113 resultados para Cardiotoxicity


Relevância:

10.00% 10.00%

Publicador:

Resumo:

Controlled delivery of anticancer drugs through osteotropic nanoparticles (NP) is a novel approach for the adjuvant therapy of osteolytic bone metastases. Doxorubicin (DXR) is widely used in chemotherapy, although its activity is restricted by dose-dependent cardiotoxicity and marrow toxicity. However, its efficacy can be improved when specific targeting at the tumor site is obtained. The aim of this study was to obtain osteotropic biodegradable NP by nanoprecipitation of a copolymer between poly(D,L-lactide-co-glycolide) (PLGA) and an osteotropic bisphosphonate, sodium alendronate (ALE). NP were subsequently characterised for their chemical-physical properties, biocompatibility, and the ability to inhibit osteoclast-mediated bone resorption, and then loaded with DXR. The effectiveness of NP-loaded DXR was investigated through in vitro and in vivo experiments, and compared to that of free DXR. For the in vitro analysis, six human cell lines were used as a representative panel of bone tumors, including breast and renal adenocarcinoma, osteosarcoma and neuroblastoma. The in vitro uptake and the inhibition of tumor cell proliferation were verified. To analyse the in vivo activity of NP-loaded DXR, osteolytic bone metastases were induced through the intratibial inoculation in BALB/c-nu/nu mice of a human breast cancer cell line, followed by the intraperitoneal administration of the free or NP-loaded DXR. In vitro, aAll of the cell lines were able to uptake both free and NP-loaded drug, and their proliferation was inhibited up to 80% after incubation either with free or NP-loaded DXR. In addition, in vivo experiments showed that NP-loaded DXR were also able to reduce the incidence of bone metastases, not only in comparison with untreated mice, but also with free DXR-treated mice. In conclusion, this research demonstrated an improvement in the therapeutic effect of the antineoplastic drug DXR, when loaded to bone-targeted NP conjugated with ALE. Osteotropic PLGA-ALE NP are suitable to be loaded with DXR and offer as a valuable tool for a tissue specific treatment of skeletal metastases.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Die in dieser Arbeit beschriebenen Untersuchungen befassen sich mit der Identifizierung des haupt-metabolisierenden Enzyms für die Reaktion von dem Zytostatikum Doxorubizin zu dem Alkohol-Metaboliten Doxorubizinol in humanem Leberzytosol. Der Metabolismus dieser Reaktion wurde in einer humanen Leberbank analysiert, welcher eine große interindividuelle Variabilität zeigte. Da die maximale Umsatzrate häufig proportional zu der Expression des umsetzenden Enzyms in Leber ist, wurden in dieser Arbeit die Expressionsmuster der Kandidatenenzyme, verschiedene Carbonyl reduzierende Enzyme (AKRs und CBRs), ermittelt und des Weiteren der Umsatz von Doxorubizin zu Doxorubizinol in verschiedenen humanen Organen gemessen. Die metabolische Clearance der Reaktion als auch die Affinität zu Doxorubizin war am höchsten in Dünndarm, Leber und Niere, während die anderen Organe wesentlich geringere Werte zeigten. Dies steht im Einklang mit der prominenten Rolle dieser Organe in der Detoxifizierung von Fremdstoffen. Auch der Umsatz von Doxorubizin zu Doxorubizinol wurde von den verschiedenen Enzymen in sehr unterschiedlichem Maß katalysiert. AKR1C3 und CBR1 zeigten einen ausgeprägten Metabolismus von Doxorubizin während AKR1A1, AKR1B1, AKR1B10 und AKR1C4 nur geringen Maximalumsatz zeigten. Auch die Affinität von CBR1 und AKR1C3 zu Doxorubizin war wesentlich höher als die der anderen Enzyme. Inhibitoruntersuchungen zeigten weiterhin, dass ein spezifischer CBR1-Inhibitor die untersuchte Reaktion durch CBR1 als auch durch humanes Leberzytosol in gleichem Maße hemmte. Dieses Ergebnis deutet auf CBR1 als Haupt-Doxorubizin-Reduktase hin. Dies wurde in anfangs erwähnter humaner Leberbank weiter untersucht. Der Gehalt von CBR1-Protein wurde bestimmt und mit der gemessenen spezifischen Aktivität korreliert. Der Proteingehalt korrelierte mit der gemessenen spezifischen Aktivität in sechs von neun Western Blots. Dieses Ergebnis unterstützt die Vermutung, dass CBR1 als Haupt-Doxorubizin-Reduktase in humaner Leber betrachtet werden kann. Die Analyse des CBR1-Gens zeigte zwei Haplotypen, die einen signifikant verschiedenen Km-Wert zeigten als die übrigen Haplotypen. Diese Haplotypen konnten jedoch nur einen geringen Teil der Variabilität des Doxorubizin-Metabolismus erklären. Da aber ein starker interindividueller Doxorubizin-Metabolismus in humaner Leber beobachtet wird, deutet dies auf eine starke epigenetische Regulation des CBR1-Gens hin. Analysen der Promoterregion zeigten viele Bindungsstellen für Transkriptionsfaktoren; eine Aktivierung durch xenobiotische Substanzen kann vermutet werden. Der Metabolismus von Doxorubizin kann zu einer Deaktivierung des Medikaments und damit zu einer verminderten chemotherapeutischen Wirkung führen. Durch die Charakterisierung der humanen Doxorubizin-Reduktase in Leber ist die gezielte Suche nach Hemmstoffen für dieses Enzym möglich. Dies eröffnet die Möglichkeit, die Bildung von Doxorubizin zu Doxorubizinol zu verringern und die Inaktivierung der antineoplastischen Wirkung zu vermeiden. Eine Dosisverminderung und bessere Verträglichkeit des Medikaments kann erreicht, die schweren Nebenwirkungen reduziert werden. Die Identifizierung der verantwortlichen hepatischen Doxorubizin-Reduktase im Menschen kann somit einen wichtigen Beitrag leisten, die Krebstherapie in Zukunft verträglicher und nebenwirkungsfreier zu gestalten und Resistenzbildungen vorzubeugen.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

A growing number of drugs have been shown to prolong cardiac repolarization, predisposing individuals to life-threatening ventricular arrhythmias known as Torsades de Pointes. Most of these drugs are known to interfere with the human ether à-gogo related gene 1 (hERG1) channel, whose current is one of the main determinants of action potential duration. Prolonged repolarization is reflected by lengthening of the QT interval of the electrocardiogram, as seen in the suitably named drug-induced long QT syndrome. Chirality (presence of an asymmetric atom) is a common feature of marketed drugs, which can therefore exist in at least two enantiomers with distinct three-dimensional structures and possibly distinct biological fates. Both the pharmacokinetic and pharmacodynamic properties can differ between enantiomers, as well as also between individuals who take the drug due to metabolic polymorphisms. Despite the large number of reports about drugs reducing the hERG1 current, potential stereoselective contributions have only been scarcely investigated. In this review, we present a non-exhaustive list of clinically important molecules which display chiral toxicity that may be related to hERG1-blocking properties. We particularly focus on methadone cardiotoxicity, which illustrates the importance of the stereoselective effect of drug chirality as well as individual variations resulting from pharmacogenetics. Furthermore, it seems likely that, during drug development, consideration of chirality in lead optimization and systematic assessment of the hERG1 current block with all enantiomers could contribute to the reduction of the risk of drug-induced LQTS.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

The clinical use of anthracyclines in cancer therapy is limited by dose-dependent cardiotoxicity that involves cardiomyocyte injury and death. We have tested the hypothesis that anthracyclines affect protein degradation pathways in adult cardiomyocytes. To this aim, we assessed the effects of doxorubicin (Doxo) on apoptosis, autophagy and the proteasome/ubiquitin system in long-term cultured adult rat cardiomyocytes. Accumulation of poly-ubiquitinated proteins, increase of cathepsin-D-positive lysosomes and myofibrillar degradation were observed in Doxo-treated cardiomyocytes. Chymotrypsin-like activity of the proteasome was initially increased and then inhibited by Doxo over a time-course of 48 h. Proteasome 20S proteins were down-regulated by higher doses of Doxo. The expression of MURF-1, an ubiquitin-ligase specifically targeting myofibrillar proteins, was suppressed by Doxo at all concentrations measured. Microtubule-associated protein 1 light chain 3B (LC3)-positive punctae and both LC3-I and -II proteins were induced by Doxo in a dose-dependent manner, as confirmed by using lentiviral expression of green fluorescence protein bound to LC3 and live imaging. The lysosomotropic drug chloroquine led to autophagosome accumulation, which increased with concomitant Doxo treatment indicating enhanced autophagic flux. We conclude that Doxo causes a downregulation of the protein degradation machinery of cardiomyocytes with a resulting accumulation of poly-ubiquitinated proteins and autophagosomes. Although autophagy is initially stimulated as a compensatory response to cytotoxic stress, it is followed by apoptosis and necrosis at higher doses and longer exposure times. This mechanism might contribute to the late cardiotoxicity of anthracyclines by accelerated aging of the postmitotic adult cardiomyocytes and to the susceptibility of the aging heart to anthracycline cancer therapy.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

The clinical manifestations of anti-cancer drug associated cardiac side effects are diverse and can range from acutely induced cardiac arrhythmias to Q-T interval prolongation, changes in coronary vasomotion with consecutive myocardial ischemia, myocarditis, pericarditis, severe contractile dysfunction, and potentially fatal heart failure. The pathophysiology of these adverse effects is similarly heterogeneous and the identification of potential mechanisms is frequently difficult since the majority of cancer patients is not only treated with a multitude of cancer drugs but might also be exposed to potentially cardiotoxic radiation therapy. Some of the targets inhibited by new anti-cancer drugs also appear to be important for the maintenance of cellular homeostasis of normal tissue, in particular during exposure to cytotoxic chemotherapy. If acute chemotherapy-induced myocardial damage is only moderate, the process of myocardial remodeling can lead to progressive myocardial dysfunction over years and eventually induce myocardial dysfunction and heart failure. The tools for diagnosing anti-cancer drug associated cardiotoxicity and monitoring patients during chemotherapy include invasive and noninvasive techniques as well as laboratory investigations and are mostly only validated for anthracycline-induced cardiotoxicity and more recently for trastuzumab-associated cardiac dysfunction.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Inhibition of ErbB2 (HER2) with monoclonal antibodies, an effective therapy in some forms of breast cancer, is associated with cardiotoxicity, the pathophysiology of which is poorly understood. Recent data suggest, that dual inhibition of ErbB1 (EGFR) and ErbB2 signaling is more efficient in cancer therapy, however, cardiac safety of this therapeutic approach is unknown. We therefore tested an ErbB1-(CGP059326) and an ErbB1/ErbB2-(PKI166) tyrosine kinase inhibitor in an in-vitro system of adult rat ventricular cardiomyocytes and assessed their effects on 1. cell viability, 2. myofibrillar structure, 3. contractile function, and 4. MAPK- and Akt-signaling alone or in combination with Doxorubicin. Neither CGP nor PKI induced cardiomyocyte necrosis or apoptosis. PKI but not CGP caused myofibrillar structural damage that was additive to that induced by Doxorubicin at clinically relevant doses. These changes were associated with an inhibition of excitation-contraction coupling. PKI but not CGP decreased p-Erk1/2, suggesting a role for this MAP-kinase signaling pathway in the maintenance of myofibrils. These data indicate that the ErbB2 signaling pathway is critical for the maintenance of myofibrillar structure and function. Clinical studies using ErbB2-targeted inhibitors for the treatment of cancer should be designed to include careful monitoring for cardiac dysfunction.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Trastuzumab, a monoclonal antibody that blocks HER-2 receptor, improves the survival of women with HER-2-positive early and advanced breast cancer when given with chemotherapy. Lapatinib, a dual tyrosine kinase inhibitor of EGFR and HER-2, is approved for the treatment of metastatic breast cancer patients after failure of prior anthracycline, taxanes and trastuzumab therapies in combination with capecitabine. Importantly, cardiac toxicity, manifested as symptomatic congestive heart failure or asymptomatic left ventricular ejection fraction decline, has been reported in some of the patients receiving these novel anti-HER-2 therapies, particularly when these drugs are used following anthracyclines, whose cardiotoxic potential has been recognized for decades. This review will focus on the incidence, natural history, underlying mechanisms, management, and areas of uncertainty regarding trastuzumab-and lapatinib-induced cardiotoxicity.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

PDGFR is an important target for novel anticancer therapeutics because it is overexpressed in a wide variety of malignancies. Recently, however, several anticancer drugs that inhibit PDGFR signaling have been associated with clinical heart failure. Understanding this effect of PDGFR inhibitors has been difficult because the role of PDGFR signaling in the heart remains largely unexplored. As described herein, we have found that PDGFR-beta expression and activation increase dramatically in the hearts of mice exposed to load-induced cardiac stress. In mice in which Pdgfrb was knocked out in the heart in development or in adulthood, exposure to load-induced stress resulted in cardiac dysfunction and heart failure. Mechanistically, we showed that cardiomyocyte PDGFR-beta signaling plays a vital role in stress-induced cardiac angiogenesis. Specifically, we demonstrated that cardiomyocyte PDGFR-beta was an essential upstream regulator of the stress-induced paracrine angiogenic capacity (the angiogenic potential) of cardiomyocytes. These results demonstrate that cardiomyocyte PDGFR-beta is a regulator of the compensatory cardiac response to pressure overload-induced stress. Furthermore, our findings may provide insights into the mechanism of cardiotoxicity due to anticancer PDGFR inhibitors.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Antiangiogenesis is a promising anti-tumor strategy through inhibition tumor vascularformation to suppress tumor growth. Targeting specific VEGF/R has been showntherapeutic benefits in many cancer types and become a first approvedantiangiogenic modalities by Food and Drug Administration (FDA) in United States.However, interruption of homeostasis in normal tissues that is likely due to theinhibition of VEGF/R signaling pathway induces unfavorable side effects. Moreover,cytostatic nature of antiangiogenic drugs frequently causes less tumor cell specifickilling activity, and cancer cells escaped from cell death induced by these drugseven gain more malignant phenotypes, resulting in tumor invasion and metastasis.To overcome these issues, we developed a novel anti-tumor therapeutic EndoCDfusion protein which linked endostatin (Endo) to cytosine deaminase-uracilvphosphoribosyl transferase (CD). Endo targets unique tumor endothelial cells toprovide tumor-specific antiangiogenesis activity and also carries CD to the localtumor area, where it serves nontoxic prodrug 5-fluorocytosine (5-FC) enzymaticconversion reaction to anti-metabolite chemotherapy drug 5-fluorouracil (5-FU). Wedemonstrated that 5-FU concentration was highly increased in tumor sites, resultingin high level of endothelial cells and tumor cells cytotoxic efficacy. Furthermore,EndoCD/5-FC therapy decreased tumor growth and colorectal liver metastasisincident compared with bevacizumab/5-FU treatment in human breast and colorectalliver metastasis orthotropic animal models. In cardiotoxicity safety profile,EndoCD/5-FC is a contrast to bevacizumab/5-FU; lower risk of cardiotoxicityinduction or heart function failure was found in EndoCD/5-FC treatment thanbevacizumab/5-FU does in mice. EndoCD/5-FC showed more potent therapeuticefficacy with high safety profile and provided stronger tumor invasion or metastasisinhibition than antiangiogenic drugs. Together, EndoCD fusion protein with 5-FCshowed dual tumor targeting activities including antiangiogenesis and tumor localchemotherapy, and it could serve as an alternative option for antiangiogenic therapy.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Purpose: Cardiomyocytes are terminally differentiated cells in the adult heart and ischemia and cardiotoxic compounds can lead to cell death and irreversible decline of cardiac function. As testing platforms, isolated organs and primary cells from rodents have been the standard in research and toxicology, but there is a need for better models that more faithfully recapitulate native human biology. Hence, a new in vitro model comprising the advantages of 3D cell culture and the availability of induced pluripotent stem cells (iPSC) from human origin was developed and characterized. Methods: Human cardiomyocytes (CMs) derived from induced pluripotent stem cells (iPSCs) were studied in standard 2D culture and as cardiac microtissues (MTs) formed in hanging drops. 2D cultures were examined using immunofluorescence microscopy and Western blotting while the cardiac MTs were subjected to immunofluorescence, contractility, and pharmacological investigations. Results: iPSC-derived CMs in 2D culture showed well-formed myofibrils, cell-cell contacts positive for connexin-43, and other typical cardiac proteins. The cells reacted to pro-hypertrophic growth factors with a substantial increase in myofibrils and sarcomeric proteins. In hanging drop cultures, iPSC-derived cardiomyocytes formed spheroidal MTs within 4 days showing a homogeneous tissue structure with well-developed myofibrils extending throughout the whole spheroid without a necrotic core. MTs showed spontaneous contractions for more than 4 weeks that were recorded by optical motion tracking, sensitive to temperature, and responsive to electrical pacing. Contractile pharmacology was tested with several agents known to modulate cardiac rate and viability. Calcium-transients underlay the contractile activity and were also responsive to electrical stimulation, caffeine-induced Ca2+-release, extracellular calcium levels. Conclusions: 3D culture using iPSC-derived human cardiomyocytes provides an organoid human-based cellular platform that is free of necrosis and recapitulates vital cardiac functionality, thereby providing new and promising relevant model for the evaluation and development of new therapies and detection of cardiotoxicity.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Approximately 6,600 people die from acute myelogenous leukemia (AML) on an annual basis. During the past 10 to 15 years, there has been gradual overall improvements in the therapy of this disease, yet the majority of patients with AML succumb to this disease. In an attempt to improve current therapeutic strategies for AML, we became interested in a commercially available drug, dexrazoxane, which protects against anthracycline-induced cardiotoxicity. We have investigated dexrazoxane's (DEX) effects on different tissue types in an effort to determine its unique mechanism of action. Colony forming assays were used to evaluate stem-cell renewal of myeloid cells in vitro and median effect analysis was used to evaluate antagonism, synergism, or additivity. The anthracyclines, doxorubicin, daunorubicin, and idarubicin were individually combined with DEX in leukemic myeloid models to determine if the combination of the two drugs resulted in a synergistic, additive or antagonistic effect. Etoposide and cytosine arabinoside were also evaluated in combination with DEX using the same in vitro model and evaluated similarly. ^ Dexrazoxane in combination with any of the anthracyclines was schedule dependent. The combination of DEX and anthracycline resulted in a greater antitumor effect than anthracycline alone except for DEX administered 24 hours before doxorubicin or daunorubicin. These data were corroborated through median effect analysis. Etoposide in combination with dexrazoxane was synergistic for all combinations, and the combination of cytosine arabinoside and DEX was schedule dependent. In contrast, using an in vivo gastrointestinal model, DEX in combination with doxorubicin was antagonistic for almost all of the ratios used, except for the highest. A Withers' assay was used to evaluate toxicity on jejunal crypt cells. No effect was apparent for the combination of idarubicin and DEX, however, as seen with RZ, DEX in addition to radiation greatly potentiated the cytotoxic effects of radiation on crypts. These paradoxical effects of dexrazoxane were initially enigmatic, but after additional investigation, we propose a model that explains our findings. We conclude that DEX in combination with anthracyclines produces an additive to synergistic antileukemic response and may have therapeutic potential clinically. Additionally, DEX protects the gastrointestinal tract from doxorubicin toxicity, which could have clinical implications for the administration of greater doses of doxorubicin. ^

Relevância:

10.00% 10.00%

Publicador:

Resumo:

The signal transducer and activator of transcription (STAT) 3, a transcriptional factor downstream of several cytokines, is activated by Janus kinase families and plays a pivotal role in cardiac hypertrophy through gp130. To determine the physiological significance of STAT3 in vivo, transgenic mice with cardiac-specific overexpression of the Stat3 gene (STAT3-TG) were generated. STAT3-TG manifested myocardial hypertrophy at 12 wk of age with increased expression of the atrial natriuretic factor (ANF), β-myosin heavy chain (MHC), and cardiotrophin (CT)-1 genes. The animals were injected i.p. with 15 mg/kg doxorubicin (Dox), an antineoplastic drug with restricted use because of its cardiotoxicity. The survival rates after 10 days were 25% (5/20) for control littermates (WT), but 80% (16/20) for STAT3-TG (P < 0.01). WT showed increased expression of β-MHC and ANF mRNAs in the hearts 1 day after Dox treatment; this expression peaked at 3 days, suggesting that the WT suffered from congestive heart failure. Although the expression of these mRNAs was elevated in STAT3-TG hearts before Dox treatment, no additional increase was observed after the treatment. Dox administration significantly reduced the expression of the cardiac α-actin and Stat3 genes in WT hearts but not in STAT3-TG. These results provide direct evidence that STAT3 transduces not only a hypertrophic signal but also a protective signal against Dox-induced cardiomyopathy by inhibiting reduction of cardiac contractile genes and inducing cardiac protective factors.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Thesis (Ph.D.)--University of Washington, 2016-06

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Drug targeting is an active area of research and nano-scaled drug delivery systems hold tremendous potential for the treatment of neoplasms. In this study, a novel cyclodextrin (CD)-based nanoparticle drug delivery system has been assembled and characterized for the therapy of folate receptor-positive [FR(+)] cancer. Water-soluble folic acid (FA)-conjugated CD carriers (FACDs) were successfully synthesized and their structures were confirmed by 1D/2D nuclear magnetic resonance (NMR), matrix-assisted laser desorption ionization time-of-flight mass spectrometer (MALDI-TOF-MS), high performance liquid chromatography (HPLC), Fourier transform infrared spectroscopy (FTIR), and circular dichroism. Drug complexes of adamatane (Ada) and cytotoxic doxorubicin (Dox) with FACD were readily obtained by mixed solvent precipitation. The average size of FACD-Ada-Dox was 1.5–2.5 nm. The host-guest association constant Ka was 1,639 M−1 as determined by induced circular dichroism and the hydrophilicity of the FACDs was greatly enhanced compared to unmodified CD. Cellular uptake and FR binding competitive experiments demonstrated an efficient and preferentially targeted delivery of Dox into FR-positive tumor cells and a sustained drug release profile was seen in vitro. The delivery of Dox into FR(+) cancer cells via endocytosis was observed by confocal microscopy and drug uptake of the targeted nanoparticles was 8-fold greater than that of non-targeted drug complexes. Our docking results suggest that FA, FACD and FACD-Ada-Dox could bind human hedgehog interacting protein that contains a FR domain. Mouse cardiomyocytes as well as fibroblast treated with FACD-Ada-Dox had significantly lower levels of reactive oxygen species, with increased content of glutathione and glutathione peroxidase activity, indicating a reduced potential for Dox-induced cardiotoxicity. These results indicate that the targeted drug complex possesses high drug association and sustained drug release properties with good biocompatibility and physiological stability. The novel FA-conjugated β-CD based drug complex might be promising as an anti-tumor treatment for FR(+) cancer.