Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 880
Filter
1.
J Nanobiotechnology ; 22(1): 464, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095755

ABSTRACT

BACKGROUND: Doxorubicin (DOX) is a first-line chemotherapeutic drug for various malignancies that causes cardiotoxicity. Plant-derived exosome-like nanovesicles (P-ELNs) are growing as novel therapeutic agents. Here, we investigated the protective effects in DOX cardiotoxicity of ELNs from Momordica charantia L. (MC-ELNs), a medicinal plant with antioxidant activity. RESULTS: We isolated MC-ELNs using ultracentrifugation and characterized them with canonical mammalian extracellular vesicles features. In vivo studies proved that MC-ELNs ameliorated DOX cardiotoxicity with enhanced cardiac function and myocardial structure. In vitro assays revealed that MC-ELNs promoted cell survival, diminished reactive oxygen species, and protected mitochondrial integrity in DOX-treated H9c2 cells. We found that DOX treatment decreased the protein level of p62 through ubiquitin-dependent degradation pathway in H9c2 and NRVM cells. However, MC-ELNs suppressed DOX-induced p62 ubiquitination degradation, and the recovered p62 bound with Keap1 promoting Nrf2 nuclear translocation and the expressions of downstream gene HO-1. Furthermore, both the knockdown of Nrf2 and the inhibition of p62-Keap1 interaction abrogated the cardioprotective effect of MC-ELNs. CONCLUSIONS: Our findings demonstrated the therapeutic beneficials of MC-ELNs via increasing p62 protein stability, shedding light on preventive approaches for DOX cardiotoxicity.


Subject(s)
Cardiotoxicity , Doxorubicin , Exosomes , Momordica charantia , NF-E2-Related Factor 2 , Animals , Cardiotoxicity/prevention & control , Cardiotoxicity/metabolism , Momordica charantia/chemistry , Exosomes/metabolism , Rats , NF-E2-Related Factor 2/metabolism , Cell Line , Kelch-Like ECH-Associated Protein 1/metabolism , Reactive Oxygen Species/metabolism , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Cell Survival/drug effects , Rats, Sprague-Dawley , Sequestosome-1 Protein/metabolism
2.
J Pharmacol Sci ; 156(2): 69-76, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39179336

ABSTRACT

Despite the widespread recognition of the global concern regarding the onset of cardiovascular diseases in a significant number of patients following cancer treatment, definitive strategies for prevention and treatment remain elusive. In this study, we established systems to evaluate the influence of anti-cancer drugs on the quality control of mitochondria, pivotal for energy metabolism, using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Osimertinib, an epidermal growth factor receptor tyrosine kinase inhibitor used for treatment in lung cancer, reportedly increases the risk of cardiovascular disease. However, its underlying mechanism is largely unknown. Here, we found that the treatment of hiPSC-CMs with osimertinib and doxorubicin, but not trastuzumab and cisplatin, revealed a concentration-dependent impairment of respiratory function accompanied by mitochondrial fission. We previously reported the significant role of sulfur metabolism in maintaining mitochondrial quality in the heart. Co-treatment with various inorganic sulfur donors (Na2S, Na2S2, Na2S3) alongside anti-cancer drugs demonstrated that Na2S attenuated the cardiotoxicity of osimertinib but not doxorubicin. Osimertinib decreased intracellular reduced sulfur levels, while Na2S treatment suppressed the sulfur leakage, suggesting its potential in mitigating osimertinib-induced cardiotoxicity. These results imply the prospect of inorganic sulfides, such as Na2S, as a seed for precision pharmacotherapy to alleviate osimertinib's cardiotoxic effects.


Subject(s)
Acrylamides , Aniline Compounds , Antineoplastic Agents , Doxorubicin , Induced Pluripotent Stem Cells , Mitochondria , Myocytes, Cardiac , Sulfides , Humans , Acrylamides/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Aniline Compounds/pharmacology , Induced Pluripotent Stem Cells/drug effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Doxorubicin/adverse effects , Sulfides/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Cells, Cultured , Mitochondrial Dynamics/drug effects , Sulfur , Indoles , Pyrimidines
4.
Open Vet J ; 14(7): 1668-1676, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39175975

ABSTRACT

Background: Doxorubicin (DOX) is a chemotherapeutic drug applied clinically for the remedy of cancer, but its possibly life-threatening cardiotoxicity effects remain a concern. Aim: After that, this study evaluates the cardioprotective impacts of Lagenaria siceraria (LSS) oil on DOX induced cardiomyopathy in rats. Methods: Wistar male rats (n = 28, weighting 190-210 g) were arbitrarily allocated into four equal groups. Group 1 control group (CTR) received normal saline orally (1 ml/kg); group 2 (DOX) received DOX (10 mg/kg); group 3 (DOLS) received DOX + 3 g of Lagenaria siceraria seeds oil/kg; group 4 (LSSO) received LSSO (3 g/kg) daily for 18 days. The serum samples were collected to determine the creatine kinase-MB (CK-MB) isoenzyme, lactate dehydrogenase (LDH), aspartate aminotransferase (AST), and Troponin I activity. At the same time, the catalase, malondialdehyde (MDA), and reduced glutathione (GSH) were assessed in heart tissues. Additionally, histopathological investigations for the heart tissue were performed. Results: Results revealed no significant change in CK-MB levels between the DOLS group compared to the CTR group (p > 0.05). DOX group confirmed a substantial increase in AST, LDH, and Troponin1 serum levels compared to DOLS and LLSO groups (p < 0.05). The study demonstrated the antioxidant activity of LSS oil against DOX-induced toxicity. The DOX group significantly reduced GSH and catalase levels, with an increase in MDA levels compared to DOLS and LLSO groups. Histopathological analysis showed protective properties of LSS oil against myocardial damage caused by DOX. Conclusion: This study highlights the favorable impacts of LSS oil in mitigating DOX-triggered cardiotoxicity in a rat model.


Subject(s)
Cardiomyopathies , Doxorubicin , Rats, Wistar , Animals , Doxorubicin/adverse effects , Male , Cardiomyopathies/chemically induced , Cardiomyopathies/veterinary , Cardiomyopathies/prevention & control , Rats , Cucurbitaceae/chemistry , Antibiotics, Antineoplastic , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Cardiotoxicity/veterinary , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Plant Oils/pharmacology , Plant Oils/administration & dosage , Plant Oils/therapeutic use
5.
Int J Pharm ; 663: 124569, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39127172

ABSTRACT

Doxorubicin (Dox) is a broad-spectrum antineoplastic chemotherapeutic agent used in clinical settings, yet it exhibits significant cardiotoxicity, which in severe cases can lead to heart failure. Research indicates that oxidative stress plays a pivotal role in Dox -induced cardiomyocyte injury. Therefore, the application of antioxidants represents an effective strategy to mitigate the cardiotoxic effects of doxorubicin. In preliminary studies, we isolated an antioxidative peptide, PHWWEYRR (8P). This study utilizes a PCM cardiomyocyte-targeting peptide-modified liposome as a carrier to deliver 8P into cardiomyocytes, aiming to prevent Dox-induced cardiac injury through its antioxidative mechanism. The results demonstrated that we prepared the 8P-loaded and PCM-targeting peptide-modified liposome (P-P-8P), which exhibited good dispersibility, encapsulation efficiency, drug loading capacity, and in vitro release, along with myocardial targeting capability. In vitro experiments showed that P-P-8P could prevent oxidative stress injury in H9C2 cells, protect mitochondrial functions, and inhibit cell apoptosis through a mitochondria-dependent pathway. In vivo experiments indicated that P-P-8P could prevent abnormalities in serum biochemical indicators, cardiac dysfunction, and myocardial pathological changes in mice. In conclusion, P-P-8P effectively delivers 8P to cardiomyocytes, offering protection against the cardiotoxic effects of Dox, and holds potential as a future preventative or therapeutic agent for drug-induced cardiomyopathy.


Subject(s)
Antioxidants , Doxorubicin , Liposomes , Myocytes, Cardiac , Oxidative Stress , Doxorubicin/administration & dosage , Animals , Antioxidants/pharmacology , Antioxidants/administration & dosage , Antioxidants/chemistry , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Male , Mice , Cell Line , Apoptosis/drug effects , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Rats , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/toxicity , Myocardium/pathology , Myocardium/metabolism , Drug Liberation , Drug Delivery Systems , Peptides/administration & dosage , Peptides/pharmacology , Peptides/chemistry
6.
Chem Biol Interact ; 400: 111179, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39089415

ABSTRACT

Oxidative stress contributes greatly to doxorubicin (DOX)-induced cardiotoxicity. Down-regulation of nuclear factor erythroid 2-related factor 2 (Nrf2) is a key factor in DOX-induced myocardial oxidative injury. Recently, we found that mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1)-dependent k48-linked ubiquitination was responsible for down-regulation of myocardial Nrf2 in DOX-treated mice. Micafungin, an antifungal drug, was identified as a potential MALT1 inhibitor. This study aims to explore whether micafungin can reduce DOX-induced myocardial oxidative injury and if its anti-oxidative effect involves a suppression of MALT1-dependent k48-linked ubiquitination of Nrf2. To establish the cardiotoxicity models in vivo and in vitro, mice were treated with a single dose of DOX (15 mg/kg, i.p.) and cardiomyocytes were incubated with DOX (1 µM) for 24 h, respectively. Using mouse model of DOX-induced cardiotoxicity, micafungin (10 or 20 mg/kg) was shown to improve cardiac function, concomitant with suppression of oxidative stress, mitochondrial dysfunction, and cell death in a dose-dependent manner. Similar protective roles of micafungin (1 or 5 µM) were observed in DOX-treated cardiomyocytes. Mechanistically, micafungin weakened the interaction between MALT1 and Nrf2, decreased the k48-linked ubiquitination of Nrf2 while elevated the protein levels of Nrf2 in both DOX-treated mice and cardiomyocytes. Furthermore, MALT1 overexpression counteracted the cardioprotective effects of micafungin. In conclusion, micafungin reduces DOX-induced myocardial oxidative injury via suppression of MALT1, which decreases the k48-linked ubiquitination of Nrf2 and elevates Nrf2 protein levels. Thus, micafungin may be repurposed for treating DOX-induced cardiotoxicity.


Subject(s)
Doxorubicin , Micafungin , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-E2-Related Factor 2 , Oxidative Stress , Ubiquitination , Animals , NF-E2-Related Factor 2/metabolism , Doxorubicin/toxicity , Ubiquitination/drug effects , Oxidative Stress/drug effects , Mice , Male , Micafungin/pharmacology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cardiotoxicity/prevention & control , Cardiotoxicity/metabolism , Cardiotoxicity/etiology , Myocardium/metabolism , Myocardium/pathology
7.
Chem Biol Interact ; 399: 111132, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-38964637

ABSTRACT

The clinical application of doxorubicin (DOX) was limited by the serious cardiotoxicity. The traditional Chinese medicine Andrographis paniculata and its principal active component (Dehydroandrographolide, DA) have been well known for their diverse cardiovascular protective effects. However, the effects of DA on DOX-induced cardiotoxicity (DIC) were still unknown. In this study, we evaluated the effects and revealed the potential mechanisms of DA on DIC both in vivo and in vitro. The effects of DA on DIC were systematically assessed by echocardiography and histological assays. Western blot and flow cytometry were used to measure apoptosis of cardiomyocytes. Transmission electron microscopy and StubRFP-SensGFP-LC3 lentivirus were further used to assay autophagic flux. Our results showed that DA administration significantly improved cardiac function and attenuated DOX-induced cardiomyocyte apoptosis. Mechanically, DA restored autophagic flux and lysosome functions via inhibiting DOX-induced mTOR signal pathway activation and increasing the translocation of TFEB to the nucleus. However, activation of mTOR or knockdown of TFEB significantly inhibited the protective effects of DA against DIC by impacting lysosomal functions and autophagic flux. In conclusion, our results revealed that DA might be a potential cardioprotective agent against DIC.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cardiotoxicity , Diterpenes , Doxorubicin , Myocytes, Cardiac , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Doxorubicin/toxicity , Autophagy/drug effects , Diterpenes/pharmacology , Diterpenes/chemistry , TOR Serine-Threonine Kinases/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Signal Transduction/drug effects , Cardiotoxicity/prevention & control , Apoptosis/drug effects , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
8.
Eur J Pharm Sci ; 200: 106849, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38992452

ABSTRACT

Doxorubicin (DOX) is an anthracycline chemotherapy drug widely employed in the treatment of various cancers, known for its potent antineoplastic properties but often associated with dose-dependent cardiotoxicity, limiting its clinical use. This review explores the complex molecular details that determine the heart-protective effectiveness of carvedilol in relation to cardiotoxicity caused by DOX. The harmful effects of DOX on heart cells could include oxidative stress, DNA damage, iron imbalance, disruption of autophagy, calcium imbalance, apoptosis, dysregulation of topoisomerase 2-beta, arrhythmogenicity, and inflammatory responses. This review carefully reveals how carvedilol serves as a strong protective mechanism, strategically reducing each aspect of cardiac damage caused by DOX. Carvedilol's antioxidant capabilities involve neutralizing free radicals and adjusting crucial antioxidant enzymes. It skillfully manages iron balance, controls autophagy, and restores the calcium balance essential for cellular stability. Moreover, the anti-apoptotic effects of carvedilol are outlined through the adjustment of Bcl-2 family proteins and activation of the Akt signaling pathway. The medication also controls topoisomerase 2-beta and reduces the renin-angiotensin-aldosterone system, together offering a thorough defense against cardiotoxicity induced by DOX. These findings not only provide detailed understanding into the molecular mechanisms that coordinate heart protection by carvedilol but also offer considerable potential for the creation of targeted treatment strategies intended to relieve cardiotoxicity caused by chemotherapy.


Subject(s)
Cardiotoxicity , Carvedilol , Doxorubicin , Carvedilol/pharmacology , Carvedilol/therapeutic use , Humans , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Doxorubicin/adverse effects , Doxorubicin/toxicity , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Carbazoles/pharmacology , Oxidative Stress/drug effects , Apoptosis/drug effects , Propanolamines/pharmacology
9.
Cells ; 13(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38995002

ABSTRACT

Doxorubicin (DOX) is an anthracycline anticancer agent that is highly effective in the treatment of solid tumors. Given the multiplicity of mechanisms involved in doxorubicin-induced cardiotoxicity, it is difficult to identify a precise molecular target for toxicity. The findings of a literature review suggest that natural products may offer cardioprotective benefits against doxorubicin-induced cardiotoxicity, both in vitro and in vivo. However, further confirmatory studies are required to substantiate this claim. It is of the utmost importance to direct greater attention towards the intricate signaling networks that are of paramount importance for the survival and dysfunction of cardiomyocytes. Notwithstanding encouraging progress made in preclinical studies of natural products for the prevention of DOX-induced cardiotoxicity, these have not yet been translated for clinical use. One of the most significant obstacles hindering the development of cardioprotective adjuvants based on natural products is the lack of adequate bioavailability in humans. This review presents an overview of current knowledge on doxorubicin DOX-induced cardiotoxicity, with a focus on the potential benefits of natural compounds and herbal preparations in preventing this adverse effect. As literature search engines, the browsers in the Scopus, PubMed, Web of Science databases and the ClinicalTrials.gov register were used.


Subject(s)
Anthracyclines , Biological Products , Cardiotoxicity , Humans , Biological Products/therapeutic use , Biological Products/pharmacology , Cardiotoxicity/prevention & control , Anthracyclines/adverse effects , Animals , Doxorubicin/adverse effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use
10.
Med Oncol ; 41(8): 196, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977536

ABSTRACT

Cardiotoxicity is a side effect of chemotherapy in human epidermal growth factor receptor 2 (HER2) positive breast cancer patients receiving both anthracyclines and trastuzumab. We looked for a possible protective effect of rosuvastatin against chemotherapy-induced cardiotoxicity. Methods: 50 newly diagnosed HER2 positive breast cancer patients were randomly allocated into two groups: 25patients in each. Group 1(control group) received doxorubicin for 4 cycles (3 months) followed by trastuzumab adjuvant therapy. Group 2 (treatment group) received doxorubicin for 4 cycles (3 months) followed by trastuzumab adjuvant therapy and 20 mg of oral rosuvastatin 24 h before the first cycle of chemotherapy and once daily for the rest of the follow-up period (6 months). Transthoracic echocardiography was done, and blood samples were collected for patients 24 h before the initiation of therapy, after 3 months and after 6 months to assess serum levels of high sensitivity cardiac troponin I (hs-cTnI), Myeloperoxidase (MPO), Interleukin-6 (IL-6) and Alanine aminotransferase (ALT). The study was retrospectively registered in Clinical Trials.gov in April 2022. Its ID is NCT05338723. Compared to control group, Rosuvastatin-treated group had a significantly lower decline in LVEF after 3 months and after 6 months. They had significantly lower Hs-cTnI and IL-6 after 3 months and after 6 months, and significantly lower MPO after 6 months. Four patients in control group experienced cardiotoxicity while no one in rosuvastatin-treated group. Rosuvastatin attenuated cardiotoxicity, so it is a promising protective agent against chemotherapy-induced cardiotoxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms , Cardiotoxicity , Doxorubicin , Receptor, ErbB-2 , Rosuvastatin Calcium , Trastuzumab , Humans , Breast Neoplasms/drug therapy , Rosuvastatin Calcium/therapeutic use , Female , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Receptor, ErbB-2/metabolism , Middle Aged , Doxorubicin/adverse effects , Trastuzumab/adverse effects , Trastuzumab/therapeutic use , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Troponin I/blood
11.
Chem Biol Interact ; 401: 111155, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39029857

ABSTRACT

Doxorubicin (Dox) is widely used as a chemotherapy drug, while anethole (AN) is primarily known as the main aromatic component in various plant species. This research focused on the impact of AN on the cardiac and renal toxicity induced by Dox and to understand the underlying mechanisms. For cardiac toxicity, Wistar rats were categorized into four groups: a Control group; a Dox group, where rats received 2.5 mg/kg of Dox intraperitoneally every other day; and two Dox + AN groups, where animals were administered Dox (2.5 mg/kg/every other day, IP) along with 125 mg/kg or 250 mg/kg of AN, respectively. The renal toxicity study included similar groups, with the Dox group receiving a single dose of 20 mg/kg of Dox intraperitoneally on the tenth day, and the Dox + AN groups receiving 125 mg/kg and 250 mg/kg of AN for two weeks, alongside the same dose of Dox (20 mg/kg, IP, once on the 10th day). Parameters assessed included ECG, cardiac injury markers (CK, CK-MB, and LDH), and kidney function tests (Cr, BUN, uric acid, LDL, Kim-1, NGAL, and CysC). Antioxidant activity, lipid peroxidation, inflammation, and apoptotic markers were also monitored in heart and renal tissues. Gene expression levels of the TLR4/MyD88/NFκB pathway, along with Bax and Bcl-2, were evaluated. Dox significantly altered ECG, elevated cardiac injury markers, and renal function markers. It also augmented gene expressions of TLR4/MyD88/NFκB, amplified oxidative stress, inflammatory cytokines and apoptotic markers. Conversely, AN reduced cardiac injury markers and kidney function tests, improved ECG, diminished TLR4/MyD88/NFκB gene expression, and alleviated oxidative stress by increasing antioxidant enzyme activities and reducing inflammatory cytokines. AN also enhanced Bcl-2 levels and inhibited Bax and the cleavage of caspase-3 and 9. AN countered the lipid peroxidation, oxidative stress, inflammation, and apoptosis induced by Dox, marking it as a potential preventive strategy against Dox-induced nephrotoxic and cardiotoxic injuries.


Subject(s)
Allylbenzene Derivatives , Anisoles , Doxorubicin , Kidney , Network Pharmacology , Rats, Wistar , Animals , Doxorubicin/toxicity , Rats , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Anisoles/pharmacology , Anisoles/toxicity , Male , Apoptosis/drug effects , Oxidative Stress/drug effects , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Heart/drug effects , Toll-Like Receptor 4/metabolism , Antibiotics, Antineoplastic/toxicity , Antioxidants/pharmacology , NF-kappa B/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Kidney Diseases/metabolism
12.
Curr Treat Options Oncol ; 25(8): 1038-1054, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39066853

ABSTRACT

OPINION STATEMENT: Anthracycline (ANT)-induced cardiotoxicity (AIC) is a particularly prominent form of cancer therapy-related cardiovascular toxicity leading to the limitations of ANTs in clinical practice. Even though AIC has drawn particular attention, the best way to treat it is remaining unclear. Updates to AIC therapy have been made possible by recent developments in research on the underlying processes of AIC. We review the current molecular pathways leading to AIC: 1) oxidative stress (OS) including enzymatic-induced and other mechanisms; 2) topoisomerase; 3) inflammatory response; 4) cardiac progenitor cell damage; 5) epigenetic changes; 6) renin-angiotensin-aldosterone system (RAAS) dysregulation. And we systematically discuss current prevention and treatment strategies and novel pathogenesis-based therapies for AIC: 1) dose reduction and change; 2) altering drug delivery methods; 3) antioxidants, dexrezosen, statina, RAAS inhibitors, and hypoglycemic drugs; 4) miRNA, natural phytochemicals, mesenchymal stem cells, and cardiac progenitor cells. We also offer a fresh perspective on the management of AIC by outlining the current dilemmas and challenges associated with its prevention and treatment.


Subject(s)
Anthracyclines , Cardiotoxicity , Humans , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Anthracyclines/adverse effects , Anthracyclines/therapeutic use , Precision Medicine/methods , Animals , Neoplasms/drug therapy , Oxidative Stress/drug effects , Disease Management , Disease Susceptibility , Renin-Angiotensin System/drug effects , Biomarkers
13.
Food Chem Toxicol ; 191: 114863, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38997059

ABSTRACT

Antioxidants given concurrently with chemotherapy offer an effective strategy for reducing the negative effects of the drug. One remaining obstacle to the use of doxorubicin (DOX) in chemotherapy is cardiotoxicity. Using vitamin E (Vit. E) as a reference standard, our study focuses on the potential preventive benefits of oxyresveratrol (ORES) and/or dapagliflozin (DAPA) against DOX-induced cardiac injury. Acute cardiotoxicity was noticed after a single intravenous injection of a male rat's tail vein with 10 mg/kg of DOX. Oral doses of ORES (80 mg/kg), DAPA (10 mg/kg), and Vit. E (1 g/kg) were given, respectively. Pretreatment of animals with Vit. E, ORES and/or DAPA revealed a considerable alleviation of heart damage, as evidenced by histopathological change mitigation and a notable drop in serum AST, LDH, CK, CK-MB, and cardiac contents of MDA and NO2-. Also, serum TAC, tissue GSH, and SOD showed substantial increases. Additionally, tissue caspase-3, serum IL-6, and TNF-α were considerably reduced. Moreover, a downregulation in cardiac gene expression of ATG-5, Keap-1, and NF-κB in addition to an upregulation of Bcl-2 gene expression and HO-1, Nrf-2, and PPAR-γ protein expression clearly appeared. Ultimately, ORES and/or DAPA have an optimistic preventive action against severe heart deterioration caused by DOX.


Subject(s)
Benzhydryl Compounds , Cardiotoxicity , Caspase 3 , Doxorubicin , Glucosides , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , NF-kappa B , PPAR gamma , Animals , Glucosides/pharmacology , Male , Rats , PPAR gamma/metabolism , PPAR gamma/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NF-kappa B/metabolism , NF-kappa B/genetics , Benzhydryl Compounds/toxicity , Cardiotoxicity/prevention & control , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , Doxorubicin/toxicity , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Caspase 3/metabolism , Caspase 3/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Stilbenes/pharmacology , Interleukin-6/metabolism , Interleukin-6/genetics , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/genetics , Cardiotonic Agents/pharmacology , Signal Transduction/drug effects , Disease Models, Animal , Plant Extracts/pharmacology , Rats, Wistar , Antioxidants/pharmacology
14.
Curr Probl Cardiol ; 49(10): 102743, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39053681

ABSTRACT

BACKGROUND: Chemotherapy associated with breast cancer often induces cardiotoxicity, which compromises patients' health and quality of life. OBJECTIVE: To verify the effect of physical exercise on chemotherapy-induced cardiotoxicity, through the assessment of cardiac function in patients with breast cancer. METHODS: A systematic review and meta-analysis of clinical studies was conducted to evaluate the effectiveness of physical training in chemotherapy-induced cardiomyopathy in the PubMed, Web of Sciences and Scopus databases. Thirteen studies were included in the systematic review and eleven studies in the data meta-analysis. RESULTS: Global longitudinal strain presents a cardioprotective effect when compared to the control group (Heterogeneity: Chi² = 12.81, df = 10 (p = 0.23); I² = 22 %.) Test for global effect: Z = 2, 13 (p = 0.03). Physical training is more effective (test for subgroup differences, p = 0.031) in attenuating the impairment of %GLS induced by chemotherapy if performed concomitantly with exposure to chemotherapy (95 % CI; Heterogeneity: Chi² = 7.49, gl = 5 (p = 0.19); I² = 33 %; Test for global effect: Z = 2.33 (p = 0.02) when compared after chemotherapy treatment, or in the long term (for 12 months or more). However, without benefits in LVEF (Heterogeneity: Chi² = 42.14, df = 10 (p < 0.00001); I² = 76 %) Test for global effect: Z = 2.51 (p = 0.01) Conclusion: Exercise training is a cardioprotective approach in breast cancer patients who experience chemotherapy-induced cardiotoxicity. Exercise during exposure to chemotherapy has greater effects on preserving cardiac function.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Cardiotoxicity , Exercise Therapy , Female , Humans , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cardiomyopathies/physiopathology , Cardiomyopathies/prevention & control , Cardiomyopathies/chemically induced , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Exercise Therapy/methods , Quality of Life
15.
Cell Physiol Biochem ; 58(3): 273-287, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38881348

ABSTRACT

BACKGROUND/AIMS: Inhaled particulate air pollution is associated with cardiotoxicity with underlying mechanisms including oxidative stress and inflammation. Carnosol, commonly found in rosemary and sage, is known to possess a broad range of therapeutic properties such as antioxidant, anti-inflammatory and antiapoptotic. However, its cardioprotective effects on diesel exhaust particles (DEPs)-induced toxicity have not been studied yet. Hence, we evaluated the potential ameliorative effects of carnosol on DEPs-induced heart toxicity in mice, and the underlying mechanisms involved. METHODS: Mice were intratracheally instilled with DEPs (1 mg/kg) or saline, and 1 hour prior to instillation they were given intraperitoneally either carnosol (20 mg/kg) or saline. Twenty-four hours after the DEPs instillation, multiple parameters were evaluated in the heart by enzyme-linked immunosorbent assay, colorimetric assay, Comet assay and Western blot technique. RESULTS: Carnosol has significantly reduced the elevation in the plasma levels of lactate hydrogenase and brain natriuretic peptide induced by DEPs. Likewise, the augmented cardiac levels of proinflammatory cytokines, lipid peroxidation, and total nitric oxide in DEPs-treated groups were significantly normalized with the treatment of carnosol. Moreover, carnosol has markedly reduced the heart mitochondrial dysfunction, as well as DNA damage and apoptosis of mice treated with DEPs. Similarly, carnosol significantly reduced the elevated expressions of phosphorylated nuclear factor-кB (NF-кB) and mitogen-activated protein kinases (MAPKs) in the hearts. Furthermore, the treatment with carnosol has restored the decrease in the expression of sirtuin-1 in the hearts of mice exposed to DEPs. CONCLUSION: Carnosol significantly attenuated DEP-induced cardiotoxicity in mice by suppressing inflammation, oxidative stress, DNA damage, and apoptosis, at least partly via mechanisms involving sirtuin-1 activation and the inhibition of NF-кB and MAPKs activation.


Subject(s)
Abietanes , Cardiotoxicity , NF-kappa B , Oxidative Stress , Vehicle Emissions , Animals , Mice , NF-kappa B/metabolism , Oxidative Stress/drug effects , Vehicle Emissions/toxicity , Abietanes/pharmacology , Abietanes/therapeutic use , Male , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cardiotoxicity/prevention & control , Cardiotoxicity/drug therapy , Cardiotoxicity/pathology , Nitrosative Stress/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation/chemically induced , MAP Kinase Signaling System/drug effects , Antioxidants/pharmacology , Apoptosis/drug effects , Signal Transduction/drug effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Sirtuin 1/metabolism , Sirtuin 1/genetics , DNA Damage/drug effects
17.
Colloids Surf B Biointerfaces ; 241: 113992, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38833960

ABSTRACT

In order to reduce the cardiotoxicity of doxorubicin (DOX) and improve its antitumor effect, dihydroartemisinin (DHA) and DOX prodrug (DOX-S-DHA) synthesized via a single sulfur bond was used with TEPP-46 to prepare nano-liposomes (DOX-S-DHA@TEPP-46 Lips). In which, TEPP-46 was expected to exert p53 bidirectional regulation to promote the synergistic antitumor effect of DOX and DHA while reducing cardiotoxicity. DOX-S-DHA@TEPP-46 Lips exhibited uniform particle size, good stability, and excellent redox-responsive activity. DOX-S-DHA@TEPP-46 Lips could significantly inhibit the proliferation of tumor cells, but had less cytotoxicity on normal cells. The presence of TEPP-46 increased the content of p53 protein, which further induced tumor cell apoptosis. DOX-S-DHA@TEPP-46 Lips had satisfactory long circulation to enhance the antitumor efficacy and reversed the cardiotoxicity of DOX in B16-F10 tumor-bearing mice. In conclusion, DOX-S-DHA@TEPP-46 Lips provides a new insight on creating sophisticated redox-sensitive nano-liposomes for cancer therapy as well as the decreased cardiotoxicity of DOX.


Subject(s)
Artemisinins , Cardiotoxicity , Doxorubicin , Liposomes , Prodrugs , Animals , Artemisinins/chemistry , Artemisinins/pharmacology , Artemisinins/administration & dosage , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacology , Mice , Liposomes/chemistry , Cardiotoxicity/prevention & control , Cell Proliferation/drug effects , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Particle Size , Nanoparticles/chemistry , Drug Delivery Systems , Mice, Inbred C57BL , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Drug Screening Assays, Antitumor , Humans , Cell Line, Tumor
18.
Breast Cancer Res Treat ; 207(2): 283-291, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38922547

ABSTRACT

PURPOSE: In Ireland, over 3000 patients are diagnosed with breast cancer annually, and 1 in 9 Irish women will be diagnosed with breast cancer in their lifetime. There is evidence that female breast cancer survivors are more likely to die of cardiovascular disease than their age-matched counterparts. Specific services for cancer patients suffering from cancer therapy related cardiovascular toxicity have led to a higher incidence of safe anti-cancer treatment completion. Such services are not widely available in our jurisdiction, and the purpose of this trial is to remedy this situation. METHODS: This protocol describes a prospective, single arm, pilot feasibility study implementing a dedicated Cardio-Oncology assessment and surveillance pathway for patients receiving multimodal breast cancer treatment. It incorporates novel biomarker and radiomic surveillance and monitoring approaches for cancer-therapy related cardiac dysfunction into routine care for breast cancer patients undergoing adjuvant systemic chemotherapy. RESULTS: Declaration of results will via peer reviewed academic journals, and communicated directly to key knowledge users both nationally and internationally. This engagement will be critical to enable to healthcare services and policy sector make informed decisions or valuable changes to clinical practice, expenditure and/or systems development to support specialized Cardio-Oncology clinical pathways. All data is to be made available upon request. CONCLUSION: Dedicated cardio-oncology services have been recommended in recent literature to improve patient outcomes. Our protocol describes a feasibility study into the provision of such services for breast cancer.


Subject(s)
Breast Neoplasms , Cardio-Oncology , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/complications , Cardio-Oncology/methods , Cardiotoxicity/diagnosis , Cardiotoxicity/epidemiology , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Feasibility Studies , Ireland/epidemiology , Pilot Projects , Prospective Studies
19.
Intern Med J ; 54(8): 1254-1263, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38874281

ABSTRACT

Cancer therapy-related cardiac dysfunction (CTRCD) is a complication of selected cancer therapy agents associated with decline in left ventricular ejection fraction (LVEF). Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) have established benefits in heart failure with reduced ejection fraction, but their efficacy for preventing CTRCD remains controversial. This narrative systematic review assessed the efficacy and safety of ACEI/ARB in the prevention of cancer therapy LVEF decline. We systematically searched PubMed, Embase and Cochrane from January 1980 to June 2022. Studies of interest were randomised controlled trials of patients with normal LVEF and active malignancy receiving cancer therapy, randomised to receive either an ACEI or ARB compared with a control group. The outcome was the change in LVEF from baseline to the end of the follow-up period. Death, clinical heart failure and adverse drug reactions were recorded. A total of 3731 search records were screened and 12 studies were included, comprising a total of 1645 participants. Nine studies assessed the prevention of anthracycline-induced LVEF decline, of which five showed a beneficial effect (1%-14% higher LVEF in treated groups), whereas four studies showed no effect. Three studies assessed the prevention of trastuzumab-induced LVEF decline, of which one showed a beneficial effect (4% higher LVEF) in a subset of participants. There are mixed data regarding the efficacy of ACEI/ARB in preventing the LVEF decline in patients undergoing anthracycline or trastuzumab therapy, with evidence suggesting no clinically meaningful benefit observed in recent studies.


Subject(s)
Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors , Anthracyclines , Antineoplastic Agents , Stroke Volume , Trastuzumab , Ventricular Dysfunction, Left , Humans , Angiotensin Receptor Antagonists/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Anthracyclines/adverse effects , Antineoplastic Agents/adverse effects , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Neoplasms/drug therapy , Randomized Controlled Trials as Topic , Stroke Volume/drug effects , Trastuzumab/adverse effects , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects
20.
Curr Probl Cardiol ; 49(9): 102722, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38908726

ABSTRACT

In recent years, the mean survival rate of children after a cancer diagnosis has significantly improved. At the same time, a growing interest in short and long-term cardiovascular (CV) complications of cancer therapy, as well as long-term CV risk in childhood cancer survivors (CCS) developed, along with proposals of protocols for the diagnosis, management, and prevention of cancer therapy-related CV toxicity (CTR-CVT) in this population. Many clinical and individual risk factors for CTR-CVT have been identified, and a non-negligible prevalence of traditional CV risk factors has been described in this population, potentially associated with a further worsening in both CTR-CVT and long-term CV risk. Physical exercise (PE) represents a promising, free-of-cost and free-of-complications, helpful therapy for primary and secondary prevention of CTR-CVT in CCS. The present narrative review aims to summarize the most critical evidence available about CTR-CVT in CCS, focusing on the role of PE in this clinical scenario.


Subject(s)
Antineoplastic Agents , Cancer Survivors , Cardiotoxicity , Exercise , Neoplasms , Humans , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Neoplasms/drug therapy , Exercise/physiology , Antineoplastic Agents/adverse effects , Child , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/etiology , Exercise Therapy/methods , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL