Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(5)2023 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-36901982

RESUMO

Photodynamic therapy (PDT) using 5-aminolevulinic acid (ALA) which is the precursor of the photosensitizer protoporphyrin IX (PpIX) is an available treatment for several diseases. ALA-PDT induces the apoptosis and necrosis of target lesions. We have recently reported the effects of ALA-PDT on cytokines and exosomes of human healthy peripheral blood mononuclear cells (PBMCs). This study has investigated the ALA-PDT-mediated effects on PBMC subsets from patients with active Crohn's disease (CD). No effects on lymphocyte survival after ALA-PDT were observed, although the survival of CD3-/CD19+ B-cells seemed slightly reduced in some samples. Interestingly, ALA-PDT clearly killed monocytes. The subcellular levels of cytokines and exosomes associated with inflammation were widely downregulated, which is consistent with our previous findings in PBMCs from healthy human subjects. These results suggest that ALA-PDT may be a potential treatment candidate for CD and other immune-mediated diseases.


Assuntos
Doença de Crohn , Exossomos , Fotoquimioterapia , Humanos , Ácido Aminolevulínico/farmacologia , Leucócitos Mononucleares , Doença de Crohn/tratamento farmacológico , Citocinas , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas , Linhagem Celular Tumoral
2.
Biomedicines ; 10(2)2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35203441

RESUMO

Photodynamic therapy (PDT) with 5-aminolevulinic acid (ALA), a precursor to the potent photosensitizer, protoporphyrin IX (PpIX), is an established modality for several malignant and premalignant diseases. This treatment is based on the light-activated PpIX in targeted lesions. Although numerous studies have confirmed the necrosis and apoptosis involved in the mechanism of action of this modality, little information is available for the change of exosome levels after treatment. We report from the first study on the effects of ALA-PDT on cytokines and exosomes of human healthy peripheral blood mononuclear cells (PBMCs). The treatment reduced the cytokines and exosomes studied, although there was variation among individual PBMC samples. This reduction is consistent with PDT-mediated survivals of subsets of PBMCs. More specifically, the ALA-PDT treatment apparently decreased all pro-inflammatory cytokines included, suggesting that this treatment may provide a strong anti-inflammatory effect. In addition, the treatment has decreased the levels of different types of exosomes, the HLA-DRDPDQ exosome in particular, which plays an important role in the rejection of organ transplantation as well as autoimmune diseases. These results may suggest future therapeutic strategies of ALA-PDT.

3.
Biomedicines ; 10(2)2022 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-35203632

RESUMO

Photodynamic therapy (PDT) is a non-invasive therapeutic modality based on the interaction between a photosensitive molecule called photosensitizer (PS) and visible light irradiation in the presence of oxygen molecule. Protoporphyrin IX (PpIX), an efficient and widely used PS, is hampered in clinical PDT by its poor water-solubility and tendency to self-aggregate. These features are strongly related to the PS hydrophilic-lipophilic balance. In order to improve the chemical properties of PpIX, a series of amphiphilic PpIX derivatives endowed with PEG550 headgroups and hydrogenated or fluorinated tails was synthetized. Hydrophilic-lipophilic balance (HLB) and log p-values were computed for all of the prepared compounds. Their photochemical properties (spectroscopic characterization, photobleaching, and singlet oxygen quantum yield) were also evaluated followed by the in vitro studies of their cellular uptake, subcellular localization, and photocytotoxicity on three tumor cell lines (4T1, scc-U8, and WiDr cell lines). The results confirm the therapeutic potency of these new PpIX derivatives. Indeed, while all of the derivatives were perfectly water soluble, some of them exhibited an improved photodynamic effect compared to the parent PpIX.

4.
Cancer Rep (Hoboken) ; 5(12): e1278, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-32737955

RESUMO

BACKGROUND: Photodynamic therapy (PDT) is a minimally invasive, clinically approved therapy with numerous advantages over other mainstream cancer therapies. 5-aminolevulinic acid (5-ALA)-PDT is of particular interest, as it uses the photosensitiser PpIX, naturally produced in the heme pathway, following 5-ALA administration. Even though 5-ALA-PDT shows high specificity to cancers, differences in treatment outcomes call for predictive biomarkers to better stratify patients and to also diversify 5-ALA-PDT based on each cancer's phenotypic and genotypic individualities. AIMS: The present study seeks to highlight key biomarkers that may predict treatment outcome and simultaneously be exploited to overcome cancer-specific resistances to 5-ALA-PDT. METHODS AND RESULTS: We submitted two glioblastoma (T98G and U87) and three breast cancer (MCF7, MDA-MB-231, and T47D) cell lines to 5-ALA-PDT. Glioblastoma cells were the most resilient to 5-ALA-PDT, while intracellular production of 5-ALA-derived protoporphyrin IX (PpIX) could not account for the recorded PDT responses. We identified the levels of expression of ABCG2 transporters, ferrochelatase (FECH), and heme oxygenase (HO-1) as predictive biomarkers for 5-ALA-PDT. GPX4 and GSTP1 expression vs intracellular glutathione (GSH) levels also showed potential as PDT biomarkers. For T98G cells, inhibition of ABCG2, FECH, HO-1, and/or intracellular GSH depletion led to profound PDT enhancement. Inhibition of ABCG2 in U87 cells was the only synergistic adjuvant to 5-ALA-PDT, rendering the otherwise resistant cell line fully responsive to 5-ALA-PDT. ABCG2 or FECH inhibition significantly enhanced 5-ALA-PDT-induced MCF7 cytotoxicity, while for MDA-MB-231, ABCG2 inhibition and intracellular GSH depletion conferred profound synergies. FECH inhibition was the only synergism to ALA-PDT for the most susceptible among the cell lines, T47D cells. CONCLUSION: This study demonstrates the heterogeneity in the cellular response to 5-ALA-PDT and identifies biomarkers that may be used to predict treatment outcome. The study also provides preliminary findings on the potential of inhibiting specific molecular targets to overcome inherent resistances to 5-ALA-PDT.


Assuntos
Glioblastoma , Fotoquimioterapia , Humanos , Ácido Aminolevulínico/farmacologia , Fotoquimioterapia/métodos , Glioblastoma/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Biomarcadores
6.
Pharmaceutics ; 13(10)2021 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-34683851

RESUMO

Extracorporeal photopheresis (ECP), an immunomodulatory therapy for the treatment of chronic graft-versus-host disease (cGvHD), exposes isolated white blood cells to photoactivatable 8-methoxypsoralen (8-MOP) and UVA light to induce the apoptosis of T-cells and, hence, to modulate immune responses. However, 8-MOP-ECP kills diseased and healthy cells with no selectivity and has limited efficacy in many cases. The use of 5-aminolevulinic acid (ALA) and light (ALA-based photodynamic therapy) may be an alternative, as ex vivo investigations show that ALA-ECP kills T-cells from cGvHD patients more selectively and efficiently than those treated with 8-MOP-ECP. The purpose of this phase I-(II) study was to evaluate the safety and tolerability of ALA-ECP in cGvHD patients. The study included 82 treatments in five patients. One patient was discharged due to the progression of the haematological disease. No significant persistent changes in vital signs or laboratory values were detected. In total, 62 adverse events were reported. Two events were severe, 17 were moderate, and 43 were mild symptoms. None of the adverse events evaluated by the internal safety review committee were considered to be likely related to the study medication. The results indicate that ALA-ECP is safe and is mainly tolerated well by cGvHD patients.

7.
Cancers (Basel) ; 13(13)2021 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-34283089

RESUMO

Photodynamic therapy (PDT) is a treatment strategy that utilizes photosensitizers (PSs) and light of a specific wavelength to kill cancer cells. However, limited tumor specificity is still a drawback for the clinical application of PDT. To increase the therapeutic efficacy and specificity of PDT, a novel human minibody (MS5) that recognizes a cell surface receptor expressed on various cancer cells was labeled with the hydrophilic phthalocyanine PS IR700 to generate an MS5-IR700 conjugate that is activated by near-infrared (NIR) light. The phototoxicity of the conjugate was mainly tested against the PC3 prostate cancer cell line. The MS5-IR700 conjugate killed PC3 cells after NIR light irradiation as compared to untreated cells or cells treated with IR700 alone. Time-course analysis of cell viability revealed a high percentage of cell death during the first hour in PC3 cells exposed to the MS5-IR700 conjugate and NIR light irradiation. After irradiation, the MS5-IR700 conjugate-treated PC3 cells displayed cellular swelling, round shape, and rupture of the cell and nuclear membranes. In a co-culture model, the MS5-IR700 conjugate killed MS5-positive Ramos lymphoma cells specifically, while leaving MS5-negative cells unaffected. In line with the data obtained with the monolayer cultures, the MS5-IR700 conjugate also killed PC3 cancer cell spheroids. The treatment induced relocation of heat shock protein 70 and calreticulin to the cell surface, implying the induction of immunogenic cell death. Overall, the data suggest that the developed MS5-IR700 conjugate is a promising therapeutic agent that warrants further preclinical studies.

8.
Int J Mol Sci ; 22(9)2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34062920

RESUMO

Radioligand therapy targeting the prostate-specific membrane antigen (PSMA) is rapidly evolving as a promising treatment for metastatic castration-resistant prostate cancer. The PSMA-targeting ligand p-SCN-Bn-TCMC-PSMA (NG001) labelled with 212Pb efficiently targets PSMA-positive cells in vitro and in vivo. The aim of this preclinical study was to evaluate the therapeutic potential of 212Pb-NG001 in multicellular tumour spheroid and mouse models of prostate cancer. The cytotoxic effect of 212Pb-NG001 was tested in human prostate C4-2 spheroids. Biodistribution at various time points and therapeutic effects of different activities of the radioligand were investigated in male athymic nude mice bearing C4-2 tumours, while long-term toxicity was studied in immunocompetent BALB/c mice. The radioligand induced a selective cytotoxic effect in spheroids at activity concentrations of 3-10 kBq/mL. In mice, the radioligand accumulated rapidly in tumours and was retained over 24 h, while it rapidly cleared from nontargeted tissues. Treatment with 0.25, 0.30 or 0.40 MBq of 212Pb-NG001 significantly inhibited tumour growth and improved median survival with therapeutic indexes of 1.5, 2.3 and 2.7, respectively. In BALB/c mice, no signs of long-term radiation toxicity were observed at activities of 0.05 and 0.33 MBq. The obtained results warrant clinical studies to evaluate the biodistribution, therapeutic efficacy and toxicity of 212Pb-NG001.


Assuntos
Neoplasias de Próstata Resistentes à Castração/radioterapia , Ensaio Radioligante , Compostos Radiofarmacêuticos/farmacologia , Esferoides Celulares/efeitos da radiação , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Xenoenxertos , Humanos , Chumbo/farmacologia , Ligantes , Masculino , Camundongos , Metástase Neoplásica , Neoplasias de Próstata Resistentes à Castração/patologia , Radioisótopos/farmacologia , Esferoides Celulares/patologia , Distribuição Tecidual/efeitos da radiação
9.
Cancers (Basel) ; 12(2)2020 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-32041351

RESUMO

Extracorporeal photopheresis (ECP), a modality that exposes isolated leukocytes to the photosensitizer 8-methoxypsoralen (8-MOP) and ultraviolet-A (UV-A) light, is used to treat conditions such as cutaneous T-cell lymphoma and graft-versus-host disease. However, the current procedure of ECP has limited selectivity and efficiency; and produces only partial response in the majority of treated patients. Additionally, the treatment is expensive and time-consuming, so the improvement for this modality is needed. In this study, we used the concept of photodynamic therapy (PDT) with 5-aminolevulinic acid (ALA), a precursor of an endogenously synthesized photosensitizer protoporphyrin IX (PpIX) in combination with blue light to explore the possibility of targeting activated human blood T cells ex vivo. With various T-cell activation protocols, a high ALA-induced PpIX production took place in activated CD3+, CD4+CD25+, and CD8+ T cell populations with their subsequent killing after blue light exposure. By contrast, resting T cells were much less damaged by the treatment. The selective and effective killing effect on the activated cells was also seen after co-cultivating activated and resting T cells. Under our clinically relevant experimental conditions, ALA-PDT killed activated T cells more selectively and efficiently than 8-MOP/UV-A. Monocyte-derived dendritic cells (DCs) were not affected by the treatment. Incubation of ALA-PDT damaged T cells with autologous DCs induced a downregulation of the co-stimulatory molecules CD80/CD86 and also upregulation of interleukin 10 (IL-10) and indoleamine 2,3-dioxygenase expression, two immunosuppressive factors that may account for the generation of tolerogenic DCs. Overall, the data support the potential use of ALA-PDT strategy for improving ECP by selective and effective killing of activated T cells and induction of immune tolerance.

10.
Int J Oncol ; 53(6): 2319-2331, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30334567

RESUMO

Protein kinase B (Akt), similar to many other protein kinases, is at the crossroads of cell death and survival, playing a pivotal role in multiple interconnected cell signaling mechanisms implicated in cell metabolism, growth and division, apoptosis suppression and angiogenesis. Akt protein kinase displays important metabolic effects, among which are glucose uptake in muscle and fat cells or the suppression of neuronal cell death. Disruptions in the Akt­regulated pathways are associated with cancer, diabetes, cardiovascular and neurological diseases. The regulation of the Akt signaling pathway renders Akt a valuable therapeutic target. The discovery process of Akt inhibitors using various strategies has led to the identification of inhibitors with great selectivity, low side­effects and toxicity. The usefulness of Akt emerges beyond cancer therapy and extends to other major diseases, such as diabetes, heart diseases, or neurodegeneration. This review presents key features of Akt structure and functions, and presents the progress of Akt inhibitors in regards to drug development, and their preclinical and clinical activity in regards to therapeutic efficacy and safety for patients.


Assuntos
Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Ensaios Clínicos como Assunto , Humanos , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/química , Transdução de Sinais/efeitos dos fármacos
11.
Oncotarget ; 8(7): 10966-10979, 2017 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-28030798

RESUMO

Recent studies have shown synergistic cytotoxic effects of simultaneous Chk1- and Wee1-inhibition. However, the mechanisms behind this synergy are not known. Here, we present a flow cytometry-based screen for compounds that cause increased DNA damage in S-phase when combined with the Wee1-inhibitor MK1775. Strikingly, the Chk1-inhibitors AZD7762 and LY2603618 were among the top candidate hits of 1664 tested compounds, suggesting that the synergistic cytotoxic effects are due to increased S-phase DNA damage. Combined Wee1- and Chk1-inhibition caused a strong synergy in induction of S-phase DNA damage and reduction of clonogenic survival. To address the underlying mechanisms, we developed a novel assay measuring CDK-dependent phosphorylations in single S-phase cells. Surprisingly, while Wee1-inhibition alone induced less DNA damage compared to Chk1-inhibition, Wee1-inhibition caused a bigger increase in S-phase CDK-activity. However, the loading of replication initiation factor CDC45 was more increased after Chk1- than Wee1-inhibition and further increased by the combined treatment, and thus correlated well with DNA damage. Therefore, when Wee1 alone is inhibited, Chk1 suppresses CDC45 loading and thereby limits the extent of unscheduled replication initiation and subsequent S-phase DNA damage, despite very high CDK-activity. These results can explain why combined treatment with Wee1- and Chk1-inhibitors gives synergistic anti-cancer effects.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Fase S/efeitos dos fármacos , Células A549 , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem/metabolismo , Dano ao DNA , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/genética , Sinergismo Farmacológico , Citometria de Fluxo , Humanos , Proteínas Nucleares/metabolismo , Compostos de Fenilureia/farmacologia , Proteínas Tirosina Quinases/metabolismo , Pirazinas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Pirimidinonas , Fase S/genética , Tiofenos/farmacologia , Ureia/análogos & derivados , Ureia/farmacologia
12.
Int J Oncol ; 48(3): 869-85, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26698230

RESUMO

Targeted cancer therapies are used to inhibit the growth, progression, and metastasis of the tumor by interfering with specific molecular targets and are currently the focus of anticancer drug development. Protein kinase B, also known as Akt, plays a central role in many types of cancer and has been validated as a therapeutic target nearly two decades ago. This review summarizes the intracellular functions of Akt as a pivotal point of converging signaling pathways involved in cell growth, proliferation, apoptotis and neo­angiogenesis, and focuses on the drug design strategies to develop potent anticancer agents targeting Akt. The discovery process of Akt inhibitors has evolved from adenosine triphosphate (ATP)­competitive agents to alternative approaches employing allosteric sites in order to overcome the high degree of structural similarity between Akt isoforms in the catalytic domain, and considerable structural analogy to the AGC kinase family. This process has led to the discovery of inhibitors with greater specificity, reduced side-effects and lower toxicity. A second generation of Akt has inhibitors emerged by incorporating a chemically reactive Michael acceptor template to target the nucleophile cysteines in the catalytic activation loop. The review outlines the development of several promising drug candidates emphasizing the importance of each chemical scaffold. We explore the pipeline of Akt inhibitors and their preclinical and clinical examination status, presenting the potential clinical application of these agents as a monotherapy or in combination with ionizing radiation, other targeted therapies, or chemotherapy.


Assuntos
Inibidores Enzimáticos/química , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Trifosfato de Adenosina/química , Sítio Alostérico , Animais , Antineoplásicos/química , Apoptose , Catálise , Domínio Catalítico , Proliferação de Células , Ensaios Clínicos como Assunto , Desenho de Fármacos , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/enzimologia , Isoformas de Proteínas/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
13.
J Control Release ; 213: 152-167, 2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26160308

RESUMO

Nanocarriers based on polymers, metals and lipids have been extensively developed for cancer therapy and diagnosis due to their ability to enhance drug accumulation in cancer cells and decrease undesired drug toxicity in healthy tissues. Overcoming multidrug resistance by designing proper drug nanocarriers will improve outcome of existing oncologic treatments such as chemotherapy and radiotherapy. In this article the relation between physicochemical properties and capacity of a nanosystem to deliver therapeutic agents into pathological sites is discussed. Most promising examples of drug delivery systems are reviewed, and, in particular, the design of a carbohydrate based matrix with entrapped gold nanoparticles is highlighted.


Assuntos
Antineoplásicos/administração & dosagem , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Humanos , Metais/química , Nanotecnologia/métodos , Polímeros/química , Polissacarídeos/química
14.
Colloids Surf B Biointerfaces ; 129: 79-86, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25829130

RESUMO

Nanoparticulates responsive to X-rays offer increased efficacy of radiation therapy. However, successful demonstrations of such nanoparticle use are limited so far due to lack of significant radiosensitizing effects or poor nanoparticle stability in a biological system. Zinc oxide (ZnO) is the most promising biocompatible material for medicinal applications. In this paper, we report preparation and characterization of scintillating ZnO/SiO2 core-shell nanoparticles. The ZnO/SiO2 nanoparticles absorb ultraviolet (UV) radiation (below 360nm) and emit green fluorescence (400-750nm, maximum 550nm). Under X-ray irradiation (200kVp), the nanoparticles scintillate emitting luminescence in the region 350-700nm (maximum 420nm). The synthesized ZnO/SiO2 nanoparticles are stable in a biologically relevant environment (water and cell growth medium). The potential of the ZnO/SiO2 nanoparticles for radiosensitization is demonstrated in human prostate adenocarcinoma cell lines (LNCaP and Du145). The nanoparticles enhance radiation-induced reduction in cell survival about 2-fold for LNCaP and 1.5-fold for Du145 cells. Radiosensitizing effect can be attributed to X-ray-induced radiocatalysis by the nanoparticles.


Assuntos
Adenocarcinoma/radioterapia , Nanopartículas Metálicas/química , Nanocompostos/química , Neoplasias da Próstata/radioterapia , Radiossensibilizantes/farmacologia , Dióxido de Silício/química , Óxido de Zinco/química , Adenocarcinoma/patologia , Sobrevivência Celular/efeitos da radiação , Humanos , Luminescência , Masculino , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas , Raios X
15.
J Biomed Nanotechnol ; 10(4): 717-23, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24734524

RESUMO

Colloidal gold nanoparticles intensify the anticancer response of the drug bortezomib, a proteasome inhibitor. Polyethylene glycol-coated gold nanoparticles and the drug show a synergistic effect in reducing the cell viability of prostate cancer cell line Du145. It was observed a significant cell viability reduction with bortezomib concentrations as low as 4 nM. The proteasome inhibitor alone had to be present at concentrations in the ranger of 120 nM to induce identical cytotoxicity response. These findings demonstrate that gold nanoparticles enhancing the permeation and retention (EPR) effect in Du145 cells and open the possibility to decrease multi-drug resistance (MDR). The in vitro results of functionalized gold nanoparticles, internalized by cancer cells, pave the way for a more efficient proteasome inhibitor delivery and release in adenocarcinoma cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Ácidos Borônicos/administração & dosagem , Ouro/administração & dosagem , Nanopartículas Metálicas/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Inibidores de Proteassoma/administração & dosagem , Pirazinas/administração & dosagem , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Bortezomib , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Masculino , Resultado do Tratamento
16.
Nanomedicine (Lond) ; 9(15): 2339-51, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24471504

RESUMO

AIM: In this study, Ce(3+)-doped lanthanum(III) fluoride (LaF3:Ce(3+)) nanoparticles were synthesized by a wet-chemistry method in dimethyl sulfoxide (DMSO) and their application as an intracellular light source for photodynamic activation was demonstrated. MATERIALS & METHODS: The LaF3:Ce(3+)/DMSO nanoparticles have a strong green emission with a peak at approximately 520 nm, which is effectively overlapped with the absorption of protoporphyrin IX (PPIX). The nanoparticles were encapsulated into poly(D,L-lactide-co-glycolide (PLGA) microspheres along with PPIX. Upon irradiation with x-rays (90 kV), energy transfer from the LaF3:Ce(3+)/DMSO nanoparticles to PPIX occurs and singlet oxygen is generated for cancer cell damage. RESULTS: The LaF3:Ce(3+)/DMSO/PLGA or LaF3:Ce(3+)/DMSO/PPIX/PLGA microspheres alone caused only sublethal cytotoxicity to the cancer cells. Upon x-ray irradiation, the LaF3:Ce(3+)/DMSO/PPIX/PLGA microspheres induced oxidative stress, mitochondrial damage and DNA fragmentation on prostate cancer cells (PC3). DISCUSSION: The results indicate that x-rays can activate LaF3:Ce(3+) and PPIX nanocomposites, which can be a novel method for cancer destruction.


Assuntos
Nanopartículas , Neoplasias/tratamento farmacológico , Fotoquimioterapia , Linhagem Celular Tumoral , Transferência de Energia , Corantes Fluorescentes , Humanos , Neoplasias/patologia , Raios X
17.
Expert Opin Drug Deliv ; 10(10): 1345-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23937147

RESUMO

BACKGROUND: Proteasome inhibition is a current therapeutic strategy used in the treatment of multiple myeloma. Drugs controlling proteasome activity are ideally suited for unidirectional manipulation of cellular pathways such as apoptosis. The first proteasome inhibitor approved in clinics was bortezomib. This drug is currently used in combination with other anticancer agents. OBJECTIVES: In this study, the enhancement of bortezomib activity was evaluated using gold nanoparticles coated with poly(ethylene glycol). The uptake mechanism of the gold nanoparticles in pancreatic cell lines, S2-013 and hTERT-HPNE, was assessed by laser scanning confocal microscopy (LSCM). RESULTS: Pancreatic cancer cells internalized the nanoparticles together with the drug in few minutes through the formation of endocytic vesicles. This rapid uptake leads to an increase in the concentration and diffusion of bortezomib in the cytoplasm yielding an increased toxicity on the cells when compared to the drug alone. CONCLUSION: Gold nanoparticles can be used as effective delivery systems to increasing the permeation and retention of drugs in cancer cells.


Assuntos
Adenocarcinoma/tratamento farmacológico , Ácidos Borônicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Ouro/química , Nanopartículas/química , Neoplasias Pancreáticas/tratamento farmacológico , Inibidores de Proteassoma/administração & dosagem , Pirazinas/administração & dosagem , Adenocarcinoma/patologia , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Bortezomib , Sinergismo Farmacológico , Células Epiteliais/efeitos dos fármacos , Humanos , Ductos Pancreáticos/citologia , Neoplasias Pancreáticas/patologia , Polietilenoglicóis/química , Células Tumorais Cultivadas
18.
Nanotechnology ; 24(32): 325103, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23868054

RESUMO

Spherical carbon nanoparticles (carbon nanodots) with a silver shell were investigated as potential sensitizing agents. The cytotoxicity of the combination of ultraviolet radiation or x-rays with the nanodots was examined in cancer cells in vitro. The cell viability decreased following the exposure to the radiation. The carbon nanodots enhanced the radiation effects by significantly reducing the amount of surviving cells compared to that of the cells exposed only to the radiation. Carbon-core silver-shell nanodots can be proposed as a bimodal sensitization platform for biological and medicinal applications employing non-ionizing or ionizing radiation.


Assuntos
Carbono/química , Fármacos Fotossensibilizantes/farmacologia , Fototerapia , Radioterapia , Prata/química , Catálise/efeitos da radiação , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Humanos , Hidrodinâmica , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Espectrometria de Fluorescência , Raios Ultravioleta
19.
Biochim Biophys Acta ; 1820(7): 870-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22507270

RESUMO

BACKGROUND: Blebbistatin is a new inhibitor of cell motility. It is used to study dynamics of cytokinesis machinery in cells. However, the potential of this inhibitor as an anticancer agent has not been studied so far. METHODS: Cytotoxicity of blebbistatin was evaluated in five human cell lines, FEMX-I melanoma, U87 glioma, androgen independent Du145 and androgen sensitive LNCaP prostate adenocarcinoma, and F11-hTERT immortalized fibroblasts. Phototoxicity of blebbistatin was assessed in these cell lines after their exposure to a blue light (390-470 nm). Photostability of blebbistatin and its reactive oxygen species (ROS) generating properties were measured during irradiation with the blue light. RESULTS: Blebbistatin at a concentration range of 10-200 µmol/L was toxic to all studied cells. Toxic concentrations (TC) were about 10-25 µmol/L corresponding to TC10, 50-100 µmol/L to TC50 and 140-190 µmol/L to TC90. Only for the U87 glioma cells TC90 could not be measured as the highest studied concentration of 200 µmol/L gave around 70% toxicity. However, after exposure to the blue light blebbistatin exhibited phototoxicity on the cells, with a cytotoxicity enhancement ratio that was greatest for the FEMX-I cells (about 9) followed by LNCaP (5), Du145 (3), U87 (2) and F11-hTERT (1.7) cells. CONCLUSIONS: Blebbistatin inhibits cell motility and viability. Under exposure to the blue light blebbistatin exhibits photodynamic action on human cancer cells. During the irradiation blebbistatin oxidizes dihydrorhodamine 123 but not Singlet Oxygen Sensor Green. GENERAL SIGNIFICANCE: Our findings offer new possibilities for blebbistatin as a potential anticancer and photodynamic agent.


Assuntos
Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Luz , Miosinas/antagonistas & inibidores , Fármacos Fotossensibilizantes/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Miosinas/metabolismo , Oxirredução , Neoplasias da Próstata/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas
20.
J Biomed Nanotechnol ; 7(5): 667-76, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22195484

RESUMO

In this study we report the effect of classical CdSe/ZnS quantum dots and novel spherical carbon dots on generation of singlet oxygen and other reactive oxygen species (ROS) in aqueous solutions in vitro. Free radicals were initiated either chemically using 2,2'-azodiisobutyramidine dihydrochloride (AAPH) or by radiation with a blue light source emitting 390-470 nm (peak 420 nm). Two reagents, dihydrorhodamine 123 (Dhr123) and singlet oxygen sensor green (SOSG), were used as radical probes. Quantum dots and carbon dots inhibited oxidation of the radical probes under decomposition of AAPH. However, when subjected to the blue light both the quantum dots and carbon dots induced oxidation of Dhr123 to a greater extent than SOSG in water. Generation of singlet oxygen was remarkably enhanced in deuterium oxide solutions while oxidation of Dhr123 remained unchanged. For comparison, traditional photosensitizer protoporphyrin IX mainly induced oxidation of SOSG in water. In conclusion, upon external radiation carbon dots or quantum dots generate reactive oxygen species acting as prooxidants. Carbon dots or quantum dots also scavenge free radicals that are generated chemically by an azo compound. Such dual properties of these nanoparticles can be used for photodynamic and photocatalytic or antioxidant applications.


Assuntos
Antioxidantes/química , Carbono/química , Pontos Quânticos , Espécies Reativas de Oxigênio/química , Amidas/química , Protoporfirinas/química , Oxigênio Singlete/química , Espectrometria de Fluorescência , Temperatura , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...