Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Clin Oral Investig ; 28(8): 435, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39028340

RESUMO

OBJECTIVES: This study aimed to synthesize and characterize colloidal chitosan-silver nanoparticles-fluoride nanocomposite (CCAgNPF) and evaluate its efficacy compared to chlorhexidine on salivary Streptococcus mutans in orthodontic patients. MATERIALS AND METHODS: AgNPs stabilized with chitosan were synthesized by chemical reduction of AgNO3. The nanoparticles were characterized with SEM, FTIR, DLS and ICP-OES. The MIC and MBC against S. mutans and IC50 concentration of CCAgNPF were obtained for antibacterial and cytotoxicity evaluations, respectively. For the clinical study, a total of 45 orthodontic patients were divided into three groups of 15 and used the following mouthwashes twice a day for 1 month: CCAgNPF, chlorhexidine 0.2% and the combination of these mouthwashes. The colony count of salivary S. mutans was evaluated before and after using the mouthwashes. The data were analyzed using One-way ANOVA and Tukey's test. RESULTS: Stabilized AgNPs were spherical with a diameter of 25.3 ± 3.3 nm. The MIC, MBC and IC50 of CCAgNPF were 4.42, 8.85 and 18.89 µg/ml. All mouthwashes reduced the salivary S. mutans of the orthodontic patients, however, no significant difference was found between the efficacy of CCAgNPF and chlorhexidine (P-value > 0.05). The best results were achieved by the combination of CCAgNPF and chlorhexidine mouthwashes (P-value < 0.05). CONCLUSION: The CCAgNPF and its combination with chlorhexidine present potent bactericidal, biocompatible and effective anti-carious mouthwashes for orthodontic patients. CLINICAL RELEVANCE: This study proved CCAgNPF as an antibacterial mouthwash with lower cytotoxicity and side effects for patients undergoing orthodontic treatments to maintain oral hygiene and reduce salivary S. mutans.


Assuntos
Antibacterianos , Quitosana , Clorexidina , Fluoretos , Nanopartículas Metálicas , Antissépticos Bucais , Nanocompostos , Prata , Streptococcus mutans , Humanos , Streptococcus mutans/efeitos dos fármacos , Quitosana/farmacologia , Quitosana/química , Prata/farmacologia , Prata/química , Antissépticos Bucais/farmacologia , Antissépticos Bucais/química , Nanocompostos/química , Nanopartículas Metálicas/química , Antibacterianos/farmacologia , Feminino , Masculino , Fluoretos/farmacologia , Fluoretos/química , Clorexidina/farmacologia , Saliva/microbiologia , Adolescente , Testes de Sensibilidade Microbiana
2.
Molecules ; 28(1)2023 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-36615606

RESUMO

Magnetic mesoporous silica nanoparticles (MMSNPs) are being widely investigated as multifunctional novel drug delivery systems (DDSs) and play an important role in targeted therapy. Here, magnetic cores were synthesized using the thermal decomposition method. Further, to improve the biocompatibility and pharmacokinetic behavior, mesoporous silica was synthesized using the sol-gel process to coat the magnetic cores. Subsequently, sunitinib (SUN) was loaded into the MMSNPs, and the particles were armed with amine-modified mucin 1 (MUC-1) aptamers. The MMSNPs were characterized using FT-IR, TEM, SEM, electrophoresis gel, DLS, and EDX. MTT assay, flow cytometry analysis, ROS assessment, and mitochondrial membrane potential analysis evaluated the nanoparticles' biological impacts. The physicochemical analysis revealed that the engineered MMSNPs have a smooth surface and spherical shape with an average size of 97.6 nm. The biological in vitro analysis confirmed the highest impacts of the targeted MMSNPs in MUC-1 overexpressing cells (OVCAR-3) compared to the MUC-1 negative MDA-MB-231 cells. In conclusion, the synthesized MMSNP-SUN-MUC-1 nanosystem serves as a unique multifunctional targeted delivery system to combat the MUC-1 overexpressing ovarian cancer cells.


Assuntos
Nanopartículas , Neoplasias Ovarianas , Humanos , Feminino , Sunitinibe , Linhagem Celular Tumoral , Apoptose , Dióxido de Silício/química , Mucina-1/genética , Espectroscopia de Infravermelho com Transformada de Fourier , Nanopartículas/química , Sistemas de Liberação de Medicamentos/métodos , Fenômenos Magnéticos , Porosidade
3.
Infection ; 50(2): 295-308, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34902115

RESUMO

PURPOSE: The Coronavirus disease 2019 (COVID-19) pandemic is one of the most devastating global problems. Regarding the lack of disease-specific treatments, repurposing drug therapy is currently considered a promising therapeutic approach in pandemic situations. Recently, the combination therapy of Janus kinase (JAK) inhibitor baricitinib has been authorized for emergency COVID-19 hospitalized patients; however, this strategy's safety, drug-drug interactions, and cellular signaling pathways remain a tremendous challenge. METHODS: In this study, we aimed to provide a deep insight into the baricitinib combination therapies in severe COVID-19 patients through reviewing the published literature on PubMed, Scopus, and Google scholar databases. We also focused on cellular and subcellular pathways related to the synergistic effects of baricitinib plus antiviral agents, virus entry, and cytokine storm (CS) induction. The safety and effectiveness of this strategy have also been discussed in moderate to severe forms of COVID-19 infection. RESULTS: The severity of COVID-19 is commonly associated with a dysregulated immune response and excessive release of pro-inflammatory agents, resulting in CS. It has been shown that baricitinib combined with antiviral agents could modulate the inflammatory response and provide a series of positive therapeutic outcomes in hospitalized adults and pediatric patients (age ≥ two years old). CONCLUSION: Baricitinib plus the standard of care treatment might be a potential strategy in hospitalized patients with severe COVID-19.


Assuntos
Tratamento Farmacológico da COVID-19 , Inibidores de Janus Quinases , Adulto , Azetidinas , Criança , Pré-Escolar , Humanos , Inibidores de Janus Quinases/uso terapêutico , Purinas , Pirazóis , SARS-CoV-2 , Sulfonamidas
4.
Int J Mol Sci ; 21(12)2020 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-32580338

RESUMO

Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) are inhibitory checkpoints that are commonly seen on activated T cells and have been offered as promising targets for the treatment of cancers. Immune checkpoint inhibitors (ICIs)targeting PD-1, including pembrolizumab and nivolumab, and those targeting its ligand PD-L1, including avelumab, atezolizumab, and durvalumab, and two drugs targeting CTLA-4, including ipilimumab and tremelimumab have been approved for the treatment of several cancers and many others are under investigating in advanced trial phases. ICIs increased antitumor T cells' responses and showed a key role in reducing the acquired immune system tolerance which is overexpressed by cancer and tumor microenvironment. However, 50% of patients could not benefit from ICIs monotherapy. To overcome this, a combination of ipilimumab and nivolumab is frequently investigated as an approach to improve oncological outcomes. Despite promising results for the combination of ipilimumab and nivolumab, safety concerns slowed down the development of such strategies. Herein, we review data concerning the clinical activity and the adverse events of ipilimumab and nivolumab combination therapy, assessing ongoing clinical trials to identify clinical outlines that may support combination therapy as an effective treatment. To the best of our knowledge, this paper is one of the first studies to evaluate the efficacy and safety of ipilimumab and nivolumab combination therapy in several cancers.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ensaios Clínicos como Assunto/estatística & dados numéricos , Neoplasias/tratamento farmacológico , Padrões de Prática Médica , Humanos , Ipilimumab/administração & dosagem , Neoplasias/patologia , Nivolumabe/administração & dosagem , Prognóstico
5.
Int J Mol Sci ; 21(14)2020 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-32708748

RESUMO

Gynecologic cancers account for approximately 11% of the newly diagnosed cancers in women in the United States and for 18% globally. The presence of tumor-infiltrating lymphocytes (TILs) influences the clinical outcome of cancer patients and immune checkpoint inhibitors (ICIs), including anti programmed cell death protein-1 (anti-PD-1), anti-programmed death-ligand 1 (anti-PD-L1), and anticytotoxic T-lymphocyte antigen 4 (anti-CTLA-4), which have been approved for treating different types of malignancies. Antibodies targeting the PD-1/PD-L1 checkpoint have shown dynamic and durable tumor regressions, suggesting a rebalancing of the host-tumor interaction. There are several the US food and drug administration (FDA)-approved ICIs targeting PD-1, including pembrolizumab and nivolumab, as well as those targeting PD-L1, including avelumab, atezolizumab, and durvalumab for melanoma, renal cell cancer, colorectal cancer, head and neck cancer, cervix cancer, urothelial cancer, and lung cancer. Current pre-clinical and clinical studies assessing PD-1/PD-L1 inhibitors in several gynecologic cancers have reported significant antitumor activity. In this review, we investigate pre-clinical and clinical studies that describe the safety and efficacy of anti-PD-1/PD-L1 antibodies, with a particular focus on ongoing clinical trials, analyzing the oncological outcome and adverse effects of ICIs in gynecologic cancers.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Neoplasias dos Genitais Femininos/tratamento farmacológico , Inibidores de Checkpoint Imunológico/uso terapêutico , Animais , Antineoplásicos Imunológicos/efeitos adversos , Feminino , Neoplasias dos Genitais Femininos/imunologia , Neoplasias dos Genitais Femininos/patologia , Neoplasias dos Genitais Femininos/terapia , Genitália Feminina/efeitos dos fármacos , Genitália Feminina/imunologia , Genitália Feminina/patologia , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos
6.
Bioimpacts ; 13(3): 255-267, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37431477

RESUMO

Introduction: Mesoporous silica nanoparticles (MSNPs) are considered innovative multifunctional structures for targeted drug delivery owing to their outstanding physicochemical characteristics. Methods: MSNPs were fabricated using the sol-gel method, and polyethylene glycol-600 (PEG600) was used for MSNPs modification. Subsequently, sunitinib (SUN) was loaded into the MSNPs, MSNP-PEG and MSNP-PEG/SUN were grafted with mucin 16 (MUC16) aptamers. The nanosystems (NSs) were characterized using FT-IR, TEM, SEM, DLS, XRD, BJH, and BET. Furthermore, the biological impacts of MSNPs were evaluated on the ovarian cancer cells by MTT assay and flow cytometry analysis. Results: The results revealed that the MSNPs have a spherical shape with an average dimension, pore size, and surface area of 56.10 nm, 2.488 nm, and 148.08 m2g-1, respectively. The cell viability results showed higher toxicity of targeted MSNPs in MUC16 overexpressing OVCAR-3 cells as compared to the SK-OV-3 cells; that was further confirmed by the cellular uptake results. The cell cycle analysis exhibited that the induction of sub-G1 phase arrest mostly occurred in MSNP-PEG/SUN-MUC16 treated OVCAR-3 cells and MSNP-PEG/SUN treated SK-OV-3 cells. DAPI staining showed apoptosis induction upon exposure to targeted MSNP in MUC16 positive OVCAR-3 cells. Conclusion: According to our results, the engineered NSs could be considered an effective multifunctional targeted drug delivery platform for the mucin 16 overexpressing cells.

7.
Expert Opin Biol Ther ; 22(7): 895-909, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35687355

RESUMO

INTRODUCTION: Melittin (MLT), a natural membrane-active component, is the most prominent cytolytic peptide from bee venom. Remarkable biological properties of MLT, including anti-inflammatory, antimicrobial, anticancer, anti-protozoan, and antiarthritic activities, make it an up-and-coming therapeutic candidate for a wide variety of human diseases. Therapeutic applications of MLT may be hindered due to low stability, high toxicity, and weak tissue penetration. Different bio-nano scale modifications hold promise for improving its functionality and therapeutic efficacy. AREAS COVERED: In the current review, we aimed to provide a comprehensive insight into strategies used for MLT conjugations and modifications, cellular delivery of modified forms, and their clinical perspectives by reviewing the published literature on PubMed, Scopus, and Google Scholar databases. We also emphasized the MLT structure modifications, mechanism of action, and cellular toxicity. EXPERT OPINION: Developing new analogs and conjugates of MLT as a natural drug with improved functions and fewer side effects is crucial for the clinical translation of this approach worldwide, especially where the chemicals and synthetic drugs are more expensive or unavailable in the healthcare system. MLT-nanoconjugation may be one of the best-optimized strategies for improving peptide delivery, increasing its therapeutic efficacy, and providing minimal nonspecific cellular lytic activity. [Figure: see text].


Assuntos
Anti-Infecciosos , Venenos de Abelha , Humanos , Meliteno/efeitos adversos , Peptídeos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa