Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Healthc Mater ; 13(7): e2302606, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37987462

RESUMEN

Induction of pyroptosis can promote anti-PD-L1 therapeutic efficacy due to the release of pro-inflammatory cytokines, but current approaches can cause off target toxicity. Herein, a phthalocyanine-conjugated mesoporous silicate nanoparticle (PMSN) is designed for amplifying sonodynamic therapy (SDT) to augment oxidative stress and induce robust pyroptosis in tumors. The sub-10 nm diameter structure and c(RGDyC)-PEGylated modification enhance tumor targeting and renal clearance. The unique porous architecture of PMSN doubles ROS yield and enhances pyroptotic cell populations in tumors (25.0%) via a cavitation effect. PMSN-mediated SDT treatment efficiently reduces tumor mass and suppressed residual tumors in treated and distant sites by synergizing with PD-L1 blockade (85.93% and 77.09%, respectively). Furthermore, loading the chemotherapeutic, doxorubicin, into PMSN intensifies SDT-pyroptotic effects and increased efficacy. This is the first report of the use of SDT regimens to induce pyroptosis in liver cancer. This noninvasive and effective strategy has potential for clinical translation.


Asunto(s)
Neoplasias Hepáticas , Nanopartículas , Terapia por Ultrasonido , Humanos , Piroptosis , Antígeno B7-H1 , Línea Celular Tumoral , Nanopartículas/química , Inmunoterapia
2.
STAR Protoc ; 4(4): 102723, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37976155

RESUMEN

Microbubbles are currently approved for diagnostic ultrasound imaging and are under evaluation in therapeutic protocols. Here, we present a protocol for in vitro sonoporation validation using non-targeted microbubbles for gene delivery. We describe steps for computational simulation, experimental calibration, reagent preparation, ultrasound treatment, validation, and gene expression analysis. This protocol uses approved diagnostic microbubbles and parameters that are applicable for human use. For complete details on the use and execution of this protocol, please refer to Bez et al. (2017).1.


Asunto(s)
Sistemas de Liberación de Medicamentos , Microburbujas , Humanos , Sistemas de Liberación de Medicamentos/métodos , Ultrasonografía/métodos , Técnicas de Transferencia de Gen
3.
Theranostics ; 13(15): 5151-5169, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37908737

RESUMEN

Rationale: Despite recent advances in the use of adeno-associated viruses (AAVs) as potential vehicles for genetic intervention of central and peripheral nervous system-associated disorders, gene therapy for the treatment of neuropathology in adults has not been approved to date. The currently FDA-approved AAV-vector based gene therapies rely on naturally occurring serotypes, such as AAV2 or AAV9, which display limited or no transport across the blood-brain barrier (BBB) if systemically administered. Recently developed engineered AAV variants have shown broad brain transduction and reduced off-target liver toxicity in non-human primates (NHPs). However, these vectors lack spatial selectivity for targeted gene delivery, a potentially critical limitation for delivering therapeutic doses in defined areas of the brain. The use of microbubbles, in conjunction with focused ultrasound (FUS), can enhance regional brain AAV transduction, but methods to assess transduction in vivo are needed. Methods: In a murine model, we combined positron emission tomography (PET) and optical imaging of reporter gene payloads to non-invasively assess the spatial distribution and transduction efficiency of systemically administered AAV9 after FUS and microbubble treatment. Capsid and reporter probe accumulation are reported as percent injected dose per cubic centimeter (%ID/cc) for in vivo PET quantification, whereas results for ex vivo assays are reported as percent injected dose per gram (%ID/g). Results: In a study spanning accumulation and transduction, mean AAV9 accumulation within the brain was 0.29 %ID/cc without FUS, whereas in the insonified region of interest of FUS-treated mice, the spatial mean and maximum reached ~2.3 %ID/cc and 4.3 %ID/cc, respectively. Transgene expression assessed in vivo by PET reporter gene imaging employing the pyruvate kinase M2 (PKM2)/[18F]DASA-10 reporter system increased up to 10-fold in the FUS-treated regions, as compared to mice receiving AAVs without FUS. Systemic injection of AAV9 packaging the EF1A-PKM2 transgene followed by FUS in one hemisphere resulted in 1) an average 102-fold increase in PKM2 mRNA concentration compared to mice treated with AAVs only and 2) a 12.5-fold increase in the insonified compared to the contralateral hemisphere of FUS-treated mice. Conclusion: Combining microbubbles with US-guided treatment facilitated a multi-hour BBB disruption and stable AAV transduction in targeted areas of the murine brain. This unique platform has the potential to provide insight and aid in the translation of AAV-based therapies for the treatment of neuropathologies.


Asunto(s)
Dependovirus , Tomografía Computarizada por Rayos X , Ratones , Animales , Dependovirus/genética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Tomografía de Emisión de Positrones , Vectores Genéticos
4.
Nat Commun ; 14(1): 6575, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37852951

RESUMEN

Manipulating gene expression in the host genome with high precision is crucial for controlling cellular function and behavior. Here, we present a precise, non-invasive, and tunable strategy for controlling the expression of multiple endogenous genes both in vitro and in vivo, utilizing ultrasound as the stimulus. By engineering a hyper-efficient dCas12a and effector under a heat shock promoter, we demonstrate a system that can be inducibly activated through thermal energy produced by ultrasound absorption. This system allows versatile thermal induction of gene activation or base editing across cell types, including primary T cells, and enables multiplexed gene activation using a single guide RNA array. In mouse models, localized temperature elevation guided by high-intensity focused ultrasound effectively triggers reporter gene expression in implanted cells. Our work underscores the potential of ultrasound as a clinically viable approach to enhance cell and gene-based therapies via precision genome and epigenome engineering.


Asunto(s)
Edición Génica , Genoma , Animales , Ratones , Genoma/genética , Terapia Genética , Epigenoma , Genes Reporteros , Sistemas CRISPR-Cas/genética
5.
Theranostics ; 12(18): 7884-7902, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36451859

RESUMEN

Background: Although combination immunotherapies incorporating local and systemic components have shown promising results in treating solid tumors, varied tumor microenvironments (TMEs) can impact immunotherapeutic efficacy. Method: We designed and evaluated treatment strategies for breast and pancreatic cancer combining magnetic resonance-guided focused ultrasound (MRgFUS) ablation and antibody therapies. With a combination of single-cell sequencing, spectral flow cytometry, and histological analyses, we profiled an immune-suppressed KPC (Kras+/LSL-G12D; Trp53+/LSL-R172H; Pdx1-Cre) pancreatic adenocarcinoma (MT4) model and a dense epithelial neu deletion (NDL) HER2+ mammary adenocarcinoma model with a greater fraction of lymphocytes, natural killer cells and activated dendritic cells. We then performed gene ontology analysis, spectral and digital cytometry to assess the immune response to combination immunotherapies and correlation with survival studies. Result: Based on gene ontology analysis, adding ablation to immunotherapy enriched immune cell migration pathways in the pancreatic cancer model and extensively enriched wound healing pathways in the breast cancer model. With CIBERSORTx digital cytometry, aCD40 + aPD-1 immunotherapy combinations enhanced dendritic cell activation in both models. In the MT4 TME, adding the combination of aCD40 antibody and checkpoint inhibitors (aPD-1 and aCTLA-4) with ablation was synergistic, increasing activated natural killer cells and T cells in distant tumors. Furthermore, ablation with immunotherapy upregulated critical Ly6c myeloid remodeling phenotypes that enhance T-cell effector function and increased granzyme and protease encoding genes by as much as 100-fold. Ablation combined with immunotherapy then extended survival in the MT4 model to a greater extent than immunotherapy alone. Conclusion: In summary, TME profiling informed a successful multicomponent treatment protocol incorporating ablation and facilitated differentiation of TMEs in which ablation is most effective.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Ratones , Animales , Neoplasias Pancreáticas/terapia , Inmunoterapia , Factores Inmunológicos , Microambiente Tumoral , Neoplasias Pancreáticas
6.
Zhongguo Dang Dai Er Ke Za Zhi ; 23(12): 1262-1266, 2021 Dec 15.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-34911610

RESUMEN

OBJECTIVES: To study the physical and neuropsychological development of children with Citrin deficiency (CD). METHODS: A total of 93 children, aged 1.9-59.8 months, who were diagnosed with CD by SLC25A13 gene analysis in the First Affiliated Hospital of Jinan University from August 2010 to August 2015, were enrolled as subjects. A retrospective analysis was performed for their birth condition and physical growth and neuropsychological development indices. Among these children, 7 underwent physical measurement and neuropsychological development assessment within 1 year old and after 1 year old, and therefore, a total of 100 cases were included for analysis. RESULTS: For the 93 children with CD, the incidence rate of failure to thrive was 25% (23 children) and the proportion of small for gestational age was 47% (44 children). For the 100 cases of CD, the incidence rates of growth retardation, underweight, emaciation, overweight, and microcephalus were 23% (23 cases), 14% (14 cases), 4% (4 cases), 8% (8 cases), and 9% (9 cases), respectively. The incidence rate of neuropsychological developmental delay was 25% (25 cases), and the incidence rates of development delay in the five domains of adaptability, gross motor, fine motor, language, and social ability were 7% (7 cases), 15% (15 cases), 7% (7 cases), 9% (9 cases), and 7% (7 cases), respectively. CONCLUSIONS: Physical and neuropsychological developmental delay can be observed in children with CD, and physical and neuropsychological development should be regularly assessed.


Asunto(s)
Desarrollo Infantil , Citrulinemia , Citrulinemia/fisiopatología , Humanos , Lactante , Proteínas de Transporte de Membrana Mitocondrial , Pruebas Neuropsicológicas , Estudios Retrospectivos
7.
Adv Ther (Weinh) ; 4(9)2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34632048

RESUMEN

Immunotherapy is an important cancer treatment strategy; nevertheless, the lack of robust immune cell infiltration in the tumor microenvironment remains a factor in limiting patient response rates. In vivo gene delivery protocols can amplify immune responses and sensitize tumors to immunotherapies, yet non-viral transfection methods often sacrifice transduction efficiency for improved safety tolerance. To improve transduction efficiency, we optimized a strategy employing low ultrasound transmission frequency-induced bubble oscillation to introduce plasmids into tumor cells. Differential centrifugation isolated size-specific microbubbles. The diameter of the small microbubble population was 1.27 ± 0.89 µm and that of larger population was 4.23 ± 2.27 µm. Upon in vitro insonation with the larger microbubble population, 29.7% of cancer cells were transfected with DNA plasmids, higher than that with smaller microbubbles (18.9%, P <0.05) or positive control treatments with a commercial transfection reagent (12%, P < 0.01). After 48 h, gene expression increased more than two-fold in tumors treated with large, as compared with small, microbubbles. Furthermore, the immune response, including tumor infiltration of CD8+ T cells and F4/80+ macrophages, was enhanced. We believe that this safe and efficacious method can improve preclinical procedures and outcomes for DNA vaccines in cancer immunotherapy in the future.

8.
Adv Drug Deliv Rev ; 178: 113906, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34333075

RESUMEN

Due to the ease of use and excellent safety profile, ultrasound is a promising technique for both diagnosis and site-specific therapy. Ultrasound-based techniques have been developed to enhance the pharmacokinetics and efficacy of therapeutic agents in cancer treatment. In particular, transfection with exogenous nucleic acids has the potential to stimulate an immune response in the tumor microenvironment. Ultrasound-mediated gene transfection is a growing field, and recent work has incorporated this technique into cancer immunotherapy. Compared with other gene transfection methods, ultrasound-mediated gene transfection has a unique opportunity to augment the intracellular uptake of nucleic acids while safely and stably modulating the expression of immunostimulatory cytokines. The development and commercialization of therapeutic ultrasound systems further enhance the potential translation. In this Review, we introduce the underlying mechanisms and ongoing preclinical studies of ultrasound-based techniques in gene transfection for cancer immunotherapy. Furthermore, we expand on aspects of therapeutic ultrasound that impact gene therapy and immunotherapy, including tumor debulking, enhancing cytokines and chemokines and altering nanoparticle pharmacokinetics as these effects of ultrasound cannot be fully dissected from targeted gene therapy. We finally explore the outlook for this rapidly developing field.


Asunto(s)
Terapia Genética , Inmunoterapia , Neoplasias/terapia , Terapia por Ultrasonido , Animales , Humanos , Nanopartículas/química
9.
Sci Rep ; 11(1): 927, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441763

RESUMEN

High intensity focused ultrasound (HIFU) rapidly and non-invasively destroys tumor tissue. Here, we sought to assess the immunomodulatory effects of MR-guided HIFU and its combination with the innate immune agonist CpG and checkpoint inhibitor anti-PD-1. Mice with multi-focal breast cancer underwent ablation with a parameter set designed to achieve mechanical disruption with minimal thermal dose or a protocol in which tumor temperature reached 65 °C. Mice received either HIFU alone or were primed with the toll-like receptor 9 agonist CpG and the checkpoint modulator anti-PD-1. Both mechanical HIFU and thermal ablation induced a potent inflammatory response with increased expression of Nlrp3, Jun, Mefv, Il6 and Il1ß and alterations in macrophage polarization compared to control. Furthermore, HIFU upregulated multiple innate immune receptors and immune pathways, including Nod1, Nlrp3, Aim2, Ctsb, Tlr1/2/4/7/8/9, Oas2, and RhoA. The inflammatory response was largely sterile and consistent with wound-healing. Priming with CpG attenuated Il6 and Nlrp3 expression, further upregulated expression of Nod2, Oas2, RhoA, Pycard, Tlr1/2 and Il12, and enhanced T-cell number and activation while polarizing macrophages to an anti-tumor phenotype. The tumor-specific antigen, cytokines and cell debris liberated by HIFU enhance response to innate immune agonists.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Animales , Neoplasias de la Mama/fisiopatología , Modelos Animales de Enfermedad , Humanos , Inmunidad , Espectroscopía de Resonancia Magnética/métodos , Ratones , Ratones Endogámicos , Neoplasias/inmunología , Oligodesoxirribonucleótidos/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Pirina/metabolismo , Ultrasonografía/métodos
10.
Proc Natl Acad Sci U S A ; 117(23): 12674-12685, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32430322

RESUMEN

Robust cytotoxic T cell infiltration has proven to be difficult to achieve in solid tumors. We set out to develop a flexible protocol to efficiently transfect tumor and stromal cells to produce immune-activating cytokines, and thus enhance T cell infiltration while debulking tumor mass. By combining ultrasound with tumor-targeted microbubbles, membrane pores are created and facilitate a controllable and local transfection. Here, we applied a substantially lower transmission frequency (250 kHz) than applied previously. The resulting microbubble oscillation was significantly enhanced, reaching an effective expansion ratio of 35 for a peak negative pressure of 500 kPa in vitro. Combining low-frequency ultrasound with tumor-targeted microbubbles and a DNA plasmid construct, 20% of tumor cells remained viable, and ∼20% of these remaining cells were transfected with a reporter gene both in vitro and in vivo. The majority of cells transfected in vivo were mucin 1+/CD45- tumor cells. Tumor and stromal cells were then transfected with plasmid DNA encoding IFN-ß, producing 150 pg/106 cells in vitro, a 150-fold increase compared to no-ultrasound or no-plasmid controls and a 50-fold increase compared to treatment with targeted microbubbles and ultrasound (without IFN-ß). This enhancement in secretion exceeds previously reported fourfold to fivefold increases with other in vitro treatments. Combined with intraperitoneal administration of checkpoint inhibition, a single application of IFN-ß plasmid transfection reduced tumor growth in vivo and recruited efficacious immune cells at both the local and distant tumor sites.


Asunto(s)
Inmunoterapia/métodos , Interferón beta/genética , Neoplasias Experimentales/terapia , Linfocitos T/inmunología , Transfección/métodos , Ondas Ultrasónicas , Animales , Línea Celular Tumoral , Membrana Celular/efectos de la radiación , Movimiento Celular , Humanos , Interferón beta/metabolismo , Ratones , Microburbujas/uso terapéutico , Linfocitos T/fisiología
11.
RSC Adv ; 9(72): 41993-41999, 2019 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-35542847

RESUMEN

The recent progress in the development of highly biocompatible nanoplatforms mostly encompasses the use of biological excipients such as red blood cells, cancer cell membranes, and also platelets. Such specialized vectors, if mimicked correctly, have intrinsic ability to navigate through the biological system and perform their intended action without eliciting any cascade of inflammatory processes. Naturally, platelets have been found to accumulate in the wound sites and also interact with circulating tumor cells (CTCs). Inspired by the targeting ability of platelets and the clinical success of ultrasound, herein we developed a novel ultrasound contrast agent (UCA) by backfilling of an insoluble gas into the platelets after lyophilization ex vivo. The as-prepared platelet-based ultrasound contrast agent (P-UCA) disguised the structural integrity of the natural platelets with an average diameter of 3.1 ± 0.4 µm, and could enhance the ultrasound signal both in vitro and in vivo. Besides, we further evaluated that such platelet particles could facilitate active loading of ICG molecules for prolonged in vivo fluorescence imaging compared to the free ICG. Taking all the results together, we established that biological structures such as platelets could be repurposed ex vivo as a"shell" to encapsulate gas and be further extended to load ICG for real-time ultrasound and fluorescence imaging respectively. This not only indicates many potential uses of these MBs in the diagnosis of platelet-related diseases, such as vascular damage, thrombosis, and atherosclerosis, but also serves as a powerful platform with multimodal theranostic capability after active loading of a variety of therapeutic and diagnostic agents.

12.
Theranostics ; 8(19): 5501-5518, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555560

RESUMEN

The limited clinical efficacy of monotherapies in the clinic has urged the development of novel combination platforms. Taking advantage of light-triggered photodynamic treatment combined together with the controlled release of nanomedicine, it has been possible to treat cancer without eliciting any adverse effects. However, the challenges imposed by limited drug loading capacity and complex synthesis process of organic nanoparticles (NPs) have seriously impeded advances in chemo-photodynamic combination therapy. In this experiment, we utilize our previously synthesized porphyrin-grafted lipid (PGL) NPs to load highly effective chemotherapeutic drug, doxorubicin (DOX) for synergistic chemo-photodynamic therapy. Methods: A relatively simple and inexpensive rapid injection method was used to prepare porphyrin-grafted lipid (PGL) NPs. The self-assembled PGL NPs were used further to encapsulate DOX via a pH-gradient loading protocol. The self-assembled liposome-like PGL NPs having a hydrophilic core were optimized to load DOX at an encapsulation efficiency (EE) of ~99%. The resultant PGL-DOX NPs were intact, highly stable and importantly these NPs successfully escaped from the endo-lysosomal compartment after laser irradiation to release DOX in the cytosol. The therapeutic efficacy of the aforementioned formulation was validated both in vitro and in vivo. Results: PGL-DOX NPs demonstrated excellent cellular uptake, chemo-photodynamic response, and fluorescence imaging ability in different cell lines. Under laser irradiation, cells treated with a low molar concentration of PGL-DOX NPs reduced cell viability significantly. Moreover, in vivo experiments conducted in a xenograft mouse model further demonstrated the excellent tumor accumulation capability of PGL-DOX NPs driven by the enhanced permeability and retention (EPR) effect. Through fluorescence imaging, the biodistribution of PGL-DOX NPs in tumor and major organs was also easily monitored in real time in vivo. The inherent ability of porphyrin to generate ROS under laser irradiation combined with the cytotoxic effect of the anticancer drug DOX significantly suppressed tumor growth in vivo. Conclusion: In summary, the PGL-DOX NPs combined chemo-photodynamic nanoplatform may serve as a potential candidate for cancer theranostics.


Asunto(s)
Antibióticos Antineoplásicos/metabolismo , Doxorrubicina/metabolismo , Portadores de Fármacos/química , Colorantes Fluorescentes/metabolismo , Liposomas/química , Porfirinas/metabolismo , Nanomedicina Teranóstica/métodos , Animales , Carcinoma/terapia , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Quimioterapia/métodos , Células HeLa , Xenoinjertos , Humanos , Terapia por Luz de Baja Intensidad , Ratones , Nanopartículas/química , Trasplante de Neoplasias , Imagen Óptica/métodos , Células PC-3 , Fotoquimioterapia/métodos , Resultado del Tratamiento
13.
Bioconjug Chem ; 29(12): 3967-3981, 2018 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-30485070

RESUMEN

Chemotherapy suffers from some limitations such as poor bioavailability, rapid clearance from blood, poor cellular uptake, low tumor accumulation, severe side effects on healthy tissues and most importantly multidrug resistance (MDR) in cancer cells. Nowadays, a series of smart drug delivery system (DDS) based on amphiphilic drug conjugates (ADCs) has been developed to solve these issues, including polymer-drug conjugate (PDC), phospholipid-mimicking prodrugs, peptide-drug conjugates (PepDCs), pure nanodrug (PND), amphiphilic drug-drug conjugate (ADDC), and Janus drug-drug conjugate (JDDC). These ADCs can self-assemble into nanoparticles (NPs) or microbubbles (MBs) for targeted drug delivery by minimizing the net amount of excipients, realizing great goals, such as stealth behavior and physical integrity, high drug loading content, no premature leakage, long blood circulation time, fixed drug combination, and controlled drug-release kinetics. Besides, these self-assembled systems can be further used to load additional therapeutic agents and imaging contrast agents for combined therapy, personalized monitoring of in vivo tumor targeting, and the pharmacokinetics of drugs for predicting the therapeutic outcome. In this review, we will summarize the latest progress in the development of ADCs based combination chemotherapy and discuss the important roles for overcoming the tumor MDR.


Asunto(s)
Nanomedicina , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Humanos , Polímeros/farmacología , Medicina de Precisión
14.
ACS Appl Mater Interfaces ; 10(35): 29385-29397, 2018 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-30096224

RESUMEN

Triple-negative breast cancer (TNBC) is a malignant and refractory disease with high morbidity and mortality. The TNBC shows no response to hormonal therapy nor targeted therapy due to the lack of known targetable biomarkers. Furthermore, the TNBC also exhibits a high degree of heterogeneity that leads to cancer evolution, drug resistance, metastatic progression, and recurrence, arising from the tumor-initiating properties of cancer stem cells (CSCs). Thus, the development of radical therapeutic regimens with high efficacy and limited side effects is crucial. In this study, we designed an innovative ternary cocktail chemotherapy by using Lovastatin (L)-loaded Janus camptothecin-floxuridine conjugate (CF) nanocapsules (NCs) with ultrahigh drug loading capacity. The obtained LCF NCs were shown to be able to suppress growth of TNBC, including inhibition of growth and metastasis of CSCs, both in vitro and in tumor-bearing mice. Moreover, in animal experiments, the LCF NCs showed sustained and synchronous drug release (half-life > 300 min), 85.2% reduction in pulmonary metastases, and no cancer recurrence during one-month observation post-treatment. Thus, this innovative LCF NC design provides a simple and synergistic strategy for the development of simultaneous triple chemotherapy and could be an efficacious, safe, and amenable choice with higher therapeutic relevance and fewer toxic complications than conventional multidrug delivery systems for TNBC treatment in the future.


Asunto(s)
Camptotecina , Floxuridina , Lovastatina , Nanocápsulas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Camptotecina/administración & dosificación , Camptotecina/química , Línea Celular Tumoral , Floxuridina/administración & dosificación , Floxuridina/química , Humanos , Lovastatina/administración & dosificación , Lovastatina/química , Ratones , Nanocápsulas/administración & dosificación , Nanocápsulas/química
15.
ACS Nano ; 12(7): 7312-7326, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29901986

RESUMEN

Multidrug resistance remains one of the main obstacles to efficient chemotherapy of colorectal cancer. Herein, an efficient combination therapeutic strategy is proposed based on porphyrin/camptothecin-floxuridine triad microbubbles (PCF-MBs) with high drug loading contents, which own highly stable co-delivery drug combinations and no premature release. The triad PCF-MBs can act not only as a contrast agent for ultrasound (US)/fluorescence bimodal imaging but also a multimodal therapeutic agent for synergistic chemo-photodynamic combination therapy. Upon local ultrasound exposure under the guidance of ultrasound imaging, in situ conversion of PCF-MBs into porphyrin/camptothecin-floxuridine nanoparticles (PCF-NPs) leads to high accumulation of chemo-drugs and photosensitizer in tumors due to the induced high permeability of the capillary wall and cell membrane temporarily via sonoporation effect, greatly reducing the risk of systemic exposure. Most importantly, it was found that the PCF-MB-mediated photodynamic therapy could significantly reduce the expression of adenosine-triphosphate (ATP)-binding cassette subfamily G member 2 (ABCG2), which is responsible for the drug resistance in chemotherapy, resulting in a prominent intracellular camptothecin increase. In vivo experiments revealed that the PCF-MBs in combination with ultrasound and laser irradiation could achieve a 90% tumor inhibition rate of HT-29 cancer with no recurrence. Therefore, such triad PCF-MB-based combination therapeutic strategy shows great promise for overcoming drug resistance of colorectal cancer and other cancers.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Microburbujas , Nanopartículas/química , Fármacos Fotosensibilizantes/farmacología , Ultrasonografía , Antineoplásicos/química , Camptotecina/química , Neoplasias Colorrectales/diagnóstico por imagen , Células HT29 , Humanos , Imagen Óptica , Tamaño de la Partícula , Fármacos Fotosensibilizantes/química , Porfirinas/química , Propiedades de Superficie
16.
J Cell Mol Med ; 22(9): 4171-4182, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29956460

RESUMEN

The presence of blood-brain barrier (BBB) greatly limits the availability of drugs and their efficacy against glioma. Focused ultrasound (FUS) can induce transient and local BBB opening for enhanced drug delivery. Here, we developed polysorbate 80-modified paclitaxel-loaded PLGA nanoparticles (PS-80-PTX-NPs, PPNP) and examined the enhanced local delivery into the brain for glioma treatment by combining with FUS. Our result showed PPNP had good stability, fast drug release rate and significant toxicity to glioma cells. Combined with FUS, PPNP showed a stronger BBB permeation efficiency both in the in vitro and in vivo BBB models. Mechanism studies revealed the disrupted tight junction, reduced P-glycoprotein expression and ApoE-dependent PS-80 permeation collectively contribute to the enhanced drug delivery, resulting in significantly stronger antitumour efficacy and longer survival time in the tumour-bearing mice. Our study provided a new strategy to efficiently and locally deliver drugs into the brain to treat glioma.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/terapia , Sistemas de Liberación de Medicamentos/métodos , Glioblastoma/terapia , Nanopartículas/química , Paclitaxel/farmacología , Polisorbatos/química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacocinética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Liberación de Fármacos , Tratamiento con Ondas de Choque Extracorpóreas/métodos , Femenino , Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Paclitaxel/farmacocinética , Análisis de Supervivencia , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología
17.
Theranostics ; 8(8): 2264-2277, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721078

RESUMEN

Rationale: Treatment for Parkinson's disease (PD) is challenged by the presence of the blood-brain barrier (BBB) that significantly limits the effective drug concentration in a patient's brain for therapeutic response throughout various stages of PD. Curcumin holds the potential for α-synuclein clearance to treat PD; however, its applications are still limited due to its low bioavailability and poor permeability through the BBB in a free form. Methods: Herein, this paper fabricated curcumin-loaded polysorbate 80-modified cerasome (CPC) nanoparticles (NPs) with a mean diameter of ~110 nm for enhancing the localized curcumin delivery into the targeted brain nuclei via effective BBB opening in combination with ultrasound-targeted microbubble destruction (UTMD). Results: The liposomal nanohybrid cerasome exhibited superior stability towards PS 80 surfactant solubilization and longer circulation lifetime (t1/2 = 6.22 h), much longer than free curcumin (t1/2 = 0.76 h). The permeation was found to be 1.7-fold higher than that of CPC treatment only at 6 h after the systemic administration of CPC NPs. Notably, motor behaviors, dopamine (DA) level and tyrosine hydroxylase (TH) expression all returned to normal, thanks to α-synuclein (AS) removal mediated by efficient curcumin delivery to the striatum. Most importantly, the animal experiment demonstrated that the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice had notably improved behavior disorder and dopamine depletion during two-week post-observation after treatment with CPC NPs (15 mg curcumin/kg) coupled with UTMD. Conclusion: This novel CPC-UTMD formulation approach could be an effective, safe and amenable choice with higher therapeutic relevance and fewer unwanted complications than conventional chemotherapeutics delivery systems for PD treatment in the near future.


Asunto(s)
Encéfalo/metabolismo , Curcumina/administración & dosificación , Curcumina/farmacología , Sistemas de Liberación de Medicamentos , Microburbujas , Enfermedad de Parkinson/diagnóstico por imagen , Polisorbatos/química , Animales , Encéfalo/efectos de los fármacos , Curcumina/química , Curcumina/farmacocinética , Dopamina/metabolismo , Hidrodinámica , Liposomas , Ratones Endogámicos C57BL , Modelos Biológicos , Actividad Motora , Neostriado/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Tamaño de la Partícula , Permeabilidad , Electricidad Estática , Distribución Tisular , Ultrasonografía
18.
ACS Appl Mater Interfaces ; 10(4): 3219-3228, 2018 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-29299917

RESUMEN

A dual-in-dual synergistic strategy was proposed based on the self-assembly of combinatorial nanocapsules (NCs) from Janus camptothecin-floxuridine (CF) conjugate and the near-infrared absorber of 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide (DiR) by introducing PEGylated phospholipid of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycerol)-2000] to increase the blood circulation time of NCs. Due to the use of amphiphilic CF and DiR themselves to form liposome-like nanocapsules, the obtained CF-DiR NCs owned a significantly high loading content, a stable co-delivery drug combinations, a no premature release, and an excellent photothermal conversion efficiency. The in vivo fluorescence imaging indicated that CF-DiR NCs could achieve a high tumor accumulation after an intravenous injection. The dual drugs of camptothecin and floxuridine could be coordinately released due to the hydrolysis of the ester bond by the esterase in tumor. The in vivo experiments showed that more cytotoxicity of the CF-DiR NCs-mediated chemo- and photothermal dual therapy to tumor cells could be clearly observed than the chemotherapy or photothermal therapy alone due to the synergistic effect, leading to no recurrence in the entire treatment. All of the results highlighted that CF-DiR NCs were highly effective theranostic agents that could be used for imaging-guided cancer chemophotothermal therapy to conquer an intrinsic resistance to chemotherapeutics.


Asunto(s)
Nanocápsulas , Camptotecina , Floxuridina , Humanos , Liposomas , Neoplasias
19.
Nanoscale ; 9(33): 11888-11901, 2017 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-28561825

RESUMEN

We have demonstrated that a typical nanothermometer was incorporated in a bovine serum albumin stabilized gold nanostar-indocyanine green (denoted as GNS-ICG-BSA) nanoprobe to realize surface-enhanced Raman scattering (SERS) imaging-based real-time sensitive monitoring of intracellular temperature in photothermal therapy (PTT), which significantly improved the spatial resolution compared to infrared thermal imaging. Herein, an exogenous thermosensitive molecule, ICG, acting as a tri-functional agent, was selected as the Raman reporter instead of direct cellular biochemical changes. The triggering of the obtained probe was unaffected by the cellular microenvironment, so it can act as a monitor of PTT in various cell types. High-resolution mass spectrometry (HRMS) was used to investigate the thermosensitive mechanism of ICG. The actively targeted GNS-ICG-BSA nanotags were used to induce SERS mapping-guided in vitro PTT of U87 glioma cells. Meanwhile, small temperature variations within a cell during PTT can be precisely monitored through the SERS fingerprint information, with a spatial resolution at the subcellular level and a sensitivity of 0.37 °C. Thus, the integrated GNS-ICG-BSA nanotags can be treated as a theranostic probe, a SERS imaging probe and an intracellular thermometer. Moreover, the good biocompatibility and the low cytotoxicity of GNS-ICG-BSA nanotags, together with their superior photothermal ablation effect on U87 glioma cells have been confirmed. This suggested that the implanted nanothermometry approach would be promising for a better understanding of the biological processes at subcellular level and provide new insights into the fabrication of a multifunctional nanoplatform. Furthermore, this study revealed that the SERS-based monitoring technique can offer great potential for theranostics as an emerging strategy.


Asunto(s)
Oro , Verde de Indocianina , Nanopartículas del Metal , Fotoquimioterapia , Espectrometría Raman , Línea Celular Tumoral , Humanos , Albúmina Sérica Bovina , Nanomedicina Teranóstica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...