Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Mar Drugs ; 21(5)2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37233473

RESUMO

Proteolytic enzymes, also known as peptidases, are critical in all living organisms. Peptidases control the cleavage, activation, turnover, and synthesis of proteins and regulate many biochemical and physiological processes. They are also involved in several pathophysiological processes. Among peptidases, aminopeptidases catalyze the cleavage of the N-terminal amino acids of proteins or peptide substrates. They are distributed in many phyla and play critical roles in physiology and pathophysiology. Many of them are metallopeptidases belonging to the M1 and M17 families, among others. Some, such as M1 aminopeptidases N and A, thyrotropin-releasing hormone-degrading ectoenzyme, and M17 leucyl aminopeptidase, are targets for the development of therapeutic agents for human diseases, including cancer, hypertension, central nervous system disorders, inflammation, immune system disorders, skin pathologies, and infectious diseases, such as malaria. The relevance of aminopeptidases has driven the search and identification of potent and selective inhibitors as major tools to control proteolysis with an impact in biochemistry, biotechnology, and biomedicine. The present contribution focuses on marine invertebrate biodiversity as an important and promising source of inhibitors of metalloaminopeptidases from M1 and M17 families, with foreseen biomedical applications in human diseases. The results reviewed in the present contribution support and encourage further studies with inhibitors isolated from marine invertebrates in different biomedical models associated with the activity of these families of exopeptidases.


Assuntos
Aminopeptidases , Leucil Aminopeptidase , Humanos , Aminopeptidases/química , Aminopeptidases/metabolismo , Leucil Aminopeptidase/química , Peptídeos/química , Antígenos CD13
2.
Int J Biol Macromol ; 229: 825-837, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36592847

RESUMO

Bufadienolides are steroids that inhibit Na+/K+-ATPase; recent evidence shows that bufalin inhibits the activity of porcine aminopeptidase N (pAPN). We evaluated the selectivity of some bufadienolides on metallo-aminopeptidases. Among the enzymes of the M1 and M17 families, pAPN and porcine aminopeptidase A (pAPA) were the only targets of some bufadienolides. ѱ-bufarenogin, telocinobufagin, marinobufagin, bufalin, cinobufagin, and bufogenin inhibited the activity of pAPN in a dose-dependent manner in the range of 10-7-10-6 M. The inhibition mechanism was classical reversible noncompetitive for telocinobufagin, bufalin and cinobufagin. Bufogenin had the lowest Ki value and a non-competitive behavior. pAPA activity was inhibited by ѱ-bufarenogin, cinobufagin, and bufogenin, with a classical competitive type of inhibition. The models of enzyme-inhibitor complexes agreed with the non-competitive type of inhibition of pAPN by telocinobufagin, bufalin, cinobufagin, and bufogenin. Since APN is a target in cancer therapy, we tested the effect of bufadienolides on the MeWo APN+ human melanoma cell line; they induced cell death, but we obtained scant evidence that inhibition of APN contributed to their effect. Thus, APN is a selective target of some bufadienolides, and we suggest that inhibition of APN activity by bufadienolides is not a major contributor to their antiproliferative properties in MeWo cells.


Assuntos
Bufanolídeos , Melanoma , Humanos , Suínos , Animais , Antígenos CD13 , Aminopeptidases , Bufanolídeos/farmacologia , Bufanolídeos/metabolismo , Inibidores Enzimáticos , Melanoma/tratamento farmacológico , Mamíferos/metabolismo
3.
Artigo em Espanhol | LILACS, CUMED | ID: biblio-1509234

RESUMO

Las vacunas SOBERANA®02 y SOBERANA® Plus contra el coronavirus tipo 2 causante del síndrome respiratorio agudo severo, recibieron autorización de uso en emergencia por la autoridad reguladora de Cuba, y de inmediato aconteció una campaña de vacunación masiva en población pediátrica, lo que devino en una inminente movilización de centros de vacunación y vigilancia de sus eventos adversos. El Centro de Inmunología Molecular realizó un estudio de farmacovigilancia intensiva que evalúo el cumplimiento del esquema heterólogo con ambas vacunas, su seguridad y la incidencia de casos positivos a COVID-19 en niños y adolescentes después de completar el esquema de inmunización. Desde el 15 de septiembre al 31 de diciembre del 2021, participaron 529 sujetos entre 2 y 18 años de edad, de ambos sexos, sin antecedentes de infección por coronavirus tipo 2 del síndrome respiratorio agudo severo, procedentes de 35 municipios y 12 provincias cubanas, quienes recibieron vacuna SOBERANA®02 (dos dosis) y SOBERANA®Plus (una dosis). Se realizó vigilancia de eventos adversos hasta 30 días después de la última dosis recibida. Se consultó la plataforma informática nacional Higia Andariego para identificar los casos positivos al virus del síndrome respiratorio agudo severo coronavirus 2, hasta 3 meses de haber completado la vacunación. El 98,5 por ciento de los participantes completó el esquema de vacunación y en el 6,6 por ciento se notificó algún evento adverso con relación consistente a la vacunación. Predominaron las reacciones locales (dolor, eritema, inflamación), sobre las reacciones sistémicas (fatiga y febrícula), de intensidad ligera o moderada. Se logró un elevado cumplimiento del esquema de inmunización, con un perfil de seguridad favorable, los sujetos con esquema completo de inmunización no enfermaron de COVID-19(AU)


The severe acute respiratory syndrome coronavirus 2 vaccines, SOBERANA®02 and SOBERANA®Plus, received authorization for emergency use by the Cuban regulatory authority; a massive vaccination campaign was immediately launched in the pediatric population, which led to an imminent mobilization of vaccination centers and surveillance of adverse events. The Molecular Immunology Center conducted an intensive pharmacovigilance study to evaluate compliance of the heterologous scheme with both vaccines, their safety, and the incidence of COVID-19 positive cases in children and adolescents after completing the immunization schedule. From September 15 to December 31, 2021, a total of 529 subjects between 2 and 18 years of age, of both sexes, without a history of infection by severe acute respiratory syndrome coronavirus type 2, from 35 municipalities and 12 Cuban provinces, who received SOBERANA®02 (two doses) and SOBERANA®Plus (one dose) vaccines, were included in the study. Surveillance for adverse events was performed up to 30 days after the last dose received. The national computer platform Higia Andariego was consulted to identify positive cases for severe acute respiratory syndrome coronavirus 2 up to 3 months after completing vaccination. According to the report, 98.5percent of the participants completed the vaccination schedule and 6.6percent of them reported some adverse event consistently related to vaccination. Local reactions (pain, erythema, inflammation) prevailed over systemic reactions (fatigue and fever), of light or moderate intensity. High compliance with the immunization schedule was achieved, with a favorable safety profile; subjects with a complete immunization schedule did not become ill with COVID-19(AU)


Assuntos
Humanos , Masculino , Feminino , Criança , Adolescente , Serviços Preventivos de Saúde/métodos , Vacinação em Massa , Vacinas contra COVID-19/uso terapêutico , Cuba , Estudo Observacional
4.
Front Oncol ; 12: 951267, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36408164

RESUMO

Background: Antitumor therapies targeting HER1/EGFR and HER2, such as monoclonal antibodies (MAbs) and tyrosine-kinase inhibitors (TKIs), have demonstrated a significant clinical benefit, but the emergence of resistance limits long-term efficacy. While secondary HER1 mutations confer tolerance to TKI, compensatory upregulation of HER2 drives resistance to anti-HER1 MAbs, which identifies MAb combinations targeting both receptors as an attractive therapeutic strategy. Nevertheless, toxicity hampers the clinical validation of this approach. Alternatively, cancer vaccines may induce antibodies directed against several antigens with less concern about induced toxicity. Methods: Polyclonal antibodies (PAbs) targeting HER1 and HER2 were induced in mice or rabbits through immunization. Recognition of different epitopes on targets by PAbs was validated by phage-display technology. Receptor downregulation was evaluated by flow cytometry, immunofluorescence, and Western blot. MTT assays assessed cytotoxicity, while the antitumor effect of PAbs was assayed in nude mice. Results: PAbs promoted degradation of HER1 and HER2 regarding clinical MAbs or their combinations. As a result, inhibition of cytotoxicity on tumor cell lines was improved, even in the presence of oncogenic mutations in HER1, as well as in cetuximab-insensitive cells. Accordingly, the antitumor effect of vaccination-induced PAbs was observed in lung tumor lines representative of sensitivity or resistance to HER1 targeting therapies. Conclusions: Immunization against HER1 and HER2 receptors offers an alternative to passive administration of combinations of MAbs, since vaccination-induced PAbs promote the downregulation of both receptors and they have a higher impact on the survival of tumor cells.

5.
Immunopharmacol Immunotoxicol ; 43(3): 283-290, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33722157

RESUMO

Purpose: A vaccine composition based on the extracellular domain of the human epidermal growth factor receptor 1 (HER1-ECD) and the combination of VSSP (very small size proteoliposomes) and Montanide ISA 51 adjuvants when used by intramuscular route, demonstrated promising results in preclinical studies. However, in order to avoid potential adverse events due to the use of Montanide, it is proposed to modify the vaccine formulation by using VSSP (very small size proteoliposomes) adjuvant alone, and to evaluate the quality of subcutaneously induced immune response. This study aimed to assess the immunotoxicological effects of HER1 vaccine in Cercopithecus aethiops.Materials and methods: Fifteen monkeys were randomized into four groups: Negative Control (Tris/NaCl, s.c.), Positive Control (200 µg HER1-ECD/VSSP/Montanide ISA-51 VG, i.m), Low Dose (200 µg HER1-ECD/VSSP/Tris NaCl, s.c.) and High Dose (800 µg HER1-ECD/VSSP/Tris NaCl, s.c). All monkeys received 7 doses and were daily inspected for clinical signs. Body weight, rectal temperature, cardiac and respiratory rates were measured during the study, and electrocardiographical and ophthalmological studies were performed. Humoral and cellular immune response and clinical pathology parameters were analyzed.Results: Animal's survival in the study was 100% (n = 15). Administration site reactions were observed in the Positive Control animals (n = 4). HER1 vaccine administered subcutaneously (High Dose Group) achieved good IgG antibody titers although lower than the Positive Control group, but with higher ability to inhibit HER1 phosphorylation. Conclusions: This suggests that the alternative of eliminating the use of Montanide in the HER1 vaccine preparation and the using subcutaneous route is feasible.


Assuntos
Vacinas Anticâncer/farmacologia , Animais , Vacinas Anticâncer/efeitos adversos , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Receptores ErbB/efeitos adversos , Receptores ErbB/farmacologia , Feminino , Injeções Subcutâneas , Masculino
6.
Semin Oncol ; 45(1-2): 75-83, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-30318087

RESUMO

The human epidermal growth factor receptor 1 (HER1) is a tumor-associated antigen that has been validated as a clinical target for several passive, non-immune therapies currently approved for the treatment of epithelial tumors. HER1 is an oncogene that not only promotes tumor progression and survival, but also immune escape. Its overexpression in some epithelial malignancies has been correlated with a poor prognosis. We developed an approach to target HER1 by specific active immunotherapy, recognizing the extracellular domain of the receptor, using a combination of VSSP and Montanide ISA 51 as adjuvants. We summarize the results obtained with this vaccine in both the preclinical and clinical settings, emphasizing the importance of the induction of both humoral and cellular responses for the success of cancer vaccines as safe therapeutic alternatives for the treatment of cancer.


Assuntos
Vacinas Anticâncer/imunologia , Receptores ErbB/imunologia , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Imunoterapia Ativa/métodos , Neoplasias/imunologia , Vacinas Anticâncer/uso terapêutico , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Modelos Imunológicos , Neoplasias/tratamento farmacológico , Resultado do Tratamento
7.
Front Pharmacol ; 8: 595, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29056908

RESUMO

Defining how epidermal growth factor receptor (EGFR)-targeting therapies influence the immune response is essential to increase their clinical efficacy. A growing emphasis is being placed on immune regulator genes that govern tumor - T cell interactions. Previous studies showed an increase in HLA class I cell surface expression in tumor cell lines treated with anti-EGFR agents. In particular, earlier studies of the anti-EGFR blocking antibody cetuximab, have suggested that increased tumor expression of HLA class I is associated with positive clinical response. We investigated the effect of another commercially available anti-EGFR antibody nimotuzumab on HLA class I expression in tumor cell lines. We observed, for the first time, that nimotuzumab increases HLA class I expression and its effect is associated with a coordinated increase in mRNA levels of the principal antigen processing and presentation components. Moreover, using 7A7 (a specific surrogate antibody against murine EGFR), we obtained results suggesting the importance of the increased MHC-I expression induced by EGFR-targeted therapies display higher in antitumor immune response. 7A7 therapy induced upregulation of tumor MHC-I expression in vivo and tumors treated with this antibody display higher susceptibility to CD8+ T cells-mediated lysis. Our results represent the first evidence suggesting the importance of the adaptive immunity in nimotuzumab-mediated antitumor activity. More experiments should be conducted in order to elucidate the relevance of this mechanism in cancer patients. This novel immune-related antitumor mechanism mediated by nimotuzumab opens new perspectives for its combination with various immunotherapeutic agents and cancer vaccines.

8.
ISRN Pharmacol ; 2012: 417515, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23227357

RESUMO

Immunocompetent mice, Fc receptor γ-chain deficient mice (Fcer1g(-/-)), and molecular tools as F(ab')(2) bivalent fragments appear as the most suitable biological models to study the mechanisms of the action of anti-epidermal growth factor receptor (EGFR) monoclonal antibodies (mAbs). In vivo experiments contrasting antitumor effects of whole Abs and their bivalent fragments commonly involve a previous comparative pharmacokinetics study. In this paper, pharmacokinetics and biodistribution of an anti-mouse EGFR Ab were assessed using immunocompetent mice. (125)I-labeled 7A7 mAb holds an elimination half-life (t(1/2)ß) of 23.1 h in C57BL/6 mice. Accumulation of mAb was found in liver, spleen, kidneys, and mostly in lungs. We used an ELISA method to determine the t(1/2)ß of a 7A7 mAb using the same experimental setting. Results from this new analysis revealed a t(1/2)ß of 23.9 h, supporting this method as a safer and easier system to evaluate pharmacokinetics parameters of mAbs targeting mouse EGFR. Using this system we also studied pharmacokinetics of 7A7 F(ab')(2) fragment. A tenfold difference between the mAb and fragment t(1/2)ß was found. These data support the use of the 7A7 F(ab')(2) fragment in in vivo studies to explore the contribution of the EGFR signaling blockade and the Fc region to the antitumor effect of 7A7 mAb in this autologous scenario.

9.
J Gene Med ; 14(3): 151-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22262303

RESUMO

BACKGROUND: Lung cancer remains a leading cause of cancer mortality, and so the aim of the present study was to develop a therapeutic vaccine protocol. METHODS: We constructed a lentiviral vector (LV) expressing the extracellular domain (ECD) of murine Her1, an antigen associated with poor prognosis in lung cancer. RESULTS: A single LV injection, followed by two Her1 protein boosts, was effective in reducing the metastatic burden of Lewis lung carcinoma in mice. The Her1 LV immunisation generated CD8+ T cells that recognised Her1 ECD presented by dendritic cells, and that also homed to Her1-expressing tumours. Protein boosting further increased the CD8+ T cell response and generated anti-Her1 antibodies; in the antibody response, Her1 LV priming increased Th1-dependent immunoglobulin G2c production. CONCLUSIONS: The ability of this vaccine protocol to break both T cell and B cell tolerance to a self-antigen likely explains its effectiveness.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer , Carcinoma Pulmonar de Lewis/imunologia , Receptores ErbB/metabolismo , Imunoterapia/métodos , Metástase Neoplásica/prevenção & controle , Animais , Anticorpos/imunologia , Receptores ErbB/genética , Vetores Genéticos/genética , Tolerância Imunológica/imunologia , Lentivirus , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA