1
|
Saroj N, Dholaniya PS, Alvi SB, Sridharan D, Soni N, Ashraf SA, Choudhry A, Ashraf YA, Mikula SK, Singla DK, Khan M. SiRNA-mediated knockdown of TOP2B protects hiPSC-derived cardiomyocytes from doxorubicin-induced toxicity. Life Sci 2025; 371:123595. [PMID: 40158615 DOI: 10.1016/j.lfs.2025.123595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
AIMS Doxorubicin (Dox) is a potent chemotherapeutic agent, but its use is limited by cardiotoxicity, primarily due to the disruption of Topoisomerase-2 beta (TOP2B) activity. Dexrazoxane (Dex), an FDA-approved cardioprotective drug, alleviates Dox-induced toxicity but lacks heart-specific targeting. This study investigates siRNA-mediated TOP2B knockdown as a more targeted strategy to protect cardiomyocytes from Dox-induced damage. MATERIALS AND METHODS Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were treated with siRNA to knock down TOP2B and were then exposed to Dox. We compared the cardioprotective effects of siRNA-mediated knockdown to Dex treatment using cell viability, cell toxicity assay and electrophysiological evaluation was performed using a multielectrode array (MEA). KEY FINDINGS Our results demonstrate that TOP2B silencing significantly decreases apoptosis and improved cell viability, as compared to the Dex treatment. Additionally, electrophysiological assays using a Multielectrode Array (MEA) demonstrated enhanced contractility and conductivity in siRNA-treated hiPSC-CMs. Furthermore, transmission electron microscopy (TEM) data revealed that TOP2B knockdown preserves mitochondrial morphology and sarcomere structure, compared to Dox and Dex-treated groups. SIGNIFICANCE These findings suggest that siRNA-mediated TOP2B inhibition could provide a safer, more specific approach to mitigate Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Neha Saroj
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Pankaj Singh Dholaniya
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India.
| | - Syed Baseeruddin Alvi
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Divya Sridharan
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Navisha Soni
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Syed Abdullah Ashraf
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Ayza Choudhry
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Yusuf Ali Ashraf
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Sarah Kathleen Mikula
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
| | - Dinender Kumar Singla
- Biomolecular Science Center, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Mahmood Khan
- Division of Basic and Translational Research, Department of Emergency Medicine, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Balough E, Ariza A, Asnani A, Hoeger CW. Cardiotoxicity of Anthracyclines. Cardiol Clin 2025; 43:111-127. [PMID: 39551553 DOI: 10.1016/j.ccl.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Anthracycline chemotherapy is associated with cardiotoxicity, predominantly manifesting as left ventricular systolic dysfunction within the first year of treatment. Early detection is possible through biomarkers and cardiovascular imaging before clinical symptoms develop. Comprehensive cardiovascular risk assessment is essential for all patients prior to anthracycline therapy to stratify their risk of cardiotoxicity. Preventive measures, including cardiovascular risk optimization, as well as anthracycline dose adjustments, the use of liposomal anthracyclines, and dexrazoxane in high-risk patients, are crucial to mitigate the risk of cardiotoxicity. Long-term follow-up and cardiovascular risk optimization are critical for cancer survivors to optimize cardiovascular outcomes.
Collapse
Affiliation(s)
- Elizabeth Balough
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, 185 Pilgrim Road, Baker 4, Boston, MA 02215, USA. https://twitter.com/ElizabethBaloug
| | - Abul Ariza
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; 3 Blackfan Circle, CLS-911, Boston, MA 02115, USA
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; 3 Blackfan Circle, CLS-911, Boston, MA 02115, USA. https://twitter.com/AartiAsnaniMD
| | - Christopher W Hoeger
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, 185 Pilgrim Road, Baker 4, Boston, MA 02215, USA.
| |
Collapse
|
3
|
Feng W, Wang Q, Tan Y, Qiao J, Liu Q, Yang B, Yang S, Cui L. Early detection of anthracycline-induced cardiotoxicity. Clin Chim Acta 2025; 565:120000. [PMID: 39401650 DOI: 10.1016/j.cca.2024.120000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Although anthracyclines are important anticancer agents, their use is limited due to various adverse effects, particularly cardiac toxicity. Mechanisms underlying anthracycline-induced cardiotoxicity (AIC) are complex. Given the irreplaceable role of anthracyclines in treatment of malignancies and other serious diseases, early monitoring of AIC is paramount. In recent years, multiple studies have investigated various biomarkers for early detection of AIC. Currently, the two most common are cardiac troponin and B-type natriuretic peptide. In addition, a range of other molecules, including RNAs, myeloperoxidase (MPO), C-reactive protein (CRP), various genes, and others, also play roles in AIC prediction. Unfortunately, current research indicates a need to validate their sensitivity and specificity of these biomarkers especially in large study populations. In this review, we summarize the mechanisms and potential biomarkers of AIC, although some remain preliminary.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
4
|
McKeown BT, Goralski KB. Pharmacological interactions of jadomycin B with topoisomerase poisons in MDA-MB-231 human breast cancer cells. Can J Physiol Pharmacol 2025; 103:36-45. [PMID: 39481122 DOI: 10.1139/cjpp-2024-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Jadomycin B, a natural product isolated from Streptomyces venezuelae, exerts an anti-cancer effect on human triple negative breast cancer cells in vitro and has anti-tumoral effects in vivo in animal models of breast cancer. One proposed mechanism for this anti-cancer effect is through interaction with topoisomerase 2 (TOP2). Based on the previously described interactions between jadomycin B and TOP2 we hypothesized that jadomycin B will act additively with TOP2 poisons and produce a similar functional outcome in eliciting cell cycle arrest. Combined treatments of jadomycin B and the TOP2 poisons doxorubicin or mitoxantrone produced moderately synergistic to additive cytotoxicity (combination index values ranging from 0.72-0.94) in MDA-MB-231 cells. In comparison, combined mitoxantrone and doxorubicin produced additive cytotoxicity (combination index values 0.96-1.11). Jadomycin B combined with the proteosome inhibitor MG132 had additive cytotoxicity (combination index values 0.76-1.18). In contrast, mitoxantrone or doxorubicin cytotoxicity was antagonized by MG132 (combination index values 1.21-2.31). Jadomycin B treatment arrested cells in S-phase (P = 0.0024) as opposed to mitoxantrone which caused G2/M-phase arrest (P < 0.0001). In conclusion, jadomycin B interacts differently than known TOP2 poisons in combination, supporting a novel pharmacological mechanism(s) of action for jadomycin B cytotoxicity.
Collapse
Affiliation(s)
- Brendan T McKeown
- Department of Pharmacology. Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Kerry B Goralski
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
- Department of Pharmacology and Department of Pediatrics, Faculty of Medicine and College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, NS, Canada
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre, Halifax, NS, Canada
| |
Collapse
|
5
|
Krüger DN, Bosman M, Van Craenenbroeck EM, De Meyer GRY, Franssen C, Guns PJ. Dexrazoxane prevents vascular toxicity in doxorubicin-treated mice. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:65. [PMID: 39367508 PMCID: PMC11451066 DOI: 10.1186/s40959-024-00270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Doxorubicin (DOX) is used for breast cancer and lymphoma, but can cause cardiotoxicity, arterial stiffness, and endothelial dysfunction. We recently reported SERPINA3N as biomarker of cardiovascular toxicity in patients and mice. Dexrazoxane (DEXRA) is an FDA-approved drug that prevents DOX-induced cardiac toxicity in high-risk patients. However, the effect of DEXRA on vascular dysfunction during DOX treatment has not been documented. Therefore, here we investigated whether DEXRA protects against DOX-induced arterial stiffness, endothelial dysfunction, and SERPINA3N upregulation in tissue and plasma from mice. METHODS Male C57BL6/J mice were treated with DOX (4 mg/kg), DEXRA (40 mg/kg), a combination (DEXRA + DOX), or VEHICLE (0.9% NaCl) weekly i.p. for 6 weeks (n = 8 per group). Cardiovascular function was measured in vivo by ultrasound imaging at baseline, weeks 2 and 6. Vascular reactivity was analyzed ex vivo in the thoracic aorta at week 6 and molecular analysis was performed. RESULTS DEXRA prevented left ventricular ejection fraction decline by DOX (DEXRA + DOX: 62 ± 2% vs DOX: 51 ± 2%). Moreover, DEXRA prevented the increase in pulse wave velocity by DOX (DEXRA + DOX: 2.1 ± 0.2 m/s vs DOX: 4.5 ± 0.3 m/s) and preserved endothelium-dependent relaxation (DEXRA + DOX: 82 ± 3% vs DOX: 62 ± 3%). In contrast to DOX-treated mice, SERPINA3N did not increase in the DEXRA + DOX group. CONCLUSION Our results not only confirm the cardioprotective effects of DEXRA against DOX-induced cardiotoxicity but also add preservation of vascular endothelial cell function as an important mechanism. Moreover, the study demonstrates the potential of SERPINA3N as a biomarker for monitoring cardiovascular complications of DOX in high-risk patients.
Collapse
Affiliation(s)
- Dustin N Krüger
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium.
| | - Matthias Bosman
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem, B-2650, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem, B-2650, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| |
Collapse
|
6
|
Xie S, Sun Y, Zhao X, Xiao Y, Zhou F, Lin L, Wang W, Lin B, Wang Z, Fang Z, Wang L, Zhang Y. An update of the molecular mechanisms underlying anthracycline induced cardiotoxicity. Front Pharmacol 2024; 15:1406247. [PMID: 38989148 PMCID: PMC11234178 DOI: 10.3389/fphar.2024.1406247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Anthracycline drugs mainly include doxorubicin, epirubicin, pirarubicin, and aclamycin, which are widely used to treat a variety of malignant tumors, such as breast cancer, gastrointestinal tumors, lymphoma, etc. With the accumulation of anthracycline drugs in the body, they can induce serious heart damage, limiting their clinical application. The mechanism by which anthracycline drugs cause cardiotoxicity is not yet clear. This review provides an overview of the different types of cardiac damage induced by anthracycline-class drugs and delves into the molecular mechanisms behind these injuries. Cardiac damage primarily involves alterations in myocardial cell function and pathological cell death, encompassing mitochondrial dysfunction, topoisomerase inhibition, disruptions in iron ion metabolism, myofibril degradation, and oxidative stress. Mechanisms of uptake and transport in anthracycline-induced cardiotoxicity are emphasized, as well as the role and breakthroughs of iPSC in cardiotoxicity studies. Selected novel cardioprotective therapies and mechanisms are updated. Mechanisms and protective strategies associated with anthracycline cardiotoxicity in animal experiments are examined, and the definition of drug damage in humans and animal models is discussed. Understanding these molecular mechanisms is of paramount importance in mitigating anthracycline-induced cardiac toxicity and guiding the development of safer approaches in cancer treatment.
Collapse
Affiliation(s)
- Sicong Xie
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuwei Sun
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Zhao
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiqun Xiao
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fei Zhou
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liang Lin
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Wang
- College of Electronic and Optical Engineering and College of Flexible Electronics, Future Technology, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Bin Lin
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Department of Pharmacy, Changxing People's Hospital, Huzhou, China
| | - Zun Wang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zixuan Fang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Wang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Department of Pharmacy, Changxing People's Hospital, Huzhou, China
| |
Collapse
|
7
|
Linders AN, Dias IB, López Fernández T, Tocchetti CG, Bomer N, Van der Meer P. A review of the pathophysiological mechanisms of doxorubicin-induced cardiotoxicity and aging. NPJ AGING 2024; 10:9. [PMID: 38263284 PMCID: PMC10806194 DOI: 10.1038/s41514-024-00135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
The population of cancer survivors is rapidly increasing due to improving healthcare. However, cancer therapies often have long-term side effects. One example is cancer therapy-related cardiac dysfunction (CTRCD) caused by doxorubicin: up to 9% of the cancer patients treated with this drug develop heart failure at a later stage. In recent years, doxorubicin-induced cardiotoxicity has been associated with an accelerated aging phenotype and cellular senescence in the heart. In this review we explain the evidence of an accelerated aging phenotype in the doxorubicin-treated heart by comparing it to healthy aged hearts, and shed light on treatment strategies that are proposed in pre-clinical settings. We will discuss the accelerated aging phenotype and the impact it could have in the clinic and future research.
Collapse
Affiliation(s)
- Annet Nicole Linders
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Itamar Braga Dias
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Teresa López Fernández
- Division of Cardiology, Cardiac Imaging and Cardio-Oncology Unit, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Federico II University, Naples, Italy
- Centre for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
- Interdepartmental Centre of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Centre (CIRIAPA), Federico II University, Naples, Italy
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Peter Van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands.
| |
Collapse
|
8
|
Mody H, Vaidya TR, Lezeau J, Taha K, Ait-Oudhia S. In vitro to clinical translation of combinatorial effects of doxorubicin and dexrazoxane in breast cancer: a mechanism-based pharmacokinetic/pharmacodynamic modeling approach. Front Pharmacol 2023; 14:1239141. [PMID: 37927589 PMCID: PMC10620511 DOI: 10.3389/fphar.2023.1239141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/18/2023] [Indexed: 11/07/2023] Open
Abstract
Dexrazoxane (DEX) is the only drug clinically approved to treat Doxorubicin-induced cardiotoxicity (DIC), however its impact on the anticancer efficacy of DOX is not extensively studied. In this manuscript, a proof-of-concept in vitro study is carried out to quantitatively characterize the anticancer effects of DOX and DEX and determine their nature of drug-drug interactions in cancer cells by combining experimental data with modeling approaches. First, we determined the static concentration-response of DOX and DEX in breast cancer cell lines, JIMT-1 and MDA-MB-468. With a three-dimensional (3D) response surface analysis using a competitive interaction model, we characterized their interaction to be modestly synergistic in MDA-MB-468 or modestly antagonistic in JIMT-1 cells. Second, a cellular-level, pharmacodynamic (PD) model was developed to capture the time-course effects of the two drugs which determined additive and antagonistic interactions for DOX and DEX in MDA-MB-468 and JIMT-1, respectively. Finally, we performed in vitro to in vivo translation by utilizing DOX and DEX clinical dosing regimen that was previously identified to be maximally cardioprotective, to drive tumor cell PD models. The resulting simulations showed that a 10:1 DEX:DOX dose ratio over three cycles of Q3W regimen of DOX results in comparable efficacy based on MDA-MB-468 (additive effect) estimates and lower efficacy based on JIMT-1 (antagonistic effect) estimates for DOX + DEX combination as compared to DOX alone. Thus, our developed cell-based PD models can be used to simulate different scenarios and better design preclinical in vivo studies to further optimize DOX and DEX combinations.
Collapse
Affiliation(s)
- Hardik Mody
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, United States
| | - Tanaya R Vaidya
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, United States
| | - Jovin Lezeau
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, United States
| | - Kareem Taha
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, United States
| | - Sihem Ait-Oudhia
- Quantitative Pharmacology and Pharmacometrics (QP2), Merck & Co., Inc., Rahway, NJ, United States
| |
Collapse
|
9
|
Mattioli R, Ilari A, Colotti B, Mosca L, Fazi F, Colotti G. Doxorubicin and other anthracyclines in cancers: Activity, chemoresistance and its overcoming. Mol Aspects Med 2023; 93:101205. [PMID: 37515939 DOI: 10.1016/j.mam.2023.101205] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023]
Abstract
Anthracyclines have been important and effective treatments against a number of cancers since their discovery. However, their use in therapy has been complicated by severe side effects and toxicity that occur during or after treatment, including cardiotoxicity. The mode of action of anthracyclines is complex, with several mechanisms proposed. It is possible that their high toxicity is due to the large set of processes involved in anthracycline action. The development of resistance is a major barrier to successful treatment when using anthracyclines. This resistance is based on a series of mechanisms that have been studied and addressed in recent years. This work provides an overview of the anthracyclines used in cancer therapy. It discusses their mechanisms of activity, toxicity, and chemoresistance, as well as the approaches used to improve their activity, decrease their toxicity, and overcome resistance.
Collapse
Affiliation(s)
- Roberto Mattioli
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy
| | - Beatrice Colotti
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Luciana Mosca
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy.
| |
Collapse
|
10
|
Marwick TH. Global Longitudinal Strain Monitoring to Guide Cardioprotective Medications During Anthracycline Treatment. Curr Oncol Rep 2022; 24:687-694. [PMID: 35239105 PMCID: PMC9054886 DOI: 10.1007/s11912-022-01242-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 11/30/2022]
Abstract
Purpose of the Review Anthracycline chemotherapy carries a risk of myocardial dysfunction and heart failure even at relatively low doses, and the clinical prediction of cancer treatment-related cardiac dysfunction (CTRCD) is inexact. Careful imaging or biomarker surveillance during chemotherapy can identify CTRCD before the development of heart failure. Currently, this surveillance is performed using ejection fraction (EF). While this is a reliable and reproducible test with three-dimensional techniques, the most widely used imaging technique is two-dimensional echocardiography, for which EF measurements have broad confidence intervals. Recent Findings The use of global myocardial strain (GLS) provides a more reliable and reproducible means of assessing global cardiac function and shows meaningful changes before a significant change of EF. Observational studies have shown that although absolute measurements of GLS, both at baseline and during therapy, are predictive of CTRCD risk, the most reliable approach is to assess the change of GLS with therapy — a meaningful relative change of 10–15% being significant. A clinical trial comparing GLS to EF surveillance did not show a significant change of EF in the overall study group, but did show that patients managed with a the GLS-guided approach were less likely to develop a meaningful change of cardiac function to an abnormal level. In at-risk patients, there is good evidence for the protective value of neurohormonal antagonists and statins: the use of GLS enables these benefits to be directed to those most likely to benefit, while minimizing their use in the majority of people, who do not need them. Summary Although GLS requires an element of training and efforts to ensure uniformity, it has proven to be a feasible, robust, and reproducible technique, ready for wide adoption.
Collapse
Affiliation(s)
- Thomas H Marwick
- Baker Heart and Diabetes Institute, PO Box 6492, Melbourne, Victoria, 3004, Australia.
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Jirkovský E, Jirkovská A, Bavlovič-Piskáčková H, Skalická V, Pokorná Z, Karabanovich G, Kollárová-Brázdová P, Kubeš J, Lenčová-Popelová O, Mazurová Y, Adamcová M, Lyon AR, Roh J, Šimůnek T, Štěrbová-Kovaříková P, Štěrba M. Clinically Translatable Prevention of Anthracycline Cardiotoxicity by Dexrazoxane Is Mediated by Topoisomerase II Beta and Not Metal Chelation. Circ Heart Fail 2021; 14:e008209. [PMID: 34551586 DOI: 10.1161/circheartfailure.120.008209] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Anthracycline-induced heart failure has been traditionally attributed to direct iron-catalyzed oxidative damage. Dexrazoxane (DEX)-the only drug approved for its prevention-has been believed to protect the heart via its iron-chelating metabolite ADR-925. However, direct evidence is lacking, and recently proposed TOP2B (topoisomerase II beta) hypothesis challenged the original concept. METHODS Pharmacokinetically guided study of the cardioprotective effects of clinically used DEX and its chelating metabolite ADR-925 (administered exogenously) was performed together with mechanistic experiments. The cardiotoxicity was induced by daunorubicin in neonatal ventricular cardiomyocytes in vitro and in a chronic rabbit model in vivo (n=50). RESULTS Intracellular concentrations of ADR-925 in neonatal ventricular cardiomyocytes and rabbit hearts after treatment with exogenous ADR-925 were similar or exceeded those observed after treatment with the parent DEX. However, ADR-925 did not protect neonatal ventricular cardiomyocytes against anthracycline toxicity, whereas DEX exhibited significant protective effects (10-100 µmol/L; P<0.001). Unlike DEX, ADR-925 also had no significant impact on daunorubicin-induced mortality, blood congestion, and biochemical and functional markers of cardiac dysfunction in vivo (eg, end point left ventricular fractional shortening was 32.3±14.7%, 33.5±4.8%, 42.7±1.0%, and 41.5±1.1% for the daunorubicin, ADR-925 [120 mg/kg]+daunorubicin, DEX [60 mg/kg]+daunorubicin, and control groups, respectively; P<0.05). DEX, but not ADR-925, inhibited and depleted TOP2B and prevented daunorubicin-induced genotoxic damage. TOP2B dependency of the cardioprotective effects was probed and supported by experiments with diastereomers of a new DEX derivative. CONCLUSIONS This study strongly supports a new mechanistic paradigm that attributes clinically effective cardioprotection against anthracycline cardiotoxicity to interactions with TOP2B but not metal chelation and protection against direct oxidative damage.
Collapse
Affiliation(s)
- Eduard Jirkovský
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic.,Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové (E.J.), Charles University, Czech Republic
| | - Anna Jirkovská
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové (A.J., V.S., J.K., T.Š.), Charles University, Czech Republic
| | - Hana Bavlovič-Piskáčková
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Králové (H.B.-P., P.Š.-K.), Charles University, Czech Republic
| | - Veronika Skalická
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové (A.J., V.S., J.K., T.Š.), Charles University, Czech Republic
| | - Zuzana Pokorná
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic
| | - Galina Karabanovich
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové (G.K., J.R.), Charles University, Czech Republic
| | - Petra Kollárová-Brázdová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic
| | - Jan Kubeš
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové (A.J., V.S., J.K., T.Š.), Charles University, Czech Republic
| | - Olga Lenčová-Popelová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic
| | - Yvona Mazurová
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové (Y.M.), Charles University, Czech Republic
| | - Michaela Adamcová
- Department of Physiology, Faculty of Medicine in Hradec Králové (M.A.), Charles University, Czech Republic
| | - Alexander R Lyon
- Department of Cardiology, Royal Brompton Hospital and Faculty of Medicine, National Heart and Lung Institute, Imperial College London, United Kingdom (A.R.L.)
| | - Jaroslav Roh
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové (G.K., J.R.), Charles University, Czech Republic
| | - Tomáš Šimůnek
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové (A.J., V.S., J.K., T.Š.), Charles University, Czech Republic
| | - Petra Štěrbová-Kovaříková
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Králové (H.B.-P., P.Š.-K.), Charles University, Czech Republic
| | - Martin Štěrba
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., Z.P., P.K.-B., O.L.-P., M.Š.), Charles University, Czech Republic
| |
Collapse
|
12
|
Sawicki KT, Sala V, Prever L, Hirsch E, Ardehali H, Ghigo A. Preventing and Treating Anthracycline Cardiotoxicity: New Insights. Annu Rev Pharmacol Toxicol 2021; 61:309-332. [PMID: 33022184 DOI: 10.1146/annurev-pharmtox-030620-104842] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Anthracyclines are the cornerstone of many chemotherapy regimens for a variety of cancers. Unfortunately, their use is limited by a cumulative dose-dependent cardiotoxicity. Despite more than five decades of research, the biological mechanisms underlying anthracycline cardiotoxicity are not completely understood. In this review, we discuss the incidence, risk factors, types, and pathophysiology of anthracycline cardiotoxicity, as well as methods to prevent and treat this condition. We also summarize and discuss advances made in the last decade in the comprehension of the molecular mechanisms underlying the pathology.
Collapse
Affiliation(s)
- Konrad Teodor Sawicki
- Division of Cardiology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Lorenzo Prever
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Hossein Ardehali
- Division of Cardiology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| |
Collapse
|
13
|
Prodrug of ICRF-193 provides promising protective effects against chronic anthracycline cardiotoxicity in a rabbit model in vivo. Clin Sci (Lond) 2021; 135:1897-1914. [PMID: 34318878 DOI: 10.1042/cs20210311] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 11/17/2022]
Abstract
The anthracycline (ANT) anticancer drugs such as doxorubicin or daunorubicin (DAU) can cause serious myocardial injury and chronic cardiac dysfunction in cancer survivors. A bisdioxopiperazine agent dexrazoxane (DEX) has been developed as a cardioprotective drug to prevent these adverse events, but it is uncertain whether it is the best representative of the class. The present study used a rabbit model of chronic ANT cardiotoxicity to examine another bisdioxopiperazine compound called GK-667 (meso-(butane-2,3-diylbis(2,6-dioxopiperazine-4,1-diyl))bis(methylene)-bis(2-aminoacetate) hydrochloride), a water-soluble prodrug of ICRF-193 (meso-4,4'-(butan-2,3-diyl)bis(piperazine-2,6-dione)), as a potential cardioprotectant. The cardiotoxicity was induced by DAU (3 mg/kg, intravenously, weekly, 10 weeks), and GK-667 (1 or 5 mg/kg, intravenously) was administered before each DAU dose. The treatment with GK-667 was well tolerated and provided full protection against DAU-induced mortality and left ventricular (LV) dysfunction (determined by echocardiography and LV catheterization). Markers of cardiac damage/dysfunction revealed minor cardiac damage in the group co-treated with GK-667 in the lower dose, whereas almost full protection was achieved with the higher dose. This was associated with similar prevention of DAU-induced dysregulation of redox and calcium homeostasis proteins. GK-667 dose-dependently prevented tumor suppressor p53 (p53)-mediated DNA damage response in the LV myocardium not only in the chronic experiment but also after single DAU administration. These effects appear essential for cardioprotection, presumably because of the topoisomerase IIβ (TOP2B) inhibition provided by its active metabolite ICRF-193. In addition, GK-667 administration did not alter the plasma pharmacokinetics of DAU and its main metabolite daunorubicinol (DAUol) in rabbits in vivo. Hence, GK-667 merits further investigation as a promising drug candidate for cardioprotection against chronic ANT cardiotoxicity.
Collapse
|
14
|
Shi H, Zeng Q, Wei Y, Yang H, Tang H, Wang D, Pu P, Feng R. Canagliflozin is a potential cardioprotective drug but exerts no significant effects on pirarubicin‑induced cardiotoxicity in rats. Mol Med Rep 2021; 24:703. [PMID: 34368866 DOI: 10.3892/mmr.2021.12342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/09/2021] [Indexed: 11/05/2022] Open
Abstract
Pirarubicin (THP), one of the anthracycline anticancer drugs, is widely used in the treatment of various types of cancer, but its cardiotoxicity cannot be ignored. Canagliflozin, the first sodium‑glucose co‑transporter‑2 inhibitor approved by the USA FDA, has been shown to have a significant effect on cardiovascular damage caused by diabetes. However, it has not been reported whether it can resist THP‑induced cardiotoxicity. The aim of the present study was to investigate the effect of canagliflozin on THP‑induced cardiotoxicity and its mechanism. A rat model of cardiotoxicity induced by THP was established and canagliflozin treatment was performed at the same time. The changes of electrocardiography, cardiac coefficient and echocardiogram were observed. The levels of lactate dehydrogenase, brain natriuretic peptide, creatine kinase MB, cardiac troponin T, superoxide dismutase (SOD) and malondialdehyde were detected. The expression of SOD2, NADPH oxidase 2, pro/cleaved‑caspase‑ and Bcl‑2/Bax were evaluated by western blotting. The primary culture of cardiomyocytes was prepared to explore the effect in vitro. After eight weeks, a series of cardiotoxicity manifestations were observed in THP rats. However, canagliflozin treatment had no significant effect on the above adverse reactions. Similarly, further studies showed that canagliflozin had no significant effect on THP‑induced cardiomyocyte injury in vitro. The present study showed that there was no significant protective effect of canagliflozin on THP‑induced cardiotoxicity and cardiomyocyte injury.
Collapse
Affiliation(s)
- Hongwei Shi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Qingfu Zeng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yunjie Wei
- Department of Cardiology, Hubei Shiyan Taihe Hospital, Wuhan, Hubei 430000, P.R. China
| | - Hong Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Yuanjiagang, Yuzhong, Chongqing 400042, P.R. China
| | - Heng Tang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Yuanjiagang, Yuzhong, Chongqing 400042, P.R. China
| | - Dan Wang
- Department of Cardiology, Chongqing Red Cross Hospital, Yuzhong, Chongqing 400020, P.R. China
| | - Peng Pu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Yuanjiagang, Yuzhong, Chongqing 400042, P.R. China
| | - Rui Feng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Yuanjiagang, Yuzhong, Chongqing 400042, P.R. China
| |
Collapse
|
15
|
Qin Y, Guo T, Wang Z, Zhao Y. The role of iron in doxorubicin-induced cardiotoxicity: recent advances and implication for drug delivery. J Mater Chem B 2021; 9:4793-4803. [PMID: 34059858 DOI: 10.1039/d1tb00551k] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
As an anthracycline antibiotic, doxorubicin (DOX) is one of the most potent and widely used chemotherapeutic agents for treating various types of tumors. Unfortunately, the clinical application of this drug results in severe side effects, particularly dose-dependent cardiotoxicity. There are multiple mechanisms involved with the cardiotoxicity caused by DOX, among which intracellular iron homeostasis plays an essential role based on a recent discovery. In this mini-review, we summarize the clinical features and symptoms of DOX-dependent cardiotoxicity, discuss the correlation between iron and cardiotoxicity, and highlight the involvement of iron-dependent ferroptotic cell death therein. Recent advances in this topic will aid the development of novel DOX delivery systems with reduced adverse effects, and expand the clinical application of anthracycline.
Collapse
Affiliation(s)
- Yan Qin
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | | | | | | |
Collapse
|
16
|
Eilenberger C, Rothbauer M, Selinger F, Gerhartl A, Jordan C, Harasek M, Schädl B, Grillari J, Weghuber J, Neuhaus W, Küpcü S, Ertl P. A Microfluidic Multisize Spheroid Array for Multiparametric Screening of Anticancer Drugs and Blood-Brain Barrier Transport Properties. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004856. [PMID: 34105271 PMCID: PMC8188192 DOI: 10.1002/advs.202004856] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/30/2021] [Indexed: 05/08/2023]
Abstract
Physiological-relevant in vitro tissue models with their promise of better predictability have the potential to improve drug screening outcomes in preclinical studies. Despite the advances of spheroid models in pharmaceutical screening applications, variations in spheroid size and consequential altered cell responses often lead to nonreproducible and unpredictable results. Here, a microfluidic multisize spheroid array is established and characterized using liver, lung, colon, and skin cells as well as a triple-culture model of the blood-brain barrier (BBB) to assess the effects of spheroid size on (a) anticancer drug toxicity and (b) compound penetration across an advanced BBB model. The reproducible on-chip generation of 360 spheroids of five dimensions on a well-plate format using an integrated microlens technology is demonstrated. While spheroid size-related IC50 values vary up to 160% using the anticancer drugs cisplatin (CIS) or doxorubicin (DOX), reduced CIS:DOX drug dose combinations eliminate all lung microtumors independent of their sizes. A further application includes optimizing cell seeding ratios and size-dependent compound uptake studies in a perfused BBB model. Generally, smaller BBB-spheroids reveal an 80% higher compound penetration than larger spheroids while verifying the BBB opening effect of mannitol and a spheroid size-related modulation on paracellular transport properties.
Collapse
Affiliation(s)
- Christoph Eilenberger
- Faculty of Technical ChemistryVienna University of TechnologyGetreidemarkt 9Vienna1060Austria
| | - Mario Rothbauer
- Faculty of Technical ChemistryVienna University of TechnologyGetreidemarkt 9Vienna1060Austria
- Karl Chiari Lab for Orthopaedic BiologyDepartment of Orthopedics and Trauma SurgeryMedical University of ViennaWähringer Gürtel 18‐20Vienna1090Austria
| | - Florian Selinger
- Faculty of Technical ChemistryVienna University of TechnologyGetreidemarkt 9Vienna1060Austria
| | - Anna Gerhartl
- AIT Austrian Institute of Technology GmbHCenter Health and BioresourcesCompetence Unit Molecular DiagnosticsGiefinggasse 4Vienna1210Austria
| | - Christian Jordan
- Faculty of Technical ChemistryVienna University of TechnologyGetreidemarkt 9Vienna1060Austria
| | - Michael Harasek
- Faculty of Technical ChemistryVienna University of TechnologyGetreidemarkt 9Vienna1060Austria
| | - Barbara Schädl
- Ludwig‐Boltzmann‐Institute for Experimental and Clinical TraumatologyDonaueschingenstraße 13Vienna1200Austria
| | - Johannes Grillari
- Ludwig‐Boltzmann‐Institute for Experimental and Clinical TraumatologyDonaueschingenstraße 13Vienna1200Austria
- Institute for Molecular BiotechnologyDepartment of BiotechnologyUniversity of Natural Resources and Life SciencesMuthgasse 18Vienna1190Austria
| | - Julian Weghuber
- School of EngineeringUniversity of Applied Sciences Upper AustriaStelzhamerstraße 23Wels4600Austria
- FFoQSI GmbH‐Austrian Competence Centre for Feed and Food QualitySafety and InnovationTechnopark 1CTulln3430Austria
| | - Winfried Neuhaus
- AIT Austrian Institute of Technology GmbHCenter Health and BioresourcesCompetence Unit Molecular DiagnosticsGiefinggasse 4Vienna1210Austria
| | - Seta Küpcü
- Institute of Synthetic BioarchitecturesDepartment of NanobiotechnologyUniversity of Natural Resources and Life SciencesVienna, Muthgasse 11Vienna1190Austria
| | - Peter Ertl
- Faculty of Technical ChemistryVienna University of TechnologyGetreidemarkt 9Vienna1060Austria
| |
Collapse
|
17
|
Jirkovská A, Karabanovich G, Kubeš J, Skalická V, Melnikova I, Korábečný J, Kučera T, Jirkovský E, Nováková L, Bavlovič Piskáčková H, Škoda J, Štěrba M, Austin CA, Šimůnek T, Roh J. Structure-Activity Relationship Study of Dexrazoxane Analogues Reveals ICRF-193 as the Most Potent Bisdioxopiperazine against Anthracycline Toxicity to Cardiomyocytes Due to Its Strong Topoisomerase IIβ Interactions. J Med Chem 2021; 64:3997-4019. [PMID: 33750129 DOI: 10.1021/acs.jmedchem.0c02157] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cardioprotective activity of dexrazoxane (ICRF-187), the only clinically approved drug against anthracycline-induced cardiotoxicity, has traditionally been attributed to its iron-chelating metabolite. However, recent experimental evidence suggested that the inhibition and/or depletion of topoisomerase IIβ (TOP2B) by dexrazoxane could be cardioprotective. Hence, we evaluated a series of dexrazoxane analogues and found that their cardioprotective activity strongly correlated with their interaction with TOP2B in cardiomyocytes, but was independent of their iron chelation ability. Very tight structure-activity relationships were demonstrated on stereoisomeric forms of 4,4'-(butane-2,3-diyl)bis(piperazine-2,6-dione). In contrast to its rac-form 12, meso-derivative 11 (ICRF-193) showed a favorable binding mode to topoisomerase II in silico, inhibited and depleted TOP2B in cardiomyocytes more efficiently than dexrazoxane, and showed the highest cardioprotective efficiency. Importantly, the observed ICRF-193 cardioprotection did not interfere with the antiproliferative activity of anthracycline. Hence, this study identifies ICRF-193 as the new lead compound in the development of efficient cardioprotective agents.
Collapse
Affiliation(s)
- Anna Jirkovská
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Galina Karabanovich
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Jan Kubeš
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Veronika Skalická
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Iuliia Melnikova
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Jan Korábečný
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 50005 Hradec Králové, Czech Republic
- Faculty of Military Health Sciences, University of Defence, Třebešská 1575, 50005 Hradec Králové, Czech Republic
| | - Tomáš Kučera
- Faculty of Military Health Sciences, University of Defence, Třebešská 1575, 50005 Hradec Králové, Czech Republic
| | - Eduard Jirkovský
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Lucie Nováková
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Hana Bavlovič Piskáčková
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Josef Škoda
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Martin Štěrba
- Department of Pharmacology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 50003 Hradec Králové, Czech Republic
| | - Caroline A Austin
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Tomáš Šimůnek
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| | - Jaroslav Roh
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 50005 Hradec Králové, Czech Republic
| |
Collapse
|
18
|
Sutormin DA, Galivondzhyan AK, Polkhovskiy AV, Kamalyan SO, Severinov KV, Dubiley SA. Diversity and Functions of Type II Topoisomerases. Acta Naturae 2021; 13:59-75. [PMID: 33959387 PMCID: PMC8084294 DOI: 10.32607/actanaturae.11058] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/09/2020] [Indexed: 11/29/2022] Open
Abstract
The DNA double helix provides a simple and elegant way to store and copy genetic information. However, the processes requiring the DNA helix strands separation, such as transcription and replication, induce a topological side-effect - supercoiling of the molecule. Topoisomerases comprise a specific group of enzymes that disentangle the topological challenges associated with DNA supercoiling. They relax DNA supercoils and resolve catenanes and knots. Here, we review the catalytic cycles, evolution, diversity, and functional roles of type II topoisomerases in organisms from all domains of life, as well as viruses and other mobile genetic elements.
Collapse
Affiliation(s)
- D. A. Sutormin
- Institute of Gene Biology RAS, Moscow, 119334 Russia
- Centre for Life Sciences, Skolkovo Institute of Science and Technology, Moscow, 121205 Russia
| | - A. K. Galivondzhyan
- Lomonosov Moscow State University, Moscow, 119991 Russia
- Institute of Molecular Genetics RAS, Moscow, 123182 Russia
| | - A. V. Polkhovskiy
- Institute of Gene Biology RAS, Moscow, 119334 Russia
- Centre for Life Sciences, Skolkovo Institute of Science and Technology, Moscow, 121205 Russia
| | - S. O. Kamalyan
- Institute of Gene Biology RAS, Moscow, 119334 Russia
- Centre for Life Sciences, Skolkovo Institute of Science and Technology, Moscow, 121205 Russia
| | - K. V. Severinov
- Centre for Life Sciences, Skolkovo Institute of Science and Technology, Moscow, 121205 Russia
- Centre for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology RAS, Moscow, 119334 Russia
- Waksman Institute for Microbiology, Piscataway, New Jersey, 08854 USA
| | - S. A. Dubiley
- Institute of Gene Biology RAS, Moscow, 119334 Russia
- Centre for Life Sciences, Skolkovo Institute of Science and Technology, Moscow, 121205 Russia
| |
Collapse
|
19
|
Olorundare O, Adeneye A, Akinsola A, Kolo P, Agede O, Soyemi S, Mgbehoma A, Okoye I, Albrecht R, Mukhtar H. Irvingia gabonensis Seed Extract: An Effective Attenuator of Doxorubicin-Mediated Cardiotoxicity in Wistar Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1602816. [PMID: 33149803 PMCID: PMC7603620 DOI: 10.1155/2020/1602816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 01/16/2023]
Abstract
Cardiotoxicity as an off-target effect of doxorubicin therapy is a major limiting factor for its clinical use as a choice cytotoxic agent. Seeds of Irvingia gabonensis have been reported to possess both nutritional and medicinal values which include antidiabetic, weight losing, antihyperlipidemic, and antioxidative effects. Protective effects of Irvingia gabonensis ethanol seed extract (IGESE) was investigated in doxorubicin (DOX)-mediated cardiotoxicity induced with single intraperitoneal injection of 15 mg/kg of DOX following the oral pretreatments of Wistar rats with 100-400 mg/kg/day of IGESE for 10 days, using serum cardiac enzyme markers (cardiac troponin I (cTI) and lactate dehydrogenase (LDH)), cardiac tissue oxidative stress markers (catalase (CAT), malonyldialdehyde (MDA), superoxide dismutase (SOD), glutathione-S-transferase (GST), glutathione peroxidase (GSH-Px), and reduced glutathione (GSH)), and cardiac histopathology endpoints. In addition, both qualitative and quantitative analyses to determine IGESE's secondary metabolites profile and its in vitro antioxidant activities were also conducted. Results revealed that serum cTnI and LDH were significantly elevated by the DOX treatment. Similarly, activities of tissue SOD, CAT, GST, and GSH levels were profoundly reduced, while GPx activity and MDA levels were profoundly increased by DOX treatment. These biochemical changes were associated with microthrombi formation in the DOX-treated cardiac tissues on histological examination. However, oral pretreatments with 100-400 mg/kg/day of IGESE dissolved in 5% DMSO in distilled water significantly attenuated increases in the serum cTnI and LDH, prevented significant alterations in the serum lipid profile and the tissue activities and levels of oxidative stress markers while improving cardiovascular disease risk indices and DOX-induced histopathological lesions. The in vitro antioxidant studies showed IGESE to have good antioxidant profile and contained 56 major secondary metabolites prominent among which are γ-sitosterol, Phytol, neophytadiene, stigmasterol, vitamin E, hexadecanoic acid and its ethyl ester, Phytyl palmitate, campesterol, lupeol, and squalene. Overall, both the in vitro and in vivo findings indicate that IGESE may be a promising prophylactic cardioprotective agent against DOX-induced cardiotoxicity, at least in part mediated via IGESE's antioxidant and free radical scavenging and antithrombotic mechanisms.
Collapse
Affiliation(s)
- Olufunke Olorundare
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Adejuwon Adeneye
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Clinical Sciences, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Akinyele Akinsola
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Phillip Kolo
- Department of Medicine, Faculty of Clinical, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Olalekan Agede
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Sunday Soyemi
- Department of Pathology and Forensic Medicine, Faculty of Basic Clinical Sciences, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Alban Mgbehoma
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Ikechukwu Okoye
- Department of Oral Pathology and Medicine, Faculty of Dentistry, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Ralph Albrecht
- Department of Animal Sciences, 1675 Observatory Drive, University of Wisconsin, Madison, WI 53706, USA
| | - Hasan Mukhtar
- Department of Dermatology, University of Wisconsin, Madison, Medical Science Center, 1300 University Avenue, Madison, WI 53706, USA
| |
Collapse
|
20
|
Petrykey K, Andelfinger GU, Laverdière C, Sinnett D, Krajinovic M. Genetic factors in anthracycline-induced cardiotoxicity in patients treated for pediatric cancer. Expert Opin Drug Metab Toxicol 2020; 16:865-883. [DOI: 10.1080/17425255.2020.1807937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Kateryna Petrykey
- Immune Diseases and Cancer, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Université De Montréal (Quebec), Montreal, Canada
| | - Gregor U. Andelfinger
- Department of Pediatrics, Université De Montréal (Quebec), Canada
- Fetomaternal and Neonatal Pathologies, Sainte-JustineUniversity Health Center (SJUHC), Montreal, Quebec, Canada
| | - Caroline Laverdière
- Immune Diseases and Cancer, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada
- Department of Pediatrics, Université De Montréal (Quebec), Canada
| | - Daniel Sinnett
- Immune Diseases and Cancer, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada
- Department of Pediatrics, Université De Montréal (Quebec), Canada
| | - Maja Krajinovic
- Immune Diseases and Cancer, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada
- Department of Pharmacology and Physiology, Université De Montréal (Quebec), Montreal, Canada
- Department of Pediatrics, Université De Montréal (Quebec), Canada
| |
Collapse
|
21
|
Costa VM, Capela JP, Sousa JR, Eleutério RP, Rodrigues PRS, Dores-Sousa JL, Carvalho RA, Lourdes Bastos M, Duarte JA, Remião F, Almeida MG, Varner KJ, Carvalho F. Mitoxantrone impairs proteasome activity and prompts early energetic and proteomic changes in HL-1 cardiomyocytes at clinically relevant concentrations. Arch Toxicol 2020; 94:4067-4084. [DOI: 10.1007/s00204-020-02874-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/12/2020] [Indexed: 11/24/2022]
|
22
|
The Role of Oxidative Stress in Physiopathology and Pharmacological Treatment with Pro- and Antioxidant Properties in Chronic Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2082145. [PMID: 32774665 PMCID: PMC7396016 DOI: 10.1155/2020/2082145] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/08/2020] [Indexed: 01/01/2023]
Abstract
Oxidative stress (OS) has the ability to damage different molecules and cellular structures, altering the correct function of organs and systems. OS accumulates in the body by endogenous and exogenous mechanisms. Increasing evidence points to the involvement of OS in the physiopathology of various chronic diseases that require prolonged periods of pharmacological treatment. Long-term treatments may contribute to changes in systemic OS. In this review, we discuss the involvement of OS in the pathological mechanisms of some chronic diseases, the pro- or antioxidant effects of their pharmacological treatments, and possible adjuvant antioxidant alternatives. Diseases such as high blood pressure, arteriosclerosis, and diabetes mellitus contribute to the increased risk of cardiovascular disease. Antihypertensive, lipid-lowering, and hypoglycemic treatments help reduce the risk with an additional antioxidant benefit. Treatment with methotrexate in autoimmune systemic inflammatory diseases, such as rheumatoid arthritis, has a dual role in stimulating the production of OS and producing mitochondrial dysfunction. However, it can also help indirectly decrease the systemic OS induced by inflammation. Medicaments used to treat neurodegenerative diseases tend to decrease the mechanisms related to the production of reactive oxygen species (ROS) and balance OS. On the other hand, immunosuppressive treatments used in cancer or human immunodeficiency virus infection increase the production of ROS, causing significant oxidative damage in different organs and systems without widely documented exogenous antioxidant administration alternatives.
Collapse
|
23
|
Kollárová-Brázdová P, Jirkovská A, Karabanovich G, Pokorná Z, Bavlovič Piskáčková H, Jirkovský E, Kubeš J, Lenčová-Popelová O, Mazurová Y, Adamcová M, Skalická V, Štěrbová-Kovaříková P, Roh J, Šimůnek T, Štěrba M. Investigation of Structure-Activity Relationships of Dexrazoxane Analogs Reveals Topoisomerase II β Interaction as a Prerequisite for Effective Protection against Anthracycline Cardiotoxicity. J Pharmacol Exp Ther 2020; 373:402-415. [PMID: 32253261 DOI: 10.1124/jpet.119.264580] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/23/2020] [Indexed: 01/23/2023] Open
Abstract
Bisdioxopiperazine agent dexrazoxane (ICRF-187) has been the only effective and approved drug for prevention of chronic anthracycline cardiotoxicity. However, the structure-activity relationships (SARs) of its cardioprotective effects remain obscure owing to limited investigation of its derivatives/analogs and uncertainties about its mechanism of action. To fill these knowledge gaps, we tested the hypothesis that dexrazoxane derivatives exert cardioprotection via metal chelation and/or modulation of topoisomerase IIβ (Top2B) activity in chronic anthracycline cardiotoxicity. Dexrazoxane was alkylated in positions that should not interfere with the metal-chelating mechanism of cardioprotective action; that is, on dioxopiperazine imides or directly on the dioxopiperazine ring. The protective effects of these agents were assessed in vitro in neonatal cardiomyocytes. All studied modifications of dexrazoxane molecule, including simple methylation, were found to abolish the cardioprotective effects. Because this challenged the prevailing mechanistic concept and previously reported data, the two closest derivatives [(±)-4,4'-(propane-1,2-diyl)bis(1-methylpiperazine-2,6-dione) and 4-(2-(3,5-dioxopiperazin-1-yl)ethyl)-3-methylpiperazine-2,6-dione] were thoroughly scrutinized in vivo using a rabbit model of chronic anthracycline cardiotoxicity. In contrast to dexrazoxane, both compounds failed to protect the heart, as demonstrated by mortality, cardiac dysfunction, and myocardial damage parameters, although the pharmacokinetics and metal-chelating properties of their metabolites were comparable to those of dexrazoxane. The loss of cardiac protection was shown to correlate with their abated potential to inhibit and deplete Top2B both in vitro and in vivo. These findings suggest a very tight SAR between bisdioxopiperazine derivatives and their cardioprotective effects and support Top2B as a pivotal upstream druggable target for effective cardioprotection against anthracycline cardiotoxicity. SIGNIFICANCE STATEMENT: This study has revealed the previously unexpected tight structure-activity relationships of cardioprotective effects in derivatives of dexrazoxane, which is the only drug approved for the prevention of cardiomyopathy and heart failure induced by anthracycline anticancer drugs. The data presented in this study also strongly argue against the importance of metal-chelating mechanisms for the induction of this effect and support the viability of topoisomerase IIβ as an upstream druggable target for effective and clinically translatable cardioprotection.
Collapse
Affiliation(s)
- Petra Kollárová-Brázdová
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Anna Jirkovská
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Galina Karabanovich
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Zuzana Pokorná
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Hana Bavlovič Piskáčková
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Eduard Jirkovský
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Jan Kubeš
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Olga Lenčová-Popelová
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Yvona Mazurová
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Michaela Adamcová
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Veronika Skalická
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Petra Štěrbová-Kovaříková
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Jaroslav Roh
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Tomáš Šimůnek
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Martin Štěrba
- Departments of Pharmacology (P.K.-B., Z.P., E.J., O.L.-P., M.Š.), Histology and Embryology (Y.M.), and Physiology (M.A.), Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic; and Departments of Biochemical Sciences (A.J., J.K., V.S., T.Š.), Organic and Bioorganic Chemistry (G.K., J.R.), Pharmaceutical Chemistry and Pharmaceutical Analysis (H.B.P., P.Š.-K.), and Pharmacology and Toxicology (E.J.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
24
|
Li J, Chang HM, Banchs J, Araujo DM, Hassan SA, Wagar EA, Yeh ETH, Meng QH. Detection of subclinical cardiotoxicity in sarcoma patients receiving continuous doxorubicin infusion or pre-treatment with dexrazoxane before bolus doxorubicin. CARDIO-ONCOLOGY 2020; 6:1. [PMID: 32154027 PMCID: PMC7048030 DOI: 10.1186/s40959-019-0056-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/28/2019] [Indexed: 12/03/2022]
Abstract
Background Continuous infusion of doxorubicin or dexrazoxane pre-treatment prior to bolus doxorubicin are proven strategies to protect against doxorubicin-induced cardiotoxicity. Recently, global longitudinal peak systolic strain (GLS) measured with speckle tracking echocardiography (STE) and high-sensitivity troponin T (hs-TnT) have been validated as sensitive indicators of doxorubicin-induced cardiotoxicity. Here, we asked whether changes in hs-TnT and/or GLS can be detected in patients who were treated with continuous infusion of doxorubicin or pre-treated with dexrazoxane followed by bolus doxorubicin. Methods Twenty-nine patients with newly diagnosed sarcoma were assigned to receive either 72-h doxorubicin infusion or dexrazoxane pre-treatment before bolus doxorubicin. Eight patients received dexrazoxane pre-treatment; eleven patients received continuous doxorubicin infusion; ten patients crossed over from continuous infusion to dexrazoxane. Bloods were collected for hs-TnT at baseline, 24 h or 72 h after initiation of doxorubicin treatment in each chemotherapy cycle. All blood samples were assayed in batch using hs-TnT kit from Roche diagnostics. 2D Echo and STE were performed before doxorubicin, after cycle 3, and at the end of chemotherapy. Results Seven patients in the cross-over group have at least one hs-TnT measurement between 5 ng/L to 10 ng/L during and after chemotherapy. Ten patients have at least one hs-TnT measurement above 10 ng/ml during and after chemotherapy (six in dexrazoxane group, three in continuous infusion group, one in cross-over group). The average hs-TnT level increases with each additional cycle of doxorubicin treatment. Eight patients had a more than 5% reduction in LVEF at the end of chemotherapy (four in dexrazoxane group, three in continuous infusion group, and one in cross-over group). Four out of these eight patients had a change of GLS by more than 15% (three in the dexrazoxane group). Conclusion Elevation in hs-TnT levels were observed in more than 59% of patients who had received either continuous doxorubicin infusion or dexrazoxane pre-treatment before bolus doxorubicin. However, changes in LVEF and GLS were less frequently observed. Thus, continuous doxorubicin infusion or dexrazoxane pre-treatment do not completely ameliorate subclinical doxorubicin-induced cardiotoxicity as detected by more sensitive techniques.
Collapse
Affiliation(s)
- Jieli Li
- 1Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Hui-Ming Chang
- 2Center for Precision Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO USA
| | - Jose Banchs
- 3Departments of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Dejka M Araujo
- 4Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Saamir A Hassan
- 3Departments of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Elizabeth A Wagar
- 1Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Edward T H Yeh
- 2Center for Precision Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO USA
| | - Qing H Meng
- 1Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
25
|
Rohilla S, Dureja H, Chawla V. Cytoprotective Agents to Avoid Chemotherapy Induced Sideeffects on Normal Cells: A Review. Curr Cancer Drug Targets 2019; 19:765-781. [PMID: 30914026 DOI: 10.2174/1568009619666190326120457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 03/01/2019] [Accepted: 03/10/2019] [Indexed: 01/16/2023]
Abstract
Anticancer agents play a vital role in the cure of patients suffering from malignancy. Though, the chemotherapeutic agents are associated with various adverse effects which produce significant toxic symptoms in the patients. But this therapy affects both the malignant and normal cells and leads to constricted therapeutic index of antimalignant drugs which adversely impacts the quality of patients’ life. Due to these adversities, sufficient dose of drug is not delivered to patients leading to delay in treatment or improper treatment. Chemoprotective agents have been developed either to minimize or to mitigate the toxicity allied with chemotherapeutic agents. Without any concession in the therapeutic efficacy of anticancer drugs, they provide organ specific guard to normal tissues.
Collapse
Affiliation(s)
- Seema Rohilla
- Department of Pharmaceutics, Hindu College of Pharmacy, Sonepat- 131001, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak-124001, India
| | - Vinay Chawla
- Institute of Pharmaceutical Sciences, Baba Farid University of Health Sciences, Faridkot-151203, India
| |
Collapse
|
26
|
Varela-López A, Battino M, Navarro-Hortal MD, Giampieri F, Forbes-Hernández TY, Romero-Márquez JM, Collado R, Quiles JL. An update on the mechanisms related to cell death and toxicity of doxorubicin and the protective role of nutrients. Food Chem Toxicol 2019; 134:110834. [PMID: 31577924 DOI: 10.1016/j.fct.2019.110834] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/10/2019] [Accepted: 09/21/2019] [Indexed: 12/11/2022]
Abstract
Doxorubicin (DOX), is a very effective chemotherapeutic agent against cancer whose clinical use is limited by toxicity. Different strategies have been proposed to attenuate toxicity, including combined therapy with bioactive compounds. This review update mechanisms of action and toxicity of doxorubicin and the role of nutrients like vitamins (A, C, E), minerals (selenium) and n-3 polyunsaturated fatty acids. Protective activities against DOX toxicity in liver, kidney, skin, bone marrow, testicles or brain have been reported, but these have not been evaluated for all of the reviewed nutrients. In most cases oxidation-related effects were present either, by reducing ROS levels and/or increasing antioxidant defenses. Antiapoptotic and anti-inflammatory mechanisms are also commonly reported. In some cases, interferences with autophagy and calcium homeostasis also have shown to be affected. Notwithstanding, there is a wide variety in duration and doses of treatment tested for both, compounds and DOX, which make difficult to compare the results of the studies. In spite of the reduction of DOX cardiotoxicity in health models, DOX anti-cancer activity in cancer cell lines or xenograft models usually did not result compromised when this has been evaluated. Importantly, clinical studies are needed to confirm all the observed effects.
Collapse
Affiliation(s)
- Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology ''José Mataix", Biomedical Research Centre, University of Granada, 18071, Granada, Spain
| | - Maurizio Battino
- Dipartimento di Scienze Cliniche Specialistiche Ed Odontostomatologiche (DISCO)-Sez, Biochimica, Facoltà di Medicina, Università Politecnica Delle Marche, 60131, Ancona, Italy; Nutrition and Food Science Group. Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, Vigo, Spain; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China
| | - María D Navarro-Hortal
- Department of Physiology, Institute of Nutrition and Food Technology ''José Mataix", Biomedical Research Centre, University of Granada, 18071, Granada, Spain
| | - Francesca Giampieri
- Dipartimento di Scienze Cliniche Specialistiche Ed Odontostomatologiche (DISCO)-Sez, Biochimica, Facoltà di Medicina, Università Politecnica Delle Marche, 60131, Ancona, Italy
| | - Tamara Y Forbes-Hernández
- Nutrition and Food Science Group. Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, Vigo, Spain
| | - José M Romero-Márquez
- Department of Physiology, Institute of Nutrition and Food Technology ''José Mataix", Biomedical Research Centre, University of Granada, 18071, Granada, Spain
| | - Ricardo Collado
- Complejo Hospitalario Universitario de Cáceres, Cáceres, Spain
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology ''José Mataix", Biomedical Research Centre, University of Granada, 18071, Granada, Spain.
| |
Collapse
|
27
|
In vitro and in vivo investigation of cardiotoxicity associated with anticancer proteasome inhibitors and their combination with anthracycline. Clin Sci (Lond) 2019; 133:1827-1844. [PMID: 31409729 DOI: 10.1042/cs20190139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/26/2019] [Accepted: 08/12/2019] [Indexed: 01/07/2023]
Abstract
Although proteasome inhibitors (PIs) are modern targeted anticancer drugs, they have been associated with a certain risk of cardiotoxicity and heart failure (HF). Recently, PIs have been combined with anthracyclines (ANTs) to further boost their anticancer efficacy. However, this raised concerns regarding cardiac safety, which were further supported by several in vitro studies on immature cardiomyocytes. In the present study, we investigated the toxicity of clinically used PIs alone (bortezomib (BTZ), carfilzomib (CFZ)) as well as their combinations with an ANT (daunorubicin (DAU)) in both neonatal and adult ventricular cardiomyocytes (NVCMs and AVCMs) and in a chronic rabbit model of DAU-induced HF. Using NVCMs, we found significant cytotoxicity of both PIs around their maximum plasma concentration (cmax) as well as significant augmentation of DAU cytotoxicity. In AVCMs, BTZ did not induce significant cytotoxicity in therapeutic concentrations, whereas the toxicity of CFZ was significant and more profound. Importantly, neither PI significantly augmented the cardiotoxicity of DAU despite even more profound proteasome-inhibitory activity in AVCMs compared with NVCMs. Furthermore, in young adult rabbits, no significant augmentation of chronic ANT cardiotoxicity was noted with respect to any functional, morphological, biochemical or molecular parameter under study, despite significant inhibition of myocardial proteasome activity. Our experimental data show that combination of PIs with ANTs is not accompanied by an exaggerated risk of cardiotoxicity and HF in young adult animal cardiomyocytes and hearts.
Collapse
|
28
|
Reimerová P, Jirkovská A, Piskáčková HB, Karabanovich G, Roh J, Šimůnek T, Štěrbová-Kovaříková P. UHPLC-MS/MS method for analysis of sobuzoxane, its active form ICRF-154 and metabolite EDTA-diamide and its application to bioactivation study. Sci Rep 2019; 9:4524. [PMID: 30872754 PMCID: PMC6418109 DOI: 10.1038/s41598-019-40928-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/26/2019] [Indexed: 11/09/2022] Open
Abstract
Sobuzoxane (MST-16) is an approved anticancer agent, a pro-drug of bisdioxopiperazine analog ICRF-154. Due to the structural similarity of ICRF-154 to dexrazoxane (ICRF-187), MST-16 deserves attention as a cardioprotective drug. This study presents for the first time UHPLC-MS/MS assay of MST-16, ICRF-154 and its metabolite (EDTA-diamide) in cell culture medium, buffer, plasma and cardiac cells and provides data on MST-16 bioactivation under conditions relevant to investigation of cardioprotection of this drug. The analysis of these compounds that differ considerably in their lipophilicity was achieved on the Zorbax SB-Aq column using a mixture of aqueous ammonium formate and methanol as a mobile phase. The biological samples were either diluted or precipitated with methanol, which was followed by acidification for the assay of MST-16. The method was validated for determination of all compounds in the biological materials. The application of the method for analysis of samples from in vitro experiments provided important findings, namely, that (1) MST-16 is quickly decomposed in biological environments, (2) the cardiac cells actively metabolize MST-16, and (3) MST-16 readily penetrates into the cardiac cells and is converted into ICRF-154 and EDTA-diamide. These data are useful for the in-depth examination of the cardioprotective potential of this drug.
Collapse
Affiliation(s)
- Petra Reimerová
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - Anna Jirkovská
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - Hana Bavlovič Piskáčková
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - Galina Karabanovich
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - Jaroslav Roh
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - Tomáš Šimůnek
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - Petra Štěrbová-Kovaříková
- Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic.
| |
Collapse
|
29
|
Jansová H, Šimůnek T. Cardioprotective Potential of Iron Chelators and Prochelators. Curr Med Chem 2019; 26:288-301. [DOI: 10.2174/0929867324666170920155439] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 06/07/2017] [Accepted: 09/12/2017] [Indexed: 02/08/2023]
Abstract
Heart is a particularly sensitive organ to iron overload and cardiomyopathy due to the excessive cardiac iron deposition causes most deaths in disorders such as beta-thalassemia major. Free or loosely bound iron ions readily cycle between ferrous and ferric states and catalyze Haber-Weiss reaction that yields highly reactive and toxic hydroxyl radicals. Treatment with iron chelators (desferrioxamine, deferiprone, and deferasirox) substantially improved cardiovascular morbidity and mortality in iron overloaded patients. Furthermore, iron chelators have been studied in various cardiovascular disorders with known or presumed oxidative stress roles (e.g., ischemia/reperfusion injury) also in patients with normal body iron contents. The pharmacodynamic and pharmacokinetic properties of these chelators are critical for effective therapy. For example, the widely clinically used but hydrophilic chelator desferrioxamine suffers from poor plasma membrane permeability, which means that high and clinically unachievable concentrations/doses must be employed to obtain cardioprotection. Therefore, small-molecular and lipophilic chelators with oral availability are more suitable for this purpose, particularly in states without systemic iron overload. Apart from agents that are already used in clinical practice, aroylhydrazone iron chelators, namely salicylaldehyde isonicotinoyl hydrazone (SIH), have provided promising results. However, the use of classical iron-chelating agents is associated with a risk of toxicity due to indiscriminate iron depletion. Recent studies have therefore focused on "masked" prochelators that have little or no affinity for iron until site-specific activation by reactive oxygen species.
Collapse
Affiliation(s)
- Hana Jansová
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, Prague, Czech Republic
| | - Tomáś Šimůnek
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
30
|
Kabir A, Tilekar K, Upadhyay N, Ramaa C. Novel Anthraquinone Derivatives as Dual Inhibitors of Topoisomerase 2 and Casein Kinase 2: In Silico Studies, Synthesis and Biological Evaluation on Leukemic Cell Lines. Anticancer Agents Med Chem 2019; 18:1551-1562. [DOI: 10.2174/1871520618666180423111309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 11/22/2022]
Abstract
Background:
Cancer being a complex disease, single targeting agents remain unsuccessful. This calls
for “multiple targeting”, wherein a single drug is so designed that it will modulate the activity of multiple protein
targets. Topoisomerase 2 (Top2) helps in removing DNA tangles and super-coiling during cellular replication,
Casein Kinase 2 (CK2) is involved in the phosphorylation of a multitude of protein targets. Thus, in the
present work, we have tried to develop dual inhibitors of Top2 and CK2.
Objective:
With this view, in the present work, 2 human proteins, Top2 and CK2 have been targeted to achieve
the anti-proliferative effects.
Methods:
Novel 1-acetylamidoanthraquinone (3a-3y) derivatives were designed, synthesized and their structures
were elucidated by analytical and spectral characterization techniques (FTIR, 1H NMR, 13C NMR and
Mass Spectroscopy). The synthesized compounds were then subjected to evaluation of cytotoxic potential by the
Sulforhodamine B (SRB) protein assay, using HL60 and K562 cell lines. Ten compounds were analyzed for
Top2, CK2 enzyme inhibitory potential. Further, top three compounds were subjected to cell cycle analysis.
Results:
The compounds 3a to 3c, 3e, 3f, 3i to 3p, 3t and 3x showed excellent cytotoxic activity to HL-60 cell
line indicating their high anti-proliferative potential in AML. The compounds 3a to 3c, 3e, 3f, 3i to 3p and 3y
have shown good to moderate activity on K-562 cell line. Compounds 3e, 3f, 3i, 3x and 3y were found more
cytotoxic than standard doxorubicin. In cell cycle analysis, the cells (79-85%) were found to arrest in the G0/G1
phase.
Conclusion:
We have successfully designed, synthesized, purified and structurally characterized 1-
acetylamidoanthraquinone derivatives. Even though our compounds need design optimization to further increase
enzyme inhibition, their overall anti-proliferative effects were found to be encouraging.
Collapse
Affiliation(s)
- Abbas Kabir
- Bharati Vidyapeeth's College of Pharmacy, Department of Pharmaceutical Chemistry, Sector 8, C. B. D. Belapur, Navi Mumbai 400614, Maharashtra, India
| | - Kalpana Tilekar
- Bharati Vidyapeeth's College of Pharmacy, Department of Pharmaceutical Chemistry, Sector 8, C. B. D. Belapur, Navi Mumbai 400614, Maharashtra, India
| | - Neha Upadhyay
- Bharati Vidyapeeth's College of Pharmacy, Department of Pharmaceutical Chemistry, Sector 8, C. B. D. Belapur, Navi Mumbai 400614, Maharashtra, India
| | - C.S. Ramaa
- Bharati Vidyapeeth's College of Pharmacy, Department of Pharmaceutical Chemistry, Sector 8, C. B. D. Belapur, Navi Mumbai 400614, Maharashtra, India
| |
Collapse
|
31
|
Tripaydonis A, Conyers R, Elliott DA. Pediatric Anthracycline-Induced Cardiotoxicity: Mechanisms, Pharmacogenomics, and Pluripotent Stem-Cell Modeling. Clin Pharmacol Ther 2019; 105:614-624. [PMID: 30460992 PMCID: PMC6590110 DOI: 10.1002/cpt.1311] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022]
Abstract
Anthracycline‐induced cardiotoxicity (ACT) is a severe adverse drug reaction for a subset of children treated with anthracyclines as part of chemotherapy protocols. The identification of genetic markers associated with increased ACT susceptibility has clinical significance toward improving patient care and our understanding of the molecular mechanisms involved in ACT. Human‐induced pluripotent stem cell–derived cardiomyocytes represent a novel approach to determine the pharmacogenomics of ACT and guide the development of genetic screening tests.
Collapse
Affiliation(s)
- Anne Tripaydonis
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, The Royal Children's Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Rachel Conyers
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, The Royal Children's Hospital, The University of Melbourne, Parkville, Victoria, Australia.,Children's Cancer Centre, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - David A Elliott
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, The Royal Children's Hospital, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
32
|
Abstract
Drug resistance is a well-known phenomenon that occurs when initially responsive to chemotherapy cancer cells become tolerant and elude further effectiveness of anticancer drugs. Based on their mechanism of action, anticancer drugs can be divided into cytotoxic-based agents and target-based agents. An important role among the therapeutics of the second group is played by drugs targeting topoisomerases, nuclear enzymes critical to DNA function and cell survival. These enzymes are cellular targets of several groups of anticancer agents which generate DNA damage in rapidly proliferating cancer cells. Drugs targeting topoisomerase I are mostly analogs of camtothecin, a natural compound isolated from the bark of a tree growing in China. Drugs targeting topoisomerase II are divided into poisons, such as anthracycline antibiotics, whose action is based on intercalation between DNA bases, and catalytic inhibitors that block topoisomerase II at different stages of the catalytic cycle. Unfortunately, chemotherapy is often limited by the induction of drug resistance. Identifying mechanisms that promote drug resistance is critical for the improvement of patient prognosis. Cancer drug resistance is a complex phenomenon that may be influenced by many factors. Here we discuss various mechanisms by which cancer cells can develop resistance to topoisomerase-directed drugs, which include enhanced drug efflux, mutations in topoisomerase genes, hypophosphorylation of topoisomerase II catalytic domain, activation of NF-κB transcription factor and drug inactivation. All these events may lead to the ineffective induction of cancer cell death. Attempts at circumventing drug resistance through the inhibition of cellular efflux pumps, use of silencing RNAs or inhibition of some important mechanisms, which can allow cancer cells to survive therapy, are also presented.
Collapse
Affiliation(s)
- Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| | - Angelika Długosz
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| |
Collapse
|
33
|
McCormack K. The cardioprotective effect of dexrazoxane (Cardioxane) is consistent with sequestration of poly(ADP-ribose) by self-assembly and not depletion of topoisomerase 2B. Ecancermedicalscience 2018; 12:889. [PMID: 30792806 PMCID: PMC6351063 DOI: 10.3332/ecancer.2018.889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Indexed: 01/12/2023] Open
Abstract
Following systematic scrutiny of the evidence in support of the hypothesis that the cardioprotective mechanism of action of dexrazoxane is mediated by a 'depletion' or 'downregulation' of Top2β protein levels in heart tissue, the author concludes that this hypothesis is untenable. In seeking to understand how dexrazoxane protects the heart, the outcomes of a customised association rule learning algorithm incorporating the use of antecedent surrogate variables (CEME, 2017 McCormack Pharma) reveal a previously unknown relationship between dexrazoxane and poly(ADP-ribose) (PAR) polymer. The author shows how this previously unknown relationship explains both acute and long-term cardioprotection in patients receiving anthracyclines. In addition, as a direct inhibitor of PAR dexrazoxane has access to the epigenome and this offers a new insight into protection by dexrazoxane against doxorubicin-induced late-onset damage [McCormack K, manuscript in preparation]. Notably, through this review article, the author illustrates the practical application of probing natural language text using an association rule learning algorithm for the discovery of new and interesting associations that, otherwise, would remain lost. Historically, the use of CEME enabled the first report of the capacity of a small molecule to catalyse the hybrid self-assembly of a nucleic acid biopolymer via canonical and non-canonical, non-covalent interactions analogous to Watson Crick and Hoogsteen base pairing, respectively.
Collapse
Affiliation(s)
- Keith McCormack
- McCormack Pharma, a division of McCormack Ltd, Stirling House, 9 Burroughs Gardens, London NW4 4AU, UK
| |
Collapse
|
34
|
Corremans R, Adão R, De Keulenaer GW, Leite-Moreira AF, Brás-Silva C. Update on pathophysiology and preventive strategies of anthracycline-induced cardiotoxicity. Clin Exp Pharmacol Physiol 2018; 46:204-215. [DOI: 10.1111/1440-1681.13036] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/31/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Affiliation(s)
| | - Rui Adão
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
| | | | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
- Faculty of Nutrition and Food Sciences; University of Porto; Porto Portugal
| |
Collapse
|
35
|
Prakash A, Garcia-Moreno JF, Brown JAL, Bourke E. Clinically Applicable Inhibitors Impacting Genome Stability. Molecules 2018; 23:E1166. [PMID: 29757235 PMCID: PMC6100577 DOI: 10.3390/molecules23051166] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 12/14/2022] Open
Abstract
Advances in technology have facilitated the molecular profiling (genomic and transcriptomic) of tumours, and has led to improved stratification of patients and the individualisation of treatment regimes. To fully realize the potential of truly personalised treatment options, we need targeted therapies that precisely disrupt the compensatory pathways identified by profiling which allow tumours to survive or gain resistance to treatments. Here, we discuss recent advances in novel therapies that impact the genome (chromosomes and chromatin), pathways targeted and the stage of the pathways targeted. The current state of research will be discussed, with a focus on compounds that have advanced into trials (clinical and pre-clinical). We will discuss inhibitors of specific DNA damage responses and other genome stability pathways, including those in development, which are likely to synergistically combine with current therapeutic options. Tumour profiling data, combined with the knowledge of new treatments that affect the regulation of essential tumour signalling pathways, is revealing fundamental insights into cancer progression and resistance mechanisms. This is the forefront of the next evolution of advanced oncology medicine that will ultimately lead to improved survival and may, one day, result in many cancers becoming chronic conditions, rather than fatal diseases.
Collapse
Affiliation(s)
- Anu Prakash
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, H91 YR71 Galway, Ireland.
| | - Juan F Garcia-Moreno
- Discipline of Surgery, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, H91 YR71 Galway, Ireland.
| | - James A L Brown
- Discipline of Surgery, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, H91 YR71 Galway, Ireland.
| | - Emer Bourke
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, H91 YR71 Galway, Ireland.
| |
Collapse
|
36
|
Koleini N, Kardami E. Autophagy and mitophagy in the context of doxorubicin-induced cardiotoxicity. Oncotarget 2018; 8:46663-46680. [PMID: 28445146 PMCID: PMC5542301 DOI: 10.18632/oncotarget.16944] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 03/17/2017] [Indexed: 12/18/2022] Open
Abstract
Doxorubicin (Dox) is a cytotoxic drug widely incorporated in various chemotherapy protocols. Severe side effects such as cardiotoxicity, however, limit Dox application. Mechanisms by which Dox promotes cardiac damage and cardiomyocyte cell death have been investigated extensively, but a definitive picture has yet to emerge. Autophagy, regarded generally as a protective mechanism that maintains cell viability by recycling unwanted and damaged cellular constituents, is nevertheless subject to dysregulation having detrimental effects for the cell. Autophagic cell death has been described, and has been proposed to contribute to Dox-cardiotoxicity. Additionally, mitophagy, autophagic removal of damaged mitochondria, is affected by Dox in a manner contributing to toxicity. Here we will review Dox-induced cardiotoxicity and cell death in the broad context of the autophagy and mitophagy processes.
Collapse
Affiliation(s)
- Navid Koleini
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada.,Department of Physiology and Pathophysiology, Winnipeg, Manitoba, Canada
| | - Elissavet Kardami
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada.,Department of Physiology and Pathophysiology, Winnipeg, Manitoba, Canada.,Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
37
|
Jirkovský E, Jirkovská A, Bureš J, Chládek J, Lenčová O, Stariat J, Pokorná Z, Karabanovich G, Roh J, Brázdová P, Šimůnek T, Kovaříková P, Štěrba M. Pharmacokinetics of the Cardioprotective Drug Dexrazoxane and Its Active Metabolite ADR-925 with Focus on Cardiomyocytes and the Heart. J Pharmacol Exp Ther 2018; 364:433-446. [PMID: 29273587 DOI: 10.1124/jpet.117.244848] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/19/2017] [Indexed: 11/22/2022] Open
Abstract
Dexrazoxane (DEX), the only cardioprotectant approved against anthracycline cardiotoxicity, has been traditionally deemed to be a prodrug of the iron-chelating metabolite ADR-925. However, pharmacokinetic profile of both agents, particularly with respect to the cells and tissues essential for its action (cardiomyocytes/myocardium), remains poorly understood. The aim of this study is to characterize the conversion and disposition of DEX to ADR-925 in vitro (primary cardiomyocytes) and in vivo (rabbits) under conditions where DEX is clearly cardioprotective against anthracycline cardiotoxicity. Our results show that DEX is hydrolyzed to ADR-925 in cell media independently of the presence of cardiomyocytes or their lysate. Furthermore, ADR-925 directly penetrates into the cells with contribution of active transport, and detectable concentrations occur earlier than after DEX incubation. In rabbits, ADR-925 was detected rapidly in plasma after DEX administration to form sustained concentrations thereafter. ADR-925 was not markedly retained in the myocardium, and its relative exposure was 5.7-fold lower than for DEX. Unlike liver tissue, myocardium homogenates did not accelerate the conversion of DEX to ADR-925 in vitro, suggesting that myocardial concentrations in vivo may originate from its distribution from the central compartment. The pharmacokinetic parameters for both DEX and ADR-925 were determined by both noncompartmental analyses and population pharmacokinetics (including joint parent-metabolite model). Importantly, all determined parameters were closer to human than to rodent data. The present results open venues for the direct assessment of the cardioprotective effects of ADR-925 in vitro and in vivo to establish whether DEX is a drug or prodrug.
Collapse
Affiliation(s)
- Eduard Jirkovský
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Anna Jirkovská
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Jan Bureš
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Jaroslav Chládek
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Olga Lenčová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Ján Stariat
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Zuzana Pokorná
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Galina Karabanovich
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Jaroslav Roh
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Petra Brázdová
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Tomáš Šimůnek
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Petra Kovaříková
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Martin Štěrba
- Department of Pharmacology, Faculty of Medicine in Hradec Králové (E.J., J.C., O.L., Z.P., P.B., M.Š.), and Departments of Biochemical Sciences (E.J., A.J., T.Š.), Pharmaceutical Chemistry and Pharmaceutical Analysis (J.B., J.S., P.K.), and Inorganic and Organic Chemistry (G.K., J.R.), Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
38
|
Delgado JL, Hsieh CM, Chan NL, Hiasa H. Topoisomerases as anticancer targets. Biochem J 2018; 475:373-398. [PMID: 29363591 PMCID: PMC6110615 DOI: 10.1042/bcj20160583] [Citation(s) in RCA: 296] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/14/2017] [Accepted: 12/21/2017] [Indexed: 12/15/2022]
Abstract
Many cancer type-specific anticancer agents have been developed and significant advances have been made toward precision medicine in cancer treatment. However, traditional or nonspecific anticancer drugs are still important for the treatment of many cancer patients whose cancers either do not respond to or have developed resistance to cancer-specific anticancer agents. DNA topoisomerases, especially type IIA topoisomerases, are proved therapeutic targets of anticancer and antibacterial drugs. Clinically successful topoisomerase-targeting anticancer drugs act through topoisomerase poisoning, which leads to replication fork arrest and double-strand break formation. Unfortunately, this unique mode of action is associated with the development of secondary cancers and cardiotoxicity. Structures of topoisomerase-drug-DNA ternary complexes have revealed the exact binding sites and mechanisms of topoisomerase poisons. Recent advances in the field have suggested a possibility of designing isoform-specific human topoisomerase II poisons, which may be developed as safer anticancer drugs. It may also be possible to design catalytic inhibitors of topoisomerases by targeting certain inactive conformations of these enzymes. Furthermore, identification of various new bacterial topoisomerase inhibitors and regulatory proteins may inspire the discovery of novel human topoisomerase inhibitors. Thus, topoisomerases remain as important therapeutic targets of anticancer agents.
Collapse
Affiliation(s)
- Justine L Delgado
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 115 S Grand Ave., S321 Pharmacy Building, Iowa City, IA 52242, U.S.A
| | - Chao-Ming Hsieh
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei City 100, Taiwan
| | - Nei-Li Chan
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei City 100, Taiwan
| | - Hiroshi Hiasa
- Department of Pharmacology, University of Minnesota Medical School, 6-120 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, U.S.A.
| |
Collapse
|
39
|
Lipshultz ER, Holt GE, Ramasamy R, Yechieli R, Lipshultz SE. Fertility, Cardiac, and Orthopedic Challenges in Survivors of Adult and Childhood Sarcoma. Am Soc Clin Oncol Educ Book 2017; 37:799-806. [PMID: 28561655 DOI: 10.1200/edbk_174708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The combination of cisplatin, doxorubicin, and methotrexate was established as the standard backbone of contemporary osteosarcoma therapy in 1986. Since then, however, further improving the survival of patients with osteosarcoma has been challenging-30% to 40% of patients with osteosarcoma still die of this disease. In addition, these patients often experience loss of fertility at a young age, short- and long-term treatment-related cardiotoxicity, and adverse orthopedic effects from surgical resection of the tumor or endoprosthetic reconstructions. Cancer treatment often markedly increases the risk of infertility later in life, causing many patients substantial distress and regret. Sperm banking and oocyte cryopreservation are standard of care and should be available to all at-risk patients. Newer techniques, such as autologous gonadal tissue transplant for prepubertal children, are being developed, and newer systemic agents have infertility risk profiles that remain undefined and warrant further study. Cost and access remain barriers to these options. The late effects of anthracycline-induced cardiotoxicity are also increasingly a problem for these patients. These effects are often progressive and can be disabling. Adding dexrazoxane to doxorubicin therapy significantly reduces the risk for most adverse cardiac outcomes without compromising the efficacy of induction chemotherapy. Limb salvage surgery remains the standard of care for treatment in the majority of patients with extremity sarcomas. Modular metal prostheses and allograft reconstructions comprised the majority of surgical procedures for limb salvage surgery. The most common mechanism of failure of these implants is infection and mechanical failure of the implant.
Collapse
Affiliation(s)
- Emma R Lipshultz
- From the Dana-Farber Cancer Institute, Boston, MA; Vanderbilt-Ingram Cancer Center, Nashville, TN; University of Miami Miller School of Medicine, Miami, FL; Wayne State University, Children's Hospital of Michigan, Karmanos Cancer Institute, Detroit, MI
| | - Ginger E Holt
- From the Dana-Farber Cancer Institute, Boston, MA; Vanderbilt-Ingram Cancer Center, Nashville, TN; University of Miami Miller School of Medicine, Miami, FL; Wayne State University, Children's Hospital of Michigan, Karmanos Cancer Institute, Detroit, MI
| | - Ranjith Ramasamy
- From the Dana-Farber Cancer Institute, Boston, MA; Vanderbilt-Ingram Cancer Center, Nashville, TN; University of Miami Miller School of Medicine, Miami, FL; Wayne State University, Children's Hospital of Michigan, Karmanos Cancer Institute, Detroit, MI
| | - Raphael Yechieli
- From the Dana-Farber Cancer Institute, Boston, MA; Vanderbilt-Ingram Cancer Center, Nashville, TN; University of Miami Miller School of Medicine, Miami, FL; Wayne State University, Children's Hospital of Michigan, Karmanos Cancer Institute, Detroit, MI
| | - Steven E Lipshultz
- From the Dana-Farber Cancer Institute, Boston, MA; Vanderbilt-Ingram Cancer Center, Nashville, TN; University of Miami Miller School of Medicine, Miami, FL; Wayne State University, Children's Hospital of Michigan, Karmanos Cancer Institute, Detroit, MI
| |
Collapse
|
40
|
Bures J, Jirkovska A, Sestak V, Jansova H, Karabanovich G, Roh J, Sterba M, Simunek T, Kovarikova P. Investigation of novel dexrazoxane analogue JR-311 shows significant cardioprotective effects through topoisomerase IIbeta but not its iron chelating metabolite. Toxicology 2017; 392:1-10. [PMID: 28941780 DOI: 10.1016/j.tox.2017.09.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/10/2017] [Accepted: 09/19/2017] [Indexed: 12/31/2022]
Abstract
Novel dexrazoxane derivative JR-311 was prepared to investigate structure-activity relationships and mechanism(s) of protection against anthracycline cardiotoxicity. Its cardioprotective, antiproliferative, iron (Fe) chelation and inhibitory and/or depletory activities on topoisomerase IIbeta (TOP2B) were examined and compared with dexrazoxane. While in standard assay, JR-311 failed in both cardioprotection and depletion of TOP2B, its repeated administration to cell culture media led to depletion of TOP2B and significant protection of isolated rat neonatal ventricular cardiomyocytes from daunorubicin-induced damage. This effect was explained by a focused analytical investigation that revealed rapid JR-311 decomposition, resulting in negligible intracellular concentrations of the parent compound but high exposure of cells to the decomposition products, including Fe-chelating JR-H2. Although chemical instability is an obstacle for the development of JR-311, this study identified a novel dexrazoxane analogue with preserved pharmacodynamic properties, contributed to the investigation of structure-activity relationships and suggested that the cardioprotection of bis-dioxopiperazines is likely attributed to TOP2B activity of the parent compound rather than Fe chelation of their hydrolytic metabolites/degradation products. Moreover, this study highlights the importance of early stability testing during future development of novel dexrazoxane analogues.
Collapse
Affiliation(s)
- Jan Bures
- Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Anna Jirkovska
- Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Vit Sestak
- Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Hana Jansova
- Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Galina Karabanovich
- Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Jaroslav Roh
- Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Martin Sterba
- Faculty of Medicine in Hradec Králové, Charles University, Šimkova 850, 500 03 Hradec Králové, Czech Republic
| | - Tomas Simunek
- Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Petra Kovarikova
- Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| |
Collapse
|
41
|
Andrei SA, Sijbesma E, Hann M, Davis J, O’Mahony G, Perry MWD, Karawajczyk A, Eickhoff J, Brunsveld L, Doveston RG, Milroy LG, Ottmann C. Stabilization of protein-protein interactions in drug discovery. Expert Opin Drug Discov 2017; 12:925-940. [DOI: 10.1080/17460441.2017.1346608] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Sebastian A. Andrei
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Michael Hann
- Platform Technology and Science, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage, UK
| | - Jeremy Davis
- Department of Chemistry, UCB Celltech, Slough, UK
| | - Gavin O’Mahony
- CVMD Medicinal Chemistry, Innovative Medicines and Early Development, AstraZeneca Gothenburg, Pepparedsleden, Mölndal, Sweden
| | - Matthew W. D. Perry
- RIA Medicinal Chemistry, Innovative Medicines and Early Development, AstraZeneca Gothenburg, Pepparedsleden, Mölndal, Sweden
| | - Anna Karawajczyk
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Dortmund, Germany
| | - Jan Eickhoff
- Assay development & screening, Lead Discovery Center GmbH, Dortmund, Germany
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Richard G. Doveston
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Lech-Gustav Milroy
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Chemistry, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
42
|
Aniogo EC, George BPA, Abrahamse H. Phthalocyanine induced phototherapy coupled with Doxorubicin; a promising novel treatment for breast cancer. Expert Rev Anticancer Ther 2017; 17:693-702. [PMID: 28657372 DOI: 10.1080/14737140.2017.1347505] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Globally, breast cancer is the most common life-threatening malignant disease among women. Adjuvant chemotherapeutic treatment of anthracycline-based chemotherapy (e.g., doxorubicin) has been shown to be more advantageous over non-anthracycline-based therapies, yet possess the tenacity of developing resistance and potential side effects which have limited its use in the clinical setting. These reasons necessitate combining doxorubicin with emerging photodynamic treatment regimens. Areas covered: In this review, the authors have concisely explained doxorubicin chemotherapy and the photobiological processes of phthalocyanine triggered photodynamic therapy (PDT). A literature search was conducted and reports demonstrating the use of doxorubicin and photodynamic therapy as a treatment modality for breast cancer were identified. More emphasis was made on studies demonstrating the efficacy and improved anticancer effect of combining chemotherapy with photodynamic therapy. However, it was concluded that for this combination therapy, still in it's infancy, it could be relevant when integrated into standard treatment. Expert Commentary: To these effects, comprehensive models based on experimental evaluations are needed for rational design of anthracycline-based chemotherapy and PDT to be integrated into the clinical setting.
Collapse
Affiliation(s)
- Eric Chekwube Aniogo
- a Laser Research Centre, Faculty of Health Sciences , University of Johannesburg , Doornfontein , South Africa
| | | | - Heidi Abrahamse
- a Laser Research Centre, Faculty of Health Sciences , University of Johannesburg , Doornfontein , South Africa
| |
Collapse
|
43
|
Statins in anthracycline-induced cardiotoxicity: Rac and Rho, and the heartbreakers. Cell Death Dis 2017; 8:e2564. [PMID: 28102848 PMCID: PMC5386353 DOI: 10.1038/cddis.2016.418] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/02/2016] [Indexed: 01/06/2023]
Abstract
Cancer patients receiving anthracycline-based chemotherapy are at risk to develop life-threatening chronic cardiotoxicity with the pathophysiological mechanism of action not fully understood. Besides the most common hypothesis that anthracycline-induced congestive heart failure (CHF) is mainly caused by generation of reactive oxygen species, recent data point to a critical role of topoisomerase II beta (TOP2B), which is a primary target of anthracycline poisoning, in the pathophysiology of CHF. As the use of the only clinically approved cardioprotectant dexrazoxane has been limited by the FDA in 2011, there is an urgent need for alternative cardioprotective measures. Statins are anti-inflammatory and anti-oxidative drugs that are clinically well established for the prevention of cardiovascular diseases. They exhibit pleiotropic beneficial properties beyond cholesterol-lowering effects that most likely rest on the indirect inhibition of small Ras homologous (Rho) GTPases. The Rho GTPase Rac1 has been shown to be a major factor in the regulation of the pro-oxidative NADPH oxidase as well as in the regulation of type II topoisomerase. Both are discussed to play an important role in the pathophysiology of anthracycline-induced CHF. Therefore, off-label use of statins or novel Rac1 inhibitors might represent a promising pharmacological approach to gain control over chronic cardiotoxicity by interfering with key mechanisms of anthracycline-induced cardiomyocyte cell death.
Collapse
|
44
|
Kuncová J, Jirkovská A, Švíglerová J, Marková M, Meireles D, Čedíková M. Neonatal capsaicin administration impairs postnatal development of the cardiac chronotropy and inotropy in rats. Physiol Res 2016; 65:S633-S642. [PMID: 28006945 DOI: 10.33549/physiolres.933540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The present study evaluated the impact of neonatal administration of capsaicin (neurotoxin from red hot pepper used for sensory denervation) on postnatal development of the heart rate and ventricular contractility. In the rats subjected to capsaicin administration (100 mg/kg) on postnatal days 2 and 3 and their vehicle-treated controls at the ages of 10 to 90 days, function of the sympathetic innervation of the developing heart was characterized by evaluation of chronotropic responses to metipranolol and atropine, norepinephrine concentrations in the heart, and norepinephrine release from the heart atria. Sensory denervation was verified by determination of calcitonin gene-related peptide levels in the heart. Direct cytotoxic effects of capsaicin were assessed on cultured neonatal cardiomyocytes. Capsaicin-treated rats displayed higher resting heart rates, lower atropine effect, but no difference in the effect of metipranolol. Norepinephrine tissue levels and release did not differ from controls. Contraction force of the right ventricular papillary muscle was lower till the age of 60 days. Significantly reduced viability of neonatal cardiomyocytes was demonstrated at capsaicin concentration 100 micromol/l. Our study suggests that neonatal capsaicin treatment leads to impaired maturation of the developing cardiomyocytes. This effect cannot be attributed exclusively to sensory denervation of the rat heart since capsaicin acts also directly on the cardiac cells.
Collapse
Affiliation(s)
- J Kuncová
- Department of Physiology, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | | | | | | | | | | |
Collapse
|
45
|
Baek N, Seo OW, Kim M, Hulme J, An SSA. Monitoring the effects of doxorubicin on 3D-spheroid tumor cells in real-time. Onco Targets Ther 2016; 9:7207-7218. [PMID: 27920558 PMCID: PMC5125797 DOI: 10.2147/ott.s112566] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recently, increasing numbers of cell culture experiments with 3D spheroids presented better correlating results in vivo than traditional 2D cell culture systems. 3D spheroids could offer a simple and highly reproducible model that would exhibit many characteristics of natural tissue, such as the production of extracellular matrix. In this paper numerous cell lines were screened and selected depending on their ability to form and maintain a spherical shape. The effects of increasing concentrations of doxorubicin (DXR) on the integrity and viability of the selected spheroids were then measured at regular intervals and in real-time. In total 12 cell lines, adenocarcinomic alveolar basal epithelial (A549), muscle (C2C12), prostate (DU145), testis (F9), pituitary epithelial-like (GH3), cervical cancer (HeLa), HeLa contaminant (HEp2), embryo (NIH3T3), embryo (PA317), neuroblastoma (SH-SY5Y), osteosarcoma U2OS, and embryonic kidney cells (293T), were screened. Out of the 12, 8 cell lines, NIH3T3, C2C12, 293T, SH-SY5Y, A549, HeLa, PA317, and U2OS formed regular spheroids and the effects of DXR on these structures were measured at regular intervals. Finally, 5 cell lines, A549, HeLa, SH-SY5Y, U2OS, and 293T, were selected for real-time monitoring and the effects of DXR treatment on their behavior were continuously recorded for 5 days. A potential correlation regarding the effects of DXR on spheroid viability and ATP production was measured on days 1, 3, and 5. Cytotoxicity of DXR seemed to occur after endocytosis, since the cellular activities and ATP productions were still viable after 1 day of the treatment in all spheroids, except SH-SY5Y. Both cellular activity and ATP production were halted 3 and 5 days from the start of the treatment in all spheroids. All cell lines maintained their spheroid shape, except SHSY-5, which behaved in an unpredictable manner when exposed to toxic concentrations of DXR. Cytotoxic effects of DXR towards SH-SY5Y seemed to cause degradation of the extracellular matrix, since all cells were dismantled from the spheroid upon cell death. On the other hand, 293T spheroids revealed retarded cellular activity and ATP productions upon DXR treatment throughout the experiment. Since 293T was the embryonic kidney cells, the fast clearance or neutralizations could have made them resistant towards DXR. In conclusion, the same degree of sensitivity from the 2D system did not translate to a 3D culture system, resulting in higher IC50 values than the 2D system. The varying sensitivities and tolerances to drugs could be better understood with a 3D cell culture system.
Collapse
Affiliation(s)
- NamHuk Baek
- Department of R & D, NanoEntek Inc., Seoul, Republic of Korea
| | - Ok Won Seo
- Department of R & D, NanoEntek Inc., Seoul, Republic of Korea
| | - MinSung Kim
- Department of R & D, NanoEntek Inc., Seoul, Republic of Korea
| | - John Hulme
- Department of BioNano Technology Gachon University, Gyeonggi-do, Republic of Korea
| | - Seong Soo A An
- Department of BioNano Technology Gachon University, Gyeonggi-do, Republic of Korea
| |
Collapse
|
46
|
Frank NE, Cusack BJ, Talley TT, Walsh GM, Olson RD. Comparative effects of doxorubicin and a doxorubicin analog, 13-deoxy, 5-iminodoxorubicin (GPX-150), on human topoisomerase IIβ activity and cardiac function in a chronic rabbit model. Invest New Drugs 2016; 34:693-700. [DOI: 10.1007/s10637-016-0388-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/22/2016] [Indexed: 01/21/2023]
|
47
|
Ezzat SM, El Gaafary M, El Sayed AM, Sabry OM, Ali ZY, Hafner S, Schmiech M, Jin L, Syrovets T, Simmet T. The Cardenolide Glycoside Acovenoside A Affords Protective Activity in Doxorubicin-Induced Cardiotoxicity in Mice. J Pharmacol Exp Ther 2016; 358:262-70. [PMID: 27247000 DOI: 10.1124/jpet.116.232652] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 11/15/2011] [Indexed: 03/08/2025] Open
Abstract
The current study aimed to investigate the protective effect of the cardenolide glycoside acovenoside A (AcoA) against doxorubicin-induced cardiotoxicity in mice. AcoA was isolated from the pericarps of Acokanthera oppositifolia to chemical homogeneity and characterized by means of one- and two-dimensional nuclear magnetic resonance spectroscopy. AcoA exhibited relatively low toxicity in mice (LD50 = 223.3 mg/kg bw). Repeated administration of doxorubicin induced cardiotoxicity manifested by reduced activity of myocardial membrane-bound ion pumps and elevated serum biomarkers of myocardial dysfunction, oxidative stress, and inflammation. Pretreatment of doxorubicin-exposed mice with AcoA (11.16 or 22.33 mg/kg bw, i.p.) for 2 weeks after 2 weeks of combined administration of AcoA and doxorubicin protected the animals dose dependently against doxorubicin-induced cardiotoxicity as indicated by normalization of the levels of different myocardial markers of oxidative stress (malondialdehyde, nitric oxide, total antioxidant capacity, and cardiac glutathione), serum myocardial diagnostic marker enzymes (serum cardiac troponin T, creatine kinase isoenzyme MB, aspartate aminotransferase, and lactate dehydrogenase), and inflammatory markers (c-reactive protein, tumor necrosis factor-α, and interleukin-6), as well as myocardial Na(+)/K(+)-ATPase activity. These effects were attributed to the negative impact of AcoA on transcription factors nuclear factor κB and interferon regulatory factor 3/7. Thus acovenoside A might act as a cardioprotective agent to prevent doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shahira M Ezzat
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Menna El Gaafary
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Abeer M El Sayed
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Omar M Sabry
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Zeinab Y Ali
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Susanne Hafner
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Michael Schmiech
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Lu Jin
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Tatiana Syrovets
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| | - Thomas Simmet
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Giza, Egypt (S.M.E., M.E.G., A.M.E.S., O.M.S.); Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt (Z.Y.A.); Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany (M.E.G., S.H., M.S., L.J., Ta.S., Th.S.)
| |
Collapse
|
48
|
Morbidelli L, Donnini S, Ziche M. Targeting endothelial cell metabolism for cardio-protection from the toxicity of antitumor agents. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2016; 2:3. [PMID: 33530139 PMCID: PMC7837145 DOI: 10.1186/s40959-016-0010-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/16/2016] [Indexed: 12/17/2022]
Abstract
The vascular endothelium plays a fundamental role in the maintenance of tissue homeostasis, regulating local blood flow and other physiological processes. Chemotherapeutic drugs and target therapies, including antiangiogenic drugs targeting vascular endothelial growth factor (VEGF) or its receptors, not only efficiently act against tumor growth, but may also induce endothelial dysfunction and cardiovascular toxicity. Continued research efforts aim to better understand, prevent and mitigate these chemotherapy associated cardiovascular diseases. Conventional chemotherapeutic agents, such as anthracyclines, platinum compounds, and taxanes, and newer targeted agents, such as bevacizumab, trastuzumab, and tyrosine kinase inhibitors, have known risk of cardiovascular toxicity, which can limit their effectiveness by promoting increased morbidity and/or mortality. This review describes a) the activity of anticancer agents in inducing endothelial dysfunction, b) the metabolic pathways and signalling cascades which may be targeted by protective agents able to maintain or restore endothelial cell function, such as endothelial nitric oxide synthase/fibroblast growth factor-2 (eNOS-FGF-2) pathway, and c) the drugs/strategies reported to improve endothelial function and to reduce the risks of cardiovascular diseases such as angiotensin converting enzyme inhibitors (ACEi) and beta blockers, that are fundamental therapies in chronic heart failure (HF), as well as non-standard HF treatments such ad nitric oxide donors and antioxidant strategies. There is increasing interest in whether ACEi, beta-blockers, and/or statins might prevent and/or therapeutically control cardiotoxic effects in cancer patients. Maintaining endothelial function during or following treatments with chemotherapeutic agents, without affecting anti-tumor drug-effectiveness, is essential for preserving or recovering cardiovascular homeostasis. In this respect, the early detection and immediate therapy of cardiovascular toxicity appear crucial for substantial recovery of cardiac function in cancer patients.
Collapse
Affiliation(s)
- Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Marina Ziche
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
49
|
Cardioprotective effects of inorganic nitrate/nitrite in chronic anthracycline cardiotoxicity: Comparison with dexrazoxane. J Mol Cell Cardiol 2015; 91:92-103. [PMID: 26724189 DOI: 10.1016/j.yjmcc.2015.12.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/11/2015] [Accepted: 12/22/2015] [Indexed: 12/29/2022]
Abstract
Dexrazoxane (DEX) is a clinically available cardioprotectant that reduces the toxicity induced by anthracycline (ANT) anticancer drugs; however, DEX is seldom used and its action is poorly understood. Inorganic nitrate/nitrite has shown promising results in myocardial ischemia-reperfusion injury and recently in acute high-dose ANT cardiotoxicity. However, the utility of this approach for overcoming clinically more relevant chronic forms of cardiotoxicity remains elusive. Hence, in this study, the protective potential of inorganic nitrate and nitrite against chronic ANT cardiotoxicity was investigated, and the results were compared to those using DEX. Chronic cardiotoxicity was induced in rabbits with daunorubicin (DAU). Sodium nitrate (1g/L) was administered daily in drinking water, while sodium nitrite (0.15 or 5mg/kg) or DEX (60mg/kg) was administered parenterally before each DAU dose. Although oral nitrate induced a marked increase in plasma NOx, it showed no improvement in DAU-induced mortality, myocardial damage or heart failure. Instead, the higher nitrite dose reduced the incidence of end-stage cardiotoxicity, prevented related premature deaths and significantly ameliorated several molecular and cellular perturbations induced by DAU, particularly those concerning mitochondria. The latter result was also confirmed in vitro. Nevertheless, inorganic nitrite failed to prevent DAU-induced cardiac dysfunction and molecular remodeling in vivo and failed to overcome the cytotoxicity of DAU to cardiomyocytes in vitro. In contrast, DEX completely prevented all of the investigated molecular, cellular and functional perturbations that were induced by DAU. Our data suggest that the difference in cardioprotective efficacy between DEX and inorganic nitrite may be related to their different abilities to address a recently proposed upstream target for ANT in the heart - topoisomerase IIβ.
Collapse
|
50
|
Kropp J, Roti Roti EC, Ringelstetter A, Khatib H, Abbott DH, Salih SM. Dexrazoxane Diminishes Doxorubicin-Induced Acute Ovarian Damage and Preserves Ovarian Function and Fecundity in Mice. PLoS One 2015; 10:e0142588. [PMID: 26544188 PMCID: PMC4636352 DOI: 10.1371/journal.pone.0142588] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 10/23/2015] [Indexed: 11/19/2022] Open
Abstract
Advances in cancer treatment utilizing multiple chemotherapies have dramatically increased cancer survivorship. Female cancer survivors treated with doxorubicin (DXR) chemotherapy often suffer from an acute impairment of ovarian function, which can persist as long-term, permanent ovarian insufficiency. Dexrazoxane (Dexra) pretreatment reduces DXR-induced insult in the heart, and protects in vitro cultured murine and non-human primate ovaries, demonstrating a drug-based shield to prevent DXR insult. The present study tested the ability of Dexra pretreatment to mitigate acute DXR chemotherapy ovarian toxicity in mice through the first 24 hours post-treatment, and improve subsequent long-term fertility throughout the reproductive lifespan. Adolescent CD-1 mice were treated with Dexra 1 hour prior to DXR treatment in a 1:1 mg or 10:1 mg Dexra:DXR ratio. During the acute injury period (2-24 hours post-injection), Dexra pretreatment at a 1:1 mg ratio decreased the extent of double strand DNA breaks, diminished γH2FAX activation, and reduced subsequent follicular cellular demise caused by DXR. In fertility and fecundity studies, dams pretreated with either Dexra:DXR dose ratio exhibited litter sizes larger than DXR-treated dams, and mice treated with a 1:1 mg Dexra:DXR ratio delivered pups with birth weights greater than DXR-treated females. While DXR significantly increased the "infertility index" (quantifying the percentage of dams failing to achieve pregnancy) through 6 gestations following treatment, Dexra pretreatment significantly reduced the infertility index following DXR treatment, improving fecundity. Low dose Dexra not only protected the ovaries, but also bestowed a considerable survival advantage following exposure to DXR chemotherapy. Mouse survivorship increased from 25% post-DXR treatment to over 80% with Dexra pretreatment. These data demonstrate that Dexra provides acute ovarian protection from DXR toxicity, improving reproductive health in a mouse model, suggesting this clinically available drug may provide ovarian protection for cancer patients.
Collapse
Affiliation(s)
- Jenna Kropp
- Department of Animal Sciences, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Elon C. Roti Roti
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Ashley Ringelstetter
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Hasan Khatib
- Department of Animal Sciences, University of Wisconsin, Madison, Wisconsin, United States of America
| | - David H. Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, Madison, Wisconsin 53715, United States of America
| | - Sana M. Salih
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|