calcium sequestration by rabbit heart, liver, and kidney mitochondria. Life Sciences, 25(12), 1055–1063.PubMedCrossRef Revis, N. W., & Marusić, N. (1979). Effects of doxorubicin and its aglycone metabolite on calcium sequestration by rabbit heart, liver, and kidney mitochondria. Life Sciences, 25(12), 1055–1063.PubMedCrossRef
60.
go back to reference Sokolove, P. M. (1994). Interactions of adriamycin aglycones with mitochondria may mediate adriamycin cardiotoxicity. International Journal of Biochemistry, 26(12), 1341–1350.PubMedCrossRef Sokolove, P. M. (1994). Interactions of adriamycin aglycones with mitochondria may mediate adriamycin cardiotoxicity. International Journal of Biochemistry, 26(12), 1341–1350.PubMedCrossRef
61.
go back to reference Clementi, M. E., et al. (2003). Doxorubicin-derived metabolites induce release of cytochrome C and inhibition of respiration on cardiac isolated mitochondria. Anticancer Research, 23(3b), 2445–2450.PubMed Clementi, M. E., et al. (2003). Doxorubicin-derived metabolites induce release of cytochrome C and inhibition of respiration on cardiac isolated mitochondria. Anticancer Research, 23(3b), 2445–2450.PubMed
62.
go back to reference Sokolove, P. M., et al. (1993). Interaction of adriamycin aglycones with isolated mitochondria. Effect of selenium deficiency. Biochemical Pharmacology, 46(4), 691–697PubMedCrossRef Sokolove, P. M., et al. (1993). Interaction of adriamycin aglycones with isolated mitochondria. Effect of selenium deficiency. Biochemical Pharmacology, 46(4), 691–697PubMedCrossRef
Metadata
Title
Comparative In Vitro Study of the Cytotoxic Effects of Doxorubicin’s Main Metabolites on Cardiac AC16 Cells Versus the Parent Drug
Authors
Ana Reis-Mendes
Cláudia Vitorino-Oliveira
Mariana Ferreira
Félix Carvalho
Fernando Remião
Emília Sousa
Maria de Lourdes Bastos
Vera Marisa Costa
Publication date
13-02-2024
Publisher
Springer US
Published in
Cardiovascular Toxicology / Issue 3/2024
Print ISSN: 1530-7905
Electronic ISSN: 1559-0259
DOI
https://doi.org/10.1007/s12012-024-09829-6

Other articles of this Issue 3/2024

Cardiovascular Toxicology 3/2024 Go to the issue