Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 20(3): 1818-1841, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36802639

RESUMEN

Photochemical internalization (PCI) is a novel, minimally invasive drug delivery technology that facilitates the delivery of therapeutic molecules into the cytosol of cells. In this work, PCI was utilized in an effort to enhance the therapeutic index of the existing anticancer drugs as well as novel nanοformulations against breast and pancreatic cancer cells. Frontline anticancer drugs were tested with bleomycin as a benchmark PCI control; namely, three vinca alkaloids (vincristine, vinorelbine, and vinblastine), two taxanes (docetaxel and paclitaxel), two antimetabolites (gemcitabine and capecitabine), a combination of taxanes with antimetabolites, and two nano-sized formulations (squalene- and polymer-bound gemcitabine derivatives) were tested in a 3D PCI in vitro model. Strikingly, we discovered that several drug molecules exhibited remarkably augmented therapeutic activity by several orders of magnitude compared to their respective controls (without PCI technology or directly compared with bleomycin controls). Nearly all drug molecules showed enhanced therapeutic efficiency, but more interestingly, we traced several drug molecules that showed multi-fold enhancement (ranging from 5000- up to 170,000-fold enhancement) in their IC70 indices. Interestingly, PCI delivery of the vinca alkaloids (especially PCI-vincristine), and some of the nanoformulations tested, was seen to perform impressively across all of the treatment outcomes of potency, efficacy, and synergy─as determined by means of a cell viability assay. The study constitutes a systematic guide for the development of future PCI-based therapeutic modalities for precision oncology.


Asunto(s)
Antineoplásicos , Neoplasias Pancreáticas , Alcaloides de la Vinca , Humanos , Vincristina , Antimetabolitos , Línea Celular Tumoral , Medicina de Precisión , Antineoplásicos/uso terapéutico , Paclitaxel , Bleomicina , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas
2.
Oncologist ; 27(6): 430-e433, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35675633

RESUMEN

BACKGROUND: Photochemical internalization (PCI) is a novel technology for light-induced enhancement of the local therapeutic effect of cancer drugs, utilizing a specially designed photosensitizing molecule (fimaporfin). The photosensitizing molecules are trapped in endosomes along with macromolecules or drugs. Photoactivation of fimaporfin disrupts the endosomal membranes so that drug molecules are released from endosomes inside cells and can reach their therapeutic target in the cell cytosol or nucleus. Compared with photodynamic therapy, the main cytotoxic effect with PCI is disruption of the endosomal membrane resulting in delivery of chemotherapy drug, and not to the photochemical reactions per se. In this study we investigated the effect of PCI with gemcitabine in patients with inoperable perihilar cholangiocarcinoma (CCA). METHODS: The in vitro cytotoxic effect of PCI with gemcitabine was studied on two CCA-derived cell lines. In a fimaporfin dose-escalation phase I clinical study, we administered PCI with gemcitabine in patients with perihilar CCA (n = 16) to establish a safe and tolerable fimaporfin dose and to get early signals of efficacy. The patients enrolled in the study had tumors in which the whole length of the tumor could be illuminated from the inside of the bile duct, using an optical fiber inserted via an endoscope (Fig. 1). Fimaporfin was administered intravenously at day 0; gemcitabine (i.v.) and intraluminal biliary endoscopic laser light application on day 4; followed by standard gemcitabine/cisplatin chemotherapy. RESULTS: Preclinical experiments showed that PCI enhanced the effect of gemcitabine. In patients with CCA, PCI with gemcitabine was well tolerated with no dose-limiting toxicities, and no unexpected safety signals. Disease control was achieved in 10 of 11 evaluable patients, with a clearly superior effect in the two highest dose groups. The objective response rate (ORR) was 42%, including two complete responses, while ORR at the highest dose was 60%. Progression-free survival at 6 months was 75%, and median overall survival (mOS) was 15.4 months, with 22.8 months at the highest fimaporfin dose. CONCLUSION: Photochemical internalization with gemcitabine was found to be safe and resulted in encouraging response and survival rates in patients with unresectable perihilar CCA.


Asunto(s)
Colangiocarcinoma , Desoxicitidina , Fotoquimioterapia , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Conductos Biliares Intrahepáticos , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/patología , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Humanos , Fotoquimioterapia/efectos adversos , Fotoquimioterapia/métodos , Gemcitabina
3.
Pharm Res ; 39(6): 1047-1064, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35619043

RESUMEN

Cytoplasmic delivery of bioactives requires the use of strategies such as active transport, electroporation, or the use of nanocarriers such as polymeric nanoparticles, liposomes, micelles, and dendrimers. It is essential to deliver bioactive molecules in the cytoplasm to achieve targeted effects by enabling organelle targeting. One of the biggest bottlenecks in the successful cytoplasmic delivery of bioactives through nanocarriers is their sequestration in the endosomes that leads to the degradation of drugs by progressing to lysosomes. In this review, we discussed mechanisms by which nanocarriers are endocytosed, the mechanisms of endosomal escape, and more importantly, the strategies that can be and have been employed for their escape from the endosomes are summarized. Like other nanocarriers, polymeric micelles can be designed for endosomal escape, however, a careful control is needed in their design to balance between the possible toxicity and endosomal escape efficiency. Keeping this in view, polyion complex micelles, and polymers that have the ability to escape the endosome, are fully discussed. Finally, we provided some perspectives for designing the polymeric micelles for efficient cytoplasmic delivery of bioactive agents through endosomal escape.


Asunto(s)
Micelas , Nanopartículas , Endocitosis , Endosomas/metabolismo , Polímeros/metabolismo
4.
Mikrochim Acta ; 189(3): 120, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35201432

RESUMEN

Although upconversion photodynamic therapy (PDT) has gained extensive interests in disease treatment, the intracellular migration pathway of upconversion photosensitizers and underlying cell-particle interaction mechanism is still largely unexplored. In this work photoswitchable upconversion nanoparticles (UCNPs) are reported  that can release orthogonal emissions excited by two near-infrared lights, i.e., red color of 980-nm and green color of 808-nm light excitation. Taking advantage of the dual-emissive property, a methodology based on Pearson's correlation analysis is proposed to verify the accuracy of upconversion luminescence signals under different excitation lights, which has been previously neglected. Meanwhile, we have designed a near-infrared mediated bioimaging nanoplatform that can generate reactive oxygen species (ROS) using one light and simultaneously track the location of upconversion photosensitizers using another excitation light. Our study not only depicts the migration pathway of upconversion photosensitizers, but also demonstrates the organelle escape of these upconversion nanoparticles via PCI (photochemical internalization) process. It is believed that our results inspire more efficient synergistic therapy by combining PDT with other modalities in a programmable manner.


Asunto(s)
Nanopartículas , Intervención Coronaria Percutánea , Fotoquimioterapia , Rayos Infrarrojos , Nanopartículas/química , Fármacos Fotosensibilizantes/química
5.
Chembiochem ; 22(23): 3277-3282, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34519410

RESUMEN

Endosomal escape continues to be a limiting factor in the therapeutic use of nanomaterials. Assays to visualize endosomal escape often do not decouple the endosomal/lysosomal disruption from the release of payload into the cytosol. Here, we discuss three approaches to directly probe endosomal/lysosomal rupture: calcein dye dilution, lysosome size quantification and endosome/lysosome membrane integrity visualized with a genetically engineered cell line. We apply the three assays to endosomes/lysosomes ruptured via osmotic pressure and photochemical internalization.


Asunto(s)
Endosomas/química , Citosol/química , Fluoresceínas/química , Humanos , Lisosomas/química , Presión Osmótica , Procesos Fotoquímicos
6.
Lasers Med Sci ; 36(5): 1031-1038, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33123852

RESUMEN

Fibrin glue (FG) has potential as a delivery vehicle for photosensitizer directly to the resection cavity, so it may bypass the blood-brain barrier (BBB) and increase the concentration of successfully delivered photosensitizer. A specialized form of photodynamic therapy (PDT), photochemical internalization (PCI), which involves both photosensitizer and chemotherapeutic agent internalization, can locally inhibit the growth of cells. This will allow the reduction of recurrence of malignant gliomas around surgical resection. This study will look at the efficacy of FG loaded with drugs in mediating both PDT and PCI in inhibiting 3-dimensional tumor spheroid growth in vitro. Experiments were conducted on spheroids comprised of F98 glioma cells using photosensitizer AlPcS2a and chemotherapeutic drug bleomycin (BLM). At 2-, 24-, 48-, and 72-h increments, supernatant covering an FG layer within a well was collected and replaced by fresh medium, then added to spheroid-containing wells, which contained the respective chemicals for PDT and PCI. The wells were then exposed to light treatment from a diode laser, and after, spheroid growth was monitored for a period of 14 days. Significant spheroid growth inhibition was observed in both PDT and PCI modalities, but was far greater in PCI. Additionally, complete growth suppression was achieved via PCI at the highest radiant exposure. Achieving a slow photosensitizer release, significant F98 spheroid inhibition was observed in FG-mediated PDT and PCI. The present study showed BLM-PCI was the most efficacious of the two modalities.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Portadores de Fármacos/química , Adhesivo de Tejido de Fibrina/química , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Transporte Biológico , Bleomicina/química , Bleomicina/metabolismo , Bleomicina/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular Tumoral , Humanos , Indoles/química , Indoles/metabolismo , Indoles/farmacología , Láseres de Semiconductores , Compuestos Organometálicos/química , Compuestos Organometálicos/metabolismo , Compuestos Organometálicos/farmacología , Fármacos Fotosensibilizantes/farmacología
7.
Lasers Med Sci ; 36(8): 1567-1571, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34185168

RESUMEN

Photochemical internalization (PCI) is a modified form of photodynamic therapy (PDT) that enhances the efficacy of therapeutic agents in a site and temporal specific manner in both in vitro and in vivo publications. The purpose of the study reported here was to evaluate the benefits of a modified PCI protocol in a 3D rat glioma spheroid model. In the modified protocol, F98 glioma cells were incubated with photosensitizer (AlPcS2a) prior to spheroid generation, as opposed to post-spheroid formation photosensitizer exposure commonly used in conventional protocols. The efficacy of both bleomycin and doxorubicin PCI was evaluated using either the conventional or modified protocols. The formed spheroids were then exposed to light treatment from a diode laser, λ= 670 nm. Spheroid growth was monitored for a period of 14 days. The results of spheroid growth assays showed that there was no statistically significant difference in PCI efficacy between the conventional and modified protocols for both of the drugs tested. The direct PDT effect was significantly reduced using the modified protocol. Therefore, due to its several advantages, the modified protocol is recommended for evaluating the efficacy of PCI in tumor spheroid models.


Asunto(s)
Glioma , Fotoquimioterapia , Animales , Bleomicina/uso terapéutico , Línea Celular Tumoral , Glioma/tratamiento farmacológico , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Ratas
8.
Nano Lett ; 20(5): 3039-3049, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32250633

RESUMEN

Combination of chemotherapy and gene therapy provides an effective strategy for cancer treatment. However, the lack of suitable codelivery systems with efficient endo/lysosomal escape and controllable drug release/gene unpacking is the major bottleneck for maximizing the combinational therapeutic efficacy. In this work, we developed a photoactivatable Pt(IV) prodrug-backboned polymeric nanoparticle system (CNPPtCP/si(c-fos)) for light-controlled si(c-fos) delivery and synergistic photoactivated chemotherapy (PACT) and RNA interference (RNAi) on platinum-resistant ovarian cancer (PROC). Upon blue-light irradiation (430 nm), CNPPtCP/si(c-fos) generates oxygen-independent N3• with mild oxidation energy for efficient endo/lysosomal escape through N3•-assisted photochemical internalization with less gene deactivation. Thereafter, along with Pt(IV) prodrug activation, CNPPtCP/si(c-fos) dissociates to release active Pt(II) and unpack si(c-fos) simultaneously. Both in vitro and in vivo results demonstrated that CNPPtCP/si(c-fos) displayed excellent synergistic therapeutic efficacy on PROC with low toxicity. This PACT prodrug-backboned polymeric nanoplatform may provide a promising gene/drug codelivery tactic for treatment of various hard-to-tackle cancers.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias Ováricas , Profármacos , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Femenino , Terapia Genética , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Fotoquimioterapia , Platino (Metal) , Polímeros/uso terapéutico , Profármacos/uso terapéutico
9.
Molecules ; 26(1)2020 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-33374732

RESUMEN

Endosomal escape in cell-penetrating peptide (CPP)-based drug/macromolecule delivery systems is frequently insufficient. The CPP-fused molecules tend to remain trapped inside endosomes and end up being degraded rather than delivered into the cytosol. One of the methods for endosomal escape of CPP-fused molecules is photochemical internalization (PCI), which is based on the use of light and a photosensitizer and relies on photoinduced endosomal membrane destabilization to release the cargo molecule. Currently, it remains unclear how this delivery strategy behaves after photostimulation. Recent findings, including our studies using CPP-cargo-photosensitizer conjugates, have shed light on the photoinduced endosomal escape mechanism. In this review, we discuss the structural design of CPP-photosensitizer and CPP-cargo-photosensitizer conjugates, and the PCI mechanism underlying their application.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Sistemas de Liberación de Medicamentos , Endosomas/metabolismo , Sustancias Macromoleculares/química , Fotoquimioterapia , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Péptidos de Penetración Celular/química , Endocitosis , Humanos , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/metabolismo
10.
Mol Pharm ; 16(4): 1633-1647, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30817164

RESUMEN

In cancer treatment, polymeric nanoparticles (NPs) can serve as a vehicle for the delivery of cytotoxic proteins that have intracellular targets but that lack well-defined mechanisms for cellular internalization, such as saporin. In this work, we have prepared PEGylated poly(lactic acid- co-glycolic acid- co-hydroxymethyl glycolic acid) (PLGHMGA) NPs for the selective delivery of saporin in the cytosol of HER2 positive cancer cells. This selective uptake was achieved by decorating the surface of the NPs with the 11A4 nanobody that is specific for the HER2 receptor. Confocal microscopy observations showed rapid and extensive uptake of the targeted NPs (11A4-NPs) by HER2 positive cells (SkBr3) but not by HER2 negative cells (MDA-MB-231). This selective uptake was blocked upon preincubation of the cells with an excess of nanobody. Nontargeted NPs (Cys-NPs) were not taken up by either type of cells. Importantly, a dose-dependent cytotoxic effect was only observed on SkBr3 cells when these were treated with saporin-loaded 11A4-NPs in combination with photochemical internalization (PCI), a technique that uses a photosensitizer and local light exposure to facilitate endosomal escape of entrapped nanocarriers and biomolecules. The combined use of saporin-loaded 11A4-NPs and PCI strongly inhibited cell proliferation and decreased cell viability through induction of apoptosis. Also the cytotoxic effect could be reduced by an excess of nanobody, reinforcing the selectivity of this system. These results suggest that the combination of the targeting nanobody on the NPs with PCI are effective means to achieve selective uptake and cytotoxicity of saporin-loaded NPs.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Nanopartículas/administración & dosificación , Polímeros/química , Receptor ErbB-2/metabolismo , Saporinas/administración & dosificación , Anticuerpos de Dominio Único/administración & dosificación , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Nanopartículas/química , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Poliésteres/química , Saporinas/química , Anticuerpos de Dominio Único/inmunología , Células Tumorales Cultivadas
11.
Nanomedicine ; 19: 49-57, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31028888

RESUMEN

Herein, a novel nano-system IF7-ROSPCNP, which is O2-evolving and reactive oxygen species (ROS)-activable, was developed for enhancing the combination chemotherapy and photodynamic therapy (PDT). The nanoparticles composed of photosensitizers (disulfonated meso-tetraphenylporphine, TPPS2a) and catalase in the inner core, doxorubicin (DOX) in the polymeric shell and functionalized on its surface with IF7 peptide, which specially bind to annexin 1. As confirmed that the structure of IF7-ROSPCNP was able to remain intact under normal physiological conditions. After IF7-ROSPCNP was selectively entrapped by the annexin 1-positive and ROS-abundant MCF-7/ADR cells, the shell of nanoparticles was ruptured and the entrapped photosensitizers were completely released out. Under irradiation, ROS was continuously produced and the DOX, which was conjugated to the terminal of polylactic copolymer (mPEG-PLA) by a ROS-cleavable linkage, was subsequently released. With such strategy, cellular uptake of drugs was dramatically improved resulted in an enhanced cytotoxicity and anti-tumor effect on drug resistant cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Oxígeno/metabolismo , Fotoquimioterapia , Especies Reactivas de Oxígeno/metabolismo , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Doxorrubicina/uso terapéutico , Liberación de Fármacos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Endocitosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Células MCF-7 , Ratones , Nanopartículas/ultraestructura , Neoplasias/patología , Poliésteres/síntesis química , Poliésteres/química , Polietilenglicoles/síntesis química , Polietilenglicoles/química , Espectroscopía de Protones por Resonancia Magnética , Distribución Tisular/efectos de los fármacos , Carga Tumoral/efectos de los fármacos
12.
Pharm Dev Technol ; 24(4): 513-520, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30136636

RESUMEN

The efficacy of surfactant mixtures of Pluronic® F127 and Tween 80 at overall concentration in the micromolar range and molar ratio 1:1, 1:10, and 10:1 in inhibiting aggregation of the photosensitizer meso-tetraphenyl chlorin disulphonate (TPCS2a) was investigated in aqueous media at pH 2.9 by means of steady-state absorption and fluorescence emission spectroscopy as well as time-resolved fluorescence analysis. Corresponding experiments were performed at pH 7.4 in the absence of surfactants to determine the spectroscopic properties of a monomeric sample. Aggregation resulted in a red shift of the Soret absorption band and in substantial fluorescence quenching. The fluorescence lifetime of TPCS2a was a particularly sensitive indicator of the aggregation state, as the monomer at pH 7.4 decayed with a ∼ 10 ns time constant, while aggregation resulted in subnanosecond decay. The critical micelle concentration (CMC) of the surfactant mixtures was determined spectrophotometrically in the presence of TPCS2a. The ability of the surfactant mixtures to prevent aggregation at acidic pH was evaluated at overall surfactant concentration below and above CMC. Solubilization of TPCS2a in Pluronic® F127/Tween 80 mixtures prevented aggregation of the photosensitizer at overall surfactant concentrations much lower than those needed for both pure Pluronic® F127 and pure Tween 80.


Asunto(s)
Poloxámero/química , Polisorbatos/química , Porfirinas/química , Poloxámero/análisis , Polisorbatos/análisis , Porfirinas/análisis , Solubilidad , Espectrometría de Fluorescencia/métodos
13.
Lasers Med Sci ; 33(8): 1747-1755, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29802587

RESUMEN

Light-based treatment modalities such as photothermal therapy (PTT) or photochemical internalization (PCI) have been well documented both experimentally and clinically to enhance the efficacy of chemotherapy. The main purpose of this study was to examine the cytotoxic effects of silica-gold nanoshell (AuNS)-loaded macrophage-mediated (MaNS) PTT and bleomycin BLM-PCI on monolayers of squamous cell carcinoma cells. The two modalities were applied separately and in simultaneous combination. Two different wavelengths of light were employed simultaneously, one to activate a highly efficient PCI photosensitizer, AlPcS2a (670 nm) and the other for the MaNS-mediated PTT (810 nm), to evaluate the combined effects of these modalities. The results clearly demonstrated that macrophages could ingest sufficient numbers of silica-gold nanoshells for efficient near infrared (NIR) activated PTT. A significant synergistic effect of simultaneously applied combined PTT and PCI, compared to each modality applied separately, was achieved. Light-driven therapies have the advantage of site specificity, non-invasive and non-toxic application, require inexpensive equipment and can be given as repetitive treatment protocols.


Asunto(s)
Hipertermia Inducida , Macrófagos/metabolismo , Fototerapia , Animales , Bleomicina , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Línea Celular Tumoral , Endocitosis , Oro/química , Humanos , Nanocáscaras/química , Fármacos Fotosensibilizantes/farmacología , Polietilenglicoles/química , Ratas
14.
Mol Pharm ; 14(5): 1760-1770, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28296410

RESUMEN

In this study, the photochemical internalization (PCI) technique was adopted in a nanoparticle-based antigen delivery system to enhance antigen-specific CD8+ T cell immune response for cancer immunotherapy. Pheophorbide A, a hydrophobic photosensitizer, grafted with polyethylenimine (PheoA-PEI) with endosome escape activity and near-infrared imaging capability was prepared. A model antigen ovalbumin (OVA) was then complexed with PheoA-PEI to form PheoA-PEI/OVA nanoparticles (PheoA-PEI/OVA NPs) that are responsive to light. Flow cytometry analysis revealed increased endocytosis in a murine dendritic cell line (DC2.4) that was treated with PheoA-PEI/OVA NPs compared to free OVA. Generation of reactive oxygen species (ROS) in DC2.4 cells was also confirmed quantitatively and qualitatively using 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA). Confocal laser scanning microscopy (CLSM) further demonstrated that the PheoA-PEI/OVA NPs enhanced cytosolic antigen release after light stimulation. Moreover, PheoA-PEI/OVA NP treated DC2.4 cells exhibited enhanced cross-presentation to B3Z T cell hybridoma in vitro after light irradiation, substantially increased compared to those treated with free OVA. Consistently, in vivo results revealed upregulation of CD3+CD8+T lymphocytes in tumors of mice treated with dendritic cells plus PheoA-PEI/OVA NPs and light irradiation. The activated T cell response is partly responsible for the inhibitory effect on E.G7 tumor growth in mice immunized with dendritic cells plus PheoA-PEI/OVA NPs and light irradiation. Our results demonstrate the feasibility to enhance antigen-specific CD8+ T cell immune response by light-responsive nanoparticle-based vaccine delivery for cancer immunotherapy.


Asunto(s)
Clorofila/análogos & derivados , Células Dendríticas/metabolismo , Inmunoterapia/métodos , Nanopartículas/química , Polietileneimina/química , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Línea Celular Tumoral , Clorofila/química , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Especies Reactivas de Oxígeno/metabolismo , Oxígeno Singlete/metabolismo
15.
J Exp Ther Oncol ; 12(2): 113-120, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29161778

RESUMEN

INTRODUCTION: Photochemical Internalization is a novel drug delivery technology for cancer treatment based on the principle of Photodynamic Treatment. Using a photosensitizer that locates in endocytic vesicles membranes of tumor cells, Photochemical internalization enables cytosolic release of endocytosed antitumor agents in a site-specific manner. The purpose of the present in-vitro study was to explore whether Photochemical Internalization is able to enhance the efficacy of Ranpirnase, a cytotoxic amphibian ribonuclease, for eradication of squamous cell carcinoma of the head and neck. METHODS: Cell viability was measured in 8 primary human cell lines of squamous cell carcinoma of the head and neck after treatment with Ranpirnase and Photochemical Internalization. For Photochemical Internalization the photosensitizer disulfonated tetraphenyl porphine was incubated with tumor cells followed by exposure to blue light (435 nm). RESULTS: Our study demonstrates significant enhancement of antitumor activity of Ranpirnase by Photochemical Internalization. Treatment responses were heterogeneous between the primary cancer cell lines. Combining Photochemical Internalization with Ranpirnase resulted in 4.6 to 1,940-fold increased cytotoxicity when compared with the ribonuclease alone (P < 0.05). CONCLUSION: Cytotoxicity of Ranpirnase can be markedly enhanced by Photochemical Internalization in squamous cell carcinoma of the head and neck.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Fotoquimioterapia/métodos , Ribonucleasas/uso terapéutico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Fotoquímica , Carcinoma de Células Escamosas de Cabeza y Cuello
16.
Mol Pharm ; 13(2): 320-9, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26704885

RESUMEN

One of the greatest pharmaceutical challenges in vaccinology is the delivery of antigens to the cytosol of antigen-presenting cells (APCs) in order to allow for the stimulation of major histocompatibility complex (MHC) class I-restricted CD8(+) T-cell responses, which may act on intracellular infections or cancer. Recently, we described a novel method for cytotoxic T-lymphocyte (CTL) vaccination by combining antigens with a photosensitizer and light for cytosolic antigen delivery. The goal of the current project was to test this immunization method with particle-based formulations. Liposomes were prepared from dipalmitoylphosphatidylcholine and cholesterol, and the antigen ovalbumin (OVA) or the photosensitizer tetraphenyl chlorine disulfonate (TPCS2a) was separately encapsulated. C57BL/6 mice were immunized intradermally with OVA liposomes or a combination of OVA and TPCS2a liposomes, and light was applied the next day for activation of the photosensitizer resulting in cytosolic release of antigen from phagosomes. Immune responses were tested both after a prime only regime and after a prime-boost scheme with a repeat immunization 2 weeks post priming. Antigen-specific CD8(+) T-cell responses and antibody responses were analyzed ex vivo by flow cytometry and ELISA methods. The physicochemical stability of liposomes upon storage and light exposure was analyzed in vitro. Immunization with both TPCS2a- and OVA-containing liposomes greatly improved CD8(+) T-cell responses as compared to immunization without TPCS2a and as measured by proliferation in vivo and cytokine secretion ex vivo. In contrast, OVA-specific antibody responses (IgG1 and IgG2c) were reduced after immunization with TPCS2a-containing liposomes. The liposomal formulation protected the photosensitizer from light-induced inactivation during storage. In conclusion, the photosensitizer TPCS2a was successfully formulated in liposomes and enabled a shift from MHC class II to MHC class I antigen processing and presentation for stimulation of strong CD8(+) T-cell responses. Therefore, photosensitive particulate vaccines may have the potential to add to current vaccine practice a new method of vaccination that, as opposed to current vaccines, can stimulate strong CD8(+) T-cell responses.


Asunto(s)
Citosol/inmunología , Liposomas/administración & dosificación , Fagosomas/inmunología , Porfirinas/administración & dosificación , Linfocitos T Citotóxicos/inmunología , Vacunas/administración & dosificación , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Citosol/efectos de los fármacos , Citosol/metabolismo , Femenino , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I , Inmunización , Inyecciones Intradérmicas , Liposomas/química , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Fagosomas/efectos de los fármacos , Fagosomas/efectos de la radiación , Fármacos Fotosensibilizantes/administración & dosificación , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/efectos de la radiación
17.
Biochim Biophys Acta ; 1840(1): 507-15, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24135456

RESUMEN

BACKGROUND: Fluorescently labeled cell-penetrating peptides can translocate into cells by endocytosis and upon light irradiation, lyse the endocytic vesicles. This photo-inducible endosomolytic activity of Fl-CPPs can be used to efficiently deliver macromolecules such as proteins and nucleic acids and other small organic molecules into the cytosol of live cells. The requirement of a light trigger to induce photolysis provides a more spatial and temporal control to the intracellular delivery process. METHODS: In this report, we examine the molecular level mechanisms by which cell-penetrating peptides such as TAT when labeled with small organic fluorophore molecules acquire a photo-induced lytic activity using a simplified model of lipid vesicles. RESULTS: The peptide TAT labeled with 5(6)-carboxytetramethylrhodamine binds to negatively charged phospholipids, thereby bringing the fluorophore in close proximity to the membrane of liposomes. Upon light irradiation, the excited fluorophore produces reactive oxygen species at the lipid bilayer and oxidation of the membrane is achieved. In addition, the fluorescent peptide causes aggregation of photo-oxidized lipids, an activity that requires the presence of arginine residues in the peptide sequence. CONCLUSIONS: These results suggest that the cell-penetrating peptide plays a dual role. On one hand, TAT targets a conjugated fluorophore to membranes. On the other hand, TAT participates directly in the destabilization of photosensitized membranes. Peptide and fluorophore therefore appear to act in synergy to destroy membranes efficiently. GENERAL SIGNIFICANCE: Understanding the mechanism behind Fl-CPP mediated membrane photodamage will help to design optimally photo-endosomolytic compounds.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Péptidos de Penetración Celular/farmacología , Luz/efectos adversos , Membrana Dobles de Lípidos/química , Fármacos Fotosensibilizantes/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Membrana Celular/efectos de la radiación , Permeabilidad de la Membrana Celular/efectos de la radiación , Péptidos de Penetración Celular/química , Endocitosis , Colorantes Fluorescentes , Humanos , Membrana Dobles de Lípidos/efectos de la radiación , Liposomas , Fotólisis , Especies Reactivas de Oxígeno/metabolismo , Rodaminas/química , Rodaminas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
18.
Small ; 11(22): 2607-13, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-25641862

RESUMEN

A micrometer-sized affinity bead (red) is (i) taken up into a cell by phagocytosis, (ii) photochemically released from phagosomes, (iii) optically trapped by the cell, and (iv) isolated by cell lysis for subsequent analysis of captured intracellular analyte (green).


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Inmunoensayo/métodos , Microfluídica/métodos , Pinzas Ópticas , Células HEK293 , Humanos
19.
Angew Chem Int Ed Engl ; 54(16): 4885-9, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25663536

RESUMEN

Photochemical internalization (PCI) has shown great promise as a therapeutic alternative for targeted drug delivery by light-harnessed activation. However, it has only been applicable to therapeutic macromolecules or medium-sized molecules. Herein we describe the use of an amphiphilic, water-soluble porphyrin-ß-cyclodextrin conjugate (mTHPP-ßCD) as a "Trojan horse" to facilitate the endocytosis of CD-guest tamoxifens into breast-cancer cells. Upon irradiation, the porphyrin core of mTHPP-ßCD expedited endosomal membrane rupture and tamoxifen release into the cytosol, as documented by confocal microscopy. The sustained complexation of mTHPP-ßCD with tamoxifen was corroborated by 2D NMR spectroscopy and FRET studies. Following the application of PCI protocols with 4-hydroxytamoxifen (4-OHT), estrogen-receptor ß-positive (Erß+, but not ERß-) cell groups exhibited extensive cytotoxicity and/or growth suspension even at 72 h after irradiation.


Asunto(s)
Portadores de Fármacos/química , Nanoconjugados/química , Tamoxifeno/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia , Humanos , Luz , Células MCF-7 , Espectroscopía de Resonancia Magnética , Microscopía Confocal , Porfirinas/química , Tamoxifeno/toxicidad , beta-Ciclodextrinas/química
20.
Small ; 10(4): 782-92, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24031023

RESUMEN

The interaction of Tat-conjugated PEGylated CdSe/ZnS quantum dots (QD) with the amphiphilic disulfonated aluminium phthalocyanine photosensitiser is investigated in aqueous solution and in a human breast cancer cell line. In aqueous solution, the QDs and phthalocyanine form stable nanocomposites. Using steady-state and time-resolved fluorescence measurements combined with singlet oxygen detection, efficient Förster resonance energy transfer (FRET) is observed with the QDs acting as donors, and the phthalocyanine photosensitiser, which mediates production of singlet oxygen, as acceptors. In cells, the Tat-conjugated QDs localise in lysosomes and the QD fluorescence lifetimes are close to values observed in aqueous solution. Strong FRET-induced quenching of the QD lifetime is observed in cells incubated with the nanocomposites using fluorescence lifetime imaging microscopy (FLIM). Using excitation of the QDs at wavelengths where phthalocyanine absorption is negligible, FRET-induced release of QDs from endo/lysosomes is confirmed using confocal imaging and FLIM, which is attributed to photooxidative damage to the endo/lysosomal membranes mediated by the phthalocyanine acceptor.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Indoles/farmacología , Espacio Intracelular/metabolismo , Nanopartículas/química , Imagen Óptica/métodos , Fármacos Fotosensibilizantes/farmacología , Puntos Cuánticos/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Isoindoles , Células MCF-7 , Microscopía Fluorescente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA