1
|
Zhang X, Wang Y, Han J, Zhao W, Zhang W, Li X, Chen J, Song W, Wang L. Cardiac-Focused Multi-Organ Chips: Advanced Disease Modeling, Drug Testing, and Inter-Organ Communication. Adv Biol (Weinh) 2025; 9:e2400512. [PMID: 39913111 DOI: 10.1002/adbi.202400512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/18/2024] [Indexed: 02/07/2025]
Abstract
Heart disease remains a leading cause of mortality worldwide, posing a significant challenge to global healthcare systems. Traditional animal models and cell culture techniques are instrumental in advancing the understanding of cardiac pathophysiology. However, these methods are limited in their ability to fully replicate the heart's intricate functions. This underscores the need for a deeper investigation into the fundamental mechanisms of heart disease. Notably, cardiac pathology is often influenced by systemic factors, with conditions in other organs contributing to disease onset and progression. Cardiac-focused multi-organ chip technology has emerged to better elucidate these complex inter-organ communications and address the limitations of current in vitro models. This technology offers a novel approach by recreating the cardiac microenvironment and integrating it with other organ systems, thereby enabling more precise disease modeling and drug toxicity assessment. This review provides a comprehensive overview of the heart's structure and function, explores the advancements in cardiac organ chip development, and highlights the applications of cardiac-focused multi-organ chips in medical research. Finally, the future potential of this technology in enhancing disease modeling and therapeutic evaluation is discussed.
Collapse
Affiliation(s)
- Xiaolong Zhang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Weilong Zhao
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai, 201 620, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250 021, China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| | - Wei Song
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250 021, China
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250 353, China
- Shandong Institute of Mechanical Design and Research, Jinan, 250 353, China
| |
Collapse
|
2
|
Bhalraam U, Veerni RB, Paddock S, Meng J, Piepoli M, López-Fernández T, Tsampasian V, Vassiliou VS. Impact of sodium-glucose cotransporter-2 inhibitors on heart failure outcomes in cancer patients and survivors: a systematic review and meta-analysis. Eur J Prev Cardiol 2025:zwaf026. [PMID: 40044419 DOI: 10.1093/eurjpc/zwaf026] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/30/2024] [Accepted: 01/28/2025] [Indexed: 03/20/2025]
Abstract
AIMS Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are recognized for their cardiovascular benefits. This systematic review and meta-analysis evaluated the impact of SGLT2i on heart failure (HF) outcomes in cancer patients and survivors, focusing on HF hospitalization and new HF diagnoses. METHODS AND RESULTS A comprehensive search of PubMed, MEDLINE, and Embase via Ovid, and the Cochrane Library was conducted up to 5 June 2024, focusing on studies involving cancer patients and survivors treated with SGLT2i. The search criterion used was [(SGLT2) OR (Sodium glucose cotransporter 2 inhibitors) OR (canagliflozin) OR (dapagliflozin) OR (empagliflozin) OR (ertugliflozin) AND (cancer)]. The primary outcomes assessed were HF hospitalization and new HF diagnoses. The search yielded 1880 studies, from which 13 studies encompassing 88 273 patients were included. SGLT2i use reduced HF hospitalizations by 51% (RR 0.49, 95% CI 0.36-0.66, I² = 28%, P < 0.01) and new HF diagnoses by 71% (RR 0.29, 95% CI 0.10-0.87, I² = 71%). Multi-variate meta-regression analysis suggested that among breast cancer populations, studies with ≥50% of patients on anthracyclines exhibited a 99% reduction in HF hospitalization risk compared with similar studies that included <50% of patients on anthracyclines (RR 0.0085, 95% CI: 0.0001-0.2645, P = 0.0081). CONCLUSION SGLT2i significantly lower the risk of HF hospitalization and new HF diagnoses among cancer patients and survivors, with particularly pronounced benefits in breast cancer patients receiving anthracycline-based chemotherapy. These findings support the need for prospective trials to further investigate the integration of SGLT2i into cancer patient management to enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- U Bhalraam
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Rathna B Veerni
- Department of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Sophie Paddock
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - James Meng
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Massimo Piepoli
- University Cardiology Department, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
- Cardiology Department, Quiron Pozuelo University Hospital, Madrid, Spain
| | - Vasiliki Tsampasian
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Vassilios S Vassiliou
- Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| |
Collapse
|
3
|
Mapuskar KA, London B, Zacharias ZR, Houtman JC, Allen BG. Immunometabolism in the Aging Heart. J Am Heart Assoc 2025; 14:e039216. [PMID: 39719411 PMCID: PMC12054428 DOI: 10.1161/jaha.124.039216] [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] [Received: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024]
Abstract
Structural, functional, and molecular-level changes in the aging heart are influenced by a dynamic interplay between immune signaling and cellular metabolism that is referred to as immunometabolism. This review explores the crosstalk between cellular metabolic pathways including glycolysis, oxidative phosphorylation, fatty acid metabolism, and the immune processes that govern cardiac aging. With a rapidly aging population that coincides with increased cardiovascular risk and cancer incidence rates, understanding the immunometabolic underpinnings of cardiac aging provides a foundation for identifying therapeutic targets to mitigate cardiac dysfunction. Aging alters the immune environment of the heart by concomitantly driving the changes in immune cell metabolism, mitochondrial dysfunction, and redox signaling. Shifts in these metabolic pathways exacerbate inflammation and impair tissue repair, creating a vicious cycle that accelerates cardiac functional decline. Treatment with cancer therapy further complicates this landscape, as aging-associated immunometabolic disruptions augment the susceptibility to cardiotoxicity. The current review highlights therapeutic strategies that target the immunometabolic axis to alleviate cardiac aging pathologies. Interventions include modulating metabolic intermediates, improving mitochondrial function, and leveraging immune signaling pathways to restore cardiac health. Advances in immunometabolism thus hold significant potential for translating preclinical findings into therapies that improve the quality of life for the aging population and underscore the need for approaches that address the immunometabolic mechanisms of cardiac aging, providing a framework for future research.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Barry London
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Internal MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Zeb R. Zacharias
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Jon C.D. Houtman
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Microbiology and ImmunologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Bryan G. Allen
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| |
Collapse
|
4
|
Gamal H, Ismail KA, Omar AMME, Teleb M, Abu-Serie MM, Huang S, Abdelsattar AS, Zamponi GW, Fahmy H. Non-small cell lung cancer sensitisation to platinum chemotherapy via new thiazole-triazole hybrids acting as dual T-type CCB/MMP-9 inhibitors. J Enzyme Inhib Med Chem 2024; 39:2388209. [PMID: 39140776 PMCID: PMC11328607 DOI: 10.1080/14756366.2024.2388209] [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: 05/09/2024] [Revised: 07/16/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
Cisplatin remains the unchallenged standard therapy for NSCLC. However, it is not completely curative due to drug resistance and oxidative stress-induced toxicity. Drug resistance is linked to overexpression of matrix metalloproteinases (MMPs) and aberrant calcium signalling. We report synthesis of novel thiazole-triazole hybrids as MMP-9 inhibitors with T-type calcium channel blocking and antioxidant effects to sensitise NSCLC to cisplatin and ameliorate its toxicity. MTT and whole cell patch clamp assays revealed that 6d has a balanced profile of cytotoxicity (IC50 = 21 ± 1 nM, SI = 12.14) and T-type calcium channel blocking activity (⁓60% at 10 μM). It exhibited moderate ROS scavenging activity and nanomolar MMP-9 inhibition (IC50 = 90 ± 7 nM) surpassing NNGH with MMP-9 over -2 and MMP-10 over -13 selectivity. Docking and MDs simulated its receptor binding mode. Combination studies confirmed that 6d synergized with cisplatin (CI = 0.69 ± 0.05) lowering its IC50 by 6.89 folds. Overall, the study introduces potential lead adjuvants for NSCLC platinum-based therapy.
Collapse
Affiliation(s)
- Hassan Gamal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Khadiga A Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Egypt
| | - A-Mohsen M E Omar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Marwa M Abu-Serie
- Department of Medical Biotechnology, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Egypt
| | - Sun Huang
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Abdalla S Abdelsattar
- Center for Microbiology and Phage Therapy, Zewail City of Sciences and Technology, October Gardens, Giza, Egypt
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Hesham Fahmy
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, South Dakota State University, Brookings, SD, USA
| |
Collapse
|
5
|
Wang Y, Qiu J, Yan H, Zhang N, Gao S, Xu N, Wang C, Lou H. The Bach1/HO-1 pathway regulates oxidative stress and contributes to ferroptosis in doxorubicin-induced cardiomyopathy in H9c2 cells and mice. Arch Toxicol 2024; 98:1781-1794. [PMID: 38573338 DOI: 10.1007/s00204-024-03697-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/31/2024] [Indexed: 04/05/2024]
Abstract
Doxorubicin (DOX) is one of the most frequently used chemotherapeutic drugs belonging to the class of anthracyclines. However, the cardiotoxic effects of anthracyclines limit their clinical use. Recent studies have suggested that ferroptosis is the main underlying pathogenetic mechanism of DOX-induced cardiomyopathy (DIC). BTB-and-CNC homology 1 (Bach1) acts as a key role in the regulation of ferroptosis. However, the mechanistic role of Bach1 in DIC remains unclear. Therefore, this study aimed to investigate the underlying mechanistic role of Bach1 in DOX-induced cardiotoxicity using the DIC mice in vivo (DOX at cumulative dose of 20 mg/kg) and the DOX-treated H9c2 cardiomyocytes in vitro (1 μM). Our results show a marked upregulation in the expression of Bach1 in the cardiac tissues of the DOX-treated mice and the DOX-treated cardiomyocytes. However, Bach1-/- mice exhibited reduced lipid peroxidation and less severe cardiomyopathy after DOX treatment. Bach1 knockdown protected against DOX-induced ferroptosis in both in vivo and in vitro models. Ferrostatin-1 (Fer-1), a potent inhibitor of ferroptosis, significantly alleviated DOX-induced cardiac damage. However, the cardioprotective effects of Bach1 knockdown were reversed by pre-treatment with Zinc Protoporphyrin (ZnPP), a selective inhibitor of heme oxygenase-1(HO-1). Taken together, these findings demonstrated that Bach1 promoted oxidative stress and ferroptosis through suppressing the expression of HO-1. Therefore, Bach1 may present as a promising new therapeutic target for the prevention and early intervention of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yanwei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
- Department of Radiology, Shandong Provincial Hospital, No. 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Jingru Qiu
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Hua Yan
- Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Nan Zhang
- Research Center of Translational Medicine, Breast Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, China
| | - Shixuan Gao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Ning Xu
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Cuiyan Wang
- Department of Radiology, Shandong Provincial Hospital, No. 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Haiyan Lou
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
6
|
Guimarães LC, Fidale TM, Pereira TCR, Lopes PR, Ferreira-Junior MD, Deconte SR, Ferreira-Neto ML, Brito WS, Gomes RM, de Souza FR, Cavalcante KVN, Herrera GC, de Moura FBR, Resende ES. Cardioprotective Effects of Leucine Supplementation against Doxorubicin-Induced Cardiotoxicit. Cardiovasc Toxicol 2024; 24:122-132. [PMID: 38165500 DOI: 10.1007/s12012-023-09817-2] [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] [Received: 08/08/2023] [Accepted: 11/18/2023] [Indexed: 01/03/2024]
Abstract
Doxorubicin is one of the most important antitumor drugs used in oncology; however, its cardiotoxic effect limits the therapeutic use and raises concerns regarding patient prognosis. Leucine is a branched-chain amino acid used in dietary supplementation and has been studied to attenuate the toxic effects of doxorubicin in animals, which increases oxidative stress. Oxidative stress in different organs can be estimated using several methods, including catalase expression analysis. This study aimed to analyze the effect of leucine on catalase levels in rat hearts after doxorubicin administration. Adult male Wistar rats were separated into two groups: Standard diet (SD) and 5% Leucine-Enriched Diet (LED). The animals had free access to diet from D0 to D28. At D14, the groups were subdivided in animals injected with Doxorubicin and animals injected with vehicle, until D28, and the groups were SD, SD + Dox, LED and LED + Dox. At D28, the animals were submitted do Transthoracic Echocardiography and euthanized. Despite Dox groups had impaired body weight gain, raw heart weight was not different between the groups. No substantial alterations were observed in macroscopic evaluation of the heart. Although, Doxorubicin treatment increased total interstitial collagen in the heart, which in addition to Type I collagen, is lower in LED groups. Western blot analysis showed that catalase expression in the heart of LED groups was lower than that in SD groups. In conclusion, leucine supplementation reduced both the precocious Dox-induced cardiac remodeling and catalase levels in the heart.
Collapse
Affiliation(s)
- Lucas C Guimarães
- Laboratory of Experimental Medicine, Department of Health Sciences - PGCS, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, Brazil.
| | - Thiago M Fidale
- Biotechnology Institute. Department of Medicine, Federal University of Catalão, Catalão, Goiás, Brazil
| | - Talita C R Pereira
- Institute of Biomedical Sciences, Department of Physiology, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Paulo R Lopes
- School of Dentistry-FOAr, Department of Physiology and Pathology, Universidade Estadual Paulista "Júlio de Mesquita Filho"-UNESP, Araraquara, SP, Brazil
| | - Marcos D Ferreira-Junior
- Laboratory of Endocrine Physiology and Metabolism, ICB, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Simone R Deconte
- UFU-ICBIM. Department of Physiology and Biophysics, Federal University of Uberlândia, Uberlândia, Brazil
| | - Marcos L Ferreira-Neto
- UFU-ICBIM. Department of Physiology and Biophysics, Federal University of Uberlândia, Uberlândia, Brazil
| | | | - Rodrigo M Gomes
- Laboratory of Endocrine Physiology and Metabolism, ICB, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Fernanda R de Souza
- Laboratory of Experimental Medicine, Department of Medicine, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Keilah V N Cavalcante
- Laboratory of Endocrine Physiology and Metabolism, ICB, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Gustavo C Herrera
- The Veterinary Hospital, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | | | - Elmiro S Resende
- Laboratory of Experimental Medicine, Department of Health Sciences - PGCS, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, Brazil
- Postgraduate Program in Health Sciences-PGCS, Faculty of Medicine, Federal University of Uberlândia, Uberlândia, MG, Brazil
| |
Collapse
|
7
|
Eslami SS, Jafari D, Ghotaslou A, Amoupour M, Asri Kojabad A, Jafari R, Mousazadeh N, Tarighi P, Sadeghizadeh M. Combined Treatment of Dendrosomal-Curcumin and Daunorubicin Synergistically Inhibit Cell Proliferation, Migration and Induce Apoptosis in A549 Lung Cancer Cells. Adv Pharm Bull 2023; 13:539-550. [PMID: 37646049 PMCID: PMC10460814 DOI: 10.34172/apb.2023.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/13/2022] [Accepted: 07/01/2022] [Indexed: 09/01/2023] Open
Abstract
Purpose Chemotherapy drugs used to treat lung cancer are associated with drug resistance and severe side effects. There have been rising demands for new therapeutic candidates and novel approaches, including combination therapy. Here, we aimed to investigate the combinatorial effect of a dendrosomal formulation of curcumin (DNC) and daunorubicin (DNR) on the A549 lung cancer cell line. Methods We performed cytotoxicity, apoptosis, cell migration, colony-formation capacity, and gene expression analysis to interpret the mechanism of action for a combination of DNC and DNR on A549 cells. Results Our results revealed that the combination of DNC and DNR could synergistically inhibit the A549 cells' growth. This synergistic cytotoxicity was further approved by flow cytometry, migration assessment, colony-forming capacity and gene expression analysis. DNR combination with DNC resulted in increased apoptosis to necrosis ratio compared to DNR alone. In addition, the migration and colony-forming capacity were at the minimal range when DNC was combined with DNR. Combined treatment decreased the expression level of MDR-1, hTERT and Bcl-2 genes significantly. In addition, the ratio of Bax/Bcl2 gene expression significantly increased. Our analysis by free curcumin, dendrosomes and DNC also showed that dendrosomes do not have any significant cytotoxic effect on the A549 cells, suggesting that this carrier has a high potential for enhancing the curcumin's biological effects. Conclusion Our observations suggest that the DNC formulation of curcumin synergistically enhances the antineoplastic effect of DNR on the A549 cell line through the modulation of apoptosis/necrosis ratio, as well as Bax/Bcl2 ratio, MDR-1 and hTERT gene expression.
Collapse
Affiliation(s)
- Seyed Sadegh Eslami
- Student Research Committee, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davod Jafari
- Student Research Committee, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abbas Ghotaslou
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Amoupour
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Asri Kojabad
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Rasool Jafari
- Department of Medical Parasitology and Mycology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Navid Mousazadeh
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
8
|
Caiati C, Stanca A, Lepera ME. Free Radicals and Obesity-Related Chronic Inflammation Contrasted by Antioxidants: A New Perspective in Coronary Artery Disease. Metabolites 2023; 13:712. [PMID: 37367870 PMCID: PMC10302379 DOI: 10.3390/metabo13060712] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/28/2023] Open
Abstract
We are surrounded by factors called free radicals (FR), which attach to the molecules our body is made of, first among them the endothelium. Even though FR are to a certain extent a normal factor, nowadays we face an escalating increase in these biologically aggressive molecules. The escalating formation of FR is linked to the increased usage of man-made chemicals for personal care (toothpaste, shampoo, bubble bath, etc.), domestic laundry and dish-washer detergents, and also an ever wider usage of drugs (both prescription and over the counter), especially if they are to be used long-term (years). In addition, tobacco smoking, processed foods, pesticides, various chronic infectious microbes, nutritional deficiencies, lack of sun exposure, and, finally, with a markedly increasing impact, electromagnetic pollution (a terribly destructive factor), can increase the risk of cancer, as well as endothelial dysfunction, owing to the increased production of FR that they cause. All these factors create endothelial damage, but the organism may be able to repair such damage thanks to the intervention of the immune system supported by antioxidants. However, one other factor can perpetuate the state of inflammation, namely obesity and metabolic syndrome with associated hyperinsulinemia. In this review, the role of FR, with a special emphasis on their origin, and of antioxidants, is explored from the perspective of their role in causing atherosclerosis, in particular at the coronary level.
Collapse
Affiliation(s)
- Carlo Caiati
- Unit of Cardiovascular Diseases, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.); (M.E.L.)
| | | | | |
Collapse
|
9
|
Kunika, Frey N, Rangrez AY. Exploring the Involvement of Gut Microbiota in Cancer Therapy-Induced Cardiotoxicity. Int J Mol Sci 2023; 24:7261. [PMID: 37108423 PMCID: PMC10138392 DOI: 10.3390/ijms24087261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Trillions of microbes in the human intestinal tract, including bacteria, viruses, fungi, and protozoa, are collectively referred to as the gut microbiome. Recent technological developments have led to a significant increase in our understanding of the human microbiome. It has been discovered that the microbiome affects both health and the progression of diseases, including cancer and heart disease. Several studies have indicated that the gut microbiota may serve as a potential target in cancer therapy modulation, by enhancing the effectiveness of chemotherapy and/or immunotherapy. Moreover, altered microbiome composition has been linked to the long-term effects of cancer therapy; for example, the deleterious effects of chemotherapy on microbial diversity can, in turn, lead to acute dysbiosis and serious gastrointestinal toxicity. Specifically, the relationship between the microbiome and cardiac diseases in cancer patients following therapy is poorly understood. In this article, we provide a summary of the role of the microbiome in cancer treatment, while also speculating on a potential connection between treatment-related microbial changes and cardiotoxicity. Through a brief review of the literature, we further explore which bacterial families or genera were differentially affected in cancer treatment and cardiac disease. A deeper understanding of the link between the gut microbiome and cardiotoxicity caused by cancer treatment may help lower the risk of this critical and potentially fatal side effect.
Collapse
Affiliation(s)
- Kunika
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Ashraf Y. Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Tang X, Kurban M, Hafiz I, Shen Q, Wang M. Preparation of hyaluronic acid-loaded Harmine polymeric micelles and in vitro effect anti-breast cancer. Eur J Pharm Sci 2023; 183:106388. [PMID: 36758771 DOI: 10.1016/j.ejps.2023.106388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 02/10/2023]
Abstract
AIMS To prepare hyaluronic acid-loaded Harmine polymeric micelles with CD44 targeting properties and to investigate their anti-breast cancer effects in vitro. METHODS The carboxyl group on hyaluronic acid is coupled to the amino group on 3,5-bis(trifluoromethyl)benzylamine by an amidation reaction. And the polymeric micelles self-assemble to encapsulate the Harmine in a hydrophobic core, characterized the polymer micelles by IR, 19F-NMR, Malvern particle sizing, release, hemolysis, and other experiments. Used CD44-positive MDA-MB-231 cells and CD44-negative MCF-7 cells as tumor models. The effect of polymer micelles on breast cancer cells in vitro by cytotoxicity assay, confocal, and flow cytometry. RESULTS The prepared polymer micelles had a uniform particle size of about 200 nm, good dispersion, PDI < 0.3, encapsulation rate up to 87%, drug loading of 4.12±0.03%, and negative charge. Hyaluronidase has a good enzymatic effect on polymeric micelles, with a hemolysis rate of less than 1%. It showed some dose-dependent toxicity to both MDA-MB-231 and MCF-7, with increased uptake of polymer micelles by CD44-positive MDA-MB-231 compared to CD44-negative MCF-7 cells and significant effects of polymer micelles on apoptosis and cycling in both cell types. These results suggest that the hyaluronic acid-loaded Harmine polymer micelles designed in this study are effective in killing breast cancer cells while at the same time reducing the toxicity of Harmine and improving its slow-release targeting.
Collapse
Affiliation(s)
- Xiaohui Tang
- Central Laboratory of Xinjiang Medical University, Urumqi 830017, China
| | - Munire Kurban
- Department of Pharmacy, Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, China
| | - Ipargul Hafiz
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Qi Shen
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Mei Wang
- School of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
11
|
Chang SK, Liu D, Mitchem J, Papageorgiou C, Kaifi J, Shyu CR. Understanding common key indicators of successful and unsuccessful cancer drug trials using a contrast mining framework on ClinicalTrials.gov. J Biomed Inform 2023; 139:104321. [PMID: 36806327 DOI: 10.1016/j.jbi.2023.104321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 02/04/2023] [Accepted: 02/11/2023] [Indexed: 02/18/2023]
Abstract
Clinical trials are essential to the process of new drug development. As clinical trials involve significant investments of time and money, it is crucial for trial designers to carefully investigate trial settings prior to designing a trial. Utilizing trial documents from ClinicalTrials.gov, we aim to understand the common characteristics of successful and unsuccessful cancer drug trials to provide insights about what to learn and what to avoid. In this research, we first computationally classified cancer drug trials into successful and unsuccessful cases and then utilized natural language processing to extract eligibility criteria information from the trial documents. To provide explainable and potentially modifiable recommendations for new trial design, contrast mining was applied to discoverhighly contrasted patterns with a significant difference in prevalence between successful (completion with advancement to the next phase) and unsuccessful (suspended, withdrawn, or terminated) groups. Our method identified contrast patterns consisting of combinations of drug categories, eligibility criteria, study organization, and study design for nine major cancers. In addition to a literature review for the qualitative validation of mined contrast patterns, we found that contrast-pattern-based classifiers using the top 200 contrast patterns as feature representations can achieve approximately 80% F1 score for eight out of ten cancer types in our experiments. In summary, aligning with the modernization efforts of ClinicalTrials.gov, our study demonstrates that understanding the contrast characteristics of successful and unsuccessful cancer trials may provide insights into the decision-making process for trial investigators and therefore facilitate improved cancer drug trial design.
Collapse
Affiliation(s)
- Shu-Kai Chang
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA
| | - Danlu Liu
- Electrical Engineering and Computer Science Department, University of Missouri, Columbia, MO 65211, USA
| | - Jonathan Mitchem
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Christos Papageorgiou
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Jussuf Kaifi
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Chi-Ren Shyu
- Institute for Data Science & Informatics, University of Missouri, Columbia, MO 65211, USA; Electrical Engineering and Computer Science Department, University of Missouri, Columbia, MO 65211, USA; Department of Medicine, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
12
|
Deng T, Luo D, Zhang R, Zhao R, Hu Y, Zhao Q, Wang S, Iqbal MZ, Kong X. DOX-loaded hydroxyapatite nanoclusters for colorectal cancer (CRC) chemotherapy: Evaluation based on the cancer cells and organoids. SLAS Technol 2023; 28:22-31. [PMID: 36328181 DOI: 10.1016/j.slast.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/21/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022]
Abstract
It is meaningful to find suitable in vitro models for preclinical toxicology and efficacy evaluation of nanodrugs and nanocarriers or drug screening and promoting clinical transformation of nanocarriers. The emergence and development of organoids technology provide a great possibility to achieve this goal. Herein, we constructed an in vitro 3D organoid model to study the inhibitory effect of nanocarriers on colorectal cancer. And designed hydroxyapatite nanoclusters (c-HAP) mediated by polydopamine (PDA) formed under alkaline conditions (pH 9.0), then used c-HAP to load DOX (c-HAP/DOX) as nanocarrier for improved chemotherapy. In vitro, drug release experiments show that c-HAP/DOX has suitable responsive to pH, can be triggered to the facile release of DOX in a slightly acidic environment (pH 6.0), and maintain specific stability in a neutral pH value (7.4) environment. c-HAP/DOX showed an excellent antitumor effect in the two-dimensional (2D) cell model and three-dimensional (3D) patient-derived colon cancer organoids (PDCCOs) model. In addition, c-HAP/DOX can release a sufficient amount of DOX to produce cytotoxicity in a slightly acidic environment, entering efficiently into the colorectal cancer cells caused endocytosis and induced apoptosis. Therefore, organoids can serve as an effective in vitro model to present the structure and function of colorectal cancer tissues and be used to evaluate the efficacy of nanocarriers for tumors.
Collapse
Affiliation(s)
- Tianhao Deng
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Dandan Luo
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China; School of Textile Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Rui Zhang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Ruibo Zhao
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Yeting Hu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, PR China
| | - Qingwei Zhao
- Research Center for Clinical Pharmacy & Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310018, PR China
| | - Shibo Wang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - M Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China.
| |
Collapse
|
13
|
RING Finger Protein 10 Regulates AP-1/Meox2 to Mediate Pirarubicin-Induced Cardiomyocyte Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7872193. [PMID: 36713029 PMCID: PMC9883094 DOI: 10.1155/2023/7872193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Accepted: 11/25/2022] [Indexed: 01/21/2023]
Abstract
Pirarubicin (THP) is one of the classic chemotherapy drugs for cancer treatment. It is often clinically limited because of its cardiotoxicity. The occurrence and development of THP-mediated chemotherapy-related cardiotoxicity (CRC) may be reversed by RING finger protein 10 (RNF10). This study was performed with the aim of evaluating the inhibitory effect of RNF10 on THP-mediated CRC and its molecular mechanism. In vivo, we found that the expression of RNF10 decreased in THP-induced CRC rats, accompanied by Meox2 inhibition and AP-1 activation, resulting in increased cardiomyocyte apoptosis. After small interfering RNA (siRNA) and lentivirus transfection (Lv) of RNF10 in vitro, the expression of RNF10, Meox2, and AP-1 proteins and the degree of cardiomyocyte apoptosis were detected. We found that overexpression of RNF10 in H9C2 cardiomyocytes significantly promoted Meox2 and inhibited AP-1, alleviated apoptosis, and showed further inhibitory activity on THP-induced cardiomyocyte toxicity. Silencing RNF10 showed the opposite result. Our study showed that RNF10 inhibited THP-induced CRC through the activity of Meox2 and AP-1 proteins. RNF10 may be the next drug target for the treatment of CRC and other related cardiovascular diseases.
Collapse
|
14
|
Li H, Lin L, Xia YL, Xie Y, Yang X. Research progress on the role of ferroptosis in cardiovascular disease. Front Cardiovasc Med 2022; 9:1077332. [PMID: 36620630 PMCID: PMC9815775 DOI: 10.3389/fcvm.2022.1077332] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
The cardiovascular disease pathogenesis is extremely complex and seriously threatens human health. Cardiomyocyte death plays a significant role in cardiovascular disease occurrence and development. In addition to the previously revealed modes of cell death (apoptosis, autophagy, and pyroptosis), ferroptosis is highly related to the development of cardiovascular diseases, including arrhythmia, atherosclerosis, and myocardial ischemia/reperfusion. Ferroptosis is a novel cell death pathway driven by lipid peroxidation and iron overload. Lipid, amino acid, and iron metabolism regulate the ferroptosis pathway. Small molecule compounds (iron chelators, antioxidants, and ferroptosis inhibitors) and genetic programming can alleviate or prevent cardiovascular disease by inhibiting the ferroptosis pathway. Ferroptosis plays a key role in various cardiovascular disease occurrence and development, and inhibiting ferroptosis in cardiomyocytes is expected to become a feasible treatment method. In this mini-review, we systematically summarize the molecular mechanisms of ferroptosis in different cardiovascular diseases, delineate the regulatory network between ferroptosis and cardiovascular diseases, and highlight its potential therapeutic targets.
Collapse
Affiliation(s)
- Han Li
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Li Lin
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yun-Long Xia
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China,Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunpeng Xie
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China,*Correspondence: Yunpeng Xie,
| | - Xiaolei Yang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China,Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China,Xiaolei Yang,
| |
Collapse
|
15
|
Mechchate H, de Castro Alves CE, Es-safi I, Amaghnouje A, Jawhari FZ, Costa de Oliveira R, de Freitas Gomes A, Conte R, Soares Pontes G, Bousta D, Grafov A. Antileukemic, Antioxidant, Anti-Inflammatory and Healing Activities Induced by a Polyphenol-Enriched Fraction Extracted from Leaves of Myrtus communis L. Nutrients 2022; 14:nu14235055. [PMID: 36501085 PMCID: PMC9740279 DOI: 10.3390/nu14235055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Natural products have offered a number of exciting approaches in cancer treatment over the years. In this study, we investigated the prophylactic and therapeutic effects of the polyphenol-enriched fraction extracted from Myrtus communis (PEMC) on acute and chronic leukemia. According to the UHPLC-MSn, the fraction is rich in flavonoids. Protective activity of the PEMC was assessed by evaluating the antioxidant, anti-inflammatory, wound healing, and hemolysis potential in a series of in vivo and in vitro assays, while the therapeutic approach consisted of the evaluation of cytotoxic activity of the PEMC against HL60 and K562 leukemia cell lines. Safety of the fraction was also evaluated on a non-cancerous Vero cell line and by an acute toxicity test performed in mice. The PEMC demonstrated a significant anti-inflammatory and healing potential. The activities found at the dose of 100 mg/kg were better than those observed using a reference drug. The PEMC demonstrated a significant antioxidant effect and a specific cytotoxicity towards HL60 (IC50 = 19.87 µM) and K562 (IC50 = 29.64 µM) cell lines being non-toxic to the Vero cell line. No hemolytic activity was observed in vitro and no toxicity effect was found in mice. Thus, the PEMC has a pharmacological potential as both preventive and therapeutic agent. However, further research is necessary to propose its mechanism of action.
Collapse
Affiliation(s)
- Hamza Mechchate
- Laboratory of Biotechnology, Environment, Agrifood, and Health, University of Sidi Mohamed Ben Abdellah (USMBA), Fez B.P. 1796, Morocco
| | - Carlos Eduardo de Castro Alves
- Post-Graduate Program in Basic and Applied Immunology, Institute of Biological Sciences, Federal University of Amazonas, Manaus 69077-000, AM, Brazil
| | - Imane Es-safi
- Laboratory of Biotechnology, Environment, Agrifood, and Health, University of Sidi Mohamed Ben Abdellah (USMBA), Fez B.P. 1796, Morocco
| | - Amal Amaghnouje
- Laboratory of Biotechnology, Environment, Agrifood, and Health, University of Sidi Mohamed Ben Abdellah (USMBA), Fez B.P. 1796, Morocco
| | - Fatima Zahra Jawhari
- Laboratory of Biotechnology, Environment, Agrifood, and Health, University of Sidi Mohamed Ben Abdellah (USMBA), Fez B.P. 1796, Morocco
| | - Regiane Costa de Oliveira
- Post-Graduate Program in Hematology, The State University of Amazonas, Foundation of Hematology and Hemotherapy of Amazonas, Manaus 69050-010, AM, Brazil
| | - Alice de Freitas Gomes
- Post-Graduate Program in Hematology, The State University of Amazonas, Foundation of Hematology and Hemotherapy of Amazonas, Manaus 69050-010, AM, Brazil
- Laboratory of Virology and Immunology, National Institute of Amazonian Research (INPA), Manaus 69067-375, AM, Brazil
| | - Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)—CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Gemilson Soares Pontes
- Post-Graduate Program in Basic and Applied Immunology, Institute of Biological Sciences, Federal University of Amazonas, Manaus 69077-000, AM, Brazil
- Post-Graduate Program in Hematology, The State University of Amazonas, Foundation of Hematology and Hemotherapy of Amazonas, Manaus 69050-010, AM, Brazil
- Laboratory of Virology and Immunology, National Institute of Amazonian Research (INPA), Manaus 69067-375, AM, Brazil
| | - Dalila Bousta
- Laboratory of Biotechnology, Environment, Agrifood, and Health, University of Sidi Mohamed Ben Abdellah (USMBA), Fez B.P. 1796, Morocco
| | - Andriy Grafov
- Materials Chemistry Division, Department of Chemistry, University of Helsinki, A.I. Virtasen aukio 1, 00560 Helsinki, Finland
- Correspondence:
| |
Collapse
|
16
|
Li J, Lin J, Huang S, Li M, Yu W, Zhao Y, Guo J, Zhang P, Huang X, Qiao Y. Functional Phosphoproteomics in Cancer Chemoresistance Using CRISPR-Mediated Base Editors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200717. [PMID: 36045417 PMCID: PMC9596822 DOI: 10.1002/advs.202200717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Selective inhibition of targeted protein kinases is an effective therapeutic approach for treatment of human malignancies, which interferes phosphorylation of cellular substrates. However, a drug-imposed selection creates pressures for tumor cells to acquire chemoresistance-conferring mutations or activating alternative pathways, which can bypass the inhibitory effects of kinase inhibitors. Thus, identifying downstream phospho-substrates conferring drug resistance is of great importance for developing poly-pharmacological and targeted therapies. To identify functional phosphorylation sites involved in 5-fluorouracil (5-FU) resistance during its treatment of colorectal cancer cells, CRISPR-mediated cytosine base editor (CBE) and adenine base editor (ABE) are utilized for functional screens by mutating phosphorylated amino acids with two libraries specifically targeting 7779 and 10 149 phosphorylation sites. Among the top enriched gRNAs-induced gain-of-function mutants, the target genes are involved in cell cycle and post-translational covalent modifications. Moreover, several substrates of RSK2 and PAK4 kinases are discovered as main effectors in responding to 5-FU chemotherapy, and combinational treatment of colorectal cancer cells with 5-FU and RSK2 inhibitor or PAK4 inhibitor can largely inhibit cell growth and enhance cell apoptosis through a RSK2/TP53BP1/γ-H2AX phosphorylation signaling axis. It is proposed that this screen approach can be used for functional phosphoproteomics in chemotherapy of various human diseases.
Collapse
Affiliation(s)
- Jianan Li
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
- Zhejiang LabHangzhouZhejiang311121China
| | - Jianxiang Lin
- Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
- Shanghai Institute of Precision MedicineShanghai200125China
| | | | - Min Li
- Precise Genome Engineering CenterSchool of Life SciencesGuangzhou UniversityGuangzhou510006China
| | - Wenxia Yu
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Yuting Zhao
- Precise Genome Engineering CenterSchool of Life SciencesGuangzhou UniversityGuangzhou510006China
| | - Junfan Guo
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Pumin Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseThe First Affiliated Hospitaland Institute of Translational MedicineZhejiang University School of MedicineHangzhou310029China
| | - Xingxu Huang
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseThe First Affiliated Hospitaland Institute of Translational MedicineZhejiang University School of MedicineHangzhou310029China
| | - Yunbo Qiao
- Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125China
- Shanghai Institute of Precision MedicineShanghai200125China
- Precise Genome Engineering CenterSchool of Life SciencesGuangzhou UniversityGuangzhou510006China
| |
Collapse
|
17
|
Terwoord JD, Beyer AM, Gutterman DD. Endothelial dysfunction as a complication of anti-cancer therapy. Pharmacol Ther 2022; 237:108116. [PMID: 35063569 PMCID: PMC9294076 DOI: 10.1016/j.pharmthera.2022.108116] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/16/2021] [Accepted: 01/12/2022] [Indexed: 12/14/2022]
Abstract
Recent strides in anti-cancer therapeutics have improved longevity and led to a growing population of cancer survivors, who are increasingly likely to die of other causes. Treatment-induced cardiotoxicity is a complication of several therapeutic agents with acute and long-term consequences for cancer patients. Vascular endothelial dysfunction is a precursor and hallmark of ischemic coronary disease and may play a role in anti-cancer therapy-induced cardiotoxicity. This review summarizes clinical evidence for endothelial dysfunction following anti-cancer therapy and extends the discussion to include the impact of therapeutic agents on conduit arteries and the microcirculation. We highlight the role of innate immune system activation and cross-talk between inflammation and oxidative stress as pathogenic mechanisms underlying anti-cancer therapy-induced vascular toxicity. Understanding the impact of anti-cancer agents on the vascular endothelium will inform therapeutic approaches to prevent or reverse treatment-induced cardiotoxicity and may serve as an important tool to predict, monitor, and prevent adverse cardiovascular outcomes in patients undergoing treatment.
Collapse
Affiliation(s)
- Janée D Terwoord
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America.
| | - Andreas M Beyer
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - David D Gutterman
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| |
Collapse
|
18
|
Yang Y, Shi X, Chen Z, Xu Y, Qian X, Zhu W. Novel seven-membered ring-fused naphthalimide derivatives with potentials for cancer theranostics. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.07.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Kadoglou NPE, Panayiotou C, Vardas M, Balaskas N, Kostomitsopoulos NG, Tsaroucha AK, Valsami G. A Comprehensive Review of the Cardiovascular Protective Properties of Silibinin/Silymarin: A New Kid on the Block. Pharmaceuticals (Basel) 2022; 15:538. [PMID: 35631363 PMCID: PMC9145573 DOI: 10.3390/ph15050538] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 12/04/2022] Open
Abstract
Silibinin/silymarin has been used in herbal medicine for thousands of years and it is well-known for its hepato-protective properties. The present comprehensive literature review aimed to critically summarize the pharmacological properties of silymarin extract and its main ingredient silibinin in relation to classical cardiovascular risk factors (e.g., diabetes mellitus, etc.). We also assessed their potential protective and/or therapeutic application in cardiovascular diseases (CVDs), based on experimental and clinical studies. Pre-clinical studies including in vitro tests or animal models have predominantly implicated the following effects of silymarin and its constituents: (1) antioxidant, (2) hypolipidemic, (3) hypoglycemic, (4) anti-hypertensive and (5) cardioprotective. On the other hand, a direct amelioration of atherosclerosis and endothelial dysfunction after silymarin administration seems weak based on scarce data. In clinical trials, the most important findings are improved (1) glycemic and (2) lipid profiles in patients with type 2 diabetes mellitus and/or hyperlipidemia, while (3) the anti-hypertensive effects of silibinin/silymarin seem very modest. Finally, the changes in clinical endpoints are not robust enough to draw a firm conclusion. There are significant limitations in clinical trial design, including the great variety in doses and cohorts, the underlying conditions, the small sample sizes, the short duration and the absence of pharmacokinetic/pharmacodynamic tests prior to study commitment. More data from well-designed and high-quality pre-clinical and clinical studies are required to firmly establish the clinical efficacy of silibinin/silymarin and its possible therapeutic application in cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Michail Vardas
- Medical School, University of Cyprus, Nicosia 2109, Cyprus; (C.P.); (M.V.); (N.B.)
| | - Nikolaos Balaskas
- Medical School, University of Cyprus, Nicosia 2109, Cyprus; (C.P.); (M.V.); (N.B.)
| | - Nikolaos G. Kostomitsopoulos
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Alexandra K. Tsaroucha
- Laboratory of Experimental Surgery and Surgical Research, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Laboratory of Bioethics, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Georgia Valsami
- Laboratory of Biopharmaceutics-Pharmacokinetics, Department of Pharmacy, School of Health Sciences, National & Kapodistrian University of Athens, 15784 Athens, Greece;
| |
Collapse
|
20
|
Cakan P, Yildiz S, Akyay A, Öncül Y. Intensive chemotherapy perturbs heart rate variability in children with cancer. Neurophysiol Clin 2021; 52:69-80. [PMID: 34973888 DOI: 10.1016/j.neucli.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES In children, cancer chemotherapy may impair the functioning of the cardiac autonomic nervous system. Moreover, it is not known whether there are any differences between intensive and maintenance phases of chemotherapy. Therefore, the aim of the current study was to assess autonomic nervous system activity using heart rate variability, in children receiving intensive or maintenance cancer chemotherapy. METHODS For that purpose, children who were healthy (healthy control, n = 30), receiving intensive chemotherapy (chemotherapy, n = 30), and receiving maintenance chemotherapy (maintenance, n = 25) were included in the study. Autonomic nervous system activity was measured by means of heart rate variability. Electrocardiogram recordings were used to calculate time- and frequency-domain heart rate variability parameters. RESULTS Time-domain parameters such as standard deviation of NN intervals and frequency-domain parameters such as total power were lower during the intensive chemotherapy but not during maintenance phase (standard deviation of NN intervals: 50±5, 33±3, and 48±3 ms, and total power: 2613±504, 1379±296 and 2295±264 ms2, respectively for healthy control, chemotherapy and maintenance groups, P<0.001 for both standard deviation of NN intervals and total power). DISCUSSION The present results indicate that intensive chemotherapy perturbs the function of heart rate variability in children, with recovery during the maintenance phase. This suggests that intensive chemotherapy is likely to affect the autonomic nervous system but this effect does not appear to be permanent.
Collapse
Affiliation(s)
- Pinar Cakan
- Department of Physiology, University of Health Sciences, Hamidiye Faculty of Medicine, Istanbul, Turkey.
| | - Sedat Yildiz
- Department of Physiology Inonu University, Faculty of Medicine, Malatya, Turkey
| | - Arzu Akyay
- Department of Pediatric Hematology, Inonu University, Faculty of Medicine, Malatya, Turkey
| | - Yurday Öncül
- Department of Pediatric Hematology, Inonu University, Faculty of Medicine, Malatya, Turkey
| |
Collapse
|
21
|
Sun L, Wang H, Xu D, Yu S, Zhang L, Li X. Lapatinib induces mitochondrial dysfunction to enhance oxidative stress and ferroptosis in doxorubicin-induced cardiomyocytes via inhibition of PI3K/AKT signaling pathway. Bioengineered 2021; 13:48-60. [PMID: 34898356 PMCID: PMC8805895 DOI: 10.1080/21655979.2021.2004980] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lapatinib (LAP) is an important anti-cancer drug and is frequently alongside doxorubicin (DOX) as a combination therapy for better anti-cancer efficacy. However, many studies have reported that LAP in combination with DOX may induce highly cardiotoxicity. Accordingly, we aimed to explore the potential mechanism involved in the synergistic effect of LAP in DOX-induced cardiotoxicity. Here, cell counting kit-8 was used to detect cell viability and lactate dehydrogenase measurement was performed to assess cell injury. Cell apoptosis was evaluated by TUNEL assay and western blot assay. Mitochondrial dysfunction was identified by JC-1 assay, adenosine triphosphate (ATP) and Cytochrome C. Moreover, the activity of ROS, SOD, CAT and GSH were measured to elucidate oxidative stress level. Ferroptosis was examined by levels of Fe2+, GPX4 and ASCL4. Expressions of PI3K/AKT signaling were identified by western blot assay. The results revealed that LAP inhibited the cell viability and exacerbated cell injury induced by Dox, as well as increased cell apoptosis. LAP aggravated DOX-induced mitochondria damage by changed mitochondrial membrane potential, decreased ATP and increased level of Cytochrome C. In addition, the combination of LAP and DOX induced oxidative stress and ferroptosis in H9c2 cells. The activation of PI3K/AKT signaling reversed the detrimental effects of LAP on DOX-induced H9c2 cells. The data in this study showed for the first time that LAP aggravated Dox-induced cardiotoxicity by promoting oxidative stress and ferroptosis in cardiomyocytes via PI3K/AKT-mediated mitochondrial dysfunction, suggesting that PI3K/AKT activation is a promising cardioprotective strategy for DOX /LAX combination therapies.
Collapse
Affiliation(s)
- Lei Sun
- Ultrasonic Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hua Wang
- Ultrasonic Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dan Xu
- Ultrasonic Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shanshan Yu
- Ultrasonic Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lin Zhang
- Ultrasonic Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaopeng Li
- Ultrasonic Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Lozahic C, Maddock H, Sandhu H. Anti-cancer Therapy Leads to Increased Cardiovascular Susceptibility to COVID-19. Front Cardiovasc Med 2021; 8:634291. [PMID: 33969006 PMCID: PMC8102732 DOI: 10.3389/fcvm.2021.634291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/09/2021] [Indexed: 12/15/2022] Open
Abstract
Anti-cancer treatment regimens can lead to both acute- and long-term myocardial injury due to off-target effects. Besides, cancer patients and survivors are severely immunocompromised due to the harsh effect of anti-cancer therapy targeting the bone marrow cells. Cancer patients and survivors can therefore be potentially extremely clinically vulnerable and at risk from infectious diseases. The recent global outbreak of the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its infection called coronavirus disease 2019 (COVID-19) has rapidly become a worldwide health emergency, and on March 11, 2020, COVID-19 was declared a global pandemic by the World Health Organization (WHO). A high fatality rate has been reported in COVID-19 patients suffering from underlying cardiovascular diseases. This highlights the critical and crucial aspect of monitoring cancer patients and survivors for potential cardiovascular complications during this unprecedented health crisis involving the progressive worldwide spread of COVID-19. COVID-19 is primarily a respiratory disease; however, COVID-19 has shown cardiac injury symptoms similar to the cardiotoxicity associated with anti-cancer therapy, including arrhythmia, myocardial injury and infarction, and heart failure. Due to the significant prevalence of micro- and macro-emboli and damaged vessels, clinicians worldwide have begun to consider whether COVID-19 may in fact be as much a vascular disease as a respiratory disease. However, the underlying mechanisms and pathways facilitating the COVID-19-induced cardiac injury in cancer and non-cancer patients remain unclear. Investigations into whether COVID-19 cardiac injury and anti-cancer drug-induced cardiac injury in cancer patients and survivors might synergistically increase the cardiovascular complications and comorbidity risk through a “two-hit” model are needed. Identification of cardiac injury mechanisms and pathways associated with COVID-19 development overlapping with anti-cancer therapy could help clinicians to allow a more optimized prognosis and treatment of cancer survivors suffering from COVID-19. The following review will focus on summarizing the harmful cardiovascular risk of COVID-19 in cancer patients and survivors treated with an anti-cancer drug. This review will improve the knowledge of COVID-19 impact in the field of cardio-oncology and potentially improve the outcome of patients.
Collapse
Affiliation(s)
- Caroline Lozahic
- Faculty Research Centre for Sport, Exercise and Life Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Helen Maddock
- Faculty Research Centre for Sport, Exercise and Life Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Hardip Sandhu
- Faculty Research Centre for Sport, Exercise and Life Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, United Kingdom
| |
Collapse
|
23
|
Gatti M, Raschi E, Poluzzi E, Martignani C, Salvagni S, Ardizzoni A, Diemberger I. The Complex Management of Atrial Fibrillation and Cancer in the COVID-19 Era: Drug Interactions, Thromboembolic Risk, and Proarrhythmia. Curr Heart Fail Rep 2020; 17:365-383. [PMID: 33025463 PMCID: PMC7537958 DOI: 10.1007/s11897-020-00485-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Cardiotoxicity by anticancer agents has emerged as a multifaceted issue and is expected to affect both mortality and morbidity. This review summarizes clinical challenges in the management of oncological patients requiring anticoagulants for atrial fibrillation (AF) also considering the current outbreak of the COVID-19 (coronavirus disease 2019) pandemic, since this infection can add challenges to the management of both conditions. Specifically, the aims are manyfold: (1) describe the evolving use of direct oral anticoagulants (DOACs) in AF patients with cancer; (2) critically appraise the risk of clinically important drug-drug interactions (DDIs) between DOACs and oral targeted anticancer agents; (3) address expected DDIs between DOACs and candidate anti-COVID drugs, with implications on management of the underlying thrombotic risk; and (4) characterize the proarrhythmic liability in cardio-oncology in the setting of COVID-19, focusing on QT prolongation. RECENT FINDINGS AF in cardio-oncology poses diagnostic and management challenges, also due to the number of anticancer drugs recently associated with AF onset/worsening. Oral targeted drugs can potentially interact with DOACs, with increased bleeding risk mainly due to pharmacokinetic DDIs. Moreover, the vast majority of oral anticancer agents cause QT prolongation with direct and indirect mechanisms, potentially resulting in the occurrence of torsade de pointes, especially in susceptible patients with COVID-19 receiving additional drugs with QT liability. Oncologists and cardiologists must be aware of the increased bleeding risk and arrhythmic susceptibility of patients with AF and cancer due to DDIs. High-risk individuals with COVID-19 should be prioritized to target preventive strategies, including optimal antithrombotic management, medication review, and stringent monitoring.
Collapse
Affiliation(s)
- Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Emanuel Raschi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Elisabetta Poluzzi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Cristian Martignani
- Cardiology Unit, Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | | | - Andrea Ardizzoni
- Medical Oncology Unit, Department of Experimental, Diagnostic and Specialty Medicine, Policlinico S. Orsola-Malpighi, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Igor Diemberger
- Cardiology Unit, Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| |
Collapse
|
24
|
Shalaby N, Zemzemi N, Elkhodary K. Simulating the effect of sodium channel blockage on cardiac electromechanics. Proc Inst Mech Eng H 2019; 234:16-27. [PMID: 31625448 DOI: 10.1177/0954411919882514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is growing interest to better understand drug-induced cardiovascular complications and to predict undesirable side effects at as early a stage in the drug development process as possible. The purpose of this paper is to investigate computationally the influence of sodium ion channel blockage on cardiac electromechanics. To do so, we implement a myofiber orientation dependent passive stress model (Holzapfel-Ogden) in the multiphysics solver Chaste to simulate an imaged physiological model of the human ventricles. A dosage of a sodium channel blocker was then applied and its inhibitory effects on the electrical propagation across ventricles were modeled. We employ the Kerckhoffs active stress model to generate electrically excited contractile behavior of myofibers. Our predictions indicate that a delay in the electrical activation of ventricular tissue caused by the sodium channel blockage translates to a delay in the mechanical biomarkers that were investigated. Moreover, sodium channel blockage was found to increase left ventricular twist. A multiphysics computational framework from the cell level to the organ level was thus used to predict the effect of sodium channel blocking drugs on cardiac electromechanics.
Collapse
Affiliation(s)
- Noha Shalaby
- Mechanical Engineering Department, The American University in Cairo, New Cairo, Egypt
| | - Nejib Zemzemi
- INRIA Bordeaux Sud-Ouest, Carmen Group, Talence, France.,IHU-LIRYC, Pessac, France
| | - Khalil Elkhodary
- Mechanical Engineering Department, The American University in Cairo, New Cairo, Egypt
| |
Collapse
|
25
|
Gill JH, Rockley KL, De Santis C, Mohamed AK. Vascular Disrupting Agents in cancer treatment: Cardiovascular toxicity and implications for co-administration with other cancer chemotherapeutics. Pharmacol Ther 2019; 202:18-31. [PMID: 31173840 DOI: 10.1016/j.pharmthera.2019.06.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 05/30/2019] [Indexed: 02/08/2023]
Abstract
Destruction of the established tumour vasculature by a class of compound termed Vascular Disrupting Agents (VDAs) is showing considerable promise as a viable approach for the management of solid tumours. VDAs induce a rapid shutdown and collapse of tumour blood vessels, leading to ischaemia and consequent necrosis of the tumour mass. Their efficacy is hindered by the persistence of a viable rim of tumour cells, supported by the peripheral normal vasculature, necessitating their co-administration with additional chemotherapeutics for maximal therapeutic benefit. However, a major limitation for the use of many cancer therapeutics is the development of life-threatening cardiovascular toxicities, with significant consequences for treatment response and the patient's quality of life. The aim of this review is to outline VDAs as a cancer therapeutic approach and define the mechanistic basis of cardiovascular toxicities of current chemotherapeutics, with the overall objective of discussing whether VDA combinations with specific chemotherapeutic classes would be good or bad in terms of cardiovascular toxicity.
Collapse
Affiliation(s)
- Jason H Gill
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK; School of Pharmacy, Faculty of Medical Sciences, Newcastle University, UK.
| | - Kimberly L Rockley
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| | - Carol De Santis
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| | - Asma K Mohamed
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| |
Collapse
|
26
|
Abdelgawad IY, Grant MKO, Zordoky BN. Leveraging the Cardio-Protective and Anticancer Properties of Resveratrol in Cardio-Oncology. Nutrients 2019; 11:nu11030627. [PMID: 30875799 PMCID: PMC6471701 DOI: 10.3390/nu11030627] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 12/25/2022] Open
Abstract
Cardio-oncology is a clinical/scientific discipline which aims to prevent and/or treat cardiovascular diseases in cancer patients. Although a large number of cancer treatments are known to cause cardiovascular toxicity, they are still widely used because they are highly effective. Unfortunately, therapeutic interventions to prevent and/or treat cancer treatment-induced cardiovascular toxicity have not been established yet. A major challenge for such interventions is to protect the cardiovascular system without compromising the therapeutic benefit of anticancer medications. Intriguingly, the polyphenolic natural compound resveratrol and its analogs have been shown in preclinical studies to protect against cancer treatment-induced cardiovascular toxicity. They have also been shown to possess significant anticancer properties on their own, and to enhance the anticancer effect of other cancer treatments. Thus, they hold significant promise to protect the cardiovascular system and fight the cancer at the same time. In this review, we will discuss the current knowledge regarding the cardio-protective and the anticancer properties of resveratrol and its analogs. Thereafter, we will discuss the challenges that face the clinical application of these agents. To conclude, we will highlight important gaps of knowledge and future research directions to accelerate the translation of these exciting preclinical findings to cancer patient care.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Marianne K O Grant
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
27
|
Suissa M, LeLorier J. Pharmacologically pertinent period of effect (PPPE). Pharmacoepidemiol Drug Saf 2019; 29 Suppl 1:61-67. [PMID: 30628139 PMCID: PMC6972608 DOI: 10.1002/pds.4714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 10/17/2018] [Accepted: 11/08/2018] [Indexed: 01/03/2023]
Abstract
Background The period of time during which a patient is exposed to a drug does not necessarily correspond to the period during which the drug produces the adverse effect under consideration. We propose the term Pharmacologically pertinent period of effect (PPPE) to address this time window. We explored the PPPE in light of the rofecoxib saga. Methods We identified the observational database studies of rofecoxib at doses 25 and 50 mg daily and thromboembolic events. We also obtained the Kaplan‐Meier curves of Vioxx Gastrointestinal Outcomes Research trial (VIGOR) and Adenomatous Polyp Prevention on Vioxx (APPROVE) trials. Results We found seven observational studies with nine analyses. All the studies only looked at current exposure. At the dose of 25 mg, only three of nine analyses were barely statistically significant. At the dose of 50 mg, the risk ratios were much higher. The visual inspection of the Kaplan‐Meier curves shows that in the APPROVE trial (25 mg), the placebo and rofecoxib curves start separating to become statistically significantly different only after 36 months. In contrast the VIGOR (50 mg), curves start separating very early and the divergence increases after 8 months. Discussion The 50 mg observational studies, looking at current exposure, correctively identified the almost immediate increase in risk evident in the VIGOR Kaplan‐Meier curves. The absence of an immediate increase in risk shown by the APPROVE trial was also correctively identified by most observational 25 mg studies. To our knowledge no observational study was done on the long‐term cardiac toxicity of the 25‐mg dose. It would thus appear that the two doses of rofecoxib have different PPPEs.
Collapse
Affiliation(s)
- Melanie Suissa
- Medical Student, Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Jacques LeLorier
- Medical Student, Department of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
28
|
Gomhor J Alqaraghuli H, Kashanian S, Rafipour R, Mahdavian E, Mansouri K. Development and characterization of folic acid-functionalized apoferritin as a delivery vehicle for epirubicin against MCF-7 breast cancer cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S847-S854. [PMID: 30449179 DOI: 10.1080/21691401.2018.1516671] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epirubicin (Epr) is an effective chemotherapeutic drug; however, the clinical amenability of Epr is limited by its highly toxic interaction with normal cells. This toxicity can be decreased by utilizing nanocarriers and targeted drug delivery systems. This work describes an approach for the delivery of Epr via encapsulation in the horse spleen apoferritin (HsAFr) cavity. The encapsulation was achieved by the disassembling of apoferritin into subunits at pH 2 followed by its reformation at pH 7.4 in the presence of Epr. The surface of HsAFr-encapsulated Epr was modified with folic acid (FA) for optimal targeting of breast cancer cells (MCF-7). The use of FA to functionalize HsAFr could enhance the cellular uptake efficiency via FA-receptor-mediated endocytosis. UV-vis spectroscopy, fluorescence spectroscopy, circular dichroism (CD) and transmission electron microscopy (TEM) were utilized for structural characterization of the HsAFr-Epr and HsAFr-Epr-FA complexes. The comparison of the anti-cancer activities across the HsAFr-Epr-FA complex and the free Epr drug was performed using the MTT viability assay on MCF-7.
Collapse
Affiliation(s)
- Hasanain Gomhor J Alqaraghuli
- a Department of Applied Chemistry, Faculty of Chemistry , Razi University , Kermanshah , Iran.,b Department of General Sciences, College of Basic Education , Al-Muthanna University , Al-Muthanna , Iraq
| | - Soheila Kashanian
- c Faculty of Chemistry , Sensor and Biosensor Research Center (SBRC) & Nanoscience and Nanotechnology Research Center (NNRC), Razi University , Kermanshah , Iran.,d Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences , Kermanshah, Iran
| | - Ronak Rafipour
- e Department of Chemistry , Kermanshah Branch, Islamic Azad University , Kermanshah , Iran
| | - Elahe Mahdavian
- f Department of Chemistry and Physics , Louisiana State University in Shreveport , Shreveport , LA , USA
| | - Kamran Mansouri
- g Medical Biology Research Center, Kermanshah University of Medical Sciences , Kermanshah , Iran
| |
Collapse
|
29
|
Barel G, Herwig R. Network and Pathway Analysis of Toxicogenomics Data. Front Genet 2018; 9:484. [PMID: 30405693 PMCID: PMC6204403 DOI: 10.3389/fgene.2018.00484] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Toxicogenomics is the study of the molecular effects of chemical, biological and physical agents in biological systems, with the aim of elucidating toxicological mechanisms, building predictive models and improving diagnostics. The vast majority of toxicogenomics data has been generated at the transcriptome level, including RNA-seq and microarrays, and large quantities of drug-treatment data have been made publicly available through databases and repositories. Besides the identification of differentially expressed genes (DEGs) from case-control studies or drug treatment time series studies, bioinformatics methods have emerged that infer gene expression data at the molecular network and pathway level in order to reveal mechanistic information. In this work we describe different resources and tools that have been developed by us and others that relate gene expression measurements with known pathway information such as over-representation and gene set enrichment analyses. Furthermore, we highlight approaches that integrate gene expression data with molecular interaction networks in order to derive network modules related to drug toxicity. We describe the two main parts of the approach, i.e., the construction of a suitable molecular interaction network as well as the conduction of network propagation of the experimental data through the interaction network. In all cases we apply methods and tools to publicly available rat in vivo data on anthracyclines, an important class of anti-cancer drugs that are known to induce severe cardiotoxicity in patients. We report the results and functional implications achieved for four anthracyclines (doxorubicin, epirubicin, idarubicin, and daunorubicin) and compare the information content inherent in the different computational approaches.
Collapse
Affiliation(s)
| | - Ralf Herwig
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
30
|
CPP-Ts: a new intracellular calcium channel modulator and a promising tool for drug delivery in cancer cells. Sci Rep 2018; 8:14739. [PMID: 30282983 PMCID: PMC6170434 DOI: 10.1038/s41598-018-33133-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022] Open
Abstract
Scorpion sting envenoming impacts millions of people worldwide, with cardiac effects being one of the main causes of death on victims. Here we describe the first Ca2+ channel toxin present in Tityus serrulatus (Ts) venom, a cell penetrating peptide (CPP) named CPP-Ts. We show that CPP-Ts increases intracellular Ca2+ release through the activation of nuclear InsP3R of cardiomyocytes, thereby causing an increase in the contraction frequency of these cells. Besides proposing a novel subfamily of Ca2+ active toxins, we investigated its potential use as a drug delivery system targeting cancer cell nucleus using CPP-Ts’s nuclear-targeting property. To this end, we prepared a synthetic CPP-Ts sub peptide14–39 lacking pharmacological activity which was directed to the nucleus of specific cancer cell lines. This research identifies a novel subfamily of Ca2+ active toxins and provides new insights into biotechnological applications of animal venoms.
Collapse
|
31
|
Using Zebrafish for Investigating the Molecular Mechanisms of Drug-Induced Cardiotoxicity. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1642684. [PMID: 30363733 PMCID: PMC6180974 DOI: 10.1155/2018/1642684] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/31/2018] [Accepted: 08/18/2018] [Indexed: 01/09/2023]
Abstract
Over the last decade, the zebrafish (Danio rerio) has emerged as a model organism for cardiovascular research. Zebrafish have several advantages over mammalian models. For instance, the experimental cost of using zebrafish is comparatively low; the embryos are transparent, develop externally, and have high fecundity making them suitable for large-scale genetic screening. More recently, zebrafish embryos have been used for the screening of a variety of toxic agents, particularly for cardiotoxicity testing. Zebrafish has been shown to exhibit physiological responses that are similar to mammals after exposure to medicinal drugs including xenobiotics, hormones, cancer drugs, and also environmental pollutants, including pesticides and heavy metals. In this review, we provided a summary for recent studies that have used zebrafish to investigate the molecular mechanisms of drug-induced cardiotoxicity. More specifically, we focused on the techniques that were exploited by us and others for cardiovascular toxicity assessment and described several microscopic imaging and analysis protocols that are being used for the estimation of a variety of cardiac hemodynamic parameters.
Collapse
|
32
|
Xie X, Tang F, Liu G, Li Y, Su X, Jiao X, Wang X, Tang B. Mitochondrial Peroxynitrite Mediation of Anthracycline-Induced Cardiotoxicity as Visualized by a Two-Photon Near-Infrared Fluorescent Probe. Anal Chem 2018; 90:11629-11635. [DOI: 10.1021/acs.analchem.8b03207] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Xilei Xie
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, PR China
| | - Fuyan Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, PR China
| | - Guangzhao Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, PR China
| | - Yong Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, PR China
| | - Xingxing Su
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, PR China
| | - Xiaoyun Jiao
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, PR China
| | - Xu Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, PR China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, PR China
| |
Collapse
|
33
|
Soliman AF, Anees LM, Ibrahim DM. Cardioprotective effect of zingerone against oxidative stress, inflammation, and apoptosis induced by cisplatin or gamma radiation in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:819-832. [PMID: 29736620 DOI: 10.1007/s00210-018-1506-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/17/2018] [Indexed: 01/24/2023]
Abstract
Despite their clinical benefits in cancer treatment, the deleterious effects on heart following chemo/radiotherapy are of increasing importance. Zingerone, a natural polyphenol, possesses multiple biological activities, such as antioxidant and anti-inflammatory. Thus, the current study was designed to assess the potential cardioprotective effects of zingerone against cisplatin or γ-radiation. Zingerone was given by intragastric intubation (25 mg/kg) daily for three successive weeks prior to the induction of cardiotoxicity using a single dose of cisplatin (20 mg/kg, i.p.) or a whole body γ-irradiation at a single dose of 6 Gy. Zingerone pre-treatment significantly reduced the abnormalities in heart histology and the increase in the cardiotoxicity indices, serum lactate dehydrogenase, and creatine kinase-MB activities, as well as plasma cardiac troponin T and B-natriuretic peptide, induced by cisplatin or γ-radiation. Further, zingerone, except for superoxide dismutase, notably ameliorated the state of oxidative stress as evidenced by a significant decrease in malondialdehyde level accompanied with a significant increase in the reduced glutathione content and catalase activity. Additionally, zingerone mitigated the increase in the inflammatory markers including serum level of tumor necrosis factor-alpha, cardiac myeloperoxidase activity, and cyclooxygenase-2 protein expression. Moreover, zingerone alleviated the elevation of caspase-3 gene expression and the prominent nuclear DNA fragmentation and attenuated the decrease in mitochondrial complexes' activities. This study sheds the light on a probable protective role of zingerone as an antioxidant, anti-inflammatory, and antiapoptotic agent against cisplatin- or γ-radiation-induced cardiotoxicity and holds a potential in regard to therapeutic intervention for chemo/radiotherapy mediated cardiac damage.
Collapse
Affiliation(s)
- Ahmed F Soliman
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Lobna M Anees
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Doaa M Ibrahim
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
34
|
Fidale TM, Antunes HKM, Alex dos Santos L, Rodrigues de Souza F, Deconte SR, Borges Rosa de Moura F, Mantovani MM, Alves Duarte PR, Roever L, Resende ES. Increased Dietary Leucine Reduces Doxorubicin-Associated Cardiac Dysfunction in Rats. Front Physiol 2018; 8:1042. [PMID: 29403386 PMCID: PMC5779071 DOI: 10.3389/fphys.2017.01042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 11/29/2017] [Indexed: 01/01/2023] Open
Abstract
Cardiotoxicity is one of the most significant adverse effects of the oncologic treatment with doxorubicin, which is responsible for a substantial morbid and mortality. The occurrence of heart failure with ventricular dysfunction may lead to severe cardiomyopathy and ultimately to death. Studies have focused on the effects of leucine supplementation as a strategy to minimize or revert the clinical condition of induced proteolysis by several clinical onsets. However, the impact of leucine supplementation in heart failure induced by doxorubicin is unknown. Therefore, the objective of this work is to evaluate the effects of leucine supplementation on the cardiotoxicity in the heart of rats treated with doxorubicin. Rats treated with a 7.5 mg/kg cumulative dose of doxorubicin for 14 days presented a dilatation of the left ventricle (LV), and a reduction of the ejection fraction (FE). The 5% supplementation of leucine in the rats' food prevented the malfunctioning of the LV when administered with doxorubicin. Some alterations in the extracellular matrix remodeling were confirmed by the increase of collagen fibers in the doxorubicin group, which did not increase when the treatment was associated with leucine supplementation. Leucine attenuates heart failure in this experimental model with doxorubicin. Such protection is followed by the maintenance of interstitial collagen fibers.
Collapse
Affiliation(s)
- Thiago M. Fidale
- Laboratory of Experimental Medicine, Federal University of Uberlândia, Uberlândia, Brazil
| | - Hanna K. M. Antunes
- Department of Bioscience, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Simone R. Deconte
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | | | | | | | - Leonardo Roever
- Laboratory of Experimental Medicine, Federal University of Uberlândia, Uberlândia, Brazil
| | - Elmiro S. Resende
- Laboratory of Experimental Medicine, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
35
|
Tang XM, Chen H, Li Q, Song Y, Zhang S, Xu XS, Xu Y, Chen S. Assessment of the cardiac safety between cetuximab and panitumumab as single therapy in Chinese chemotherapy-refractory mCRC. Onco Targets Ther 2017; 11:123-129. [PMID: 29343971 PMCID: PMC5749385 DOI: 10.2147/ott.s149716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE The cardiac safety of cetuximab and panitumumab, particularly as single agents, has not been investigated extensively. This trial was designed to specifically evaluate the cardiac safety of cetuximab and panitumumab as single therapy in Chinese chemotherapy-refractory metastatic colorectal cancer (mCRC) patients. PATIENTS AND METHODS Sixty-one patients received cetuximab at an initial dose of 400 mg/m2 intravenously over 120 minutes on day 1 (week 1), followed by a maintenance dose of 250 mg/m2 intravenously over 60 minutes on day 1 of each 7-day cycle. Forty-three patients received panitumumab at a dose of 6 mg/kg intravenously every 14 days. Routine laboratory tests and electrocardiogram (ECG) were performed at baseline, during therapy and after the treatment (4th and 10th months). The incidence of elevation of troponin I ultra (TNI Ultra), abnormal ECGs, cardiac events and noncardiac adverse events (AEs) were recorded and analyzed. RESULTS The incidence of elevation of TNI Ultra between the two groups had no significance (p=0.681), and TNI Ultra+ was observed more frequently in patients with metastases to more than three organs and they received fourth or above lines of chemotherapy. The most frequent abnormal ECG manifestations were nonspecific ST changes and QTc prolongation in the two groups. At 10 months after treatment, most of the abnormal ECG manifestations were reversed. The most common cardiac AEs of cetuximab and panitumumab included palpitations, dyspnea, chest pain and arrhythmias requiring treatment. Most of the events were mild and transient. The incidence of cardiac AEs had no significant difference between the two groups. Rash was still the most common noncardiac AE in both groups. CONCLUSION Cetuximab and panitumumab showed favorable cardiac safety as single agents for Chinese chemotherapy-refractory mCRC patients. But monitoring for cardiac AEs is still necessary throughout the entire treatment process.
Collapse
Affiliation(s)
- Xue-miao Tang
- Department of Ultrasound and Electrocardiogram
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
| | - Hao Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou
| | - Qing Li
- Department of Ultrasound and Electrocardiogram
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
| | - Yiling Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou
| | - Shuping Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou
| | - Xiao-Shuan Xu
- School of Laboratory Medicine, Guangdong Medical University
| | - Yiwei Xu
- Department of Clinical Laboratory, The Cancer Hospital of Shantou University Medical College
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Guangdong, China
| | - Shulin Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou
| |
Collapse
|
36
|
Xu C, Zhang C, Wang Y, Li L, Li L, Whittaker AK. Controllable synthesis of a novel magnetic core-shell nanoparticle for dual-modal imaging and pH-responsive drug delivery. NANOTECHNOLOGY 2017; 28:495101. [PMID: 29019341 DOI: 10.1088/1361-6528/aa929b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In this study, novel magnetic core-shell nanoparticles Fe3O4@La-BTC/GO have been synthesized by the layer-by-layer self-assembly (LBL) method and further modified by attachment of amino-modified PEG chains. The nanoparticles were thoroughly characterized by x-ray diffraction, FTIR, scanning electron microscopy and transmission electron microscopy. The core-shell structure was shown to be controlled by the LBL method. The drug loading of doxorubicin (DOX) within the Fe3O4@La-BTC/GO-PEG nanoparticles with different numbers of deposited layers was investigated. It was found that DOX loading increased with increasing number of metal organic framework coating layers, indicating that the drug loading can be controlled through the controllable LBL method. Cytotoxicity assays indicated that the Fe3O4@La-BTC/GO-PEG nanoparticles were biocompatible. The DOX was released rapidly at pH 3.8 and pH 5.8, but at pH 7.4 the rate and extent of release was greatly attenuated. The nanoparticles therefore demonstrate an excellent pH-triggered drug release. In addition, the particles could be tracked by magnetic resonance imaging (MRI) and fluorescence optical imaging (FOI). A clear dose-dependent contrast enhancement in T 2-weighted MR images and fluorescence images indicate the potential of these nanoparticles as dual-mode MRI/FOI contrast agents.
Collapse
Affiliation(s)
- Chen Xu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Hubei University 430062, People's Republic of China. Ministry-of-Education Key Laboratory for the Synthesis and Application of Organic Function Molecules, Hubei University, Xueyuan Road 11#, Wuchang, Wuhan City, Hubei Province 430062, People's Republic of China
| | | | | | | | | | | |
Collapse
|
37
|
Mostarda C, Castro-Filha J, Reis AD, Sevílio M, Dias CJ, Silva-Filho AC, Garcia JBS, do Desterro Nascimento M, Coelho-Junior HJ, Rodrigues B. Short-term combined exercise training improves cardiorespiratory fitness and autonomic modulation in cancer patients receiving adjuvant therapy. J Exerc Rehabil 2017; 13:599-607. [PMID: 29114536 PMCID: PMC5667608 DOI: 10.12965/jer.1735048.524] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/16/2017] [Indexed: 01/15/2023] Open
Abstract
The present study aimed to investigate the impact of a short-term exercise training (ET) on the cardiorespiratory fitness and autonomic modulation of women with breast cancer who were receiving adjuvant radiotherapy, chemotherapy or hormonotherapy. Eighteen women previously diagnosed with breast cancer receiving adjuvant radiotherapy, chemotherapy or hormone therapy were randomly allocated into breast cancer nonexercise (BC) and exercise groups (BC+Ex). Moreover, nine healthy physically inactive volunteers were recruited to compose the noncancer control group (CG). The BC+Ex group was underwent to a combined ET program, which was based on resistance, aerobic and flexibility exercises. ET was performed 3 times a week, on nonconsecutive days, for 4 weeks at the hospital room under the professional supervision. In turn, BC and CG remained without be engaged in physical exercise programs. Volunteers were evaluated regarding their cardiorespiratory fitness and autonomic modulation (i.e., time, frequency domains, and nonlinear [symbolic analysis]) before and after the end of the ET program. A priori, data indicate that women patients with breast cancer showed impaired exercise tolerance, as well as autonomic dysfunction in comparison with age-matched healthy control subjects. However, a 1-month combined ET program could reverse such impairments, so that after the intervention, BC+Ex and CG showing similar results in the cardiorespiratory test and heart rate variability analysis. In conclusion, data of the current study indicate that 1 month of ET is able to reverse impaired cardiorespiratory fitness and autonomic modulation in women with breast cancer receiving adjuvant therapy.
Collapse
Affiliation(s)
- Cristiano Mostarda
- Physical Education Department, Universidade Federal do Maranhão (UFMA), São Luis, Brazil.,Laboratory of Cardiovascular Adaptations to Exercise (LACORE), São Luis, Brazil
| | - Jurema Castro-Filha
- Physical Education Department, Universidade Federal do Maranhão (UFMA), São Luis, Brazil
| | - Andréa Dias Reis
- Physical Education Department, Universidade Federal do Maranhão (UFMA), São Luis, Brazil
| | - Mário Sevílio
- Physical Education Department, Universidade Federal do Maranhão (UFMA), São Luis, Brazil
| | - Carlos José Dias
- Physical Education Department, Universidade Federal do Maranhão (UFMA), São Luis, Brazil.,Laboratory of Cardiovascular Adaptations to Exercise (LACORE), São Luis, Brazil
| | - Antonio Carlos Silva-Filho
- Physical Education Department, Universidade Federal do Maranhão (UFMA), São Luis, Brazil.,Laboratory of Cardiovascular Adaptations to Exercise (LACORE), São Luis, Brazil
| | | | | | | | - Bruno Rodrigues
- Faculty of Physical Education, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
38
|
Polonchuk L, Chabria M, Badi L, Hoflack JC, Figtree G, Davies MJ, Gentile C. Cardiac spheroids as promising in vitro models to study the human heart microenvironment. Sci Rep 2017; 7:7005. [PMID: 28765558 PMCID: PMC5539326 DOI: 10.1038/s41598-017-06385-8] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/26/2017] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional in vitro cell systems are a promising alternative to animals to study cardiac biology and disease. We have generated three-dimensional in vitro models of the human heart ("cardiac spheroids", CSs) by co-culturing human primary or iPSC-derived cardiomyocytes, endothelial cells and fibroblasts at ratios approximating those present in vivo. The cellular organisation, extracellular matrix and microvascular network mimic human heart tissue. These spheroids have been employed to investigate the dose-limiting cardiotoxicity of the common anti-cancer drug doxorubicin. Viability/cytotoxicity assays indicate dose-dependent cytotoxic effects, which are inhibited by the nitric oxide synthase (NOS) inhibitor L-NIO, and genetic inhibition of endothelial NOS, implicating peroxynitrous acid as a key damaging agent. These data indicate that CSs mimic important features of human heart morphology, biochemistry and pharmacology in vitro, offering a promising alternative to animals and standard cell cultures with regard to mechanistic insights and prediction of toxic effects in human heart tissue.
Collapse
Affiliation(s)
- Liudmila Polonchuk
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Mamta Chabria
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Laura Badi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Jean-Christophe Hoflack
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Gemma Figtree
- Sydney Medical School, University of Sydney, Sydney, 2000, Australia
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Carmine Gentile
- Sydney Medical School, University of Sydney, Sydney, 2000, Australia.
- Heart Research Institute, Newtown, 2041, Australia.
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| |
Collapse
|
39
|
Comparision of doxorubicin-induced cardiotoxicity in the ICR mice of different sources. Lab Anim Res 2017; 33:165-170. [PMID: 28747983 PMCID: PMC5527143 DOI: 10.5625/lar.2017.33.2.165] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 01/05/2023] Open
Abstract
Doxorubicin is a widely used chemotherapeutic agents and is now part of standard therapeutic regimens for a variety of cancers (eg, hematopoietic malignancies and advanced solid tumors of the breast, ovary, thyroid, and bone). However, a potentially lethal and dose-dependent cardiotoxicity that appears within a short time after treatment limits the usage of doxorubicin in cancer patients. Although the mechanism of doxorubicin-induced cardiotoxicity is not completely understood, it is thought that free radical-induced oxidative stress and excessive production of reactive oxygen species are primary drivers of its toxicity. In this study, we compared the doxorubicin-induced cardiotoxicity of ICR mice obtained from three different sources and evaluated the utility of Korl:ICR stock established by the Korean FDA. Because doxorubicin-induced cardiotoxicity is thought to involve the excessive generation of ROS followed by oxidative stress, we determined the representative tissue index of oxidation, lipid peroxidation, and antioxidant, glutathione (GSH), as well as the parameters of heart injury. Doxorubicin treatment successfully induced cardiotoxicity as evidenced by histological examination and serum parameters (eg, levels of LDH and CK activities) in ICR mice. It was accompanied by increased lipid peroxidation and a decrease in both cysteine and GSH, further supporting previous reports that oxidative stress is a potential mechanism of doxorubicin-induced cardiotoxicity. Of interest, we did not observe a significant difference in doxorubicin-induced cardiotoxicity among mice of different origins. Collectively, our results suggest that Korl:ICR strain may be useful in the research of doxorubicin-induced cardiotoxicity.
Collapse
|
40
|
A Functional Iron Oxide Nanoparticles Modified with PLA-PEG-DG as Tumor-Targeted MRI Contrast Agent. Pharm Res 2017; 34:1683-1692. [PMID: 28608138 DOI: 10.1007/s11095-017-2165-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/25/2017] [Indexed: 12/20/2022]
Abstract
PURPOSE Tumor targeting could greatly promote the performance of magnetic nanomaterials as MRI (Magnetic Resonance Imaging) agent for tumor diagnosis. Herein, we reported a novel magnetic nanoparticle modified with PLA (poly lactic acid)-PEG (polyethylene glycol)-DG (D-glucosamine) as Tumor-targeted MRI Contrast Agent. METHODS In this work, we took use of the D-glucose passive targeting on tumor cells, combining it on PLA-PEG through amide reaction, and then wrapped the PLA-PEG-DG up to the Fe3O4@OA NPs. The stability and anti phagocytosis of Fe3O4@OA@PLA-PEG-DG was tested in vitro; the MRI efficiency and toxicity was also detected in vivo. RESULTS These functional magnetic nanoparticles demonstrated good biocompatibility and stability both in vitro and in vivo. Cell experiments showed that Fe3O4@OA@PLA-PEG-DG nanoparticles exist good anti phagocytosis and high targetability. In vivo MRI images showed that the contrast effect of Fe3O4@OA@PLA-PEG-DG nanoparticles prevailed over the commercial non tumor-targeting magnetic nanomaterials MRI agent at a relatively low dose. CONCLUSIONS The DG can validly enhance the tumor-targetting effect of Fe3O4@OA@PLA-PEG nanoparticle. Maybe MRI agents with DG can hold promise as tumor-targetting development in the future.
Collapse
|
41
|
Dokmanovic M, King KE, Mohan N, Endo Y, Wu WJ. Cardiotoxicity of ErbB2-targeted therapies and its impact on drug development, a spotlight on trastuzumab. Expert Opin Drug Metab Toxicol 2017; 13:755-766. [PMID: 28571477 DOI: 10.1080/17425255.2017.1337746] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Trastuzumab, a therapeutic monoclonal antibody directed against ErbB2, is often noted as a successful example of targeted therapy. Trastuzumab improved outcomes for many patients with ErbB2-positive breast and gastric cancers, however, cardiac side effects [e.g., left ventricular dysfunction and congestive heart failure (CHF)] were reported in the early phase clinical studies. This finding, subsequently corroborated by multiple clinical studies, raised concerns that the observed cardiotoxicity induced by trastuzumab might adversely impact the clinical development of other therapeutics targeting ErbB family members. Areas covered: In this review we summarize both basic research and clinical findings regarding trastuzumab-induced cardiotoxicity and assess if there has been an impact of trastuzumab-induced cardiotoxicity on the development of other agents targeting ErbB family members. Expert opinion: There are a number of scientific gaps that are critically important to address for the continued success of HER2-targeted agents. These include: 1) elucidating the molecular mechanisms contributing to cardiotoxicity; 2) developing relevant preclinical testing systems for predicting cardiotoxicity; 3) developing clinical strategies to identify patients at risk of cardiotoxicity; and 4) enhancing management of clinical symptoms of cardiotoxicity.
Collapse
Affiliation(s)
- Milos Dokmanovic
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| | - Kathryn E King
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| | - Nishant Mohan
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| | - Yukinori Endo
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| | - Wen Jin Wu
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| |
Collapse
|
42
|
Smith JM, Flexner C. The challenge of polypharmacy in an aging population and implications for future antiretroviral therapy development. AIDS 2017; 31 Suppl 2:S173-S184. [PMID: 28471948 DOI: 10.1097/qad.0000000000001401] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
: It is estimated that by 2030 nearly three-quarters of persons living with HIV will be 50 years and older. The aging HIV population presents a new clinical concern for HIV providers: adverse effects from polypharmacy. An aging population means more comorbidities and potentially more drug-drug interactions for providers to manage. This review discusses major comorbidities including cardiovascular disease, anticoagulation, hypertension, diabetes mellitus and malignancy and considerations for drug-interactions with antiretrovirals.
Collapse
|
43
|
Cuni R, Parrini I, Asteggiano R, Conte MR. Targeted Cancer Therapies and QT Interval Prolongation: Unveiling the Mechanisms Underlying Arrhythmic Complications and the Need for Risk Stratification Strategies. Clin Drug Investig 2017; 37:121-134. [PMID: 27638052 DOI: 10.1007/s40261-016-0460-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The care and treatment of cancer patients has significantly changed in the last decade with a remarkable shift towards novel targeted therapies. These promising new drugs may represent effective and potentially life-saving therapeutic options in cancer patients, but are also emerging in the cardiotoxicity scenario for their arrhythmogenic potential due to their QT-prolonging activity. In this article we review the mechanisms underlying drug-induced QT interval prolongation and the classes of anticancer-targeted therapies most frequently responsible for this adverse event, with a particular focus on tyrosine kinase-targeting molecules. Since up to 49 % of serious adverse drug reactions (ADRs) and 58 % of potentially fatal ADRs may not appear on initial drug safety labels, we also review and discuss data from the post-marketing VigiBase® safety reporting system, the World Health Organization's global database of ADRs. Finally, we discuss arrhythmic risk stratification and prevention strategies in the complex multiple-risk setting of cancer patients, paying particular attention to drug-drug interactions with common antimicrobial, psychotropic and antiemetic supportive care, and we also provide an electrocardiographic QT monitoring algorithm for patients who are candidates for targeted cancer therapies.
Collapse
Affiliation(s)
- Rezarta Cuni
- Department of Cardiology, Azienda Ospedaliera Ordine Mauriziano, Largo Filippo Turati nr. 62, 10128, Turin, Italy.
| | - Iris Parrini
- Department of Cardiology, Azienda Ospedaliera Ordine Mauriziano, Largo Filippo Turati nr. 62, 10128, Turin, Italy
| | - Riccardo Asteggiano
- Azienda Sanitaria Locale Torino 2 and Torino 3, Out of Hospital Cardiology Service, Turin, Italy
| | - Maria Rosa Conte
- Department of Cardiology, Azienda Ospedaliera Ordine Mauriziano, Largo Filippo Turati nr. 62, 10128, Turin, Italy
| |
Collapse
|
44
|
Yoon HJ, Kim KH, Kim JY, Park HJ, Cho JY, Hong YJ, Park HW, Kim JH, Ahn Y, Jeong MH, Cho JG, Park JC. Chemotherapy-Induced Left Ventricular Dysfunction in Patients with Breast Cancer. J Breast Cancer 2016; 19:402-409. [PMID: 28053628 PMCID: PMC5204046 DOI: 10.4048/jbc.2016.19.4.402] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 09/02/2016] [Indexed: 01/03/2023] Open
Abstract
Purpose As the numbers of cancer cases and survivors increase, the incidence and natural history of chemotherapy-induced cardiotoxicities in patients with breast cancer may also be expected to change. The present study aimed to investigate the incidence and predictors of chemotherapy-induced left ventricular dysfunction (LVD) in patients with breast cancer. Methods From 2003 to 2010, 712 female patients with breast cancer (55.7±10.7 years) were enrolled and divided into the LVD group (n=82, 56.7±10.1 years) and the non-LVD group (n=630, 55.6±10.8 years). Baseline clinical and treatment-related variables were compared. Results Chemotherapy-induced LVD developed in 82 cases (11.4%). Low body mass index (BMI), low triglyceride level, advanced cancer stage, and the use of doxorubicin, paclitaxel, trastuzumab, or radiotherapy were significant predictors of LVD in a univariate analysis. In a multivariate analysis, low BMI, advanced cancer stage, and the use of target therapy with trastuzumab were independent predictors of chemotherapy-induced LVD. Chemotherapy-induced LVD was recovered in 53 patients (64.6%), but left ventricular function was not recovered in 29 patients (35.4%). Conclusion Chemotherapy-induced LVD was not uncommon and did not reduce in many of our patients with breast cancer. Low BMI, advanced cancer stage, and the use of trastuzumab were independent predictors of chemotherapy-induced LVD in patients with breast cancer. The development of chemotherapy-induced LVD should be carefully monitored in patients with breast cancer who are receiving trastuzumab therapy, have poor nutritional status, and advanced cancer stage.
Collapse
Affiliation(s)
- Hyun Ju Yoon
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Kye Hun Kim
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Jong Yoon Kim
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Hyuk Jin Park
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Jae Yeong Cho
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Young Joon Hong
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Hyung Wook Park
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Ju Han Kim
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Youngkeun Ahn
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Myung Ho Jeong
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Jeong Gwan Cho
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| | - Jong Chun Park
- Department of Cardiovascular Medicine, Chonnam National University Hospital, and Research Institute of Medical Science, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
45
|
Changes in plasma hormones and heart rate variability in patients receiving the cardiotoxic anti-cancer agent bevacizumab. Int J Cardiol 2016; 219:25-6. [PMID: 27257852 DOI: 10.1016/j.ijcard.2016.05.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/12/2016] [Indexed: 11/22/2022]
|
46
|
Razavi-Azarkhiavi K, Iranshahy M, Sahebkar A, Shirani K, Karimi G. The Protective Role of Phenolic Compounds Against Doxorubicin-induced Cardiotoxicity: A Comprehensive Review. Nutr Cancer 2016; 68:892-917. [PMID: 27341037 DOI: 10.1080/01635581.2016.1187280] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although doxorubicin (DOX) is among the most widely used anticancer agents, its clinical application is hampered owing to its cardiotoxicity. Adjuvant therapy with an antioxidant has been suggested as a promising strategy to reduce DOX-induced adverse effects. In this context, many phenolic compounds have been reported to protect against DOX-induced cardiotoxicity. The cardioprotective effects of phenolic compounds are exerted via multiple mechanisms including inhibition of reactive oxygen species generation, apoptosis, NF-κB, p53, mitochondrial dysfunction, and DNA damage. In this review, we present a summary of the in vitro, in vivo, and clinical findings on the protective mechanisms of phenolic compounds against DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Kamal Razavi-Azarkhiavi
- a Department of Pharmacodynamy and Toxicology , Faculty of Pharmacy, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Milad Iranshahy
- b Biotechnology Research Center and School of Pharmacy, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Amirhossein Sahebkar
- c Biotechnology Research Center, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Kobra Shirani
- d Department of Pharmacodynamy and Toxicology , Faculty of Pharmacy, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Gholamreza Karimi
- e Department of Pharmacodynamy and Toxicology , Faculty of Pharmacy, Mashhad University of Medical Sciences , Mashhad , Iran.,f Pharmaceutical Research Center and Pharmacy School, Mashhad University of Medical Sciences
| |
Collapse
|
47
|
Cardiotoxicity in anthracycline therapy: Prevention strategies. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2016. [DOI: 10.1016/j.repce.2015.12.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
48
|
Cruz M, Duarte-Rodrigues J, Campelo M. Cardiotoxicity in anthracycline therapy: Prevention strategies. Rev Port Cardiol 2016; 35:359-71. [PMID: 27255173 DOI: 10.1016/j.repc.2015.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 12/03/2015] [Accepted: 12/20/2015] [Indexed: 11/18/2022] Open
Abstract
The increasing use of anthracyclines, together with the longer survival of cancer patients, means the toxic effects of these drugs need to be monitored. In order to detect, prevent or mitigate anthracycline-induced cardiomyopathy, it is essential that all patients undergo a rigorous initial cardiovascular assessment, followed by close monitoring. Several clinical trials have shown the cardioprotective effect of non-pharmacological measures such as exercise, healthy lifestyles, control of risk factors and treatment of comorbidities; a cardioprotective effect has also been observed with pharmacological measures such as beta-blockers, angiotensin-converting enzyme inhibitors, angiotensin receptor antagonists, statins, dexrazoxane and liposomal formulations. However, there are currently no guidelines for managing prevention in these patients. In this review the authors discuss the state of the art of the assessment, monitoring, and, above all, the prevention of anthracycline-induced cardiotoxicity.
Collapse
Affiliation(s)
- Margarida Cruz
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | | | - Manuel Campelo
- Serviço de Cardiologia, Hospital de S. João, Porto, Portugal; Centro de Investigação em Tecnologias e Serviços de Saúde (CINTESIS), Faculdade de Medicina, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
49
|
Li S, Wang A, Gu F, Wang Z, Tian C, Qian Z, Tang L, Gu Y. Novel harmine derivatives for tumor targeted therapy. Oncotarget 2016; 6:8988-9001. [PMID: 25940702 PMCID: PMC4496197 DOI: 10.18632/oncotarget.3276] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 04/10/2015] [Indexed: 12/14/2022] Open
Abstract
Harmine is a beta-carboline alkaloid found in medicinal plant PeganumHarmala, which has served as a folk anticancer medicine. However, clinical applications of harmine were limited by its low pharmacological effects and noticeable neurotoxicity. In this study, we modified harmine to increase the therapeutic efficacy and to decrease the systemic toxicity. Specifically, two tumor targeting harmine derivatives 2DG-Har-01 and MET-Har-02 were synthesized by modifying substituent in position-2, -7 and -9 of harmine ring with two different targeting group2-amino-2-deoxy-D-glucose (2DG) and Methionine (Met), respectively. Their therapeutic efficacy and toxicity were investigated both in vitro and in vivo. Results suggested that the two newharmine derivatives displayed much higher therapeutic effects than non-modified harmine. In particular, MET-Har-02 was more potent than 2DG-Har-01 with promising potential for targeted cancer therapy.
Collapse
Affiliation(s)
- Siwen Li
- Department of Biomedical Engineering, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Arlington, TX, USA
| | - Aqin Wang
- Department of Biomedical Engineering, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Arlington, TX, USA
| | - Fan Gu
- Department of Biomedical Engineering, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Arlington, TX, USA
| | - Zhaohui Wang
- Department of Biomedical Engineering, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Arlington, TX, USA
| | - Caiping Tian
- Department of Biomedical Engineering, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Arlington, TX, USA
| | - Zhiyu Qian
- Department of Biomedical Engineering, School of Automation, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Liping Tang
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Yueqing Gu
- Department of Biomedical Engineering, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Arlington, TX, USA
| |
Collapse
|
50
|
Rosa GM, Gigli L, Tagliasacchi MI, Di Iorio C, Carbone F, Nencioni A, Montecucco F, Brunelli C. Update on cardiotoxicity of anti-cancer treatments. Eur J Clin Invest 2016; 46:264-284. [PMID: 26728634 DOI: 10.1111/eci.12589] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/30/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Anti-cancer treatments markedly improved the prognosis of patients, but unfortunately might be hampered by cardiotoxicity. Both symptomatic and asymptomatic clinical forms of heart failure have been reported, which may be reversible or irreversible. The aim of this review is to provide an overview of the antineoplastic agents associated with cardiac toxicity and of the available diagnostic techniques. METHODS AND METHODS This narrative review is based on material from MEDLINE and PUBMED up to November 2015. We looked at the terms antineoplastic drugs and cardiac toxicity in combination with echocardiography, troponins, cardiac magnetic resonance, and positron emission tomography. RESULTS Anthracyclines, monoclonal antibodies, fluoropyrimidines, taxanes, alkylating agents, vinka alkaloids were reported to induce different clinical manifestations of cardioxicity. Chest radiotherapy is also associated with various forms of cardiac damage, which are indistinguishable from those found in patients with heart disease of other aetiologies and that may even appear several years after administration. Among diagnostic techniques, echocardiography is a noninvasive, cost-effective, and widely available imaging tool. Nuclear imaging and cardiac magnetic resonance may be used but are not so widely available and are more difficult to perform. Finally, some biomarkers, such as troponins, may be used to evaluate cardiac damage, but establishing the optimal timing of troponin assessment remains unclear and defining the cut-off point for positivity is still an important goal. CONCLUSIONS Cardiotoxicity of anti-cancer treatments is associated with development of heart failure. Novel diagnostic tools might be relevant to early recognize irreversible forms cardiac diseases.
Collapse
Affiliation(s)
- Gian Marco Rosa
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Lorenzo Gigli
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Maria Isabella Tagliasacchi
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Cecilia Di Iorio
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Federico Carbone
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, Geneva University, Geneva, Switzerland
| | - Alessio Nencioni
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST, Istituto Nazionale per la Ricerca Sul Cancro, Genoa, Italy
| | - Fabrizio Montecucco
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, Geneva University, Geneva, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Claudio Brunelli
- Division of Cardiology, Department of Internal Medicine, University of Genoa - IRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| |
Collapse
|