Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.734
Filter
1.
Georgian Med News ; (349): 54-59, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38963202

ABSTRACT

Doxorubicin is the common chemotherapeutic agent that has been harnessed for the treatment of various types of malignancy including the treatment of soft tissue and osteosarcoma and cancers of the vital organs like breast, ovary, bladder, and thyroid. It is also used to treat leukaemia and lymphoma, however, this is an obstacle because of their prominent side effects including cardiotoxicity and lung fibrosis, we do aim to determine the role of CoQ10 as an antioxidant on the impeding the deleterious impacts of doxorubicin on tissue degenerative effects. To do so, 27 rats were subdivided into 3 groups of 9 each; CoQ10 exposed group, Doxorubicin exposed group, and CoQ10 plus Doxorubicin group. At the end of the study, the animals were sacrificed and lungs with hearts were harvested, and slides were prepared for examination under a microscope. The results indicated that doxorubicin induced abnormal cellular structure resulting in damaging cellular structures of the lung and heart while CoQ10 impeded these damaging effects and nearly restoring normal tissue structure. As a result, CoQ10 will maintain normal tissue of the lung and heart.


Subject(s)
Doxorubicin , Lung , Ubiquinone , Animals , Doxorubicin/adverse effects , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Rats , Lung/drug effects , Lung/pathology , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity , Myocardium/pathology , Male , Antioxidants/pharmacology , Cardiotoxicity/etiology , Cardiotoxicity/pathology , Heart/drug effects
2.
Vascul Pharmacol ; 155: 107324, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38985581

ABSTRACT

Doxorubicin (DOX) is a highly effective chemotherapeutic agent whose clinical use is hindered by the onset of cardiotoxic effects, resulting in reduced ejection fraction within the first year from treatment initiation. Recently it has been demonstrated that DOX accumulates within mitochondria, leading to disruption of metabolic processes and energetic imbalance. We previously described that phosphoinositide 3-kinase γ (PI3Kγ) contributes to DOX-induced cardiotoxicity, causing autophagy inhibition and accumulation of damaged mitochondria. Here we intend to describe the maladaptive metabolic rewiring occurring in DOX-treated hearts and the contribution of PI3Kγ signalling to this process. Metabolomic analysis of DOX-treated WT hearts revealed an accumulation of TCA cycle metabolites due to a cycle slowdown, with reduced levels of pyruvate, unchanged abundance of lactate and increased Acetyl-CoA production. Moreover, the activity of glycolytic enzymes was upregulated, and fatty acid oxidation downregulated, after DOX, indicative of increased glucose oxidation. In agreement, oxygen consumption was increased in after pyruvate supplementation, with the formation of cytotoxic ROS rather than energy production. These metabolic changes were fully prevented in KD hearts. Interestingly, they failed to increase glucose oxidation in response to DOX even with autophagy inhibition, indicating that PI3Kγ likely controls the fuel preference after DOX through an autophagy-independent mechanism. In vitro experiments showed that inhibition of PI3Kγ inhibits pyruvate dehydrogenase (PDH), the key enzyme of Randle cycle regulating the switch from fatty acids to glucose usage, while decreasing DOX-induced mobilization of GLUT-4-carrying vesicles to the plasma membrane and limiting the ensuing glucose uptake. These results demonstrate that PI3Kγ promotes a maladaptive metabolic rewiring in DOX-treated hearts, through a two-pronged mechanism controlling PDH activation and GLUT-4-mediated glucose uptake.


Subject(s)
Cardiotoxicity , Doxorubicin , Energy Metabolism , Fatty Acids , Glucose , Oxidation-Reduction , Animals , Doxorubicin/toxicity , Glucose/metabolism , Fatty Acids/metabolism , Energy Metabolism/drug effects , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Glycolysis/drug effects , Autophagy/drug effects , Male , Signal Transduction/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Citric Acid Cycle/drug effects , Mice, Inbred C57BL , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/prevention & control , Heart Diseases/physiopathology , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Mitochondria, Heart/enzymology , Mice, Knockout , Disease Models, Animal , Reactive Oxygen Species/metabolism , Glucose Transporter Type 4/metabolism , Antibiotics, Antineoplastic/toxicity , Antibiotics, Antineoplastic/adverse effects
7.
BMC Cardiovasc Disord ; 24(1): 350, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987722

ABSTRACT

BACKGROUND: Antineoplastic medications, including doxorubicin, idarubicin, and epirubicin, have been found to adversely affect the heart due to oxidative stress - mitochondrial dysfunction - ferroptosis (ORMFs), which act as contributing attributes to anthracycline-induced cardiotoxicity. To better understand this phenomenon, the time-resolved measurements of ORMFS genes were analyzed in this study. METHODS: The effect of three anthracycline drugs on ORMFs genes was studied using a human 3D cardiac microtissue cell model. Transcriptome data was collected over 14 days at two doses (therapeutic and toxic). WGCNA identified key module-related genes, and functional enrichment analysis investigated the biological processes quantified by ssGSEA, such as immune cell infiltration and angiogenesis. Biopsies were collected from heart failure patients and control subjects. GSE59672 and GSE2965 were collected for validation. Molecular docking was used to identify anthracyclines's interaction with key genes. RESULTS: The ORMFs genes were screened in vivo or in vitro. Using WGCNA, six co-expressed gene modules were grouped, with MEblue emerging as the most significant module. Eight key genes intersecting the blue module with the dynamic response genes were obtained: CD36, CDH5, CHI3L1, HBA2, HSD11B1, OGN, RPL8, and VWF. Compared with control samples, all key genes except RPL8 were down-regulated in vitro ANT treatment settings, and their expression levels varied over time. According to functional analyses, the key module-related genes were engaged in angiogenesis and the immune system pathways. In all ANT-treated settings, ssGSEA demonstrated a significant down-regulation of angiogenesis score and immune cell activity, including Activated CD4 T cell, Immature B cell, Memory B cell, Natural killer cell, Type 1 T helper cell, and Type 2 T helper cell. Molecular docking revealed that RPL8 and CHI3L1 show significant binding affinity for anthracyclines. CONCLUSION: This study focuses on the dynamic characteristics of ORMFs genes in both human cardiac microtissues and cardiac biopsies from ANT-treated patients. It has been highlighted that ORMFs genes may contribute to immune infiltration and angiogenesis in cases of anthracycline-induced cardiotoxicity. A thorough understanding of these genes could potentially lead to improved diagnosis and treatment of the disease.


Subject(s)
Cardiotoxicity , Ferroptosis , Molecular Docking Simulation , Oxidative Stress , Humans , Oxidative Stress/drug effects , Ferroptosis/drug effects , Ferroptosis/genetics , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Heart/genetics , Gene Regulatory Networks , Time Factors , Transcriptome , Epirubicin/adverse effects , Doxorubicin , Antibiotics, Antineoplastic/adverse effects , Case-Control Studies , Idarubicin , Heart Failure/chemically induced , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Gene Expression Profiling , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Longitudinal Studies , Anthracyclines/adverse effects , Gene Expression Regulation , Signal Transduction
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.
Clin Epigenetics ; 16(1): 91, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014511

ABSTRACT

BACKGROUND: Doxorubicin (Dox) is an effective chemotherapeutic drug for various cancers, but its clinical application is limited by severe cardiotoxicity. Dox treatment can transcriptionally activate multiple cardiotoxicity-associated genes in cardiomyocytes, the mechanisms underlying this global gene activation remain poorly understood. METHODS AND RESULTS: Herein, we integrated data from animal models, CUT&Tag and RNA-seq after Dox treatment, and discovered that the level of H3K27ac (a histone modification associated with gene activation) significantly increased in cardiomyocytes following Dox treatment. C646, an inhibitor of histone acetyltransferase, reversed Dox-induced H3K27ac accumulation in cardiomyocytes, which subsequently prevented the increase of Dox-induced DNA damage and apoptosis. Furthermore, C646 alleviated cardiac dysfunction in Dox-treated mice by restoring ejection fraction and reversing fractional shortening percentages. Additionally, Dox treatment increased H3K27ac deposition at the promoters of multiple cardiotoxic genes including Bax, Fas and Bnip3, resulting in their up-regulation. Moreover, the deposition of H3K27ac at cardiotoxicity-related genes exhibited a broad feature across the genome. Based on the deposition of H3K27ac and mRNA expression levels, several potential genes that might contribute to Dox-induced cardiotoxicity were predicted. Finally, the up-regulation of H3K27ac-regulated cardiotoxic genes upon Dox treatment is conservative across species. CONCLUSIONS: Taken together, Dox-induced epigenetic modification, specifically H3K27ac, acts as a molecular switch for the activation of robust cardiotoxicity-related genes, leading to cardiomyocyte death and cardiac dysfunction. These findings provide new insights into the relationship between Dox-induced cardiotoxicity and epigenetic regulation, and identify H3K27ac as a potential target for the prevention and treatment of Dox-induced cardiotoxicity.


Subject(s)
Cardiotoxicity , Doxorubicin , Histones , Myocytes, Cardiac , Doxorubicin/adverse effects , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Histones/metabolism , Histones/genetics , Mice , Cardiotoxicity/genetics , Cardiotoxicity/etiology , Apoptosis/drug effects , Apoptosis/genetics , Epigenesis, Genetic/drug effects , DNA Damage/drug effects , Antibiotics, Antineoplastic/adverse effects , Male , Humans
10.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39000584

ABSTRACT

Cardiotoxicity is the main side effect of several chemotherapeutic drugs. Doxorubicin (Doxo) is one of the most used anthracyclines in the treatment of many tumors, but the development of acute and chronic cardiotoxicity limits its clinical usefulness. Different studies focused only on the effects of long-term Doxo administration, but recent data show that cardiomyocyte damage is an early event induced by Doxo after a single administration that can be followed by progressive functional decline, leading to overt heart failure. The knowledge of molecular mechanisms involved in the early stage of Doxo-induced cardiotoxicity is of paramount importance to treating and/or preventing it. This review aims to illustrate several mechanisms thought to underlie Doxo-induced cardiotoxicity, such as oxidative and nitrosative stress, inflammation, and mitochondrial dysfunction. Moreover, here we report data from both in vitro and in vivo studies indicating new therapeutic strategies to prevent Doxo-induced cardiotoxicity.


Subject(s)
Cardiotoxicity , Doxorubicin , Inflammation , Oxidative Stress , Doxorubicin/adverse effects , Oxidative Stress/drug effects , Humans , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Animals , Inflammation/metabolism , Inflammation/chemically induced , Inflammation/pathology , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
11.
Pharmacogenomics J ; 24(4): 21, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951505

ABSTRACT

There is a known genetic susceptibility to anthracycline-induced cardiac dysfunction in childhood cancer survivors, but this has not been adequately shown in adolescent and young adult (AYA) patients. Our aim was to determine if the previously identified variants associated with cardiac dysfunction in childhood cancer patients affect AYA cancer patients similarly. Forty-five variants were selected for analysis in 253 AYAs previously treated with anthracyclines. We identified four variants that were associated with cardiac dysfunction: SLC10A2:rs7319981 (p = 0.017), SLC22A17:rs4982753 (p = 0.019), HAS3:rs2232228 (p = 0.023), and RARG:rs2229774 (p = 0.050). HAS3:rs2232228 and SLC10A2:rs7319981 displayed significant effects in our AYA cancer survivor population that were in the opposite direction than that reported in childhood cancer survivors. Genetic variants in the host genes were further analyzed for additional associations with cardiotoxicity in AYA cancer survivors. The host genes were then evaluated in a panel of induced pluripotent stem cell-derived cardiomyocytes to assess changes in levels of expression when treated with doxorubicin. Significant upregulation of HAS3 and SLC22A17 expression was observed (p < 0.05), with non-significant anthracycline-responsivity observed for RARG. Our study demonstrates that there is a genetic influence on cardiac dysfunction in AYA cancer patients, but there may be a difference in the role of genetics between childhood and AYA cancer survivors.


Subject(s)
Anthracyclines , Cancer Survivors , Cardiotoxicity , Genetic Predisposition to Disease , Humans , Adolescent , Anthracyclines/adverse effects , Young Adult , Male , Female , Cardiotoxicity/genetics , Adult , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Heart Diseases/chemically induced , Heart Diseases/genetics , Antibiotics, Antineoplastic/adverse effects , Risk Factors
12.
Heart Fail Rev ; 29(5): 969-988, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38990214

ABSTRACT

Cancer is the leading cause of death worldwide, and the number of cancer-related deaths is expected to increase. Common types of cancer include skin, breast, lung, prostate, and colorectal cancers. While clinical research has improved cancer therapies, these treatments often come with significant side effects such as chronic fatigue, hair loss, and nausea. In addition, cancer treatments can cause long-term cardiovascular complications. Doxorubicin (DOX) therapy is one example, which can lead to decreased left ventricle (LV) echocardiography (ECHO) parameters, increased oxidative stress in cellular level, and even cardiac fibrosis. The apelinergic system, specifically apelin and its receptor, together, has shown properties that could potentially protect the heart and mitigate the damages caused by DOX anti-cancer treatment. Studies have suggested that stimulating the apelinergic system may have therapeutic benefits for heart damage induced by DOX. Further research in chronic preclinical models is needed to confirm this hypothesis and understand the mechanism of action for the apelinergic system. This review aims to collect and present data on the effects of the apelinergic system on doxorubicin-induced cardiotoxicity.


Subject(s)
Antibiotics, Antineoplastic , Apelin , Cardiotoxicity , Doxorubicin , Humans , Doxorubicin/adverse effects , Cardiotoxicity/etiology , Antibiotics, Antineoplastic/adverse effects , Apelin/metabolism , Animals , Oxidative Stress/drug effects , Neoplasms/drug therapy , Apelin Receptors/metabolism
13.
J Pharmacol Sci ; 156(1): 9-18, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39068035

ABSTRACT

Necroptosis, necrosis characterized by RIPK3-MLKL activation, has been proposed as a mechanism of doxorubicin (DOX)-induced cardiomyopathy. We showed that rapamycin, an mTORC1 inhibitor, attenuates cardiomyocyte necroptosis. Here we examined role of MLKL in DOX-induced myocardial damage and protective effects of rapamycin. Cardiomyopathy was induced in mice by intraperitoneal injections of DOX (10 mg/kg, every other day) and followed for 7 days. DOX-treated mice showed a significant decline in LVEF assessed by cardiac MRI (45.5 ± 5.1% vs. 65.4 ± 4.2%), reduction in overall survival rates, and increases in myocardial RIPK3 and MLKL expression compared with those in vehicle-treated mice, and those changes were prevented by administration of rapamycin (0.25 mg/kg) before DOX injection. In immunohistochemical analyses, p-MLKL signals were detected in the cardiomyocytes of DOX-treated mice, and the signals were reduced by rapamycin. Mlkl+/- and Mlkl-/- mice were similarly resistant to DOX-induced cardiac dysfunction, indicating that a modest reduction in MLKL level is sufficient to prevent the development of DOX-induced cardiomyopathy. However, evidence of cardiomyocyte necrosis assessed by C9 immunostaining, presence of replacement fibrosis, and electron microscopic analyses was negligible in the myocardium of DOX-treated mice. Thus, MLKL-mediated signaling contributes to DOX-induced cardiac dysfunction primarily by a necrosis-independent mechanism, which is inhibitable by rapamycin.


Subject(s)
Cardiomyopathies , Doxorubicin , Mice, Inbred C57BL , Myocytes, Cardiac , Necroptosis , Protein Kinases , Receptor-Interacting Protein Serine-Threonine Kinases , Sirolimus , Animals , Doxorubicin/adverse effects , Protein Kinases/metabolism , Sirolimus/pharmacology , Cardiomyopathies/chemically induced , Cardiomyopathies/prevention & control , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Necroptosis/drug effects , Male , Mice , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity
14.
Arq Bras Cardiol ; 121(5): e20210352, 2024.
Article in Portuguese, English | MEDLINE | ID: mdl-38922271

ABSTRACT

BACKGROUND: The introduction of anthracyclines in the treatment of children and adolescents with cancer has promoted a significant increase in survival, but also in morbidity and mortality rates due to cardiovascular (CV) complications. OBJECTIVES: To determine the cardiovascular profile of pediatric patients treated with anthracyclines at a cancer center in Brazil and the incidence of CV complications. METHODS: The following data were collected from the medical records of patients of both sexes, aged younger than 19 years - frequency and form of clinical presentation of general CV complications (G1) and CV complications related to ventricular dysfunction (G2) - and correlated with risk factors, age range and vital status, cardiovascular and cardioprotective medications. A p<0.05 was considered statistically significant. RESULTS: A total of 326 patients were included, 214 (65.6%) were younger than 10 years and 192 (58.9%) of male sex. G1 complications occurred in 141 (43.3%) patients, and the most frequent was systemic arterial hypertension; G2 complications occurred in 84 patients (25.8%). Cumulative dose (CD) of anthracyclines > 250mg/m2 was used in 26.7% of patients and the association of G2 complications with this CD was not statistically significant (p=0.305; OR=1.330 and [95% CI = 0.770- 2.296]). The most used cardiac medications were diuretics (34.7% of patients). CONCLUSIONS: In accordance with literature, the study showed a high incidence of CV complications in the treatment of children and adolescents with cancer, with general CV complications as the most prevalent.


FUNDAMENTO: A introdução das antraciclinas no tratamento do câncer infantojuvenil propiciou um aumento significativo na sobrevida, mas também nas taxas de morbimortalidade devido às complicações cardiovasculares (CVs). OBJETIVOS: Conhecer o perfil cardiológico de pacientes pediátricos tratados com antraciclinas em um centro oncológico no Brasil e a incidência das complicações CVs. MÉTODOS: Foram coletados, de prontuários de pacientes de ambos os sexos com idade até 19 anos ­ frequência e forma de apresentação clínica das complicações CVs Gerais (G1) e relacionadas à Disfunção Ventricular (G2) ­ e correlacionados com fatores de risco, faixa etária e estado vital, medicações cardiológicas e cardioprotetoras. Um valor de p < 0,05 foi considerado significativo. RESULTADOS: Foram incluídos 326 pacientes, destes, 214 (65,6%) eram menores de 10 anos e 192 (58,89%) do sexo masculino. As complicações do G1 ocorreram em 141 (43,3%) pacientes e a mais frequente foi a hipertensão arterial sistêmica; as complicações do G2 ocorreram em 84 pacientes (25,76%). Uma Dose Cumulativa (DC) das antraciclinas > 250mg/m2 foi usada em 26,7% dos pacientes e a associação de complicações do G2 com essa DC não mostrou significância estatística (p=0,305; RC=1,330 e [95% IC= 0,770- 2,296]). As medicações cardiológicas mais usadas foram os diuréticos em 34,7% dos pacientes. CONCLUSÕES: O estudo mostrou, como na literatura, uma alta incidência de complicações CVs no tratamento do câncer infantojuvenil, sendo as do G1 as mais frequentes.


Subject(s)
Anthracyclines , Cardiovascular Diseases , Humans , Male , Female , Child , Anthracyclines/adverse effects , Brazil/epidemiology , Adolescent , Child, Preschool , Incidence , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/epidemiology , Risk Factors , Infant , Neoplasms/drug therapy , Neoplasms/complications , Retrospective Studies , Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/etiology , Sex Distribution , Young Adult
15.
J Ovarian Res ; 17(1): 135, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943148

ABSTRACT

BACKGROUND: This study aimed to investigate the mitigating effect of N-acetylcysteine (NAC) on doxorubicin (DOX)-induced ovarian and uterine toxicity in rats using laboratory tests, ultrasonographic (US) imaging, and histopathology analysis. METHODS: Forty-eight rats were divided into six groups (n = 8) as follows: Group A (control) (0.5 mL saline administered intraperitoneally [IP]), Group B (a single 10 mg/kg dose of DOX administered IP on day 1), Group C (a single 10 mg/kg dose of DOX administered IP 24 h before sacrifice), Group D (100 mg/kg of NAC administered IP for 21 days), Group E ( a single 10 mg/kg dose of DOX administered IP on day 1 and 100 mg/kg of NAC administered IP for 21 days), and Group F (100 mg/kg of NAC administered IP for 21 days and a single 10 mg/kg dose of DOX administered IP 24 h before sacrifice). The ovaries were examined using B-mode US on days 1, 14, and 21, and the histopathological examinations of the ovaries and the uterus were undertaken after sacrifice on day 22. RESULTS: Histomorphological analyses showed that ovarian weight decreased after DOX administration in Group B but not in Group E. US revealed a transient increase in ovarian size in Group B and E, reverting to baseline levels over time, as well as a progressive increase in peritoneal fluid in Groups B and E. Group B exhibited a significant decrease in the thickness of the endometrium and myometrium and uterine cornual length, which was not observed in Group E. Histopathological examination showed that DOX caused a decline in follicular count, especially in primordial, secondary, and Graafian follicles, and resulted in follicular atresia, predominantly in Group B. Destructive degeneration/necrosis and vascular changes were most prominently seen in the corpus luteum of Groups C and B. In NAC-treated rats (Groups E and F), although germ cell damage was present, atretic follicles and vascular changes, such as hyperemia and congestion, were reduced. The anti-müllerian hormone (AMH) level was the highest in Group F. CONCLUSIONS: NAC, an antioxidant, attenuated DOX-induced gonadotoxicity in rats.


Subject(s)
Acetylcysteine , Doxorubicin , Ovary , Ultrasonography , Uterus , Animals , Female , Doxorubicin/toxicity , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Rats , Ovary/drug effects , Ovary/pathology , Ovary/diagnostic imaging , Uterus/drug effects , Uterus/pathology , Uterus/diagnostic imaging , Antibiotics, Antineoplastic/toxicity , Antibiotics, Antineoplastic/adverse effects
16.
Life Sci ; 350: 122765, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38830506

ABSTRACT

BACKGROUND: Malnutrition is a common problem in developing countries, and the impact of severe malnutrition on optimal treatment outcomes of chemotherapy in pediatric cancer patients is well documented. However, despite being a more prevalent and distinct entity, moderate malnutrition is until now unexplored for its effects on treatment outcomes. AIMS: In this study we aimed to investigate the molecular basis of altered pharmacokinetics and cardiotoxicity of doxorubicin observed in early-life chronic moderate protein deficiency malnutrition. MATERIALS AND METHODS: We developed an animal model of early-life moderate protein-deficiency malnutrition and validated it using clinical samples. This model was used to study pharmacokinetic and toxicity changes and was further utilized to study the molecular changes in liver and heart to get mechanistic insights. KEY FINDINGS: Here we show that moderate protein-deficiency malnutrition in weanling rats causes changes in drug disposition in the liver by modification of hepatic ABCC3 and MRP2 transporters through the TNFα signalling axis. Furthermore, malnourished rats in repeat-dose doxorubicin toxicity study showed higher toxicity and mortality. A higher accumulation of doxorubicin in the heart was observed which was associated with alterations in cardiac metabolic pathways and increased cardiotoxicity. SIGNIFICANCE: Our findings indicate that moderate malnutrition causes increased susceptibility towards toxic side effects of chemotherapy. These results may necessitate further investigations and new guidelines on the dosing of chemotherapy in moderately malnourished pediatric cancer patients.


Subject(s)
Cardiotoxicity , Doxorubicin , Animals , Doxorubicin/pharmacokinetics , Doxorubicin/adverse effects , Rats , Cardiotoxicity/etiology , Male , Weaning , Liver/metabolism , Protein-Energy Malnutrition/metabolism , Humans , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity , Female , Disease Models, Animal , Rats, Wistar
17.
Rev Assoc Med Bras (1992) ; 70(suppl 1): e2024S106, 2024.
Article in English | MEDLINE | ID: mdl-38865526

ABSTRACT

BACKGROUND: Chemotherapy with doxorubicin may lead to left ventricular dysfunction. There is a controversial recommendation that biomarkers can predict ventricular dysfunction, which is one of the most feared manifestations of anthracycline cardiotoxicity. OBJECTIVE: The aim of this study was to evaluate the behavior of biomarkers such as Troponin I, type B natriuretic peptide, creatine phosphokinase fraction MB, and myoglobin in predicting cardiotoxicity in a cohort of women with breast cancer undergoing chemotherapy with anthracycline. METHODS: This is an observational, prospective, longitudinal, unicentric study, which included 40 women with breast cancer, whose therapeutic proposal included treatment with doxorubicin. The protocol had a clinical follow-up of 12 months. Biomarkers such as Troponin I, type B natriuretic peptide, creatine phosphokinase fraction MB, and myoglobin were measured pre-chemotherapy and after the first, third, fourth, and sixth cycles of chemotherapy. RESULTS: There was a progressive increase in type B natriuretic peptide and myoglobin values in all chemotherapy cycles. Although creatine phosphokinase fraction MB showed a sustained increase, this increase was not statistically significant. Troponin, type B natriuretic peptide, myoglobin, and creatine phosphokinase fraction MB were the cardiotoxicity markers with the earliest changes, with a significant increase after the first chemotherapy session. However, they were not able to predict cardiotoxicity. CONCLUSION: Troponin I, type B natriuretic peptide, myoglobin, and creatine phosphokinase fraction MB are elevated during chemotherapy with doxorubicin, but they were not able to predict cardiotoxicity according to established clinical and echocardiographic criteria. The incidence of subclinical cardiotoxicity resulting from the administration of doxorubicin was 12.5%.


Subject(s)
Biomarkers , Breast Neoplasms , Cardiotoxicity , Doxorubicin , Myoglobin , Troponin I , Humans , Female , Breast Neoplasms/drug therapy , Prospective Studies , Troponin I/blood , Doxorubicin/adverse effects , Cardiotoxicity/etiology , Middle Aged , Biomarkers/blood , Myoglobin/blood , Adult , Antibiotics, Antineoplastic/adverse effects , Natriuretic Peptide, Brain/blood , Aged , Creatine Kinase, MB Form/blood , Longitudinal Studies , Anthracyclines/adverse effects , Ventricular Dysfunction, Left/chemically induced , Predictive Value of Tests
19.
BMC Cardiovasc Disord ; 24(1): 260, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769516

ABSTRACT

INTRODUCTION: Use of doxorubicin, an anthracycline chemotherapeutic agent has been associated with late-occurring cardiac toxicities. Detection of early-occurring cardiac effects of cancer chemotherapy is essential to prevent occurrence of adverse events including toxicity, myocardial dysfunction, and death. OBJECTIVE: To investigate the prevalence of elevated cardiac troponin T (cTnT) and associated factors of myocardial injury in children on doxorubicin cancer chemotherapy. METHODS: Design: A cross-sectional study. SETTING AND SUBJECTS: A hospital-based study conducted on children aged 1-month to 12.4-years who had a diagnosis of cancer and were admitted at Kenyatta National Hospital (KNH). INTERVENTIONS AND OUTCOMES: The patients underwent Echocardiography (ECHO) before their scheduled chemotherapy infusion. Twenty-four (24) hours after the chemotherapy infusion the patients had an evaluation of the serum cardiac troponin T (cTnT) and a repeat ECHO. Myocardial injury was defined as cTnT level > 0.014 ng/ml or a Fractional Shortening (FS) of < 29% on ECHO. RESULTS: One hundred (100) children were included in the final analysis. Thirty-two percent (32%) of the study population had an elevated cTnT. A cumulative doxorubicin dose of > 175 mg/m2 was significantly associated with and elevated cTnT (OR, 10.76; 95% CI, 1.18-97.92; p = 0.035). Diagnosis of nephroblastoma was also associated with an elevated cTnT (OR, 3.0; 95% CI, 1.23-7.26) but not statistically significant (p = 0.105). Nine percent (9%) of the participants had echocardiographic evidence of myocardial injury. CONCLUSION: When compared to echocardiography, elevated levels of cTnT showed a higher association with early-occurring chemotherapy-induced myocardial injury among children on cancer treatment at a tertiary teaching and referral hospital in Kenya.


Subject(s)
Antibiotics, Antineoplastic , Biomarkers , Cardiotoxicity , Doxorubicin , Neoplasms , Tertiary Care Centers , Troponin T , Humans , Cross-Sectional Studies , Male , Female , Doxorubicin/adverse effects , Child , Kenya/epidemiology , Troponin T/blood , Child, Preschool , Antibiotics, Antineoplastic/adverse effects , Infant , Neoplasms/drug therapy , Neoplasms/blood , Risk Factors , Biomarkers/blood , Prevalence , Time Factors , Up-Regulation , Heart Diseases/chemically induced , Heart Diseases/epidemiology , Heart Diseases/diagnostic imaging , Heart Diseases/diagnosis , Heart Diseases/blood , Age Factors , Risk Assessment , Echocardiography
20.
Cardiovasc Toxicol ; 24(6): 563-575, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38700665

ABSTRACT

Anthracycline antibiotic is one of the most effective anti-tumor drugs used to manage certain types of breast cancers, lymphomas, and leukemias. However, anthracyclines induce a dose-dependent cardiotoxicity that may progress to heart failure. Thus, using a sensitive predictor of early cardiac dysfunction in patients treated with anthracyclines can help detect subclinical cardiac dysfunction early and help initiate interventions to protect these patients. Among parameters of myocardial measure, cardiac magnetic resonance (CMR)-measured native myocardial T1 mapping is considered a sensitive and accurate quantitative measure of early subclinical cardiac changes, particularly cardiac inflammation and fibrosis. However, to understand the quality and the validity of the current evidence supporting the use of these measures in patients treated with anthracyclines, we aimed to conduct a systematic review of clinical studies of this measure to detect early myocardial changes in cancer patients treated with anthracyclines. The primary outcome was the level of native T1 mapping. We performed fixed-effects meta-analyses and assessed certainty in effect estimates. Of the 1780 publications reviewed (till 2022), 23 were retrieved, and 9 articles met the inclusion criteria. Our study showed that exposure to anthracycline was associated with a significant elevation of native myocardial T1 mapping from baseline (95% CI 0.1121 to 0.5802; p = 0.0037) as well as compared to healthy control patients (95% CI 0.2925 to 0.7448; p < 0.0001). No significant publication bias was noted on the assessment of the funnel plot and Egger's test. According to the Q test, there was no significant heterogeneity in the included studies (I2 = 0.0000% versus healthy controls and I2 = 14.0666% versus baseline). Overall, our study suggests that native myocardial T1 mapping is useful for detecting anthracycline-induced cardiotoxicity in patients with cancer.


Subject(s)
Anthracyclines , Antibiotics, Antineoplastic , Cardiotoxicity , Heart Diseases , Neoplasms , Predictive Value of Tests , Humans , Anthracyclines/adverse effects , Neoplasms/drug therapy , Antibiotics, Antineoplastic/adverse effects , Female , Heart Diseases/chemically induced , Heart Diseases/diagnostic imaging , Heart Diseases/physiopathology , Heart Diseases/diagnosis , Male , Middle Aged , Early Diagnosis , Risk Factors , Adult , Aged , Risk Assessment , Magnetic Resonance Imaging , Ventricular Function, Left/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL