Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada

Detalhes bibliográficos
Autor(a) principal: Ferreira, Natália Helen
Data de Publicação: 2021
Tipo de documento: Tese
Idioma: por
Título da fonte: Repositório do Centro Universitário Braz Cubas
Texto Completo: https://repositorio.cruzeirodosul.edu.br/handle/123456789/1955
Resumo: Melanoma represents the smallest number of skin cancer cases; however, it is the most lethal type among skin cancers. This is due to factors such as their highly metastatic nature, unexpected evolution and inefficiency of current systemic treatments, mainly due to the high toxicity and resistance acquired to these treatments. In this sense, there is a need to find additional treatments that directly address tumor survival, targeting apoptotic pathways that may increase tumor cell sensitivity to drugs and minimize toxicity, which is often associated with conventional therapies. Indomethacin (IND) is known to promote mitochondrial oxidative stress and the generation of reactive oxygen species, which are known to modulate intrinsic signaling of mitochondria-mediated apoptosis. Despite its wide distribution and therapeutic use, IND has undesirable gastrointestinal effects, such as ulcerations, hemorrhages, inhibiting platelet aggregation, is associated with renal toxicity and central nervous system disorders, and its toxicity is dose dependent. In order to reduce the toxic effects of this drug, the present study investigated the antitumor efficiency of IND incorporated in mesoporous silica nanoparticles (MSNPs+IND), as well as the toxic potential of treatments in syngeneic murine B16 melanoma model. Male C57BL/6 mice were implanted subcutaneously with B16F10 cells. The animals received once daily, for five consecutive days, treatments with IND (2.5 and 5.0 mg/kg body weight [b.w.]) and MSNPs+IND (16.66 and 33.33 mg/kg b.w. [15% of IND incorporated into the MSNPs]). Evaluation of antitumor activity was performed by measuring size, tumor weight and histopathological analysis. Possible molecular signaling pathways involved in antitumor activity were analyzed by Western blot assays in liver tissue and immunohistochemistry in tumor tissue. At the same time, the potential toxicity of the treatments was evaluated by varying body mass and organs, quantifying the biochemical indicators of renal damage (creatinine and urea) and bone marrow genotoxicity. The results of the present study demonstrated that treatments with MSNPs+IND 2.5 and 5.0 showed antitumor activity, leading to 54.18 and 70.09% inhibition of tumor growth, respectively. Histopathological analysis revealed a decrease in the frequency of mitosis in the tumor tissues of the groups treated with MSNPs+IND 2.5 and 5.0, being 37.95 and 35.90% lower, respectively, compared to the IND 2.5 and 5 groups. .0. The observed antitumor activity may be related to the inhibition of COX-2, the main mechanism of action of IND. In hepatic tissue, COX-2 levels decreased significantly after treatment with MSNPs+IND and IND. In this context, it is shown that MSNPs were able to control IND delivery at the tumor site, and gradual release of the drug was responsible for the antitumor activity, inhibiting COX-2 and blocking cell proliferation. Additionally, the observed antitumor activity may also be related to the increased expression of cleaved caspase-3, which increased by 156.25 and 137.50% for the groups treated with MSNPs+IND and IND, respectively, which possibly induced tumor cells to apoptosis. Regarding toxic effects, there were no significant differences in weight gain and animal organ weight between the different treatment groups. Serum creatinine and urea levels did not differ significantly, indicating no nephrotoxicity for the different treatment groups. Finally, in the bone marrow genotoxic analysis, an increase in the frequency of micronuclei was observed for the group treated with IND 5.0, indicating genotoxicity. However, no significant differences were observed for the MSNPs+IND groups, indicating that the MSNPs were efficient in preventing the genotoxic effect of IND in this parameter. Thus, MSNPs nanocarriers may therefore be promising for future applications in cancer therapy. Keywords: Indomethacin; Mesoporous silica nanoparticles; Anti-melanoma activity; COX-2; Apoptosis.
id CUB_5aedef02cb88279cb25928d5b7bc753c
oai_identifier_str oai:repositorio.cruzeirodosul.edu.br:123456789/1955
network_acronym_str CUB
network_name_str Repositório do Centro Universitário Braz Cubas
repository_id_str
spelling Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturadaAnti-melanoma activity of indomethacin incorporated into nanostructured silica particlesApoptose - OncologiaCaspase-3 clivadaCiclo-oxigenase-2 (COX-2) - InibiçãoCrescimento tumoral - InibiçãoCNPQ::CIENCIAS EXATAS E DA TERRA::QUIMICAMelanoma represents the smallest number of skin cancer cases; however, it is the most lethal type among skin cancers. This is due to factors such as their highly metastatic nature, unexpected evolution and inefficiency of current systemic treatments, mainly due to the high toxicity and resistance acquired to these treatments. In this sense, there is a need to find additional treatments that directly address tumor survival, targeting apoptotic pathways that may increase tumor cell sensitivity to drugs and minimize toxicity, which is often associated with conventional therapies. Indomethacin (IND) is known to promote mitochondrial oxidative stress and the generation of reactive oxygen species, which are known to modulate intrinsic signaling of mitochondria-mediated apoptosis. Despite its wide distribution and therapeutic use, IND has undesirable gastrointestinal effects, such as ulcerations, hemorrhages, inhibiting platelet aggregation, is associated with renal toxicity and central nervous system disorders, and its toxicity is dose dependent. In order to reduce the toxic effects of this drug, the present study investigated the antitumor efficiency of IND incorporated in mesoporous silica nanoparticles (MSNPs+IND), as well as the toxic potential of treatments in syngeneic murine B16 melanoma model. Male C57BL/6 mice were implanted subcutaneously with B16F10 cells. The animals received once daily, for five consecutive days, treatments with IND (2.5 and 5.0 mg/kg body weight [b.w.]) and MSNPs+IND (16.66 and 33.33 mg/kg b.w. [15% of IND incorporated into the MSNPs]). Evaluation of antitumor activity was performed by measuring size, tumor weight and histopathological analysis. Possible molecular signaling pathways involved in antitumor activity were analyzed by Western blot assays in liver tissue and immunohistochemistry in tumor tissue. At the same time, the potential toxicity of the treatments was evaluated by varying body mass and organs, quantifying the biochemical indicators of renal damage (creatinine and urea) and bone marrow genotoxicity. The results of the present study demonstrated that treatments with MSNPs+IND 2.5 and 5.0 showed antitumor activity, leading to 54.18 and 70.09% inhibition of tumor growth, respectively. Histopathological analysis revealed a decrease in the frequency of mitosis in the tumor tissues of the groups treated with MSNPs+IND 2.5 and 5.0, being 37.95 and 35.90% lower, respectively, compared to the IND 2.5 and 5 groups. .0. The observed antitumor activity may be related to the inhibition of COX-2, the main mechanism of action of IND. In hepatic tissue, COX-2 levels decreased significantly after treatment with MSNPs+IND and IND. In this context, it is shown that MSNPs were able to control IND delivery at the tumor site, and gradual release of the drug was responsible for the antitumor activity, inhibiting COX-2 and blocking cell proliferation. Additionally, the observed antitumor activity may also be related to the increased expression of cleaved caspase-3, which increased by 156.25 and 137.50% for the groups treated with MSNPs+IND and IND, respectively, which possibly induced tumor cells to apoptosis. Regarding toxic effects, there were no significant differences in weight gain and animal organ weight between the different treatment groups. Serum creatinine and urea levels did not differ significantly, indicating no nephrotoxicity for the different treatment groups. Finally, in the bone marrow genotoxic analysis, an increase in the frequency of micronuclei was observed for the group treated with IND 5.0, indicating genotoxicity. However, no significant differences were observed for the MSNPs+IND groups, indicating that the MSNPs were efficient in preventing the genotoxic effect of IND in this parameter. Thus, MSNPs nanocarriers may therefore be promising for future applications in cancer therapy. Keywords: Indomethacin; Mesoporous silica nanoparticles; Anti-melanoma activity; COX-2; Apoptosis.O melanoma representa o menor número de casos de câncer de pele; no entanto, é o tipo mais letal entre as neoplasias cutâneas. Isso se deve a fatores como sua natureza altamente metastática, evolução inesperada e ineficiência dos tratamentos sistêmicos atuais, principalmente devido à alta toxicidade e resistência adquirida a esses tratamentos. Neste sentido, há uma necessidade de encontrar tratamentos adicionais que se direcionem diretamente a sobrevida do tumor, visando às vias apoptóticas, que possam aumentar a sensibilidade das células tumorais às drogas e minimizar a toxicidade, que é frequentemente associada a terapias convencionais. A indometacina (IND) é conhecida por promover o estresse oxidativo mitocondrial e a geração de espécies reativas de oxigênio, que são conhecidas por modularem a sinalização intrínseca da apoptose mediada por mitocôndrias. Apesar de sua ampla distribuição e uso terapêutico, a IND tem efeitos gastrointestinais indesejáveis, como ulcerações, hemorragias, atua inibindo a agregação plaquetária, está associada à toxicidade renal e a distúrbios do sistema nervoso central, e sua toxicidade é dosedependente. Com o objetivo de reduzir os efeitos tóxicos deste fármaco, o presente estudo investigou a eficiência antitumoral da IND incorporada em nanopartículas de sílica mesoporosa (NPSM+IND), bem como o potencial tóxico dos tratamentos em modelo singênico de melanoma B16 murino. Camundongos C57BL/6 machos foram implantados subcutaneamente com células B16F10. Os animais receberam uma vez ao dia, durante cinco dias consecutivos, tratamentos com IND (2,5 e 5,0 mg/kg m.c.) e NPSM+IND (16,66 e 33,33 mg/kg m.c. [15% de IND incorporado às NPSM]). A avaliação da atividade antitumoral foi realizada por meio das medições do tamanho, peso do tumor e análise histopatológica. As possíveis vias de sinalização molecular envolvidas na atividade antitumoral foram analisadas através dos ensaios Western blot no tecido hepático e imunohistoquímica no tecido tumoral. Paralelamente, a potencial toxicidade dos tratamentos foi avaliada por meio da variação da massa corpórea e dos órgãos, da quantificação dos indicadores bioquímicos de danos renais (creatinina e ureia) e da genotoxicidade em medula óssea. Os resultados do presente estudo demonstraram que os tratamentos com NPSM+IND 2,5 e 5,0 apresentaram atividade antitumoral, levando a 54,18 e 70,09% de inibição do crescimento tumoral, respectivamente. A análise histopatológica revelou a diminuição na frequência de mitose nos tecidos tumorais dos grupos tratados com NPSM+IND 2,5 e 5,0, sendo 37,95 e 35,90% menores, respectivamente, em relação aosgrupos IND 2,5 e 5,0. A atividade antitumoral observada pode estar relacionada à inibição da COX-2, principal mecanismo de ação da IND. No tecido hepático, os níveis de COX-2 diminuíram significativamente após tratamento com NPSM+IND e IND Nesse contexto, mostra-se que as NPSM foram capazes de controlar a entrega da IND no local do tumor, e a liberação gradativa do fármaco foi responsável pela atividade antitumoral, inibindo a COX-2 e bloqueando a proliferação celular. Adicionalmente, a atividade antitumoral observada também pode estar relacionada ao aumento da expressão de caspase-3 clivada, que apresentou elevação em 156,25 e 137,50% para os grupos tratados com NPSM+IND e IND, respectivamente, o que possivelmente induziu as células tumorais à apoptose. Em relação aos efeitos tóxicos, não houve diferenças significativas no ganho de peso e no peso dos órgãos dos animais entre os diferentes grupos de tratamento. Os níveis séricos de creatinina e ureia não diferiram significativamente, indicando ausência de nefrotoxicidade para os diferentes grupos de tratamento. Por fim, na análise genotóxica na medula óssea observou-se aumento na frequência de micronúcleos para o grupo tratado com IND 5.0, indicando genotoxicidade. Entretanto, não foram observadas diferenças significativas para os grupos NPSM+IND, indicando que os NPSM foram eficientes na prevenção do efeito genotóxico da IND neste parâmetro. Dessa forma, os nanocarreadores NPSM podem, portanto, ser promissores para futuras aplicações na terapia do câncer. Palavras-chave: Indometacina; Nanopartículas de sílica mesoporosa; Atividade anti-melanoma; COX-2; Apoptose.Universidade de FrancaBrasilPós-GraduaçãoPrograma de Doutorado em CiênciasUNIFRANTavares, Denise Crispim5342181863190744http://lattes.cnpq.br/5342181863190744Rocha, Lucas Alonso3010767093603300http://lattes.cnpq.br/3010767093603300Veneziani, Rodrigo Cassio Sola7198067266796866http://lattes.cnpq.br/7198067266796866Lima, Ildercílio Mota de Souza5715288813015613http://lattes.cnpq.br/5715288813015613Ferreira, Natália Helen2021-04-27T17:19:50Z2021-04-27T17:19:50Z2021-02-22info:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/doctoralThesisapplication/pdfFERREIRA, Natália Helen. Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada. Franca, SP, 2021. 98 f. Tese (Doutorado em Ciências) - Universidade de Franca. 2021https://repositorio.cruzeirodosul.edu.br/handle/123456789/1955porinfo:eu-repo/semantics/openAccessreponame:Repositório do Centro Universitário Braz Cubasinstname:Centro Universitário Braz Cubas (CUB)instacron:CUB2021-04-27T17:20:50Zoai:repositorio.cruzeirodosul.edu.br:123456789/1955Repositório InstitucionalPUBhttps://repositorio.brazcubas.edu.br/oai/requestbibli@brazcubas.edu.bropendoar:2021-04-27T17:20:50Repositório do Centro Universitário Braz Cubas - Centro Universitário Braz Cubas (CUB)false
dc.title.none.fl_str_mv Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada
Anti-melanoma activity of indomethacin incorporated into nanostructured silica particles
title Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada
spellingShingle Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada
Ferreira, Natália Helen
Apoptose - Oncologia
Caspase-3 clivada
Ciclo-oxigenase-2 (COX-2) - Inibição
Crescimento tumoral - Inibição
CNPQ::CIENCIAS EXATAS E DA TERRA::QUIMICA
title_short Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada
title_full Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada
title_fullStr Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada
title_full_unstemmed Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada
title_sort Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada
author Ferreira, Natália Helen
author_facet Ferreira, Natália Helen
author_role author
dc.contributor.none.fl_str_mv Tavares, Denise Crispim
5342181863190744
http://lattes.cnpq.br/5342181863190744
Rocha, Lucas Alonso
3010767093603300
http://lattes.cnpq.br/3010767093603300
Veneziani, Rodrigo Cassio Sola
7198067266796866
http://lattes.cnpq.br/7198067266796866
Lima, Ildercílio Mota de Souza
5715288813015613
http://lattes.cnpq.br/5715288813015613
dc.contributor.author.fl_str_mv Ferreira, Natália Helen
dc.subject.por.fl_str_mv Apoptose - Oncologia
Caspase-3 clivada
Ciclo-oxigenase-2 (COX-2) - Inibição
Crescimento tumoral - Inibição
CNPQ::CIENCIAS EXATAS E DA TERRA::QUIMICA
topic Apoptose - Oncologia
Caspase-3 clivada
Ciclo-oxigenase-2 (COX-2) - Inibição
Crescimento tumoral - Inibição
CNPQ::CIENCIAS EXATAS E DA TERRA::QUIMICA
description Melanoma represents the smallest number of skin cancer cases; however, it is the most lethal type among skin cancers. This is due to factors such as their highly metastatic nature, unexpected evolution and inefficiency of current systemic treatments, mainly due to the high toxicity and resistance acquired to these treatments. In this sense, there is a need to find additional treatments that directly address tumor survival, targeting apoptotic pathways that may increase tumor cell sensitivity to drugs and minimize toxicity, which is often associated with conventional therapies. Indomethacin (IND) is known to promote mitochondrial oxidative stress and the generation of reactive oxygen species, which are known to modulate intrinsic signaling of mitochondria-mediated apoptosis. Despite its wide distribution and therapeutic use, IND has undesirable gastrointestinal effects, such as ulcerations, hemorrhages, inhibiting platelet aggregation, is associated with renal toxicity and central nervous system disorders, and its toxicity is dose dependent. In order to reduce the toxic effects of this drug, the present study investigated the antitumor efficiency of IND incorporated in mesoporous silica nanoparticles (MSNPs+IND), as well as the toxic potential of treatments in syngeneic murine B16 melanoma model. Male C57BL/6 mice were implanted subcutaneously with B16F10 cells. The animals received once daily, for five consecutive days, treatments with IND (2.5 and 5.0 mg/kg body weight [b.w.]) and MSNPs+IND (16.66 and 33.33 mg/kg b.w. [15% of IND incorporated into the MSNPs]). Evaluation of antitumor activity was performed by measuring size, tumor weight and histopathological analysis. Possible molecular signaling pathways involved in antitumor activity were analyzed by Western blot assays in liver tissue and immunohistochemistry in tumor tissue. At the same time, the potential toxicity of the treatments was evaluated by varying body mass and organs, quantifying the biochemical indicators of renal damage (creatinine and urea) and bone marrow genotoxicity. The results of the present study demonstrated that treatments with MSNPs+IND 2.5 and 5.0 showed antitumor activity, leading to 54.18 and 70.09% inhibition of tumor growth, respectively. Histopathological analysis revealed a decrease in the frequency of mitosis in the tumor tissues of the groups treated with MSNPs+IND 2.5 and 5.0, being 37.95 and 35.90% lower, respectively, compared to the IND 2.5 and 5 groups. .0. The observed antitumor activity may be related to the inhibition of COX-2, the main mechanism of action of IND. In hepatic tissue, COX-2 levels decreased significantly after treatment with MSNPs+IND and IND. In this context, it is shown that MSNPs were able to control IND delivery at the tumor site, and gradual release of the drug was responsible for the antitumor activity, inhibiting COX-2 and blocking cell proliferation. Additionally, the observed antitumor activity may also be related to the increased expression of cleaved caspase-3, which increased by 156.25 and 137.50% for the groups treated with MSNPs+IND and IND, respectively, which possibly induced tumor cells to apoptosis. Regarding toxic effects, there were no significant differences in weight gain and animal organ weight between the different treatment groups. Serum creatinine and urea levels did not differ significantly, indicating no nephrotoxicity for the different treatment groups. Finally, in the bone marrow genotoxic analysis, an increase in the frequency of micronuclei was observed for the group treated with IND 5.0, indicating genotoxicity. However, no significant differences were observed for the MSNPs+IND groups, indicating that the MSNPs were efficient in preventing the genotoxic effect of IND in this parameter. Thus, MSNPs nanocarriers may therefore be promising for future applications in cancer therapy. Keywords: Indomethacin; Mesoporous silica nanoparticles; Anti-melanoma activity; COX-2; Apoptosis.
publishDate 2021
dc.date.none.fl_str_mv 2021-04-27T17:19:50Z
2021-04-27T17:19:50Z
2021-02-22
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/doctoralThesis
format doctoralThesis
status_str publishedVersion
dc.identifier.uri.fl_str_mv FERREIRA, Natália Helen. Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada. Franca, SP, 2021. 98 f. Tese (Doutorado em Ciências) - Universidade de Franca. 2021
https://repositorio.cruzeirodosul.edu.br/handle/123456789/1955
identifier_str_mv FERREIRA, Natália Helen. Atividade anti-melanoma da indometacina incorporada em partículas de sílica nanoestruturada. Franca, SP, 2021. 98 f. Tese (Doutorado em Ciências) - Universidade de Franca. 2021
url https://repositorio.cruzeirodosul.edu.br/handle/123456789/1955
dc.language.iso.fl_str_mv por
language por
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.publisher.none.fl_str_mv Universidade de Franca
Brasil
Pós-Graduação
Programa de Doutorado em Ciências
UNIFRAN
publisher.none.fl_str_mv Universidade de Franca
Brasil
Pós-Graduação
Programa de Doutorado em Ciências
UNIFRAN
dc.source.none.fl_str_mv reponame:Repositório do Centro Universitário Braz Cubas
instname:Centro Universitário Braz Cubas (CUB)
instacron:CUB
instname_str Centro Universitário Braz Cubas (CUB)
instacron_str CUB
institution CUB
reponame_str Repositório do Centro Universitário Braz Cubas
collection Repositório do Centro Universitário Braz Cubas
repository.name.fl_str_mv Repositório do Centro Universitário Braz Cubas - Centro Universitário Braz Cubas (CUB)
repository.mail.fl_str_mv bibli@brazcubas.edu.br
_version_ 1798311360445022208