• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 30
  • 20
  • 5
  • 3
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 82
  • 33
  • 25
  • 23
  • 16
  • 15
  • 13
  • 10
  • 9
  • 9
  • 8
  • 8
  • 8
  • 8
  • 7
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
31

Mecanismos de Cardiotoxicidade da Doxorrubicina em Ratos Wistar e Potencial Cardioprotetor da Alda-1

Souza, Leonardo da Cunha Menezes January 2019 (has links)
Orientador: Daisy Maria Fávero Salvadori / Resumo: A cardiotoxicidade induzida pela doxorrubicina (DOX), antraciclina isolada da actinobacteria Streptomyces peucetius e amplamente utilizada na terapia antineoplásica, corresponde a um dos mais importantes eventos patofisiológicos que limitam sua aplicação clínica. No entanto, não são completamente conhecidos todos os mecanismos envolvidos nessa toxicidade, o que diminui as possiblidades de intervenção e a redução dos efeitos colaterais para os pacientes sob tratamento. Uma das hipósteses é que os aldeídos gerados pela ação da DOX atuam sobre membranas mitocondriais, alterando o estado redox e formando adutos com proteínas, os quais prejudicam o correto funcionamento da organela. Atividades deletérias da DOX sobre outros componentes celulares, como, por exemplo, os ácidos ribonucleicos, são, também, possíveis mecanismos de toxicidade do antineoplásico. Várias estratégias têm sido utilizadas para minimizar os efeitos adversos da DOX. Uma delas, é a busca por compostos que possam proteger as células da ação citotóxica. Nesse sentido, a Alda-1, pertencente ao grupo das chaperonas e agonista da enzima aldeído desidrogenase mitocondrial (ALDH2), vem sendo testada com o objetivo de reduzir os efeitos adversos dos metabólitos e radicais gerados pelo antineoplásico. Para investigar outros possíveis mecanismos de ação da DOX e o efeito cardiprotetor da Alda-1, este estudo foi delineado utilizando duas abordagens distintas: experimentos in vivo, com ratos Wistar machos submetidos a trata... (Resumo completo, clicar acesso eletrônico abaixo) / Abstract: The cardiotoxicity induced by doxorubicin (DOX), anthracycline isolated from the actinobacteria Streptomyces peucetius, and widely used as an antineoplastic drug, is one of the most important pathophysiological events that limit its clinical application. However, all the mechanisms involved in this toxicity are not fully understood. One hypothesis is that the aldehydes generated by DOX act on mitochondrial membranes, modifying the redox state and proting adducts with proteins. DOX activities on other cellular components, such as ribonucleic acids, are also possible mechanisms of toxicity. Several strategies have been used to reduce the DOX adverse effects. One of them is the identification of compounds that can protect cells against cytotoxic. Alda-1, which belongs to a group of chaperones and is an agonist of the mitochondrial aldehyde dehydrogenase (ALDH2), has been tested to reduce the adverse effects of metabolites and radicals generated by DOX. To investigate other possible DOX mechanisms of action, and the cardioprotective activity of Alda-1, this study was designed using two different approaches: in vivo, with male Wistar rats submitted to acute and chronic treatments; and, in vitro, in mice fibroblasts and cybrids with ND5 (gene that encodes the mitochondrial Complex I subunit) mitochondrial gene heteroplasmy. The expression profiling of genes related to beta oxidation pathways, Bax, Bcl-2, C1QBP, ALDH2 and miR34a microRNA (ALDH2 expression regulator), and the lipoper... (Complete abstract click electronic access below) / Doutor
32

AvaliaÃÃo do feito citoprotetor da amifostina na cardiotoxicidade aguda induzida por doxorubicina. / Evaluation of citoprotector effect of amifostine on the doxorubicin-induced acute cardiotoxicity.

Rosemayre Souza Freire 22 July 2008 (has links)
nÃo hà / Conselho Nacional de Desenvolvimento CientÃfico e TecnolÃgico / IntroduÃÃo: A Doxorubicina (DOX), antineoplÃsico antracÃclico, à largamente utilizada no tratamento dos mais diversos tipos de tumores sÃlidos e neoplasias hematolÃgicas. A Cardiotoxicidade provocada pelo uso de antibiÃticos antracÃclicos tem sido observada hà algumas dÃcadas como fator agravante e limitante do uso terapÃutico da DOX. Apesar do conhecimento de inÃmeros fatores que podem mediar a induÃÃo da cardiotoxicidade pela DOX, os mecanismos fisiopatolÃgicos continuam nÃo esclarecidos. Objetivo: Avaliar o efeito da amifostina e glutationa na cardiotoxicidade aguda induzida por doxorubicina e estudar a morfologia do tecido cardÃaco de camundongos tratados com doxorubicina por microscopia de forÃa atÃmica. Material e MÃtodos: Camundongos C57BL/6 fÃmeas (n=8) foram tratados com doxorubicina (25mg/Kg i.p.) ou salina (0,2mL i.p.) e sacrificados 96 horas apÃs tratamento. Outro grupo experimental foi tratado com amifostina (AMF 25, 50 e 100 mg/Kg s.c.), glutationa (GLT 125, 250 e 500mg/Kg s.c.) ou salina 30 mim antes da injeÃÃo de doxorubicina, no caso da glutationa a administraÃÃo foi diÃria atà o dia do sacrifÃcio. Os parÃmetros analisados foram: eletrocardiograma, Ãndices cardÃaco e esplÃnico, dosagem de grupos sulfidrilas nÃo protÃicos, dosagem de CK e CK-MB, parÃmetros histolÃgicos, dosagem por ELISA de TNF-&#61537;, IL-1&#61538;&#61484; expressÃo por imunohistoquÃmica de TNF-&#61537;, IL-1&#61538;, iNOS, apoptose e anÃlise por microscopia de forÃa atÃmica. Resultados: O tratamento com AMF nas doses de 50 e 100mg/Kg e GLT 250 e 500 mg/Kg foi capaz de aumentar a percentagem de sobrevivÃncia dos animais que foram submetidos a cardiotoxicidade aguda induzida por DOX (25 mg/Kg) quando comparados com o grupo injetado somente com DOX. A AMF e GLT tambÃm foram capazes de prevenir, em comparaÃÃo ao grupo DOX (p<0,05), as alteraÃÃes nos valores eletrocardiogrÃficos, (aumento do QRS e QTc e diminuiÃÃo da amplitude de R), as alteraÃÃes nos Ãndices cardÃacos e esplÃnicos, a elevaÃÃo dos nÃveis sÃricos das enzimas CK e CK-MB, a reduÃÃo dos nÃveis de grupos sulfidrilas nÃo protÃicos no tecido cardÃaco e as alteraÃÃes histolÃgicas (degeneraÃÃo hidrÃpica e vacuolizaÃÃo, focos de hialinizaÃÃo de fibras cardÃacas, picnose e necrose) induzidas pela DOX (25mg/Kg). A DOX induziu aumento da marcaÃÃo imunohistoquÃmica para cÃlulas apoptÃticas e expressÃo de iNOS e diminuiu a expressÃo de TNF-&#61537;. A AMF foi capaz de prevenir estas alteraÃÃes, sendo esta prevenÃÃo apenas discreta para a expressÃo de TNF-&#61537;. A microscopia de forÃa atÃmica revelou alteraÃÃes morfolÃgicas nÃo vistas pela microscopia Ãptica e mostrou ser uma ferramenta valiosa na avaliaÃÃo de efeitos de drogas. ConclusÃo: Nossos resultados sugerem o efeito citoprotetor da amifostina pelo aumento da atividade da glutationa peroxidase no tecido cardÃaco e que esta se mostra tÃo eficiente quanto a droga de referencia dexrazoxane. A utilizaÃÃo da microscopia atÃmica introduz uma ferramenta de anÃlise comparativa em escala nanomÃtrica, tornando possÃvel observar a destruiÃÃo membranar cardÃaco condizente com dano oxidativo. / IntroduÃÃo: A Doxorubicina (DOX), antineoplÃsico antracÃclico, à largamente utilizada no tratamento dos mais diversos tipos de tumores sÃlidos e neoplasias hematolÃgicas. A Cardiotoxicidade provocada pelo uso de antibiÃticos antracÃclicos tem sido observada hà algumas dÃcadas como fator agravante e limitante do uso terapÃutico da DOX. Apesar do conhecimento de inÃmeros fatores que podem mediar a induÃÃo da cardiotoxicidade pela DOX, os mecanismos fisiopatolÃgicos continuam nÃo esclarecidos. Objetivo: Avaliar o efeito da amifostina e glutationa na cardiotoxicidade aguda induzida por doxorubicina e estudar a morfologia do tecido cardÃaco de camundongos tratados com doxorubicina por microscopia de forÃa atÃmica. Material e MÃtodos: Camundongos C57BL/6 fÃmeas (n=8) foram tratados com doxorubicina (25mg/Kg i.p.) ou salina (0,2mL i.p.) e sacrificados 96 horas apÃs tratamento. Outro grupo experimental foi tratado com amifostina (AMF 25, 50 e 100 mg/Kg s.c.), glutationa (GLT 125, 250 e 500mg/Kg s.c.) ou salina 30 mim antes da injeÃÃo de doxorubicina, no caso da glutationa a administraÃÃo foi diÃria atà o dia do sacrifÃcio. Os parÃmetros analisados foram: eletrocardiograma, Ãndices cardÃaco e esplÃnico, dosagem de grupos sulfidrilas nÃo protÃicos, dosagem de CK e CK-MB, parÃmetros histolÃgicos, dosagem por ELISA de TNF-&#61537;, IL-1&#61538;&#61484; expressÃo por imunohistoquÃmica de TNF-&#61537;, IL-1&#61538;, iNOS, apoptose e anÃlise por microscopia de forÃa atÃmica. Resultados: O tratamento com AMF nas doses de 50 e 100mg/Kg e GLT 250 e 500 mg/Kg foi capaz de aumentar a percentagem de sobrevivÃncia dos animais que foram submetidos a cardiotoxicidade aguda induzida por DOX (25 mg/Kg) quando comparados com o grupo injetado somente com DOX. A AMF e GLT tambÃm foram capazes de prevenir, em comparaÃÃo ao grupo DOX (p<0,05), as alteraÃÃes nos valores eletrocardiogrÃficos, (aumento do QRS e QTc e diminuiÃÃo da amplitude de R), as alteraÃÃes nos Ãndices cardÃacos e esplÃnicos, a elevaÃÃo dos nÃveis sÃricos das enzimas CK e CK-MB, a reduÃÃo dos nÃveis de grupos sulfidrilas nÃo protÃicos no tecido cardÃaco e as alteraÃÃes histolÃgicas (degeneraÃÃo hidrÃpica e vacuolizaÃÃo, focos de hialinizaÃÃo de fibras cardÃacas, picnose e necrose) induzidas pela DOX (25mg/Kg). A DOX induziu aumento da marcaÃÃo imunohistoquÃmica para cÃlulas apoptÃticas e expressÃo de iNOS e diminuiu a expressÃo de TNF-&#61537;. A AMF foi capaz de prevenir estas alteraÃÃes, sendo esta prevenÃÃo apenas discreta para a expressÃo de TNF-&#61537;. A microscopia de forÃa atÃmica revelou alteraÃÃes morfolÃgicas nÃo vistas pela microscopia Ãptica e mostrou ser uma ferramenta valiosa na avaliaÃÃo de efeitos de drogas. ConclusÃo: Nossos resultados sugerem o efeito citoprotetor da amifostina pelo aumento da atividade da glutationa peroxidase no tecido cardÃaco e que esta se mostra tÃo eficiente quanto a droga de referencia dexrazoxane. A utilizaÃÃo da microscopia atÃmica introduz uma ferramenta de anÃlise comparativa em escala nanomÃtrica, tornando possÃvel observar a destruiÃÃo membranar cardÃaco condizente com dano oxidativo. / Introduction: Doxorubicin (DOX) is an antineoplasic anthracyclic agent used on the treatment of several solid tumors and hematological cancers. DOX-induced cardiotoxicity has been studied for decades as a limiting factor on the anticancer therapy with this drug. Despite the current knowledge concerning the mechanisms of DOX-induced cardotoxicity, its pathophysiology is still not clear. Purpose: To evaluate of the amifostine and glutathione citoprotective effect on the DOX-induced acute cardiotoxicity and to study the morphology of cardiac tissue through the use of atomic force microscopy as a tool. Materials and Methods: C57BL/6 female mice were treated with doxorubicin (25mg/Kg i.p.) or saline (0,2mL i.p.) and sacrificed 96 hours after treatment. In another experimental setting, mice were given amifostine (AMF 25, 50 e 100 mg/Kg s.c.), glutathione (GLT 125, 250 e 500mg/Kg s.c.) or vehicle, 30 mim before the administration of DOX, except glutathione that was injected daily. Analitical parameters included: electrocardiograms, cardiac and spleen indices, non protein suphidrils groups levels, CK and CK-MB cardiac enzymes levels, histological analysis, cardiac levels of (TNF-&#61537;, IL-1&#61538;, iNOS) determined by ELISA, immunohistochemistry for TNF-&#61537;, IL-1&#61538;, iNOS, apoptosis and atomic force microscopy tissue analysis. Results: AMF (100mg/Kg) and GLT (250 e 500 mg/Kg) treatments were able of improve the survival rate of animals in spite of the injection of DOX (25 mg/Kg) in comparison to DOX-treated only group (p<0.05). A AMF e GLT were also able to prevent electrocardiographic changes (rising of QRS e QTc and reduced R amplitude), changes in the cardiac and spleen indices, the augmentation of blood levels of CK e CK-MB, reduction of non proteic suphidrils groups levels, and histological changes induced by DOX (25mg/Kg). DOX induced the augmentation of the immunostaining for apoptotic cells and iNOS what was prevented by the administration of amifostine. The atomic force microscopy reveals morphological changes on the tissue organizational structure which is not possible to be observed through optical microscopy. Conclusion: Our results suggest that the amifostine citoprotective effect on DOX-induced acute cardiotoxicity is due the rising of glutathione peroxidase activity in the cardiac tissue. The citoprotective effect of amifostine is as efficient as the reference drug dexrazoxane. The use of atomic force microscopy as a new pharmacological tool for comparative analysis in nanometric scale allow us to observe DOX-induced membrane destruction what is suggestive of oxidative stress process. / Introduction: Doxorubicin (DOX) is an antineoplasic anthracyclic agent used on the treatment of several solid tumors and hematological cancers. DOX-induced cardiotoxicity has been studied for decades as a limiting factor on the anticancer therapy with this drug. Despite the current knowledge concerning the mechanisms of DOX-induced cardotoxicity, its pathophysiology is still not clear. Purpose: To evaluate of the amifostine and glutathione citoprotective effect on the DOX-induced acute cardiotoxicity and to study the morphology of cardiac tissue through the use of atomic force microscopy as a tool. Materials and Methods: C57BL/6 female mice were treated with doxorubicin (25mg/Kg i.p.) or saline (0,2mL i.p.) and sacrificed 96 hours after treatment. In another experimental setting, mice were given amifostine (AMF 25, 50 e 100 mg/Kg s.c.), glutathione (GLT 125, 250 e 500mg/Kg s.c.) or vehicle, 30 mim before the administration of DOX, except glutathione that was injected daily. Analitical parameters included: electrocardiograms, cardiac and spleen indices, non protein suphidrils groups levels, CK and CK-MB cardiac enzymes levels, histological analysis, cardiac levels of (TNF-&#61537;, IL-1&#61538;, iNOS) determined by ELISA, immunohistochemistry for TNF-&#61537;, IL-1&#61538;, iNOS, apoptosis and atomic force microscopy tissue analysis. Results: AMF (100mg/Kg) and GLT (250 e 500 mg/Kg) treatments were able of improve the survival rate of animals in spite of the injection of DOX (25 mg/Kg) in comparison to DOX-treated only group (p<0.05). A AMF e GLT were also able to prevent electrocardiographic changes (rising of QRS e QTc and reduced R amplitude), changes in the cardiac and spleen indices, the augmentation of blood levels of CK e CK-MB, reduction of non proteic suphidrils groups levels, and histological changes induced by DOX (25mg/Kg). DOX induced the augmentation of the immunostaining for apoptotic cells and iNOS what was prevented by the administration of amifostine. The atomic force microscopy reveals morphological changes on the tissue organizational structure which is not possible to be observed through optical microscopy. Conclusion: Our results suggest that the amifostine citoprotective effect on DOX-induced acute cardiotoxicity is due the rising of glutathione peroxidase activity in the cardiac tissue. The citoprotective effect of amifostine is as efficient as the reference drug dexrazoxane. The use of atomic force microscopy as a new pharmacological tool for comparative analysis in nanometric scale allow us to observe DOX-induced membrane destruction what is suggestive of oxidative stress process.
33

Découverte et mise en évidence des effets cardioprotecteurs du premier agoniste non-peptidique du récepteur-1 des prokinéticines / Discovery and cardioprotective effects of the first non-peptide agonists of the G protein-coupled prokineticin receptor-1

Gasser, Adeline 17 October 2016 (has links)
Les prokinéticines sont des hormones angiogéniques qui exercent leurs fonctions biologiques par l’intermédiaire de deux récepteurs couplés aux protéines G : PKR1 et PKR2. PKR1 a été révélé comme crucial dans l’homéostasie cardiovasculaire. L’objectif de ce projet de thèse était de développer un nouvel agoniste non–peptidique à PKR1 pour la cardioprotection et la régénération cardiaque. Les premiers résultats ont permis de caractériser le premier ligand spécifique à PKR1 : IS20. L’étude in vivo a démontré qu’IS20 est capable de prévenir les lésions après induction d’un infarctus du myocarde chez la souris. Ce composé améliore les fonctions cardiaques en activant la prolifération de cellules progénitrices cardiaques et la néovascularisation (Gasser et al, PlosOne, 2015). Dans une deuxième étude, nous avons évalué le potentiel cardioprotecteur d’IS20 face à la toxicité induite par la doxorubicine (DOX), un anticancéreux de la famille des anthracyclines très efficace mais cardiotoxique. Les résultats montrent qu’IS20 atténue l’apoptose des cardiomyocytes H9c2 et des cellules progénitrices humaines de types EPDC, induite par la doxorubicine, sans affecter la cytotoxicité de la doxorubicine sur les cellules cancéreuses. In vivo, le traitement par IS20 atténue la diminution de la prolifération provoquée par la doxorubicine dans un modèle de cardiotoxicité juvénile. Dans un modèle de cardiotoxicité chronique, IS20 maintient l’intégrité cellulaire et tissulaire des vaisseaux et protège des défaillances produites par DOX. Par ses effets cytoprotecteurs des cardiomyocytes et des cellules progénitrices cardiaques, l’IS20 présente un potentiel thérapeutique prometteur pour protéger les patients cancéreux des effets cardiotoxiques des anthracyclines. / Prokineticins are angiogenic hormones that activate two G protein-coupled receptors (GPCRs): PKR1 and PKR2. PKR1 has emerged as a critical mediator of cardiovascular homeostasis and cardioprotection. The aim of this thesis project was to develop a first non-peptide PKR1 agonist stimulates cardioprotection and cardiac regeneration in mouse model of myocardial infarction (MI) or anti-cancer drug mediated cardiotoxicity. Collaboration with chemist and biomodelization team, we characterized the first selective/specific PKR1 agonist, named IS20. In vivo study demonstrated IS20 prevented cardiac lesion formation and improved cardiac function after myocardial infarction in mice, promoting proliferation of cardiac progenitor cells and neovasculogenesis (Gasser et al., 2015). Since use of a very potent anthracycline chemotherapeutic, Doxorubicin (DOX) is limited by cardiotoxicity, we hypothesized that IS20 could protect heart against DOX-mediated cardiotoxicity. Indeed, IS20 attenuated apoptosis induced by DOX in H9c2 cardiomyocytes and human epicardial progenitors in vitro. However, IS20 did not affect antineoplastic or cytostatic effect of DOX in cancer cell lines. In vivo, in the juvenile model of cardiotoxicity, IS20 significantly attenuated DOX-induced decrease in viability and proliferation cardiac progenitor cells. In the chronic cardiotoxicity model by DOX, IS20 improves heart structure and function by the activation of cardiac progenitor cells, diminishing cardiac cell death, improving vascular stability. IS20 has translational potential for cardioprotection in patients with cancer receiving anthracyclines.
34

Melatonin's Protection against DNA Damage by the Iron (III)-Adriamycin Complex.

Campbell, Sharon E. 01 August 2001 (has links)
Adriamycin is a first line cancer treatment drug for breast cancer and many soft tissue carcinomas. Adriamycin is a highly effective anti-cancer drug. It has a fused ring system that is planar, hydrophobic and electron rich. These features allow intercalation into DNA which, along with its topoisomerase inhibition, results in its anti-cancer effectiveness. Despite its success, its use is limited by a dose-dependent cardiotoxicity. Adriamycin forms tight complexes with iron in varying iron:drug ratios. The chelated complex [Fe+3-Adriamycin (1:2)] oxidatively cleaves DNA via the generation of reactive oxygen species (ROS). Enzymes associated specifically with heart mitochondria increase ROS formation explaining why adriamycin is selectively cardiotoxic. Pretreatment of mice with the neural hormone melatonin eliminates the cardiotoxic effectiveness of adriamycin therapy without reducing the drugÆs antitumor effect (Wahab et al. 2000, Tumori 86: 157-162). This has recently been verified in human cancer patients (Lissoni et al. 1999, European Journal of Cancer 35: (12) 1688-1692). Melatonin is soluble in both lipid and aqueous environments and has no known side-effects. This study analyzes the mechanistic features of DNA damage by Fe+3-Adriamycin and how melatonin ameliorates this damage. An Fe+3-Adriamycin (1:2) complex + glutathione cleaves DNA; this oxidative DNA cleavage does not occur with adriamycin +/- glutathione or with FeADR in the absence of glutathione. Melatonin reduces this oxidative DNA cleavage by 67%. Using a supercoiled-to-nicked-circular-conversion assay in conjunction with spectroscopic analyses give results revealing : 1) Fe+3-Adriamycin (1:2) complex + glutathione forms a reactive intermediate. 2) Melatonin protects the DNA from cleavage by this reactive intermediate. 3) Enzymatic assays show that H2O2 is the primary ROS formed with downstream production of the hydroxy radical via the Fenton reaction. 4) Fe+3-Adriamycin (1:2) intercalates into DNA to the same degree as uncomplexed adriamycin. 5) Melatonin also binds to DNA but not by intercalation. These experiments indicate that the cardioprotective effect of melatonin in adriamycin therapy may stem from melatoninÆs interaction with a reactive intermediate from Fe+3-Adriamycin(1:2) complex + glutathione and/or a direct interaction of melatonin with DNA.
35

Impaired Cardiorespiratory Fitness Following Thoracic Radiotherapy

Canada, Justin M 01 January 2018 (has links)
Cancer (CA) is the second leading cause of death in the United States preceded only by cardiovascular disease (CVD). Over the past 30 years, the 5-year survival rate for all cancers combined has increased by more than 20%. This improved survival rate is due to early diagnosis and advances in treatment involving a multimodality treatment approach that includes radiotherapy [RT] with about half of all CA patients receiving some type of RT sometime during the course of their treatment. Cardiotoxicity is one of the most important adverse reactions of RT and leads to a meaningful risk of CVD-related morbidity and mortality. Radiotherapy-related cardiotoxicity is a heterogeneous clinical syndrome characterized by symptoms related to impaired cardiac function due to radiation-injury to one or more cardiac structures. Furthermore, the relative risk of CVD increases with increasing incidental radiation dose to the heart. There is not a unified consensus on the definition of CA-related cardiotoxicity although most trials have focused on changes in resting systolic function, and/or development of cardiac symptoms.Commonly used tools to assess cardiac function are insensitive to minor injury hence subtle changes may go unnoticed for many years. Cardiotoxicity definitions should include a dynamic functional assessment of the CV system. This may allow detection of latent CV abnormalities before the precipitous decline of resting myocardial function or the development of CV symptomology that may impact quality of life. Cardiopulmonary exercise testing (CPET) including measurement of peak oxygen consumption (VO2) is the gold standard for the assessment of cardiorespiratory fitness (CRF). Cardiorespiratory fitness is a strong, independent predictor of mortality, CVD-related mortality, HF-related morbidity and mortality, CA-related mortality and may be involved in the pathophysiologic link between anti-CA related treatments and the increased risk of late CVD events. Emerging evidence indicates CRF may be reduced in CA survivors and have utility to detect subclinical cardiotoxicity, but this has not been evaluated in CA survivors treated with RT with significant heart involvement. This dissertation consists of one literature review and one comprehensive paper that will examine the ability of CPET to detect subclinical cardiotoxicity.
36

Detecção e manejo de disfunções cardíacas em pacientes com esclerose sistêmica tratados com altas doses de ciclofosfamida seguidas por transplante de células-tronco hematopoéticas / Detection and management of cardiac disfunction in patients with systemic sclerosis treated with high dose cyclophosphamide followed by hematopoietic stem cell transplantation

Leopoldo, Vanessa Cristina 17 December 2018 (has links)
O transplante autólogo de células-tronco hematopoéticas (TACTH) é efetivo para o tratamento da esclerose sistêmica (ES), com controle do acometimento pulmonar, e melhora da fibrose cutânea, da qualidade de vida e aumento da sobrevida global, quando comparado a pacientes não transplantados. Neste tratamento, utiliza-se ciclofosfamida em altas doses, uma droga imunossupressora associada a cardiotoxicidade, potencialmente fatal. A avaliação de potenciais fatores de risco e monitorização clínica durante o procedimento podem contribuir para identificar precocemente a lesão cardíaca aguda e, assim, melhorar o desfecho clínico do paciente. O objetivo deste estudo foi identificar a ocorrência de disfunções cardíacas e avaliar os fatores de risco clínicos e laboratoriais para o desenvolvimento de toxicidade cardíaca aguda induzida pela ciclofosfamida, em pacientes com ES submetidos ao TACTH. Trata-se de um estudo longitudinal e prospectivo, conduzido no Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto, em pacientes com ES com idade superior a 18 anos, no período de novembro de 2016 a maio de 2018, aprovado por Comitê de Ética em Pesquisa. Os pacientes foram avaliados quanto aos dados clínicos e laboratoriais, incluindo dosagem de peptídeo natriurético, no período pré, durante e 6 meses após o transplante. Foram incluídas 16 mulheres com ES, das quais uma optou por descontinuar a participação no estudo, tendo seus dados excluídos. Nenhum dos participantes apresentava história de uso de substâncias psicoativas ou diagnóstico de diabetes mellitus. Uma paciente era tabagista passiva e uma era tabagista ativa há 24 anos, abstinente desde há três meses antes do início do transplante. Duas pacientes eram hipertensas, controladas com medicamentos anti-hipertensivos. Durante o transplante, 7 (46,7%) pacientes apresentaram alterações cardíacas associadas à ciclofosfamida. As pacientes apresentaram taquicardia, ganho ponderal, aumento de pressão venosa central e dispneia iniciados em menos de 24 horas e até 4 dias após o término da infusão da dose total de ciclofosfamida (200mg/kg) do transplante. Em três, de cinco pacientes submetidas a ecocardiografia, foram detectadas alterações sugestivas de disfunção cardíaca, corroborando os achados clínicos. Uma paciente evoluiu com choque refratário e posterior óbito por falência múltipla de órgãos. Uma paciente necessitou de pericardiocentese de alívio e, nos demais, o manejo com medicações reverteu as alterações clínicas. A dosagem dos níveis séricos de peptídeo natriurético mostrouse mais elevada (p<0,0005) nos pacientes que apresentaram sinais de toxicidade cardíaca. Concluímos que as avaliações clínicas sistematizadas por equipe de enfermagem permitiram a detecção de disfunções cardíacas pós-infusão de altas doses de ciclofosfamida, quadros retrospectivamente comprovados por elevação dos níveis sérico de peptídeo natriurético. O número reduzido de participantes não permitiu fazer análises estatísticas preditoras de cardiotoxicidade e foi uma limitação do estudo. Futuramente, objetivamos aumentar o número de pacientes do estudo e identificar marcadores preditivos de toxicidade cardíaca antes e durante o transplante / Autologous hematopoietic stem cell transplantation (AHSCT) is effective for the treatment of systemic sclerosis (SSc), with stabilization of pulmonary involvement and improvement of cutaneous fibrosis, quality of life and overall survival, when compared to non-transplanted patients. Transplant includes high dose cyclophosphamide, an immunosuppressive drug associated with potentially fatal cardiotoxicity. The evaluation of potential risk factors and clinical monitoring during the procedure may contribute to early identification of acute cardiac injury and thus improve the patient\'s clinical outcome. The aim of this study was to detect cardiac dysfunctions and to evaluate clinical and laboratory risk factors for the development of acute cardiac toxicity induced by cyclophosphamide in SSc patients submitted to AHSCT. This is a longitudinal and prospective study, conducted in the University Hospital of the Ribeirão Preto Medical School (Brazil), in patients with SSc, older than 18 years of age, from November 2016 to May 2018. The protocol has been approved by the Institutional Research Ethics Committee and all patients signed informed consent. Patients were evaluated for clinical and laboratory data, including natriuretic peptide dosage before, during and at the 6 months post-transplant time point Sixteen women with SSc were included, one of whom chose to discontinue participation in the study, having their data excluded. None of the participants had a history of psychoactive substance abuse or a diagnosis of diabetes mellitus. One patient was a passive smoker and another, an active smoker for 24 years, not smoking in the three months before transplantation. Two patients had their blood pressure controlled with antihypertensive drugs. During transplantation, 7 (46.7%) patients had cardiac changes associated with cyclophosphamide. The patients presented tachycardia, weight gain, increased central venous pressure and dyspnoea initiated in less than 24 hours and up to 4 days after the end of the infusion of the total cyclophosphamide dose (200mg / kg) of the transplant. In three of five patients investigated by echocardiography, alterations suggestive of cardiac dysfunction were detected, corroborating the clinical findings of cardiac dysfunction. One patient evolved with refractory shock and subsequent death due to multiple organ failure. One patient required pericardiocentesis due to cardiac tamponade and, in the others, management with medications reversed the clinical alterations. Serum natriuretic peptide levels were higher (p<0,0005) in the patients with than in the patients without any signs of cardiac toxicity. Clinical evaluations by the nursing staff allowed the detection of cardiac dysfunctions after infusion of high dose cyclophosphamide, retrospectively confirmed by elevation of serum levels of natriuretic peptide. The reduced number of participants did not allow for statistical analyzes to predict cardiotoxicity and was a limitation of the study. In the future, we aim to increase the number of patients in the study and to identify predictive markers of cardiotoxicity before and during transplant procedure
37

The cardioprotective role of NACA in the prevention of Doxorubicin and Trastuzumab mediated cardiac dysfunction

Goyal, Vineet 04 September 2015 (has links)
Rationale: In the breast cancer setting, anti-cancer therapies, including Doxorubicin (DOX) and Trastuzumab (TRZ), are associated with an increased risk of cardiotoxicity. There is a need to develop prophylactic cardioprotective agents to mitigate the cardiotoxic side effects of these common anti-cancer drugs. Objective: To investigate whether the anti-oxidant, N-acetylcysteine amide (NACA), can attenuate the drug-induced heart failure caused by DOX+TRZ in a murine model. Methods: A total of 100 female mice received one of the following drug regimens: i) saline; ii) NACA; iii) DOX; iv) TRZ; v) DOX+TRZ; vi) NACA+DOX; vii) NACA+TRZ; and viii) NACA+DOX+TRZ. Serial echocardiography was performed over a 10-day study period, after which the mice were euthanized for histological and biochemical analyses. Results: In mice receiving DOX, left ventricular ejection fraction (LVEF) decreased from 73±4% to 43±2% at day 10. In mice receiving DOX+TRZ, LVEF decreased from 72±3% to 32±2% at day 10. Prophylactic administration of NACA to mice receiving DOX or DOX+TRZ was cardio-protective with an LVEF of 62±3% and 55±3% at day 10, respectively. Histological and biochemical analyses demonstrated loss of cellular integrity, increased oxidative stress (OS), and increased cardiac apoptosis in mice treated with DOX+TRZ which was attenuated by the prophylactic administration of NACA. Conclusion: NACA attenuates the cardiotoxic side effects of DOX+TRZ in a murine model of chemotherapy induced cardiac dysfunction by decreasing OS and apoptosis. / October 2015
38

Treatment of local anaesthetic induced cardiotoxicity with lipid infusion / Behandlung der Lokalanästhetikatoxicität mit Lipidinfusion

Keil, Meike 24 November 2009 (has links)
No description available.
39

Early detection of broken hearts in cancer: Bevacizumab and Sunitinib mediated cardiotoxicity

Bordun, Kimberly-Ann 26 August 2014 (has links)
Background: Although Bevacizumab (BVZ) and Sunitinib (SNT) prolong survival in cancer patients, an unanticipated side-effect is cardiotoxicity. Early indices of left ventricular (LV) systolic dysfunction would be useful to address the cardiac safety of anti-cancer drugs. Objective: Whether cardiac biomarkers, tissue velocity imaging (TVI), and/or strain rate (SR) can detect early cardiac dysfunction. Methods: A total of 95 C57Bl/6 mice received one of the following drug regimens: i) 0.9% saline; ii) BVZ; or iii) SNT and followed for 14 days. Serial blood pressure, high sensitivity troponin I (hsTnI), and echocardiography were performed. Results: BVZ- and SNT-treated mice demonstrated an increase in mean arterial blood pressure, hsTnI, cardiac apoptosis, and loss of cell integrity. TVI and SR values confirmed early LV systolic dysfunction at day 8, compared to conventional LVEF at day 13. Conclusions: Novel imaging techniques can detect early LV systolic dysfunction in a model of drug-mediated cardiomyopathy.
40

Efeito antitumoral, citotoxicidade e farmacocinética da doxorrubicina incorporada em nanopartículas poliméricas e comparada a doxorrubicina comercializada na forma cloridrato e lipossomal. /

Candido, Caroline Damico. January 2018 (has links)
Orientador: Rosangela Gonçalves Peccinini / Resumo: A Doxorrubicina (Dox) é uma antraciclina modelo amplamente utilizada em diversos tipos de neoplasias e, assim como a maioria dos quimioterápicos antineoplásicos, a Dox apresenta uma série de efeitos adversos, dentre eles os mais significativos são os efeitos cardiovasculares como hipotensão, taquicardia e insuficiência cardíaca congestiva (ICC), que limitam a terapia com este fármaco. A cardiotoxicidade é favorecida pela sua ampla distribuição no miocárdio, e a Dox tem sidoconsiderada a de maior efeito tóxico sobre o tecido cardíaco. A utilização de recursos em tecnologia farmacêutica que modifiquem o perfil farmacocinético e a toxicidade é uma importante ferramenta para a introdução de um novo produto no mercado cuja resulte em desfecho clínico mais favorável ao paciente. Pesquisadores da Universidade Federal do Rio Grande do Norte (UFRN) desenvolveram um sistema de nanopartículas (Np) para a veiculação de Dox com o intuito de modificar suas características farmacocinéticas e de toxicidade. No presente trabalho o objetivo foi avaliar a nova formulação proposta e compará-la às formulações comerciais de Dox (na forma de cloridrato e lipossomal) quanto a atividade antitumoral in vitro e in vivo e quanto às suas características farmacocinéticas. Foi abordada a atividade in vitro em ensaios de citotoxicidade como o ensaio de Resazurina (RSZ), sulforrubina B (SRB) e a determinação de Trisfosfato de Adenosina (ATP) realizados em cardiomiblasto (H9c2 (2-1), ATCC® CRL 1446™) e célula... (Resumo completo, clicar acesso eletrônico abaixo) / Doutor

Page generated in 0.0433 seconds