Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Anticancer Res ; 41(12): 5945-5951, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34848448

RESUMO

BACKGROUND: Melanoma is the deadliest variant of skin cancer and its incidence continues to increase. There are limited treatment options for advanced and metastatic cases of melanoma, despite advances in immunotherapy and chemotherapy. Melanoma is notorious as a radioresistant tumor. Previous studies found that phytochemicals, such as resveratrol and those found in green tea and blueberry, can sensitize various cancer cells, including melanoma, to radiotherapy. Our previous study also revealed that kiwifruit extract (KE) has antitumor activity to melanoma cells. This study was designed to expand upon our previous investigation and determine KE's potential as a radiosensitizer on CRL-11147 melanoma cancer cells and elucidate the possible mechanisms behind its potential. MATERIALS AND METHODS: Proliferation and apoptosis of CRL-11147 melanoma cells under radiation therapy (RT) plus KE versus RT alone were investigated using Proliferative cell nuclear antigen (PCNA) staining, quick cell proliferation assay, clonogenic assay, and caspase-3 activity assay. Reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC) were then used to investigate the mechanisms behind the observed results. RESULTS: The percentage of CRL-11147 colonies, PCNA staining intensity, and the optic density value of CRL-11147 cells decreased with RT/KE vs. RT alone. Relative caspase-3 activity was increased with RT/KE vs. RT alone. Increased expression of the anti-proliferative molecule p27 and pro-apoptotic molecule TRAILR1 correlated with the anti-tumor effect seen in the RT/KE group versus the RT alone group. CONCLUSION: KE augments radiosensitivity of CRL-11147 by up-regulating both p27 and TRAILR1 to inhibit proliferation and increase apoptosis, respectively.


Assuntos
Actinidia/química , Frutas/química , Extratos Vegetais/farmacologia , Radiossensibilizantes/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Melanoma/genética , Melanoma/metabolismo , Extratos Vegetais/química , Radiossensibilizantes/química
2.
Tumour Biol ; 43(1): 225-247, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34542050

RESUMO

BACKGROUND: The limitations of surgery, radiotherapy, and chemotherapy in cancer treatment and the increase in the application of nanomaterials in the field of biomedicine have promoted the use of nanomaterials in combination with radiotherapy for cancer treatment. OBJECTIVE: To improve the efficiency of cancer treatment, curcumin-naringenin loaded dextran-coated magnetic nanoparticles (CUR-NAR-D-MNPs) were used as chemotherapy and in combination with radiotherapy to verify their effectiveness in treating tumors. METHODS: CUR-NAR-D-MNPs were prepared and studied by several characterization methods. Median inhibitory concentration (IC50) and cellular toxicity were evaluated by 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) assay. The cell death and radiosensitization were studied by acridine orange/ethidium bromide dual staining of MCF-7 human breast cancer cells. RESULTS: CUR-NAR-D-MNPs induce apoptosis and inhibited cell proliferation through reactive oxygen species (ROS) generation. CUR-NAR-D-MNPs used alone had a certain therapeutic effect on tumors. CUR-NAR-D-MNPs plus radiotherapy significantly reduced the tumor volume and led to cell cycle arrest and induction of apoptosis through modulation of P53high, P21high, TNF-αlow, CD44low, and ROShigh signalingCONCLUSIONS:CUR-NAR-D-MNPs are effective in the treatment of tumors when combined with radiotherapy, and show radiosensitization effects against cancer proliferation in vitro and in vivo.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/terapia , Curcumina/química , Flavanonas/química , Nanopartículas de Magnetita/química , Radiossensibilizantes/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiorradioterapia , Curcumina/farmacologia , Curcumina/uso terapêutico , Dextranos/química , Feminino , Flavanonas/farmacologia , Flavanonas/uso terapêutico , Humanos , Células MCF-7 , Nanopartículas de Magnetita/uso terapêutico , Camundongos , Radiossensibilizantes/química , Radiossensibilizantes/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-34502158

RESUMO

Cancer stem cells (CSCs) play a critical role in radiation resistance and recurrence. Thus, drugs targeting CSCs can be combined with radiotherapy to improve its antitumor efficacy. Here, we investigated whether a gallotannin extract from Bouea macrophylla seed (MPSE) and its main bioactive compound, pentagalloyl glucose (PGG), could suppress the stemness trait and further confer the radiosensitivity of head and neck squamous cell carcinoma (HNSCC) cell lines. In this study, we evaluate the effect of MPSE or PGG to suppress CSC-like phenotypes and radiosensitization of HNSCC cell lines using a series of in vitro experiments, tumorsphere formation assay, colony formation assay, apoptosis assay, and Western blotting analysis. We demonstrate that MPSE or PGG is able to suppress tumorsphere formation and decrease protein expression of cancer stem cell markers. MPSE or PGG also enhanced the radiosensitivity in HNSCC cells. Pretreatment of cells with MPSE or PGG increased IR-induced DNA damage (γ-H2Ax) and enhanced radiation-induced cell death. Notably, we observed that pretreatment with MPSE or PGG attenuated the IR-induced stemness-like properties characterized by tumorsphere formation and the CD44 CSC marker. Our findings describe a novel strategy for increasing therapeutic efficacy for head and neck cancer patients using the natural products MPSE and PGG.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Taninos Hidrolisáveis/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos da radiação , Extratos Vegetais/farmacologia , Radiossensibilizantes/farmacologia , Sementes/química , Anacardiaceae/química , Animais , Antineoplásicos Fitogênicos/química , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço , Humanos , Taninos Hidrolisáveis/química , Camundongos , Estrutura Molecular , Células-Tronco Neoplásicas/metabolismo , Extratos Vegetais/química , Radiossensibilizantes/química , Sementes/anatomia & histologia
4.
Mol Biol Rep ; 48(2): 1081-1091, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33511511

RESUMO

Melanoma is aggressive, highly metastatic, and potentially fatal. In the case of patients with advanced melanoma, it is difficult to expect a good prognosis, since this cancer has low sensitivity to chemotherapy and radiation therapy. The use of natural ingredients may enhance existing therapies. Centipedegrass extract (CGE) which contains phenolic structures and C-glycosyl flavones, has been shown to have anti-inflammatory effects and anti-cancer effects. The purpose of this study was to evaluate the radio sensitizing effects of CGE in combination with ionizing radiation (IR). Two melanoma cell lines were exposed to IR after treatment with CGE at concentrations that were not toxic alone. The effects of CGE + IR on cell survival, cell cycle, and apoptotic cell death were examined using MTT and Muse® Cell Analyzer, and fluorescence microscopy. Molecular signaling mechanisms were explored by western blots. Our findings showed that co-treatment of CGE + IR reduced the survival of melanoma cells more than IR alone. Also, cell cycle arrest in CGE-treated cells was enhanced and these cells became more radiosensitive. CGE + IR increased apoptotic cell death more than IR alone. Western blot results showed that the effect of CGE + IR involved MAPKs (ERK1/2, p38, and JNK) pathway. Our study suggests that CGE + IR treatment enhanced radio-sensitization and cell death of melanoma cells via cell cycle arrest and the MAPKs pathway.


Assuntos
Melanoma/tratamento farmacológico , Extratos Vegetais/farmacologia , Poaceae/química , Radiossensibilizantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos da radiação , Humanos , Melanoma/patologia , Melanoma/radioterapia , Extratos Vegetais/química , Tolerância a Radiação/efeitos dos fármacos , Radiação Ionizante , Radiossensibilizantes/química
5.
Molecules ; 27(1)2021 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-35011360

RESUMO

Anticancer treatment is largely affected by the hypoxic tumor microenvironment (TME), which causes the resistance of the tumor to radiotherapy. Combining radiosensitizer compounds and O2 self-enriched moieties is an emerging strategy in hypoxic-tumor treatments. Herein, we engineered GdW10@PDA-CAT (K3Na4H2GdW10O36·2H2O, GdW10, polydopamine, PDA, catalase, CAT) composites as a radiosensitizer for the TME-manipulated enhancement of radiotherapy. In the composites, Gd (Z = 64) and W (Z = 74), as the high Z elements, make X-ray gather in tumor cells, thereby enhancing DNA damage induced by radiation. CAT can convert H2O2 to O2 and H2O to enhance the X-ray effect under hypoxic TME. CAT and PDA modification enhances the biocompatibility of the composites. Our results showed that GdW10@PDA-CAT composites increased the efficiency of radiotherapy in HT29 cells in culture. This polyoxometalates and O2 self-supplement composites provide a promising radiosensitizer for the radiotherapy field.


Assuntos
Gadolínio/química , Nanocompostos/química , Radiossensibilizantes/química , Hipóxia Tumoral/efeitos da radiação , Tungstênio/química , Ânions/química , Materiais Biocompatíveis/química , Catalase/metabolismo , Linhagem Celular Tumoral , Células HT29 , Humanos , Peróxido de Hidrogênio/metabolismo , Indóis/química , Oxigênio/metabolismo , Polieletrólitos/química , Polímeros/química , Radiossensibilizantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral
6.
ACS Appl Bio Mater ; 4(5): 4280-4291, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35006840

