Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
In Vivo ; 34(4): 2061-2066, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32606183

RESUMO

BACKGROUND: Peritoneal carcinomatosis (PC) is a common manifestation of many gastrointestinal (GI) malignancies and is an advanced stage that is often associated with disseminated disease. Considerable progress has been made to achieve safe elimination of macroscopic disease using cytoreductive surgery (CRS) and more recently in combination with hyperthermic intraperitoneal chemotherapy (HIPEC) for the treatment of microscopic disease or disease with minimal volume. The aim of this study was to assess the effects of such procedures on the quality of life (QoL), the long-term benefit and the functional status of the treated patients. PATIENTS AND METHODS: Data from patients who underwent CRS-HIPEC for peritoneal metastasis (PM) at our center from November 2016 to November 2018 were analyzed retrospectively. The drugs administered were mitomycin and cisplatin. Quality of life (QoL) was assessed using the Euroquol-5D-5L and National Comprehensive Cancer Network Functional Assessment of Cancer Therapy-Breast Cancer Symptom Index v2 questionnaires before CRS-HIPEC, and 1, 3 and 6 months after were administered. RESULTS: In our series, the survival efficacy of CRS plus HIPEC was confirmed in the treatment of primary and secondary peritoneal pathologies, particularly in ovarian cancer, although larger studies are needed to investigate its role in the pathology of gastric, colonic and rectal cancer. The QoL data were promising, with essentially stable values between the preoperative and the 1-month follow-up, but with incremental benefits from the second to the third month.


Assuntos
Hipertermia Induzida , Neoplasias Peritoneais , Protocolos de Quimioterapia Combinada Antineoplásica , Terapia Combinada , Procedimentos Cirúrgicos de Citorredução , Feminino , Humanos , Itália , Neoplasias Peritoneais/terapia , Qualidade de Vida , Estudos Retrospectivos , Taxa de Sobrevida
2.
Biomed Microdevices ; 21(2): 39, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30949852

RESUMO

Effective migration of dendritic cells into the lymphatic system organs is the prerequisite for a functional dendritic cell vaccine. We have previously developed a porous silicon microparticle (PSM)-based therapeutic dendritic cell vaccine (Nano-DC vaccine) where PSM serves both as the vehicle for antigen peptides and an adjuvant. Here, we analyzed parameters that determined dendritic cell uptake of PSM particles and Nano-DC vaccine accumulation in lymphatic tissues in a murine model of HER2-positive breast cancer. Our study revealed a positive correlation between sphericity of the PSM particles and their cellular uptake by circulating dendritic cells. In addition, the intravenously administered vaccines accumulated more in the spleens and inguinal lymph nodes, while the intradermally inoculated vaccines got enriched in the popliteal lymph nodes. Furthermore, mice with large tumors received more vaccines in the lymph nodes than those with small to medium size tumors. Information from this study will provide guidance on design and optimization of future therapeutic cancer vaccines.


Assuntos
Vacinas Anticâncer/química , Vacinas Anticâncer/metabolismo , Células Dendríticas/metabolismo , Nanomedicina , Animais , Transporte Biológico , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacocinética , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Camundongos , Microesferas , Fagócitos/imunologia , Silício/química , Distribuição Tecidual , Carga Tumoral/imunologia
3.
Clin Cancer Res ; 24(19): 4900-4912, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29921732

RESUMO

Purpose: Triple-negative breast cancer (TNBC) is a clinically aggressive disease with poor prognosis. Conventional chemotherapeutics are generally able to shrink the tumor mass, but often fail to completely eradicate cancer stem-like cells (CSCs) that are responsible for high risk of relapse and frequent metastases. In this study, we examined thermal sensibility of CSCs, developed an approach that enabled concurrent elimination of both the bulk of cancer cells and CSCs, and investigated the underlying mechanism.Experimental Design: We designed a platform consisting of gold nanoparticle-coated porous silicon microparticle (AuPSM) that was also loaded with docetaxel micelles (mDTXs) to enable concurrent killing of the bulk of cancer cells by released mDTX and CSCs by mild hyperthermia upon stimulation of AuPSM with near infrared. In addition, we examined the role of heat shock proteins in sensitizing CSC killing. Finally, we applied mDTX-loaded AuPSM to treat mice with SUM159 and 4T1 orthotopic tumors and evaluated tumor growth and tumor metastasis.Results: MDA-MB-231 and SUM159 TNBC cells treated with mDTX-loaded AuPSM and mild hyperthermia displayed significantly reduced efficiencies in mammosphere formation than those treated with mDTX alone or mild hyperthermia alone. Combination treatment also completely inhibited SUM159 orthotopic tumor growth and 4T1 tumor metastasis. Mechanistically, DTX treatment suppressed expression of heat shock protein 27 in cancer cells including the CSCs, rendering cells sensitive to mild hyperthermia.Conclusions: Our results indicate that chemotherapy sensitizes CSC to mild hyperthermia. We have developed an effective therapeutic approach to eliminate therapy-resistant cells in TNBC. Clin Cancer Res; 24(19); 4900-12. ©2018 AACR.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteínas de Choque Térmico HSP27/genética , Recidiva Local de Neoplasia/terapia , Neoplasias de Mama Triplo Negativas/terapia , Animais , Terapia Combinada , Docetaxel/química , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Ouro/química , Proteínas de Choque Térmico HSP27/antagonistas & inibidores , Humanos , Hipertermia Induzida/métodos , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/química , Camundongos , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Curr Drug Deliv ; 13(5): 711-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26502889

RESUMO

Hesperetin is a compound from citrus fruit that has previously been found to exert anticancer activity through a variety of mechanisms. However, the application of hesperetin to cancer therapy has been hampered by its hydrophobicity, necessitating the use of toxic solubilizing agents. Here, we have developed the first liposome-based delivery system for hesperetin. Liposomes were fabricated using the thin-layer evaporation technique and physical and pharmacological parameters were measured. The liposomes remained stable for prolonged periods of time in serum and under storage conditions, and displayed anticancer efficacy in both H441 lung cancer cells and MDA-MB-231 breast cancer cells. Furthermore, the anticancer activity was not impaired in cells expressing the multidrug resistance protein 1 (MDR-1). In conclusion, the encapsulation of hesperetin in liposomes does not interfere with therapeutic efficacy and provides a biocompatible alternative to toxic solubilizing agents, thereby enabling future clinical use of this compound for cancer therapy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Colesterol/química , Hesperidina/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Fosfatidilcolinas/química , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/química , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Composição de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Hesperidina/administração & dosagem , Hesperidina/química , Humanos , Cinética , Lipossomos , Neoplasias Pulmonares/patologia , Polietilenoglicóis/química , Solubilidade , Transfecção
5.
Nanoscale ; 7(46): 19438-47, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26376843

RESUMO

Tumor Initiating Cells (TICs) are resistant to radiotherapy and chemotherapy, and are believed to be responsible for tumor recurrence and metastasis. Combination therapies can overcome the limitation of conventional cancer treatments, and have demonstrated promising application in the clinic. Here, we show that dual modality radiotherapy (RT) and photothermal therapy (PTT) mediated by a single compartment nanosystem copper-64-labeled copper sulfide nanoparticles ([(64)Cu]CuS NPs) could suppress breast tumor metastasis through eradication of TICs. Positron electron tomography (PET) imaging and biodistribution studies showed that more than 90% of [(64)Cu]CuS NPs was retained in subcutaneously grown BT474 breast tumor 24 h after intratumoral (i.t.) injection, indicating the NPs are suitable for the combination therapy. Combined RT/PTT therapy resulted in significant tumor growth delay in the subcutaneous BT474 breast cancer model. Moreover, RT/PTT treatment significantly prolonged the survival of mice bearing orthotopic 4T1 breast tumors compared to no treatment, RT alone, or PTT alone. The RT/PTT combination therapy significantly reduced the number of tumor nodules in the lung and the formation of tumor mammospheres from treated 4T1 tumors. No obvious side effects of the CuS NPs were noted in the treated mice in a pilot toxicity study. Taken together, our data support the feasibility of a therapeutic approach for the suppression of tumor metastasis through localized RT/PTT therapy.


Assuntos
Neoplasias da Mama/terapia , Radioisótopos de Cobre , Neoplasias Pulmonares/terapia , Nanopartículas/química , Sulfetos , Animais , Neoplasias da Mama/patologia , Radioisótopos de Cobre/química , Radioisótopos de Cobre/farmacologia , Feminino , Humanos , Hipertermia Induzida/métodos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/transplante , Fototerapia/métodos , Radioterapia/métodos , Sulfetos/química , Sulfetos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cell Rep ; 11(6): 957-966, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25937283

RESUMO

Micro- and nanometer-size particles have become popular candidates for cancer vaccine adjuvants. However, the mechanism by which such particles enhance immune responses remains unclear. Here, we report a porous silicon microparticle (PSM)-based cancer vaccine that greatly enhances cross-presentation and activates type I interferon (IFN-I) response in dendritic cells (DCs). PSM-loaded antigen exhibited prolonged early endosome localization and enhanced cross-presentation through both proteasome- and lysosome-dependent pathways. Phagocytosis of PSM by DCs induced IFN-I responses through a TRIF- and MAVS-dependent pathway. DCs primed with PSM-loaded HER2 antigen produced robust CD8 T cell-dependent anti-tumor immunity in mice bearing HER2+ mammary gland tumors. Importantly, this vaccination activated the tumor immune microenvironment with elevated levels of intra-tumor IFN-I and MHCII expression, abundant CD11c+ DC infiltration, and tumor-specific cytotoxic T cell responses. These findings highlight the potential of PSM as an immune adjuvant to potentiate DC-based cancer immunotherapy.


Assuntos
Apresentação Cruzada/imunologia , Imunidade Inata , Interferon Tipo I/imunologia , Microesferas , Neoplasias/imunologia , Silício/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Feminino , Neoplasias Mamárias Animais/patologia , Camundongos Endogâmicos C57BL , Neoplasias/patologia , Ovalbumina/imunologia , Fagocitose , Porosidade , Receptor ErbB-2/metabolismo , Transdução de Sinais , Linfócitos T Citotóxicos/imunologia , Microambiente Tumoral
7.
Adv Healthc Mater ; 4(7): 1092-103, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25721343

RESUMO

Obstructive biological barriers limit the transport and efficacy of cancer nanotherapeutics. Creative manipulation of tumor microenvironment provides promising avenues towards improving chemotherapeutic response. Such strategies include the use of mechanical stimuli to overcome barriers, and increase drug delivery and therapeutic efficacy. The rational use of gold nanorod-mediated mild hyperthermia treatment (MHT) alters tumor transport properties, increases liposomal gemcitabine (Gem Lip) delivery, and antitumor efficacy in pancreatic cancer CAPAN-1 tumor model. MHT treatment leads to a threefold increase in accumulation of 80-nm liposomes and enhances spatial interstitial distribution. I.v. injection of Gem Lip and MHT treatment lead to a threefold increase in intratumor gemcitabine concentration compared to chemotherapeutic infusion alone. Furthermore, combination of MHT treatment with infusion of 12 mg kg(-1) Gem Lip leads to a twofold increase in therapeutic efficacy and inhibition of CAPAN-1 tumor growth when compared to equimolar chemotherapeutic treatment alone. Enhanced therapeutic effect is confirmed by reduction in tumor size and increase in apoptotic index where MHT treatment combined with 12 mg kg(-1) Gem Lip achieves similar therapeutic efficacy as the use of 60 mg kg(-1) free gemcitabine. In conclusion, improvements in vivo efficacy are demonstrated resulting from MHT treatment that overcome transport barriers, promote delivery, improve efficacy of nanomedicines.


Assuntos
Antineoplásicos/administração & dosagem , Desoxicitidina/análogos & derivados , Febre/fisiopatologia , Lipossomos/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/terapia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Terapia Combinada/métodos , Desoxicitidina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Ouro/administração & dosagem , Hipertermia Induzida/métodos , Camundongos , Nanotubos , Neoplasias Pancreáticas/fisiopatologia , Gencitabina
8.
Nanoscale ; 7(12): 5240-8, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25707848

RESUMO

General adoption of advanced treatment protocols such as chronotherapy will hinge on progress in drug delivery technologies that provide precise temporal control of therapeutic release. Such innovation is also crucial to future medicine approaches such as telemedicine. Here we present a nanofluidic membrane technology capable of achieving active and tunable control of molecular transport through nanofluidic channels. Control was achieved through application of an electric field between two platinum electrodes positioned on either surface of a 5.7 nm nanochannel membrane designed for zero-order drug delivery. Two electrode configurations were tested: laser-cut foils and electron beam deposited thin-films, configurations capable of operating at low voltage (≤1.5 V), and power (100 nW). Temporal, reproducible tuning and interruption of dendritic fullerene 1 (DF-1) transport was demonstrated over multi-day release experiments. Conductance tests showed limiting currents in the low applied potential range, implying ionic concentration polarization (ICP) at the interface between the membrane's micro- and nanochannels, even in concentrated solutions (≤1 M NaCl). The ability of this nanotechnology platform to facilitate controlled delivery of molecules and particles has broad applicability to next-generation therapeutics for numerous pathologies, including autoimmune diseases, circadian dysfunction, pain, and stress, among others.


Assuntos
Preparações de Ação Retardada/síntese química , Sistemas de Liberação de Medicamentos/instrumentação , Fulerenos/administração & dosagem , Fulerenos/química , Membranas Artificiais , Microeletrodos , Preparações de Ação Retardada/efeitos da radiação , Dendrímeros/química , Campos Eletromagnéticos , Desenho de Equipamento , Análise de Falha de Equipamento , Fulerenos/efeitos da radiação , Cinética
9.
Adv Healthc Mater ; 3(10): 1629-37, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24692076

RESUMO

Cancer is a complex disease that usually requires several treatment modalities. A multifunctional nanotherapeutic system is designed, incorporating small interfering RNA (siRNA) and gold nanorods (Au NRs) for photothermal therapy. Surface-engineered Au NRs with polyethylenimine are synthesized using a layer-by-layer assembly and siRNA is absorbed on the surface. The siRNA is efficiently delivered into breast cancer cells, resulting in subsequent gene silencing. Cells are then irradiated with near-infrared (NIR) light, causing heat-induced anticancer activity. The combination of gene silencing and photothermal therapy results in effective inhibition of cell proliferation.


Assuntos
Neoplasias da Mama/terapia , Terapia Genética/métodos , Ouro/química , Hipertermia Induzida/métodos , Nanotubos/química , Fototerapia/métodos , RNA Interferente Pequeno/administração & dosagem , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Feminino , Inativação Gênica , Ouro/administração & dosagem , Humanos , Macrófagos/metabolismo , Camundongos , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética
10.
PLoS One ; 9(2): e86489, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24558362

RESUMO

BACKGROUND: Hyperthermia treatment has been explored as a strategy to overcome biological barriers that hinder effective drug delivery in solid tumors. Most studies have used mild hyperthermia treatment (MHT) to target the delivery of thermo-sensitive liposomes carriers. Others have studied its application to permeabilize tumor vessels and improve tumor interstitial transport. However, the role of MHT in altering tumor vessel interfacial and adhesion properties and its relationship to improved delivery has not been established. In the present study, we evaluated effects of MHT treatment on tumor vessel flow dynamics and expression of adhesion molecules and assessed enhancement in particle localization using mesoporous silicon vectors (MSVs). We also determined the optimal time window at which maximal accumulation occur. RESULTS: In this study, using intravital microscopy analyses, we showed that temporal mild hyperthermia (∼1 W/cm(2)) amplified delivery and accumulation of MSVs in orthotopic breast cancer tumors. The number of discoidal MSVs (1000×400 nm) adhering to tumor vasculature increased 6-fold for SUM159 tumors and 3-fold for MCF-7 breast cancer tumors. By flow chamber experiments and Western blotting, we established that a temporal increase in E-selectin expression correlated with enhanced particle accumulation. Furthermore, MHT treatment was shown to increase tumor perfusion in a time-dependent fashion. CONCLUSIONS: Our findings reveal that well-timed mild hyperthermia treatment can transiently elevate tumor transport and alter vascular adhesion properties and thereby provides a means to enhance tumor localization of non-thermally sensitive particles such as MSVs. Such enhancement in accumulation could be leveraged to increase therapeutic efficacy and reduce drug dosing in cancer therapy.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Selectina E/metabolismo , Hipertermia Induzida/métodos , Silício/química , Animais , Vasos Sanguíneos/metabolismo , Neoplasias da Mama/terapia , Células Endoteliais/citologia , Eritrócitos/citologia , Feminino , Humanos , Lipossomos/química , Células MCF-7 , Camundongos , Camundongos Nus , Microscopia , Transplante de Neoplasias , Perfusão
11.
Nanomedicine ; 10(7): 1487-96, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24262998

RESUMO

The abnormal tumor vasculature presents a major challenge to the adequate delivery of chemotherapeutics, often limiting efficacy. We developed a nanoparticle-based technique to deliver localized mild hyperthermia (MHT) used to transiently alter tumor vascular transport properties and enhance transport of macromolecules into tumor interstitium. The strategy involved administering and localizing accumulation of stealth gold nanorods (GNRs, 103 µg of GNRs/g of tumor), and irradiating tumor with a low-photon laser flux (1 W/cm(2)) to generate MHT. The treatment increased vascular permeability within 24 h after treatment, allowing enhanced transport of macromolecules up to 54 nm in size. A mathematical model is used to describe changes in tumor mass transport properties where the rate of macromolecular exchange between interstitial and vascular region (R) and maximum dye enhancement (Ymax) of 23-nm dextran dye is analytically solved. During enhanced permeability, R increased by 200% while Ymax increased by 30% relative to untreated group in pancreatic CAPAN-1 tumors. MHT treatment also enhanced transport of larger dextran dye (54 nm) as assessed by intravital microscopy, without causing occlusive cellular damage. Enhanced vascular transport was prolonged for up to 24 h after treatment, but reversible with transport parameters returning to basal levels after 36 h. This study indicates that localized mild hyperthermia treatment opens a transient time-window with which to enable and augment macromolecule transport and potentially improve therapeutic efficacy. From the clinical editor: In this study, local intra-tumor mild hyperthermia is induced using a nanoparticle-based approach utilizing stealth gold nanorods and irradiating the tumor with low-photon laser flux, resulting in locally increased vascular permeability enabling enhanced delivery of therapeutics, including macromolecules up to 54 nm in size. Similar approaches would be very helpful in addressing treatment-resistant malignancies in clinical practice.


Assuntos
Vasos Sanguíneos/metabolismo , Permeabilidade Capilar , Hipertermia Induzida , Neoplasias Pancreáticas/irrigação sanguínea , Animais , Transporte Biológico , Camundongos , Camundongos Nus , Nanotubos , Neoplasias Pancreáticas/terapia
12.
Adv Healthc Mater ; 1(1): 84-9, 2012 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23184690

RESUMO

Hollow gold nanoshells are more efficient in heat generation triggered by near infrared laser when they are loaded into porous silicon particles, which results in effective cancer-cell killing in vitro and in vivo. Collective electromagnetic coupling of nanoconfined hollow gold nanoshells leads to dramatic enhancement of thermal ablation.


Assuntos
Hipertermia Induzida/métodos , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Animais , Linhagem Celular Tumoral , Camundongos , Nanopartículas/ultraestrutura , Tamanho da Partícula , Resultado do Tratamento
13.
Lab Chip ; 11(15): 2526-34, 2011 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-21677944

RESUMO

Recent work has elucidated the potential of important new therapeutic paradigms, including metronomic delivery and chronotherapy, in which the precise timing and location of therapeutic administration has a significant impact on efficacy and toxicity. New drug delivery architectures are needed to not only release drug continuously at precise rates, but also synchronize their release with circadian cycles. We present an actively controlled nanofluidic membrane that exploits electrophoresis to control the magnitude, duration, and timing of drug release. The membrane, produced using high precision silicon fabrication techniques, has platinum electrodes integrated at the inlet and outlet that allow both amplification and reversal of analyte delivery with low applied voltage (at or below 2 VDC). Device operation was demonstrated with solutions of both fluorescein isothiocyanate conjugated bovine serum albumin and lysozyme using fluorescence spectroscopy, fluorescence microscopy, and a lysozyme specific bio-assay and has been characterized for long-term molecular release and release reversibility. Through a combination of theoretical and experimental analysis, the relative contributions of electrophoresis and electroosmosis have been investigated. The membrane's clinically relevant electrophoretic release rate at 2 VDC exceeds the passive release by nearly one order of magnitude, demonstrating the potential to realize the therapeutic paradigm goal.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Sistemas de Liberação de Medicamentos/métodos , Membranas Artificiais , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas Analíticas Microfluídicas/métodos , Animais , Bovinos , Eletroforese/instrumentação , Eletroforese/métodos , Microscopia de Fluorescência/métodos , Muramidase/química , Muramidase/farmacologia , Soroalbumina Bovina/química , Soroalbumina Bovina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA