11 research outputs found

    Hepatitis C Worldwide and in Brazil: Silent Epidemic—Data on Disease including Incidence, Transmission, Prevention, and Treatment

    Get PDF
    Hepatitis C virus (HCV) is endemic worldwide and according to the World Health Organization (WHO), there are about 150 million chronic carriers worldwide. The infection is a leading cause of liver diseases like cirrhosis and hepatocellular carcinoma (HCC); thus, HCV infection constitutes a critical public health problem. There are increasing efforts worldwide in order to reduce the global impact of hepatitis C through the implementation of programmatic actions that may increase the awareness of viral hepatitis and also improve surveillance, prevention, and treatment. In Brazil, about 1,5 million people have been chronically infected with HCV. The country has a vast territory with uneven population density, and hepatitis C incidence rates are variable with the majority of cases concentrated in the most populated areas. Currently, the main priorities of Brazilian Ministry of Health's strategies for viral hepatitis management include the prevention and early diagnosis of viral hepatitis infections; strengthening of the healthcare network and lines of treatment for sexually transmitted diseases, viral hepatitis, and AIDS; improvement and development of surveillance, information, and research; and promotion of universal access to medication. This review aims to summarize the available data on hepatitis C epidemiology and current status of efforts in prevention and infection control around the world and in Brazil

    Pharmacological potential of Tetradenia riparia plant and its derivatives: A scoping review

    Get PDF
    TCC(graduação) - Universidade Federal de Santa Catarina. Centro de Ciências da Saúde. Farmácia.Esta revisão de escopo para aumentar o potencial farmacológico da planta e sua derivados para a saúde. Conduzimos este estudo de acordo com as recomendações PRISMA e registrado no OSF (doi: 10.17605 / OSF.IO / 3QFBG). Procuramos em três bancos de dados usando os termos livres combinados com operadores booleanos: Tetradenia riparia OU Iboza OU Moschosma. A estratégia PICOS foi aplicada para formular a pesquisa questão, critérios de elegibilidade (inclusão e exclusão) e extração de dados. Dois revisores independentes e cegos conduziram a fase de seleção e extração de dados, e um especialista removeu as discrepâncias. Um total de 24 estudos foram incluídos. O folhas e óleos essenciais foram estudados em sua maioria, seguindo extratos brutos e frações. ou compostos isolados. Uma planta foi obtida em regiões subtropicais, principalmente ao sul de Brasil. A atividade antimicrobiana foi relatada para alguns estudos in vitro, como antileishmania, bactéria anti-tuberculose e ação helmíntica. Além disso, T. riparia mostrou antioxidante efeitos acaricidas e imunomoduladores. O uso terapêutico desta planta e seus derivados para tratar doenças infecciosas e inflamatórias eram potenciais, mas limitados desde quanto à citotoxicidade e poucos estudos in vivo e humanos publicados.This scoping review to raise the pharmacological potential of the plant and its derivatives for health. We conducted this study according to PRISMA recommendations and registered in OSF (doi:10.17605/OSF.IO/3QFBG). We searched in three databases using the free terms combined with boolean operators: Tetradenia riparia OR Iboza OR Moschosma. PICOS strategy was applied to formulate the search question, eligibility criteria (inclusion and exclusion) and data extraction. Two independent and blinded reviewers conducted the selection and data extraction phase, and an expert removed the discrepancies. A total of 24 studies were included. The leaves and essential oils were majority studied, following crude extracts and fractions or isolated compounds. A plant was obtained in subtropical regions, mainly South of Brazil. Antimicrobial activity was reported for some in vitro studies, as antileishmanial, anti-tuberculosis bacteria, and helminthic action. Also, T. riparia showed antioxidant effects, acaricidal and immunomodulatory. The therapeutic use of this plant and its derivatives to treat infectious and inflammatory diseases was potential, but limited since regarding the cytotoxicity and few studies in vivo and humans published

    Combined use of simvastatin and paclitaxel associated to a lipidic nanoemulsion in cancer treatment

    No full text
    Uma nova alternativa para o tratamento do câncer foi proposta em estudos anteriores, consistindo no uso de uma nanoemulsão lipídica como transportadora de agentes quimioterápicos às células neoplásicas. A redução da toxicidade da quimioterapia promovida pelo direcionamento específico de quimioterápicos às células tumorais nos levou a testar o potencial de aplicação do sistema de nanopartículas lipídicas na terapêutica combinada do paclitaxel com a sinvastatina, um agente hipolipemiante que pode ser empregado como coadjuvante no tratamento do câncer. Nos dias 11, 14 e 19 após a inoculação de células de melanoma B16F10, camundongos C57BL/6J receberam pela via intraperitoneal soluções de oleato de paclitaxel associado à nanoemulsão lipídica 17,5µmol/kg (Nano-paclitaxel), formulação comercial do paclitaxel 17,5µmol/kg, nanoemulsão lipídica (Nanoemulsão) e solução salina (Controle). A sinvastatina 50mg/kg/dia foi administrada por gavagem do 11° ao 19° dia após a inoculação do tumor em um dos grupos de animais tratados com o Nano-paclitaxel (Nano-paclitaxel + Sinva), no grupo tratado com a formulação comercial do paclitaxel (Paclitaxel + Sinva) e como monoterapia (Sinva). Camundongos Balb-c saudáveis receberam os mesmos tratamentos para avaliação dos possíveis efeitos tóxicos dos diferentes tratamentos. A terapia combinada Nano-paclitaxel + Sinva apresentou toxicidade negligível em comparação com a terapia combinada Paclitaxel + Sinva que provocou perda de peso e mielossupressão nos animais. Nos animais portadores de tumor, o tratamento Nano-paclitaxel + Sinva inibiu 95% do crescimento tumoral, comparado à inibição de 44% promovida pelo tratamento Paclitaxel + Sinva. Além disso, apenas 37% dos animais portadores de melanoma submetidos ao tratamento com Nano-paclitaxel + Sinva apresentaram metástases, em contraste com 90% dos tratados com Paclitaxel + Sinva. A probabilidade de sobrevida também foi maior nos camundongos tratados com o Nano-paclitaxel + Sinva em comparação aos tratados com Paclitaxel + Sinva. A análise de amostras de tumores por citometria de fluxo mostrou que somente nos grupos de animais tratados com Sinva, Nano-paclitaxel ou com a combinação Nano-paclitaxel + Sinva houve aumento na expressão de p21 em comparação ao grupo Controle. Da mesma forma, apenas nos grupos Sinva e Nano-paclitaxel + Sinva houve redução na expressão de ciclina D1 em comparação ao grupo Controle. O teste de viabilidade celular com rodamina 123 mostrou despolarização da membrana mitocondrial com redução no número de células tumorais viáveis em todos os grupos de tratamentos em comparação aos grupos Nanoemulsão e Controle. A avaliação histológica dos tumores demonstrou que os grupos Nanoemulsão e Controle apresentaram alta densidade de células tumorais, diferentemente dos demais grupos de tratamento e que apenas os tumores do grupo Nano-paclitaxel + Sinva apresentaram aumento na presença de fibras de colágeno tipo I e III. Em comparação ao grupo Controle, os tumores dos grupos Sinva, Paclitaxel + Sinva, Nano-paclitaxel e Nano-paclitaxel + Sinva apresentaram redução na expressão imunohistoquímica de ICAM, MCP-1 e MMP-9 sendo que o grupo Nano-paclitaxel + Sinva apresentou a menor porcentagem de área marcada positivamente para a MMP-9. A terapia combinada com Nano-paclitaxel + Sinva é menos tóxica e mais efetiva na inibição do crescimento tumoral do que a mesma terapia com a formulação comercial do paclitaxel.In previous studies we have proposed a novel approach for cancer treatment consisting of the use of a lipid nanoemulsion as a vehicle to direct chemotherapeutic agents to neoplastic cells. Reduction of chemotherapy toxicity promoted by specific targeting of antineoplastic agents to tumor cells led us to test the application of the lipidic nanoparticle system in combined treatment with paclitaxel and simvastatin, a cholesterol-lowering drug that can be used as coadjuvant in cancer treatment. On days 11, 14 and 19 after B16F10 melanoma cells inoculation, C57BL/6J mice were intraperitoneally injected with paclitaxel oleate associated to the lipidic nanoemulsion 17.5 µmol/kg (Nano-paclitaxel), commercial formulation of paclitaxel 17.5 µmol/kg, lipidic nanoemulsion (Nanoemulsion) or saline solution (Control). Simvastatin 50 mg/kg/day was administered by gavage from days 11 to 19 after tumor inoculation in one group of animals treated with Nano-paclitaxel (Nano-paclitaxel + Simva), in the group treated with commercial formulation of paclitaxel (Paclitaxel + Simva) and as monotherapy (Simva). Evaluation of possible toxic effects of the treatments was accessed in healthy Balb-c mice. Combined therapy with Nano-paclitaxel + Simva showed negligible toxicity as compared with the combination of Paclitaxel + Simva which resulted in animal weight loss and myelosuppression. In tumor-bearing animals, treatment with Nano-paclitaxel + Simva resulted in a remarkable tumor growth inhibition rate of 95%, compared to a 44% inhibition rate promoted by treatment with Paclitaxel + Simva. Moreover, only 37% of melanoma bearing animals treated with Nano-paclitaxel + Simva developed metastasis, in contrast to 90% of those treated with Paclitaxel + Simva. Survival rates were also higher in mice treated with Nano-paclitaxel + Simva in comparison to Paclitaxel + Simva treated animals. Analysis of tumor samples by flow cytometry showed that only animals treated with Simva, Nano-paclitaxel or Nano-paclitaxel + Simva increased the expression of p21 in comparison to Control group. Also, tumors from animals treated with Simva or Nano-paclitaxel + Simva presented a decrease in the expression of cyclin D1 in comparison to Control group. Cell viability test with rhodamine 123 showed mitochondrial membrane depolarization with reduction of tumor viable cells in all treatment groups in comparison to Nanoemulsion and Control groups. The histological study revealed that in contrast to drugs treated groups, tumors from Nanoemulsion and Control groups presented high tumor cell density and only Nano-paclitaxel + Simva treated animals presented tumors with increased presence of collagen fibers I and III. In comparison to Control group, tumors from groups Simva, Paclitaxel + Simva, Nano-paclitaxel and Nano-paclitaxel + Simva showed a reduction in immunohistochemical expression of ICAM, MCP-1 and MMP-9 and the group Nano-paclitaxel + Simva presented the lowest percentage of area positively stained for MMP-9. Combined therapy with Nano-paclitaxel + Simva was less toxic and more effective in promoting tumor growth inhibiton than the same combined therapy with the commercial formulation of paclitaxel

    Antitumor Combined Therapy of paclitaxel and etoposide derivatives associated to a cholesterol-rich nanoemulsion LDE

    No full text
    A LDE é uma nanoemulsão rica em colesterol com composição semelhante às lipoproteínas naturais. Devido a sua capacidade de ligação aos receptores da lipoproteína de baixa densidade (LDL) cuja expressão é aumentada em células neoplásicas, a LDE pode ser usada como veículo de agentes quimioterápicos concentrando os mesmos no tecido tumoral. A base racional da quimioterapia combinada é utilizar medicamentos que atuem em diferentes partes dos processos metabólicos da célula, aumentando dessa forma a probabilidade de destruição de uma maior quantidade de células tumorais. O principal fator limitante do uso combinado dos quimioterápicos paclitaxel e etoposídeo é o efeito mielossupressor. O direcionamento específico do fármaco às células tumorais promovido pela LDE, tornaria possível a terapia antitumoral combinada LDE-oleato de paclitaxel e LDEoleato de etoposídeo, sem produzir os efeitos colaterais observados com o uso combinado das formulações comerciais. O presente estudo tinha como objetivo avaliar os efeitos da terapia antitumoral combinada LDE-oleato de paclitaxel e LDEoleato de etoposídeo em comparação com a terapia dos respectivos fármacos isoladamente, bem como com a terapia combinada do paclitaxel e etoposídeo comerciais. A atividade antitumoral foi determinada em camundongos portadores de melanoma após a administração de intraperitoneal de LDE-oleato de paclitaxel 15 mg/kg e LDE-oleato de etoposídeo 10 mg/kg. Nos grupos dos fármacos comerciais injetou-se paclitaxel 3,75 mg/kg e etoposídeo 2,5 mg/Kg, ou paclitaxel 7,5 mg/Kg e etoposídeo 5 mg/kg. Os fármacos foram administrados em dois esquemas terapêuticos: 11º, 13º, 15º; e 11°, 14°, 19° dias após a inoculação do tumor. Todos os grupos apresentaram aumento na taxa de sobrevida em comparação ao grupo controle. Por outro lado, nos grupos LDE-fármacos foi observada uma redução dos efeitos de mielossupressão, visto que as doses administradas nestes grupos foram 2 e 4 vezes maiores do que nos demais. Os tratamentos do melanoma nos dois protocolos terapêuticos mostraram-se eficazes na redução da massa tumoral, sendo seu efeito inibitório acima de 90% nos grupos LDE-fármacos e na combinação dos quimioterápicos comerciais de maior dose. O número de metástases foi menor nos grupos de combinação LDE-Fármacos. Os resultados mostraram que a terapia antitumoral combinada LDE-oleato de paclitaxel e LDE-oleato de etoposídeo foi mais eficaz que as terapias isoladas destes fármacos e que a terapia combinada com o paclitaxel e etoposídeo comerciais.LDE is a cholesterol-rich emulsion with similar composition to the natural lipoproteins. Due to its ability of binding to the Low Density Lipoprotein (LDL) receptors and concentrate in neoplastic cells which present overexpression of these receptors, LDE may be used as vehicle to target antineoplastic drugs against cancer cells. Combined chemotherapy is a therapeutic strategy against cancer that usually uses drugs that act in different parts of cells metabolism, increasing the chances of destruction of the cancer cells. In addition, adverse effects can be reduced when combining agents with different toxicities at lower doses than the usual in single therapy. The major limiting factor in combining the chemotherapeutic agents paclitaxel and etoposide is their side effects such as leukopenia, thrombocytopenia and anemia. The ability of LDE to carry the drug into the cell may bring a strong possibility of combining LDE- paclitaxel oleate and LDE-etoposide oleate without producing the toxic effects observed in the combined use of the commercial formulations of these drugs. The current study was designed to evaluate the effects of combined antitumoral therapy of LDE- paclitaxel oleate and LDE-etoposide oleate in comparison to the combination of commercial paclitaxel and etoposide formulations and to the single therapy with the same LDEagents. Antitumoral activity was determined in melanoma-bearing mice after injection of LDE-paclitaxel oleate 15 mg/kg and LDE-etoposide oleate 10 mg/kg, or commercial paclitaxel 3.75 mg/kg and etoposide 2.5 mg/kg, or commercial paclitaxel 7,5 mg/kg and etoposide 5 mg/kg. Drugs were administered in two protocols: 11º, 13º, 15º; and 11°, 14°, 19° days after tumor implantation. All groups had an increase in the survival rate in comparison to the control group, however LDE combination groups showed reduction in the bone marrow toxicity, since the doses used were 2 and 4 fold greater that in the commercial drugs groups. The tumor growth inhibition rate was greater in both LDE-drugs combination groups and in the higher dosage of paclitaxel and etoposide combination (over 90% in comparisson to the control). There were also great reduction of metastatic nodes in LDE-drugs combination groups in comparison to the commercial drugs combination groups. Our results showed that the antitumor combined therapy of LDE-paclitaxel oleate and LDE-etoposide oleate was more effective than the therapies with LDE-paclitaxel oleate and LDE-etoposide oleate alone, and the combined therapy of commercial paclitaxel and etoposide formulations

    Antitumor Combined Therapy of paclitaxel and etoposide derivatives associated to a cholesterol-rich nanoemulsion LDE

    No full text
    A LDE é uma nanoemulsão rica em colesterol com composição semelhante às lipoproteínas naturais. Devido a sua capacidade de ligação aos receptores da lipoproteína de baixa densidade (LDL) cuja expressão é aumentada em células neoplásicas, a LDE pode ser usada como veículo de agentes quimioterápicos concentrando os mesmos no tecido tumoral. A base racional da quimioterapia combinada é utilizar medicamentos que atuem em diferentes partes dos processos metabólicos da célula, aumentando dessa forma a probabilidade de destruição de uma maior quantidade de células tumorais. O principal fator limitante do uso combinado dos quimioterápicos paclitaxel e etoposídeo é o efeito mielossupressor. O direcionamento específico do fármaco às células tumorais promovido pela LDE, tornaria possível a terapia antitumoral combinada LDE-oleato de paclitaxel e LDEoleato de etoposídeo, sem produzir os efeitos colaterais observados com o uso combinado das formulações comerciais. O presente estudo tinha como objetivo avaliar os efeitos da terapia antitumoral combinada LDE-oleato de paclitaxel e LDEoleato de etoposídeo em comparação com a terapia dos respectivos fármacos isoladamente, bem como com a terapia combinada do paclitaxel e etoposídeo comerciais. A atividade antitumoral foi determinada em camundongos portadores de melanoma após a administração de intraperitoneal de LDE-oleato de paclitaxel 15 mg/kg e LDE-oleato de etoposídeo 10 mg/kg. Nos grupos dos fármacos comerciais injetou-se paclitaxel 3,75 mg/kg e etoposídeo 2,5 mg/Kg, ou paclitaxel 7,5 mg/Kg e etoposídeo 5 mg/kg. Os fármacos foram administrados em dois esquemas terapêuticos: 11º, 13º, 15º; e 11°, 14°, 19° dias após a inoculação do tumor. Todos os grupos apresentaram aumento na taxa de sobrevida em comparação ao grupo controle. Por outro lado, nos grupos LDE-fármacos foi observada uma redução dos efeitos de mielossupressão, visto que as doses administradas nestes grupos foram 2 e 4 vezes maiores do que nos demais. Os tratamentos do melanoma nos dois protocolos terapêuticos mostraram-se eficazes na redução da massa tumoral, sendo seu efeito inibitório acima de 90% nos grupos LDE-fármacos e na combinação dos quimioterápicos comerciais de maior dose. O número de metástases foi menor nos grupos de combinação LDE-Fármacos. Os resultados mostraram que a terapia antitumoral combinada LDE-oleato de paclitaxel e LDE-oleato de etoposídeo foi mais eficaz que as terapias isoladas destes fármacos e que a terapia combinada com o paclitaxel e etoposídeo comerciais.LDE is a cholesterol-rich emulsion with similar composition to the natural lipoproteins. Due to its ability of binding to the Low Density Lipoprotein (LDL) receptors and concentrate in neoplastic cells which present overexpression of these receptors, LDE may be used as vehicle to target antineoplastic drugs against cancer cells. Combined chemotherapy is a therapeutic strategy against cancer that usually uses drugs that act in different parts of cells metabolism, increasing the chances of destruction of the cancer cells. In addition, adverse effects can be reduced when combining agents with different toxicities at lower doses than the usual in single therapy. The major limiting factor in combining the chemotherapeutic agents paclitaxel and etoposide is their side effects such as leukopenia, thrombocytopenia and anemia. The ability of LDE to carry the drug into the cell may bring a strong possibility of combining LDE- paclitaxel oleate and LDE-etoposide oleate without producing the toxic effects observed in the combined use of the commercial formulations of these drugs. The current study was designed to evaluate the effects of combined antitumoral therapy of LDE- paclitaxel oleate and LDE-etoposide oleate in comparison to the combination of commercial paclitaxel and etoposide formulations and to the single therapy with the same LDEagents. Antitumoral activity was determined in melanoma-bearing mice after injection of LDE-paclitaxel oleate 15 mg/kg and LDE-etoposide oleate 10 mg/kg, or commercial paclitaxel 3.75 mg/kg and etoposide 2.5 mg/kg, or commercial paclitaxel 7,5 mg/kg and etoposide 5 mg/kg. Drugs were administered in two protocols: 11º, 13º, 15º; and 11°, 14°, 19° days after tumor implantation. All groups had an increase in the survival rate in comparison to the control group, however LDE combination groups showed reduction in the bone marrow toxicity, since the doses used were 2 and 4 fold greater that in the commercial drugs groups. The tumor growth inhibition rate was greater in both LDE-drugs combination groups and in the higher dosage of paclitaxel and etoposide combination (over 90% in comparisson to the control). There were also great reduction of metastatic nodes in LDE-drugs combination groups in comparison to the commercial drugs combination groups. Our results showed that the antitumor combined therapy of LDE-paclitaxel oleate and LDE-etoposide oleate was more effective than the therapies with LDE-paclitaxel oleate and LDE-etoposide oleate alone, and the combined therapy of commercial paclitaxel and etoposide formulations

    Combined use of simvastatin and paclitaxel associated to a lipidic nanoemulsion in cancer treatment

    No full text
    Uma nova alternativa para o tratamento do câncer foi proposta em estudos anteriores, consistindo no uso de uma nanoemulsão lipídica como transportadora de agentes quimioterápicos às células neoplásicas. A redução da toxicidade da quimioterapia promovida pelo direcionamento específico de quimioterápicos às células tumorais nos levou a testar o potencial de aplicação do sistema de nanopartículas lipídicas na terapêutica combinada do paclitaxel com a sinvastatina, um agente hipolipemiante que pode ser empregado como coadjuvante no tratamento do câncer. Nos dias 11, 14 e 19 após a inoculação de células de melanoma B16F10, camundongos C57BL/6J receberam pela via intraperitoneal soluções de oleato de paclitaxel associado à nanoemulsão lipídica 17,5µmol/kg (Nano-paclitaxel), formulação comercial do paclitaxel 17,5µmol/kg, nanoemulsão lipídica (Nanoemulsão) e solução salina (Controle). A sinvastatina 50mg/kg/dia foi administrada por gavagem do 11° ao 19° dia após a inoculação do tumor em um dos grupos de animais tratados com o Nano-paclitaxel (Nano-paclitaxel + Sinva), no grupo tratado com a formulação comercial do paclitaxel (Paclitaxel + Sinva) e como monoterapia (Sinva). Camundongos Balb-c saudáveis receberam os mesmos tratamentos para avaliação dos possíveis efeitos tóxicos dos diferentes tratamentos. A terapia combinada Nano-paclitaxel + Sinva apresentou toxicidade negligível em comparação com a terapia combinada Paclitaxel + Sinva que provocou perda de peso e mielossupressão nos animais. Nos animais portadores de tumor, o tratamento Nano-paclitaxel + Sinva inibiu 95% do crescimento tumoral, comparado à inibição de 44% promovida pelo tratamento Paclitaxel + Sinva. Além disso, apenas 37% dos animais portadores de melanoma submetidos ao tratamento com Nano-paclitaxel + Sinva apresentaram metástases, em contraste com 90% dos tratados com Paclitaxel + Sinva. A probabilidade de sobrevida também foi maior nos camundongos tratados com o Nano-paclitaxel + Sinva em comparação aos tratados com Paclitaxel + Sinva. A análise de amostras de tumores por citometria de fluxo mostrou que somente nos grupos de animais tratados com Sinva, Nano-paclitaxel ou com a combinação Nano-paclitaxel + Sinva houve aumento na expressão de p21 em comparação ao grupo Controle. Da mesma forma, apenas nos grupos Sinva e Nano-paclitaxel + Sinva houve redução na expressão de ciclina D1 em comparação ao grupo Controle. O teste de viabilidade celular com rodamina 123 mostrou despolarização da membrana mitocondrial com redução no número de células tumorais viáveis em todos os grupos de tratamentos em comparação aos grupos Nanoemulsão e Controle. A avaliação histológica dos tumores demonstrou que os grupos Nanoemulsão e Controle apresentaram alta densidade de células tumorais, diferentemente dos demais grupos de tratamento e que apenas os tumores do grupo Nano-paclitaxel + Sinva apresentaram aumento na presença de fibras de colágeno tipo I e III. Em comparação ao grupo Controle, os tumores dos grupos Sinva, Paclitaxel + Sinva, Nano-paclitaxel e Nano-paclitaxel + Sinva apresentaram redução na expressão imunohistoquímica de ICAM, MCP-1 e MMP-9 sendo que o grupo Nano-paclitaxel + Sinva apresentou a menor porcentagem de área marcada positivamente para a MMP-9. A terapia combinada com Nano-paclitaxel + Sinva é menos tóxica e mais efetiva na inibição do crescimento tumoral do que a mesma terapia com a formulação comercial do paclitaxel.In previous studies we have proposed a novel approach for cancer treatment consisting of the use of a lipid nanoemulsion as a vehicle to direct chemotherapeutic agents to neoplastic cells. Reduction of chemotherapy toxicity promoted by specific targeting of antineoplastic agents to tumor cells led us to test the application of the lipidic nanoparticle system in combined treatment with paclitaxel and simvastatin, a cholesterol-lowering drug that can be used as coadjuvant in cancer treatment. On days 11, 14 and 19 after B16F10 melanoma cells inoculation, C57BL/6J mice were intraperitoneally injected with paclitaxel oleate associated to the lipidic nanoemulsion 17.5 µmol/kg (Nano-paclitaxel), commercial formulation of paclitaxel 17.5 µmol/kg, lipidic nanoemulsion (Nanoemulsion) or saline solution (Control). Simvastatin 50 mg/kg/day was administered by gavage from days 11 to 19 after tumor inoculation in one group of animals treated with Nano-paclitaxel (Nano-paclitaxel + Simva), in the group treated with commercial formulation of paclitaxel (Paclitaxel + Simva) and as monotherapy (Simva). Evaluation of possible toxic effects of the treatments was accessed in healthy Balb-c mice. Combined therapy with Nano-paclitaxel + Simva showed negligible toxicity as compared with the combination of Paclitaxel + Simva which resulted in animal weight loss and myelosuppression. In tumor-bearing animals, treatment with Nano-paclitaxel + Simva resulted in a remarkable tumor growth inhibition rate of 95%, compared to a 44% inhibition rate promoted by treatment with Paclitaxel + Simva. Moreover, only 37% of melanoma bearing animals treated with Nano-paclitaxel + Simva developed metastasis, in contrast to 90% of those treated with Paclitaxel + Simva. Survival rates were also higher in mice treated with Nano-paclitaxel + Simva in comparison to Paclitaxel + Simva treated animals. Analysis of tumor samples by flow cytometry showed that only animals treated with Simva, Nano-paclitaxel or Nano-paclitaxel + Simva increased the expression of p21 in comparison to Control group. Also, tumors from animals treated with Simva or Nano-paclitaxel + Simva presented a decrease in the expression of cyclin D1 in comparison to Control group. Cell viability test with rhodamine 123 showed mitochondrial membrane depolarization with reduction of tumor viable cells in all treatment groups in comparison to Nanoemulsion and Control groups. The histological study revealed that in contrast to drugs treated groups, tumors from Nanoemulsion and Control groups presented high tumor cell density and only Nano-paclitaxel + Simva treated animals presented tumors with increased presence of collagen fibers I and III. In comparison to Control group, tumors from groups Simva, Paclitaxel + Simva, Nano-paclitaxel and Nano-paclitaxel + Simva showed a reduction in immunohistochemical expression of ICAM, MCP-1 and MMP-9 and the group Nano-paclitaxel + Simva presented the lowest percentage of area positively stained for MMP-9. Combined therapy with Nano-paclitaxel + Simva was less toxic and more effective in promoting tumor growth inhibiton than the same combined therapy with the commercial formulation of paclitaxel

    The prevalence of hepatitis B virus infection markers and socio-demographic risk factors in HIV-infected patients in Southern Brazil

    No full text
    IntroductionHepatitis B virus (HBV) and human immunodeficiency virus (HIV) infections are two of the world's most important infectious diseases. Our objective was to determine the hepatitis B surface antigen (HBsAg) and hepatitis B core antibody (anti-HBc) prevalences among adult HIV-infected patients and identify the associations between socio-demographic variables and these HBV infection markers.MethodsThis study was performed from October 2012 to March 2013. Three hundred HIV-seropositive patients were monitored by the Clinical Analysis Laboratory of Professor Polydoro Ernani de São Thiago University Hospital, Santa Catarina, Brazil. The blood tests included HBsAg, anti-HBc immunoglobulin M (IgM) and total anti-HBc. Patients reported their HIV viral loads and CD4+ T-cell counts using a questionnaire designed to collect sociodemographic data.ResultsThe mean patient age was 44.6 years, the mean CD4 T-cell count was 525/mm3, the mean time since beginning antiretroviral therapy was 7.6 years, and the mean time since HIV diagnosis was 9.6 years. The overall prevalences of HBsAg and total anti-HBc were 2.3% and 29.3%, respectively. Among the individuals analyzed, 0.3% were positive for HBsAg, 27.3% were positive for total anti-HBc, and 2.0% were positive either for HBsAg or total anti-HBc and were classified as chronically HBV-infected. Furthermore, 70.3% of the patients were classified as never having been infected. Male gender, age >40 years and Caucasian ethnicity were associated with an anti-HBc positive test.ConclusionsThe results showed an intermediate prevalence of HBsAg among the studied patients. Moreover, the associations between the anti-HBc marker and socio-demographic factors suggest a need for HBV immunization among these HIV-positive individuals, who are likely to have HIV/HBV coinfection

    Vaccination coverage and immunity against hepatitis B among HIV-infected patients in South Brazil

    No full text
    Evidence-based strategies to improve the hepatitis B virus (HBV) vaccination coverage rates might help to reduce the burden caused by co-infection with HBV and human immuno-deficiency virus (HIV). In this study, the aim was to evaluate the vaccination coverage and immunity against HBV among HIV-infected individuals in South Brazil, and identify factors that are associated with compliance patterns and antibody reactivity. Three hundred HIV-infected men and women were included in this survey. The patients answered a standardized questionnaire, and vaccination cards were checked in order to assess hepatitis B vaccine status. A blood sample was collected for quantitative determination of antibody to hepatitis B virus surface antigen (anti-HBs). Participants were also evaluated for their CD4 cell count and HIV viral load. The overall vaccination coverage of HBV vaccination found in this study (57.4%) was lower than that was previously reported in South Brazil. Anti-HBs levels >10 IU/L were observed in 47.0% of the studied population. A significant inequality in the coverage rates and antibody reactivity was found in favor of patients with better economic status. In conclusion, the results indicate the need for improvement in the HBV vaccination coverage among HIV carriers, in particular focusing on low-income individuals

    Serum from morbidly obese patients affects melanoma cell behavior in vitro

    No full text
    Here we examined whether serum from obese patients could create a growth-enhancing microenvironment that alters gene expression in BRAF- and NRAS-mutated melanoma cell lines. SK-Mel-28 (BRAF-mutated) and Sk-Mel-147 (NRAS-mutated) cells were treated with pooled serum from 10 severely obese patients (BMI > 40 kg/m2), pooled serum from 6 healthy lean individuals (BMI = 18.5-24.9 kg/m2), or recombinant TNF-α. We found that obese patient serum enhanced migration capacity and increased NRAS expression levels in both BRAF- and NRAS-mutated melanoma cells. Although TNF-α is the major obesity-related cytokine and TNF-α levels were found to be increased in the serum of obese individuals, this cytokine made only a modest contribution to the migration capacity of melanoma cells. These results indicate that other components present in the serum of severely obese patients may be responsible for enhancing the migration capacity of melanoma cells. As TNF-α alone did not seem to significantly affect tumor cell behavior, anti-tumor strategies aimed at blocking TNF-α should be considered with caution in future studies, particularly when in vitro models are used as screening platforms for antitumor activity
    corecore