RESUMO

The integration of multiple therapeutic and diagnostic functions into a single nanoplatform for image-guided cancer therapy has been an emerging trend in nanomedicine. We show here that multifunctional theranostic nanostructures consisting of superparamagnetic iron oxide (SPIO) and gold nanoparticles (AuNPs) scaffolded within graphene oxide nanoflakes (GO-SPIO-Au NFs) can be used for dual photo/radiotherapy by virtue of the near-infrared (NIR) absorbance of GO for photothermal therapy (PTT) and the Z element radiosensitization of AuNPs for enhanced radiation therapy (RT). At the same time, this nanoplatform can also be detected by magnetic resonance (MR) imaging because of the presence of SPIO NPs. Using a mouse carcinoma model, GO-SPIO-Au NF-mediated combined PTT/RT exhibited a 1.85-fold and 1.44-fold higher therapeutic efficacy compared to either NF-mediated PTT or RT alone, respectively, resulting in a complete eradication of tumors. As a sensitive multifunctional theranostic platform, GO-SPIO-Au NFs appear to be a promising nanomaterial for enhanced cancer imaging and therapy.


Assuntos
Antineoplásicos/farmacologia , Materiais Biocompatíveis/farmacologia , Carcinoma/tratamento farmacológico , Imageamento por Ressonância Magnética , Fototerapia , Radiossensibilizantes/farmacologia , Nanomedicina Teranóstica , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/química , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Compostos Férricos/química , Compostos Férricos/farmacologia , Ouro/química , Ouro/farmacologia , Grafite/química , Grafite/farmacologia , Masculino , Teste de Materiais , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Tamanho da Partícula , Radiossensibilizantes/síntese química , Radiossensibilizantes/química , Espécies Reativas de Oxigênio/metabolismo
7.
ACS Appl Mater Interfaces ; 12(52): 57768-57781, 2020 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-33326213

RESUMO

Solid tumors inevitably develop radioresistance due to low oxygen partial pressure in the tumor microenvironment. Despite numerous attempts, there are still few effective ways to avoid the hypoxia-induced poor radiotherapeutic effect. To overcome this problem, platinum (Pt) nanodots were fabricated into a mesoporous bismuth (Bi)-based nanomaterial to construct a biodegradable nanocomposite BiPt-folic acid-modified amphiphilic polyethylene glycol (PFA). BiPt-PFA could act as a radiosensitizer to enhance the absorption of X-rays at the tumor site and simultaneously trigger response behaviors related to the tumor microenvironment due to the enrichment of materials in the tumor area. During this process, the Bi-based component consumed glutathione via coordination, thus altering the oxidative stress balance, while Pt nanoparticles catalyzed the decomposition of hydrogen peroxide to generate oxygen, thereby relieving tumor hypoxia. Both Pt and Bi thus co-modulated the tumor microenvironment to improve the radiotherapeutic effect. In addition, Pt dots in BiPt-PFA had strong near-infrared absorption ability and created an intensive photothermal therapeutic effect. Modulation of the tumor microenvironment could thus improve the therapeutic effect in hypoxic tumors by a combination of photothermal therapy and enhanced radiotherapy. BiPt-PFA, as a biodegradable nanocomposite, may thus modulate the tumor microenvironment to enhance the hypoxic tumor therapeutic effect by thermoradiotherapy.


Assuntos
Bismuto/química , Nanocompostos/química , Radiossensibilizantes/química , Hipóxia Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Humanos , Hipertermia Induzida , Nanopartículas Metálicas/química , Camundongos , Platina/química , Polietilenoglicóis/química , Porosidade , Radiossensibilizantes/farmacologia , Segurança , Solubilidade , Hipóxia Tumoral/efeitos da radiação , Microambiente Tumoral/efeitos da radiação , Água/química
8.
Exp Oncol ; 42(3): 204-207, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32996742

RESUMO

AIM: In order to develop fundamentally new technologies for non-invasive and safer diagnosis of cancer, we aimed to detect non-contact magnetic signals from a malignant tumor in animals treated or not-treated with the ferromagnetic nanocomposite Ferroplat. MATERIALS AND METHODS: Guerin's carcinoma was used as a model of tumor growth. The biomagnetism of the tumor was evaluated in the dynamics of its growth. Ten days after tumor transplantation, Ferroplat was administered intravenously to half of the animals with the tumor and to half of the control animals. The magnitude of the magnetic signals was determined 1 h and every two days after administration of the nanocomposite using a Superconducting Quantum Interference Device magnetometer of the original design. RESULTS: We have found that the magnetic signals coming from the tumor are significantly higher compared to control tumor-free animals. Intravenous administration of a ferromagnetic nanocomposite (Ferroplat: Fe3O4 + cisplatinum) led to a significant increase of the magnetic signal, especially in the tumor tissue, and inhibition of Guerin's carcinoma growth. Ferromagnetic nanoparticles (32.7 nm) are retained in malignant cells for a longer time than in normal ones. CONCLUSION: Tumor cells accumulate iron nanoparticles more intensively than normal ones. Nanocomposite Ferroplat can be used for a targeted delivery of cisplatin to malignant cells.


Assuntos
Fenômenos Biofísicos , Carcinoma/diagnóstico , Imãs , Nanocompostos , Animais , Carcinoma/tratamento farmacológico , Cisplatino/química , Feminino , Magnetometria/instrumentação , Magnetometria/métodos , Magnetometria/normas , Neoplasias Experimentais , Radiossensibilizantes/química , Ratos , Razão Sinal-Ruído
9.
Drug Des Devel Ther ; 14: 1779-1798, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32440103

RESUMO

BACKGROUND: Radiotherapy has an ameliorative effect on a wide variety of tumors, but hepatocellular carcinoma (HCC) is insensitive to this treatment. Overactivated mammalian target of rapamycin (mTOR) plays an important part in the resistance of HCC to radiotherapy; thus, mTOR inhibitors have potential as novel radiosensitizers to enhance the efficacy of radiotherapy for HCC. METHODS: A lead compound was found based on pharmacophore modeling and molecular docking, and optimized according to the differences between the ATP-binding pockets of mTOR and PI3K. The radiosensitizing effect of the optimized compound (2a) was confirmed by colony formation assays and DNA double-strand break assays in vitro. The discovery and preclinical characteristics of this compound are described. RESULTS: The key amino acid residues in mTOR were identified, and a precise virtual screening model was constructed. Compound 2a, with a 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidine scaffold, exhibited promising potency against mTOR (mTOR IC50=7.1 nmol/L (nM)) with 126-fold selectivity over PI3Kα. Moreover, 2a significantly enhanced the sensitivity of HCC to radiotherapy in vitro in a dose-dependent manner. CONCLUSION: A new class of selective mTOR inhibitors was developed and their radiosensitization effects were confirmed. This study also provides a basis for developing mTOR-specific inhibitors for use as radiosensitizers for HCC radiotherapy.


Assuntos
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Pirimidinonas/farmacologia , Radiossensibilizantes/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Modelos Moleculares , Estrutura Molecular , Pirimidinonas/síntese química , Pirimidinonas/química , Radiossensibilizantes/síntese química , Radiossensibilizantes/química , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR/metabolismo
10.
ACS Appl Mater Interfaces ; 11(44): 41127-41139, 2019 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-31610123

RESUMO

Theranostic nanoplatforms that integrate therapy and diagnosis in a single composite have become increasingly attractive in the field of precise and efficient tumor treatment. Herein, a novel oxygen-deficient zirconia (ZrO2-x) nanosystem based on the conjugation of thiol-polyethylene glycol-amine (SH-PEG-NH2) and chlorin e6 (Ce6) was elaborately designed and established for efficacious photothermal/photodynamic therapy (PTT/PDT) and fluorescence/photoacoustic (FL/PA) bimodal imaging for the first time. The crystalline-disordered, PEGylated ZrO2-x nanoparticles (ZP NPs) displayed strong optical absorption in the near-infrared (NIR) window and were featured with significant photothermal conversion capacity. The ZP NPs were further covalently conjugated with Ce6 to form ZrO2-x@PEG/Ce6 (ZPC) NPs, which displayed a long circulatory half-life, efficient tumor accumulation, and outstanding FL/PA imaging performance. Moreover, the nanocomposites effectively generated cytotoxic intracellular reactive oxygen species (ROS) responsive to laser activation. Both cell studies and animal experiments explicitly demonstrated that ZPC NPs mediated remarkable tumor ablation with minimal systemic toxicity thanks to their tumor-specific PTT/PDT effect. Collectively, these findings may open up new avenues to broaden the application of oxygen-deficient ZrO2-x nanostructures as high-performance photothermal agents in tumor theranostics through rational design and accurate control of their physiochemical properties.


Assuntos
Raios Infravermelhos , Nanopartículas/química , Neoplasias/terapia , Zircônio/química , Aminas/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Clorofilídeos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/metabolismo , Nanopartículas/uso terapêutico , Nanopartículas/toxicidade , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Oxigênio/química , Fotoquimioterapia , Fototerapia , Polietilenoglicóis/química , Porfirinas/química , Porfirinas/farmacologia , Porfirinas/uso terapêutico , Radiossensibilizantes/química , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Compostos de Sulfidrila/química , Distribuição Tecidual
11.
Artif Cells Nanomed Biotechnol ; 47(1): 3832-3838, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31556316

RESUMO

High atomic number Z, nanoparticles are able to enhance the photoelectric and Compton effects under X-Ray irradiation resulting the increase of radiation therapy efficacy. To achieve enhanced radiation therapy, Bi2S3 biocompatible particles coated with bovine serum albumin (BSA) (Bi2S3@BSA HNPs) were prepared through a BSA-mediated biomineralization procedure under green conditions. Then, to achieve improved chemo-radiation therapy against HT-29 cancer cells, curcumin (CUR) as natural anti-cancer therapy agent loaded on the Bi2S3@BSA (Bi2S3@BSA@CUR HNPs). Next, this synthesized nanodrug was evaluated for physical and chemical properties and in vitro cytotoxicity studies. Here, in vitro enhanced chemo-radiation combination therapy power was evaluated against HT-29 cell line under 2 Gy and 6 Gy X-ray irradiation doses. The Bi2S3@BSA HNPs without irradiation rarely affect cell viability which shown the non-toxicity of Bi2S3@BSA HNPs. The result of this study proved that Bi2S3@BSA@CUR HNPs can be used as both proficient vehicles for effective delivery of CUR and radiosensitizer in the treatment of cancer. In addition, the result of this study confirmed that the combination of high Z-element nanoradiosensitizer, Bi2S3@BSA HNPs, with a natural anti-cancer drug, CUR, enhanced therapeutic power against HT-29 cells.


Assuntos
Bismuto/farmacologia , Quimiorradioterapia , Minerais/química , Soroalbumina Bovina/química , Sulfetos/síntese química , Sulfetos/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Bismuto/química , Bovinos , Técnicas de Química Sintética , Materiais Revestidos Biocompatíveis/síntese química , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Curcumina/química , Curcumina/farmacologia , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Liberação Controlada de Fármacos , Química Verde , Células HT29 , Humanos , Nanopartículas/química , Tamanho da Partícula , Radiossensibilizantes/síntese química , Radiossensibilizantes/química , Radiossensibilizantes/farmacologia , Sulfetos/química
12.
ACS Appl Mater Interfaces ; 11(37): 33628-33636, 2019 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-31433160

RESUMO

The multifunctional effect of a single molecule for therapeutic functionalities on a single theranostic nanosystem has a great significance to enhance the accuracy of diagnosis and improve the efficacy of therapy. Herein, a biocompatible multistep phototherapeutic system (Ppa-Cy7-PEG-biotin) that contains a photosensitizer pyropheophorbide A (Ppa) with the covalent conjunction of a near-infrared (NIR) cyanine dye (Cy7) was successfully fabricated and functionalized with biotin for flexible specific tumor-targeting phototherapy. These theranostic micelles will disaggregate after NIR irradiation via the photodegradation of cyanine accompanied by the photothermal conversion and the optically controlled release for the restoration of photodynamic function of quenched Ppa. Consecutively, promoted treatments of photosensitive molecules greatly prolonged the tumor retention time and treatment efficiency, having a multistep antitumor effect both in vitro and in vivo. Different from the simple phototherapeutic configurations that only act on the superficial areas of tumors at mild doses, the multistep therapy can be competent for broadly damaging the superficial and deeper regions of tumors at the same dose. Therefore, as opposed to the general combination phototherapeutic approach, this strategy presents a photoactivation-based multistep phototheranostic platform with an enormous potential in enhanced combined phototherapy for cancer.


Assuntos
Carbocianinas , Micelas , Nanopartículas , Neoplasias Experimentais/terapia , Fototerapia , Radiossensibilizantes , Células A549 , Animais , Carbocianinas/química , Carbocianinas/farmacologia , Células Hep G2 , Humanos , Camundongos , Camundongos Nus , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Radiossensibilizantes/química , Radiossensibilizantes/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Mol Pharm ; 16(9): 3831-3841, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31381351

RESUMO

Boron neutron capture therapy (BNCT) is a therapeutic modality which has been used for the treatment of cancers, including brain and head and neck tumors. For effective treatment via BNCT, efficient and selective delivery of a high boron dose to cancer cells is needed. Prostate-specific membrane antigen (PSMA) is a target for prostate cancer imaging and drug delivery. In this study, we conjugated boronic acid or carborane functional groups to a well-established PSMA inhibitor scaffold to deliver boron to prostate cancer cells and prostate tumor xenograft models. Eight boron-containing PSMA inhibitors were synthesized. All of these compounds showed a strong binding affinity to PSMA in a competition radioligand binding assay (IC50 from 555.7 to 20.3 nM). Three selected compounds 1a, 1d, and 1f were administered to mice, and their in vivo blocking of 68Ga-PSMA-11 uptake was demonstrated through a positron emission tomography (PET) imaging and biodistribution experiment. Biodistribution analysis demonstrated boron uptake of 4-7 µg/g in 22Rv1 prostate xenograft tumors and similar tumor/muscle ratios compared to the ratio for the most commonly used BNCT compound, 4-borono-l-phenylalanine (BPA). Taken together, these data suggest a potential role for PSMA targeted BNCT agents in prostate cancer therapy following suitable optimization.


Assuntos
Antígenos de Superfície/metabolismo , Terapia por Captura de Nêutron de Boro/métodos , Ácidos Borônicos/química , Ácidos Borônicos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Glutamato Carboxipeptidase II/antagonistas & inibidores , Glutamato Carboxipeptidase II/metabolismo , Neoplasias da Próstata/radioterapia , Animais , Compostos de Boro/química , Compostos de Boro/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ácido Edético/análogos & derivados , Ácido Edético/farmacocinética , Isótopos de Gálio , Radioisótopos de Gálio , Humanos , Concentração Inibidora 50 , Ligantes , Masculino , Camundongos , Camundongos Nus , Oligopeptídeos/farmacocinética , Fenilalanina/análogos & derivados , Fenilalanina/química , Fenilalanina/farmacocinética , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias da Próstata/patologia , Radiossensibilizantes/química , Radiossensibilizantes/farmacocinética , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Biomater Sci ; 7(8): 3450-3459, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31268067

RESUMO

Bismuth-containing nanoparticles (BNPs) are potential enhancers for tumor radiotherapy. Improving the bioavailability and developing synergistic therapeutic regimens benefit the drug transformation of BNPs. In the present study, we prepare a mesoporous silica-coated bismuth nanorod (BMSNR) camouflaged by a platelet membrane (PM). This biomimetic material is termed BMSNR@PM. The PM camouflage enhances the immune escape of the BMSNRs by lowering endocytosis by macrophages in the reticuloendothelial system. Additionally, the PM camouflage strengthens the material tumor-targeting capacity and leads to better radiotherapeutic efficacy compared with bare BMSNRs. Owing to the photothermal effect, BMSNR@PMs alters the cell cycle of 4T1 cancer cells post-treatment with 808 nm near-infrared irradiation (NIR). The proportions of S phase and G2/M phase cells decrease and increase, respectively, which explains the synergistic effect of NIR on BMSNR@PM-based radiotherapy. BMSNR@PMs efficiently eradicates cancer cells by the combined action of photothermal therapy (PTT) and radiotherapy in vivo and markedly improves the survival of 4T1-tumor-bearing mice. The synergistic therapeutic effect is superior to the outcomes of PTT and radiotherapy performed alone. Our study demonstrates a versatile bismuth-containing nanoplatform with tumor-targeting, immune escape, and radiosensitizing functionalities using an autologous cell membrane biomimetic concept that may promote the development of radiotherapy enhancers.


Assuntos
Bismuto/química , Bismuto/farmacologia , Plaquetas/citologia , Neoplasias da Mama/terapia , Membrana Celular/metabolismo , Nanotubos/química , Fototerapia , Sulfetos/química , Sulfetos/farmacologia , Animais , Bismuto/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/radioterapia , Linhagem Celular Tumoral , Terapia Combinada , Endocitose , Feminino , Humanos , Macrófagos/metabolismo , Camundongos , Nanocompostos/química , Porosidade , Células RAW 264.7 , Radiossensibilizantes/química , Radiossensibilizantes/metabolismo , Radiossensibilizantes/farmacologia , Dióxido de Silício/química , Sulfetos/metabolismo
15.
Molecules ; 24(12)2019 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-31216782

RESUMO

Pavetta indica L. is used in traditional medicine for the treatment of various diseases including hemorrhoids, headache, urinary conditions, ulcerated nose, and dropsy. However, no study has evaluated the anticancer effect of P. indica L. In this study, we found that a methanol extract of the leaves and branches of P. indica L. (MEPI) caused cellcycle arrest at the sub-G1 phase and induced apoptosis, as indicated by the activation of caspase-8, -3, -7, and c-PARP. Western blotting revealed that MEPI significantly reduced the levels of markers of the epithelial-mesenchymal transition, such as Vimentin, Snail, Slug, and matrix metallopeptidase 9. Notably, the expression of multidrug resistance-associated protein 1 in triple negative breast cancer (TNBC) was significantly decreased by MEPI. Moreover, the co-treatment with MEPI and doxorubicin resulted in a synergistic reduction in cell viability. MEPI also induced radiation sensitization of TNBC cells. Gas chromatography-mass spectrometry analysis revealed that 5,6-dehydrokawain (DK) is the major constituent of MEPI. Interestingly, DK exerted significant anti-invasive and anti-metastatic effects. Our results provide a strong rationale for investigating the molecular mechanisms of action of MEPI in TNBC.


Assuntos
Componentes Aéreos da Planta/química , Extratos Vegetais/farmacologia , Radiossensibilizantes/farmacologia , Rubiaceae/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Metanol , Extratos Vegetais/química , Radiossensibilizantes/química , Solventes , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
16.
Nanoscale ; 11(15): 7157-7165, 2019 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-30919835

RESUMO

The development of a new multifunctional nanomedicine capable of enhancing radiosensitization by photo-induced hyperthermia for the inhibition of cancer growth and metastasis is highly required for efficient treatment of cancer cells. Compared to the first near-infrared (NIR) window, the second NIR window light could provide a maximum penetration depth as well as minimizing autofluorescence due to its low scattering and energy absorption. Here, we report a new versatile theranostic agent based on ternary Cu3BiSe3 nanoparticles (NPs) modified by poly(vinylpyrollidone) (PVP-Cu3BiSe3). Benefiting from their preferable X-ray attenuation ability and strong NIR absorbance in the second NIR biological window, PVP-Cu3BiSe3 NPs can not only deposit more radiation doses to destroy the cancer cells, but also conduct the optical energy into hyperthermia for thermal eradication of tumor tissues and the improvement of the tumor oxygenation to overcome the hypoxia-associated radio-resistance of tumors. According to both in vitro and in vivo results, exposure to an X-ray plus 1064 nm laser completely kills cancer cells and even inhibits tumor metastasis, displaying no warning signs of a relapse. On the other hand, PVP-Cu3BiSe3 NPs can be used as a multi-model imaging agent for X-ray computer tomography (CT) and photoacoustic tomography (PAT) imaging. These demonstrate the potential of PVP-Cu3BiSe3 NPs in multimodal imaging-guided synergetic radiophotothermal therapy of deep-seated tumors and effective inhibition of their metastasis.


Assuntos
Hipertermia Induzida , Raios Infravermelhos , Nanopartículas Metálicas , Neoplasias Experimentais , Técnicas Fotoacústicas , Fototerapia , Radiossensibilizantes , Tomografia Computadorizada por Raios X , Animais , Células HeLa , Humanos , Masculino , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Radiossensibilizantes/química , Radiossensibilizantes/farmacologia
17.
ACS Nano ; 13(2): 1342-1353, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30707555

RESUMO

The small difference between tumor and normal tissues in their responses to ionizing radiation has been a significant issue for radiotherapy of tumors. Herein, we report that dumbbell-shaped heterogeneous copper selenide-gold nanocrystals can serve as an efficient radiosensitizer for enhanced radiotherapy. The mean lethal dose of X-rays to 4T1 tumor cells can be drastically decreased about 40%, that is, decreasing from 1.81 to 1.10 Gy after culture with heterostructures. Due to the synergetic effect of heterostructures, the dose of X-rays is also much lower than those obtained from mixture of Cu2- xSe + Au nanoparticles (1.78 Gy), Cu2- xSe nanoparticles (1.72 Gy) and Au nanoparticles (1.50 Gy), respectively. We demonstrate that the sensitivity enhancement ratio of Cu2- xSe nanoparticles was significantly improved 45% ( i. e., from 1.1 to 1.6) after the formation of heterostructures with gold. We also show that the heteronanocrystals exhibit an enhanced photothermal conversion efficiency, due to the synergetic interactions of localized surface plasmon resonance. These properties highly feature them as a multimodal imaging contrast agent (particularly for photoacoustic imaging, computed tomography imaging, and single photon emission computed tomography after labeled with radioisotopes) and as a radiosensitizer for imaging guided synergetic radiophotothermal treatment of cancer. The research provides insights for engineering low- Z nanomaterials with high- Z elements to form heteronanostructures with enhanced synergetic performance for tumor theranostics.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/terapia , Cobre/química , Nanopartículas Metálicas/química , Nanopartículas/química , Nanoestruturas/química , Radiossensibilizantes/química , Selênio/química , Animais , Linhagem Celular Tumoral , Feminino , Ouro/química , Humanos , Camundongos , Temperatura
18.
J Photochem Photobiol B ; 191: 123-127, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30616036

RESUMO

Selinium nanoparticles (SeNPs) with minimal toxicity and efficient antioxidant properties were reported earlier for their anti-carcinogenic influence against various types of cancers, thus elevating its potential. In the present study, the anti-carcinogenic effect of selenium nanoparticles against lung cancer was studied. Selenium nanoparticles were biosynthesized and were characterized using UV- Vis absorption spectroscopy. A decrease in the absorption intensity was recorded with the increase in time, which represented the protein consumption during the reduction of SeO32- to Se0. The calculated average crystalline size from XRD studies of the synthesized selenium nanoparticles was found to be 88.89 nm which was in accordance with the TEM analysis while the SAED pattern has disclosed hexagonal ring structure with diffraction ring pattern.MTT assay was performed to evaluate the radio-sensitizing effect of selenium nanoparticles under the X-ray influence against cancer as well as healthy cell lines. SeNPs showed potent cytotoxicity effect in cancer cells whereas it showed relatively less toxic effect in normal healthy cells. However, caspase-3 activity was even more elevated when subjected to X-ray exposure than in the absence. These findings apparently revealed the cytotoxic potential of SeNPs + X-ray combination in the lung cancer cell lines.


Assuntos
Doenças Pulmonares Intersticiais/terapia , Neoplasias Pulmonares/terapia , Nanopartículas/química , Selênio/uso terapêutico , Antioxidantes/farmacologia , Caspase 3/metabolismo , Linhagem Celular , Humanos , Doenças Pulmonares Intersticiais/tratamento farmacológico , Doenças Pulmonares Intersticiais/radioterapia , Neoplasias Pulmonares/radioterapia , Nanopartículas/uso terapêutico , Radiossensibilizantes/química , Radiossensibilizantes/uso terapêutico , Raios X
19.
ACS Nano ; 12(12): 12401-12415, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30407787

RESUMO

Cancer radiotherapy suffers from drawbacks such as radiation resistance of hypoxic cells, excessive radiation that causes damage of adjacent healthy tissues, and concomitant side effects. Hence, radiotherapy sensitizers with improved radiotherapeutic performance and requiring a relatively small radiation dose are highly desirable. In this study, a nanosystem based on poly(lactic- co-glycolic acid) (PLGA) and ultrasmall black phosphorus quantum dots (BPQDs) is designed and prepared to accomplish precise tumor radiosensitization. The PLGA nanoparticles act as carriers to package the BPQDs to avoid off-target release and rapid degradation during blood circulation. The nanosystem that targets the polypeptide peptide motif Arg-Gly-Asp-Gys actively accumulates in tumor tissues. The 2,3-dimethylmaleic anhydride shell decomposes in an acidic microenvironment, and the nanoparticles become positively charged, thereby favoring cellular uptake. Furthermore, glutathione (GSH) deoxidizes the disulfide bond of cystamine and sequentially triggers release of BPQDs, rendering tumor cells sensitive to radiotherapy. The treatment utilizing the PLGA-SS-D@BPQDs nanosystem and X-ray induces cell apoptosis triggered by overproduction of reactive oxygen species. In the in vivo study, the nanosystem shows excellent radiotherapy sensitization efficiency but negligible histological damage of the major organs. This study provides insights into the design and fabrication of surface-charge-switching and pH-responsive nanosystems as potent radiosensitizers to achieve excellent radiotherapy sensitization efficacy and negligible toxic side effects.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Melanoma/terapia , Fósforo/química , Pontos Quânticos/química , Radiossensibilizantes/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Melanoma/metabolismo , Melanoma/patologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Nus , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Radiossensibilizantes/síntese química , Radiossensibilizantes/química , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo , Propriedades de Superfície , Células Tumorais Cultivadas
20.
Oncol Rep ; 40(5): 2886-2895, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30226618

RESUMO

The use of plant­based compounds derived from traditional medicine to improve human diseases has been gaining momentum, due to their high bioavailability and moderate adverse effects. Sinomenine is one such biomonomer alkali compound derived from Sinomenium acutum and is known for its anti­inflammatory and antitumor effects. However, the molecular mechanism(s) of its antitumor properties are not fully characterized. In the present study, we evaluated the radiosensitizing effects of the water­soluble sinomenine, sinomenine hydrochloride (SH) in human cervical cancer cell line (HeLa). SH sensitized HeLa cells to ionizing radiation (IR) by promoting accumulation of IR­induced DNA double­strand breaks (DSBs) and also by interfering with DNA damage checkpoint activation. We then investigated the molecular mechanisms underlying the SH­mediated cellular sensitization to IR and found that SH inhibited the expression of DNA damage response (DDR) factors Ku80 and Rad51 at the transcription level. Finally, the radiosensitizing activity of SH was confirmed in a cervical cancer mouse xenograft model. The combinatorial treatment of SH and IR significantly slowed the tumor growth rate compared with IR alone. Collectively, our study not only provides molecular insights into the novel role of SH in cellular response to IR, but also suggests a therapeutic potential of SH as a radiosensitizer in cervical cancer therapy.


Assuntos
Morfinanos/farmacologia , Radiossensibilizantes/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/radioterapia , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Feminino , Células HeLa , Humanos , Camundongos , Morfinanos/química , Tolerância a Radiação/genética , Radiação Ionizante , Radiossensibilizantes/química , Sinomenium/química , Neoplasias do Colo do Útero/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA