Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 28(3)2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36770830

RESUMEN

Superparamagnetic iron oxide nanoparticles (SPIONs) may act as an excellent theragnostic tool if properly coated and stabilized in a biological environment, even more, if they have targeting properties towards a specific cellular target. Humanized Archaeoglobus fulgidus Ferritin (HumAfFt) is an engineered ferritin characterized by the peculiar salt-triggered assembly-disassembly of the hyperthermophile Archaeoglobus fulgidus ferritin and is successfully endowed with the human H homopolymer recognition sequence by the transferrin receptor (TfR1 or CD71), overexpressed in many cancer cells in response to the increased demand of iron. For this reason, HumAfFt was successfully used in this study as a coating material for 10 nm SPIONs, in order to produce a new magnetic nanocarrier able to discriminate cancer cells from normal cells and maintain the potential theragnostic properties of SPIONs. HumAfFt-SPIONs were exhaustively characterized in terms of size, morphology, composition, and cytotoxicity. The preferential uptake capacity of cancer cells toward HumAfFt-SPIONs was demonstrated in vitro on human breast adenocarcinoma (MCF7) versus normal human dermal fibroblast (NHDF) cell lines.


Asunto(s)
Nanopartículas de Magnetita , Neoplasias , Humanos , Ferritinas , Línea Celular , Nanopartículas Magnéticas de Óxido de Hierro , Fenómenos Magnéticos
2.
Chemistry ; 28(43): e202104618, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35604769

RESUMEN

A transfection vector that can home in on tumors is reported. Whereas previous vectors that allow moderately cell selective gene transfection used larger systems, this small-molecule approach paved the way for precise structure-activity relationship optimization. For this, biotin, which mediates cell selectivity, was combined with the potent DNA-binding motif tetralysine-guanidinocarbonypyrrol via a hydrophilic linker, thus enabling SAR-based optimization. The new vector mediated biotin receptor (BR)-selective transfection of cell lines with different BR expression levels. Computer-based analyses of microscopy images revealed a preference of one order of magnitude for the BR-positive cell lines over the BR-negative controls.


Asunto(s)
Vectores Genéticos , Neoplasias , Biotina/metabolismo , Línea Celular , Humanos , Transfección
3.
Bioorg Med Chem ; 46: 116361, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34411983

RESUMEN

Epidermal growth factor receptor (EGFR) is overexpressed in many cancers and therefore serves as an excellent target for prodrug activation. Functionalised trans-cyclooctenes (TCO) were conjugated to an EGFR antibody (cetuximab), providing a reagent for pre-targeting and localisation of the bioorthogonal reagent. The TCOs react with a 4-azidobenzyl carbamate doxorubicin prodrug via a [3 + 2]-cycloaddition and subsequent self-immolation leads to release of doxorubicin (click-and-release). In vitro cell-based assays demonstrated proof-of-concept, that cetuximab conjugated to highly strained TCO (AB-d-TCO) could bind to the EGFR in a melanoma cell line, and selectively activate the doxorubicin prodrug. In a non-EGFR expressing melanoma cell line, no significant prodrug activation was observed. In vivo experiments using this combination of AB-d-TCO and the azido-doxorubicin prodrug in a murine melanoma model revealed no significant anti-tumour activity or increased survival, suggesting there was insufficient prodrug activation and drug release at the tumour site.


Asunto(s)
Alquenos/farmacología , Antibióticos Antineoplásicos/farmacología , Azidas/farmacología , Doxorrubicina/farmacología , Profármacos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Alquenos/química , Animales , Antibióticos Antineoplásicos/síntesis química , Antibióticos Antineoplásicos/química , Azidas/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/síntesis química , Doxorrubicina/química , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Profármacos/síntesis química , Profármacos/química , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
4.
Cytometry A ; 97(3): 279-287, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31809002

RESUMEN

Understanding the intricacies of particle-cell interactions is essential for many applications such as imaging, phototherapy, and drug/gene delivery, because it is the key to accurate control of the particle properties for the improvement of their therapeutic and diagnostic efficiency. Recently, high-throughput methods have emerged for the detailed investigation of these interactions. For example, imaging flow cytometry (IFC) collects up to 60,000 images of cells per second (in 12 optical channels) and provides information about morphology and organelle localization in combination with fluorescence and side scatter intensity data. However, analysis of IFC data is extremely difficult to perform using conventional methods that calculate integral parameters or use mask-based object recognition. Here, we show application of a convolutional neural network (CNN) for precise quantitative analysis of particle targeting of cells using IFC data. CNN provides high-throughput object detection with almost human precision but avoids the subjective choice of image processing parameters that often leads to incorrect data interpretation. The method allows accurate counting of cell-bound particles with reliable discrimination from the nonbound particles in the field of view. The proposed method expands capabilities of spot counting applications (such as organelle counting, quantification of cell-cell and cell-bacteria interactions) and is going to be useful not only for high-throughput analysis of IFC data but also for other imaging techniques. © 2019 International Society for Advancement of Cytometry.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Redes Neurales de la Computación , Diagnóstico por Imagen , Citometría de Flujo , Humanos
5.
Molecules ; 25(8)2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32340382

RESUMEN

The development of synthetic ways to fabricate nanosized materials with a well-defined shape, narrow-sized distribution, and high stability is of great importance to a rapidly developing area of nanotechnology. Here, we report an unusual reaction between amorphous two-line ferrihydrite and concentrated sulfuric or other mineral and organic acids. Instead of the expected dissolution, we observed the formation of new narrow-distributed brick-red nanoparticles (NPs) of hematite. Different acids produce similar nanoparticles according to scanning (SEM) and transmission electron microscopy (TEM), selected area electron diffraction (SAED), X-ray diffraction (XRD), infrared spectroscopy (FTIR), and energy-dispersive X-ray spectroscopy (EDX). The reaction demonstrates new possibilities for the synthesis of acid-resistant iron oxide nanoparticles and shows a novel pathway for the reaction of iron hydroxide with concentrated acids. The biomedical potential of the fabricated nanoparticles is demonstrated by the functionalization of the particles with polymers, fluorescent labels, and antibodies. Three different applications are demonstrated: i) specific targeting of the red blood cells, e.g., for red blood cell (RBC)-hitchhiking; ii) cancer cell targeting in vitro; iii) infrared ex vivo bioimaging. This novel synthesis route may be useful for the development of iron oxide materials for such specificity-demanding applications such as nanosensors, imaging, and therapy.


Asunto(s)
Ácidos/química , Materiales Biomédicos y Dentales , Compuestos Férricos/química , Nanopartículas Magnéticas de Óxido de Hierro/química , Materiales Biomédicos y Dentales/química , Humanos , Nanopartículas Magnéticas de Óxido de Hierro/ultraestructura , Análisis Espectral
6.
Mol Pharm ; 16(7): 3275-3283, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31120760

RESUMEN

The lack of specificity of traditional cytostatics and increasing resistance of cancer cells represent important challenges in cancer therapy. One of the characteristics of cancer cells is their intrinsic oxidative stress caused by higher metabolic activity, mitochondrial malfunction, and oncogene stimulation. This feature can be exploited in the pursuit of more selective cancer therapy, as there is increasing evidence that cancer cells are more sensitive to elevated concentrations of reactive oxygen species than normal cells. In this study, we demonstrate a new concept for cancer cell targeting by in situ production of radicals under physiological conditions. The biologically active radicals are produced in the milieu of cancer cells by enzymatic conversion from an inactive precursor, 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)diammonium salt, by using miniature bioreactors represented by cell-sized microgels containing immobilized laccase. We utilize the pH-dependent activity of laccase to generate radicals only at a lower pH (5.7-6.1) that is characteristic of the tumor microenvironment. The composition of the microgels was optimized so as to allow sufficient substrate and radical diffusion, high enzyme activity, and stability under physiological conditions. The functionality of this system was evaluated on three cancer cell lines (HeLa, HT-29, and DLD1) and the cytotoxicity of in situ-produced radicals was successfully proven in all cases. These results demonstrate that cancer cell targeting by in situ-generated radicals using miniature enzymatic reactors may represent an alternative to traditional cytostatics. In particular, the pH-dependence of radical generation and their short-lived nature can ensure localized functionality in the tumor microenvironment and thereby reduce systemic side-effects.


Asunto(s)
Reactores Biológicos , Microgeles/química , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , Microambiente Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HT29 , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Lacasa/metabolismo , Estrés Oxidativo , Tamaño de la Partícula
7.
Appl Microbiol Biotechnol ; 100(14): 6403-6413, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27063011

RESUMEN

Immunotoxins are a new class of antibody-targeted therapy in clinical development. Traditional immunotoxins that are constructed from the toxins of plants or bacteria need to be internalized to the cytoplasm and thus have limited antitumor efficacy. In the present study, we combined a recently reported sea anemone cytolysin Gigantoxin-4 with an anti-HER2/neu single-chain variable fragment 4D5 scFv to construct a novel immunotoxin. We fused a SUMO tag to the N-terminus of Gigantoxin-4-4D5 scFv and it was successfully expressed in Escherichia coli strain BL21 (DE3) in a soluble form. After purification, the purity of Gigantoxin-4-4D5 scFv reached 96 % and the yield was 14.3 mg/L. Our results demonstrated that Gigantoxin-4-4D5 scFv exerted a highly cytotoxic effect on the HER2/neu-positive ovarian carcinoma SK-OV-3 cell line. And the hemolytic activity was weaker, making it safe for normal cells. The results of immunofluorescence analysis showed that this novel immunotoxin could specifically bind to SK-OV-3 cells with no recognition of human embryonic kidney 293 cells. Scanning electron microscope observations and extracellular lactate dehydrogenase activity indicated that it could induce necrosis in SK-OV-3 cells by disrupting the cell membrane. Moreover, it could also mediate apoptosis of SK-OV-3 cells.


Asunto(s)
Venenos de Cnidarios/farmacología , Inmunotoxinas/farmacología , Neoplasias Ováricas/patología , Anticuerpos de Cadena Única/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunotoxinas/genética , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
8.
Biochem Biophys Res Commun ; 456(3): 768-73, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25498500

RESUMEN

Exosomes, the natural vehicles of various biological molecules, have been examined in several research fields including drug delivery. Although understanding of the biological functions of exosomes has increased, how exosomes are transported between cells remains unclear. We hypothesized that cell tropism is important for effective exosomal intercellular communication and that parental cells regulate exosome movement by modulating constituent exosomal molecules. Herein, we demonstrated the strong translocation of glioblastoma-derived exosomes (U251exo) into their parental (U251) cells, breast cancer (MDA-MB-231) cells, and fibrosarcoma (HT-1080). Furthermore, disruption of proteins of U251exo by enzymatic treatment did not affect their uptake. Therefore, we focused on lipid molecules of U251exo with the expectation that they are crucial for effective incorporation of U251exo by cancer cells. Phosphatidylethanolamine was identified as a unique lipid component of U251-MG cell-derived extracellular vesicles. From these results, valuable insight is provided into the targeting of U251exo to cancer cells, which will help to develop a cancer-targeted drug delivery system.


Asunto(s)
Sistemas de Liberación de Medicamentos , Exosomas/química , Exosomas/metabolismo , Neoplasias/metabolismo , Fosfatidiletanolaminas/análisis , Comunicación Celular , Línea Celular Tumoral , Humanos
9.
Int J Biol Macromol ; 275(Pt 2): 133738, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38992536

RESUMEN

Pancreatic cancer cells highly resistance to conventional chemo drugs, resulting low survival rates. The aim of the study was to design and develop dual targeting polymersomes (DTPS) loaded with phyto alkaloid agent i.e., piperlongumine (PL) for effective pancreatic cancer treatment. Here, hyaluronic acid (HA) was functionalized with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (DSPEPEG-NH2), poly(ethylene glycol) bis (amine) (PEG), and phenylboronic acid (PBA) moieties. The designed DTPS could selectively recognize CD44/sialic acid (SA) and deliver PL to MIA PaCa-2 pancreatic cancer cells, facilitated via HA-CD44 and PBA-SA interactions. Drug release and stability results implied sustained PL release profile and pH sensitivity. DTPS could be more efficiently bound with SA than other sugars based on fluorescence spectroscopy. The anticancer efficacy of designed polymersomes was tested with H6C7 normal pancreas cells and SA/CD44-overexpressed MIA PaCa-2 pancreatic cancer cells. DTPS showed both SA and CD44-mediated higher cellular uptake while single-targeted polymersomes showed CD44-mediated cellular uptake. The PL-loaded DTPS efficiently uptake by MIA PaCa-2 cancer cells, causing up to 80 % cell growth inhibition, reduced cell spheroids volume and increased dead cells by 58.3 %. These results indicate that the newly developed DTPS can effectively serve as a pH-responsive drug delivery system for efficient treatment of cancer.


Asunto(s)
Ácidos Borónicos , Dioxolanos , Ácido Hialurónico , Neoplasias Pancreáticas , Humanos , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Dioxolanos/farmacología , Dioxolanos/química , Línea Celular Tumoral , Ácidos Borónicos/química , Ácidos Borónicos/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Liberación de Fármacos , Receptores de Hialuranos/metabolismo , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Polímeros/química , Supervivencia Celular/efectos de los fármacos , Piperidonas
10.
Top Curr Chem (Cham) ; 382(1): 6, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38400859

RESUMEN

The kinetically inert, six coordinated, octahedral Pt(IV) complexes are termed dual-, triple-, or multi-action prodrugs based on the nature of the axially substituted ligands. These ligands are either inert or biologically active, where the nature of these axial ligands provides additional stability, synergistic biological activity or cell-targeting ability. There are many literature reports from each of these classes, mentioning the varied nature of these axial ligands. The ligands comprise drug molecules such as chlorambucil, doxorubicin, valproic acid, ethacrynic acid, biologically active chalcone, coumarin, combretastatin, non-steroidal anti-inflammatory drugs (NSAIDs) and many more, potentiating the anti-proliferative profile or reducing the side effects associated with cisplatin therapy. The targeting and non-targeting nature of these moieties exert additive or synergistic effects on the anti-cancer activity of Pt(II) moieties. Herein, we discuss the effects of these axially oriented ligands and the changes in the non-leaving am(m)ine groups and in the leaving groups on the biological activity. In this review, we have presented the latest developments in the field of Pt(IV) complexes that display promising activity with a reduced resistance profile. We have discussed the structure activity relationship (SAR) and the effects of the ligands on the biological activity of Pt(IV) complexes with cisplatin, oxaliplatin, carboplatin and the Pt core other than approved drugs. This literature work will help researchers to get an idea about Pt(IV) complexes that have been classified based on the aspects of their biological activity.


Asunto(s)
Antineoplásicos , Profármacos , Platino (Metal)/química , Cisplatino/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Profármacos/farmacología , Profármacos/química , Profármacos/uso terapéutico , Clorambucilo
11.
Small ; 9(22): 3834-44, 2013 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-23606568

RESUMEN

Nanosized drug carriers functionalized with moieties specifically targeting tumor cells are promising tools in cancer therapy, due to their ability to circulate in the bloodstream for longer periods and their selectivity for tumor cells, enabling the sparing of healthy tissues. Because of its biocompatibility, high bioresorbability, and responsiveness to pH changes, synthetic biomimetic nanocrystalline apatites are used as nanocarriers to produce multifunctional nanoparticles, by coupling them with the chemotherapeutic drug doxorubicin (DOXO) and the DO-24 monoclonal antibody (mAb) directed against the Met/Hepatocyte Growth Factor receptor (Met/HGFR), which is over-expressed on different types of carcinomas and thus represents a useful tumor target. The chemical-physical features of the nanoparticles are fully investigated and their interaction with cells expressing (GTL-16 gastric carcinoma line) or not expressing (NIH-3T3 fibroblasts) the Met/HGFR is analyzed. Functionalized nanoparticles specifically bind to and are internalized in cells expressing the receptor (GTL-16) but not in the ones that do not express it (NIH-3T3). Moreover they discharge DOXO in the targeted GTL-16 cells that reach the nucleus and display cytotoxicity as assessed in an MTT assay. Two different types of ternary nanoparticles are prepared, differing for the sequence of the functionalization steps (adsorption of DOXO first and then mAb or vice versa), and it is found that the ones in which mAb is adsorbed first are more efficient under all the examined aspects (binding, internalization, cytotoxicity), possibly because of a better mAb orientation on the nanoparticle surface. These multifunctional nanoparticles could thus be useful instruments for targeted local or systemic drug delivery, allowing a reduction in the therapeutic dose of the drug and thus adverse side effects. Moreover, this work opens new perspectives in the use of nanocrystalline apatites as a new platform for theranostic applications in nanomedicine.


Asunto(s)
Apatitas/química , Biomimética/métodos , Portadores de Fármacos/química , Nanopartículas/química , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Portadores de Fármacos/administración & dosificación , Humanos , Ratones , Células 3T3 NIH , Nanopartículas/administración & dosificación
12.
ACS Appl Bio Mater ; 6(2): 483-493, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36651801

RESUMEN

A nanodelivery system based on palladium nanoparticles (PdNP) and cisplatin (CisPt) was developed by physisorption of the drug onto the PdNP synthesized via a green redox process, using d-glucose and polyvinylpyrrolidone (PVP) as reducing and stabilizing/capping agents, respectively. UV-vis analysis and H2-evolution measurements were carried out to prove the nanoparticles' capability to act as bimodal theranostic nanomedicine, i.e., having both plasmonic and photocatalytic properties. XPS, XRD, and TEM allowed light to be shed on the chemical composition and morphology of the PdNP. The analysis of the UV-visible spectra evidenced plasmonic peak changes for the hybrid nanoparticle-drug assembly (Pd@CisPt), which pointed to a significant interaction of CisPt with the NP surface. The drug loading was quantitatively estimated by ICP-OES measurements, while DLS and AFM confirmed the strong association of the drug with the nanoparticle surface. The test of SOD-like activity in a cell-free environment proved the maintenance of the antioxidant capability of PdNP also in the Pd@CisPt systems. Finally, Pd@CisPt tested in prostate cancer cells (PC-3 line) unveiled the antitumoral action of the developed nanomedicine, related to reactive oxygen species (ROS) generation, with a condition of protein misfolding/unfolding and DNA damage, as evidenced by cytotoxicity and MitoSOX assays, as well as Raman microspectroscopy, respectively. Cell imaging by confocal microscopy evidenced cellular uptake of the nanoparticles, as well as dynamic processes of copper ion accumulation at the level of subcellular compartments. Finally, cell migration studies upon treatment with Pd@CisPt evidenced a tunable response between the inhibitory effect of CisPt and the enhanced rate of cell migration for the metal NP alone, which pointed out the promising potential of the developed theranostic nanomedicine in tissue regeneration.


Asunto(s)
Antineoplásicos , Nanopartículas del Metal , Masculino , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Nanopartículas del Metal/uso terapéutico , Nanopartículas del Metal/química , Nanomedicina Teranóstica/métodos , Paladio/farmacología , Paladio/química , Cisplatino/química
13.
Colloids Surf B Biointerfaces ; 227: 113357, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37210795

RESUMEN

Functionalized metal oxide nanoparticles (NPs) have demonstrated specific binding affinity to antigens or receptors presented on the cancer cell surface, favouring selective targeting and minimizing side effects during the chemotherapy. Placenta-specific protein 1 (PLAC-1) is a small cell surface protein overexpressed in certain types of breast cancer (BC); therefore, it can be used as a therapeutic target. The objective of this study is to develop NPs that can bind PLAC-1 and hence can inhibit the progression and metastatic potential of BC cells. Zinc oxide (ZnO) NPs were coated with a peptide (GILGFVFTL), which possesses a strong binding ability to PLAC-1. The physical attachment of the peptide to ZnO NPs was verified through various physicochemical and morphological characterization techniques. The selective cytotoxicity of the designed NPs was investigated using PLAC-1-bearing MDA-MB 231 human BC cell line and compared to LS-180 cells that do not express PLAC-1. The anti-metastatic and pro-apoptotic effects of the functionalized NPs on MDA-MB 231 cells were examined. Confocal microscopy was used to investigate the mechanism of NPs uptake by MDA-MB 231 cells. Compared to non-functionalized NPs, peptide functionalization significantly improved the targeting and uptake of the designed NPs by PLAC-1-expressing cancer cells with significant pro-apoptotic and anti-metastatic effects. The uptake of peptide functionalized ZnO NPs (ZnO-P NPs) occurred via peptide-PLAC1 interaction-assisted clathrin-mediated endocytosis. These findings highlight the potential targeted therapy of ZnO-P NPs against PLAC-1-expressing breast cancer cells.


Asunto(s)
Neoplasias de la Mama , Nanopartículas del Metal , Nanopartículas , Proteínas Gestacionales , Óxido de Zinc , Humanos , Femenino , Óxido de Zinc/farmacología , Óxido de Zinc/química , Línea Celular Tumoral , Neoplasias de la Mama/tratamiento farmacológico , Nanopartículas/química , Nanopartículas del Metal/química , Péptidos/farmacología
14.
Acta Pharm Sin B ; 13(3): 1303-1317, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36970207

RESUMEN

In situ and real-time monitoring of responsive drug release is critical for the assessment of pharmacodynamics in chemotherapy. In this study, a novel pH-responsive nanosystem is proposed for real-time monitoring of drug release and chemo-phototherapy by surface-enhanced Raman spectroscopy (SERS). The Fe3O4@Au@Ag nanoparticles (NPs) deposited graphene oxide (GO) nanocomposites with a high SERS activity and stability are synthesized and labeled with a Raman reporter 4-mercaptophenylboronic acid (4-MPBA) to form SERS probes (GO-Fe3O4@Au@Ag-MPBA). Furthermore, doxorubicin (DOX) is attached to SERS probes through a pH-responsive linker boronic ester (GO-Fe3O4@Au@Ag-MPBA-DOX), accompanying the 4-MPBA signal change in SERS. After the entry into tumor, the breakage of boronic ester in the acidic environment gives rise to the release of DOX and the recovery of 4-MPBA SERS signal. Thus, the DOX dynamic release can be monitored by the real-time changes of 4-MPBA SERS spectra. Additionally, the strong T2 magnetic resonance (MR) signal and NIR photothermal transduction efficiency of the nanocomposites make it available for MR imaging and photothermal therapy (PTT). Altogether, this GO-Fe3O4@Au@Ag-MPBA-DOX can simultaneously fulfill the synergistic combination of cancer cell targeting, pH-sensitive drug release, SERS-traceable detection and MR imaging, endowing it great potential for SERS/MR imaging-guided efficient chemo-phototherapy on cancer treatment.

15.
Pharmaceutics ; 14(1)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35057088

RESUMEN

The sustained release of small, tumor-targeted cytotoxic drugs is an unmet need in cancer therapies, which usually rely on punctual administration regimens of non-targeted drugs. Here, we have developed a novel concept of protein-drug nanoconjugates, which are packaged as slow-releasing chemically hybrid depots and sustain a prolonged secretion of the therapeutic agent. For this, we covalently attached hydrophobic molecules (including the antitumoral drug Monomethyl Auristatin E) to a protein targeting a tumoral cell surface marker abundant in several human neoplasias, namely the cytokine receptor CXCR4. By this, a controlled aggregation of the complex is achieved, resulting in mechanically stable protein-drug microparticles. These materials, which are mimetics of bacterial inclusion bodies and of mammalian secretory granules, allow the slow leakage of fully functional conjugates at the nanoscale, both in vitro and in vivo. Upon subcutaneous administration in a mouse model of human CXCR4+ lymphoma, the protein-drug depots release nanoconjugates for at least 10 days, which accumulate in the tumor with a potent antitumoral effect. The modification of scaffold cell-targeted proteins by hydrophobic drug conjugation is then shown as a novel transversal platform for the design of slow releasing protein-drug depots, with potential application in a broad spectrum of clinical settings.

16.
Adv Drug Deliv Rev ; 180: 114044, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34774552

RESUMEN

Extensive research has been undertaken in the pursuit of anticancer therapeutics. Many anticancer drugs require specificity of delivery to cancer cells, whilst sparing healthy tissue. Cell-penetrating peptides (CPPs), now well established as facilitators of intracellular delivery, have in recent years advanced to incorporate target specificity and thus possess great potential for the targeted delivery of anticancer cargoes. Though none have yet been approved for clinical use, this novel technology has already entered clinical trials. In this review we present CPPs, discuss their classification, mechanisms of cargo internalization and highlight strategies for conjugation to anticancer moieties including their incorporation into therapeutic proteins. As the mainstay of this review, strategies to build specificity into tumor targeting CPP constructs through exploitation of the tumor microenvironment and the use of tumor homing peptides are discussed, whilst acknowledging the extensive contribution made by CPP constructs to target specific protein-protein interactions integral to intracellular signaling pathways associated with tumor cell survival and progression. Finally, antibody/antigen CPP conjugates and their potential roles in cancer immunotherapy and diagnostics are considered. In summary, this review aims to harness the potential of CPP-aided drug delivery for future cancer therapies and diagnostics whilst highlighting some of the most recent achievements in selective delivery of anticancer drugs, including cytostatic drugs, to a range of tumor cells both in vitro and in vivo.


Asunto(s)
Antineoplásicos/administración & dosificación , Péptidos de Penetración Celular/administración & dosificación , Sistemas de Liberación de Medicamentos , Animales , Antineoplásicos/farmacología , Péptidos de Penetración Celular/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas/metabolismo , Microambiente Tumoral
17.
J Colloid Interface Sci ; 607(Pt 1): 488-501, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34509120

RESUMEN

HYPOTHESIS: Naturally derived or synthetic anticancer peptides (ACPs) have emerged as a new generation of anticancer agents with higher selectivity for cancer cells and less propensity for drug resistance. Despite the structural diversity of ACPs, α-helix is the most common secondary structure among them. Herein we report the development of a new library of short cationic amphiphilic α-helical ACPs with selective cytotoxicity against colorectal and cervical cancer. EXPERIMENTS: The peptides had a general formula C(XXYY)3 with C representing amino acid cysteine (providing a -SH group for molecular conjugation), X representing hydrophobic amino acids (isoleucine (I) or leucine (L)), and Y representing cationic amino acids (arginine (R) or lysine (K)). Two variants of the peptides were synthesized by adding additional Isoleucine residues to the C-terminal and replacing the N-terminal cysteine with LC-propargylglycine (LC-G) to investigate the effect of N-terminal and C-terminal variation on the anticancer activity. The structure and physicochemical properties of the peptides were determined by RP-HPLC, LC-MS and CD spectroscopy. The cytotoxicity of the peptides in different cell lines was assessed by MTT test, cell proliferation assay and mitochondrial damage assay. The mechanism of cell selectivity of the peptides was investigated by studying their interfacial behaviour at the air/water and lipid/water interface using Langmuir trough. FINDINGS: The peptides consisting of K residues in their hydrophilic domains exhibited more selective anticancer activity whereas the peptides containing R exhibited strong toxicity in normal cells. The anticancer activity of the peptides was a function of their helical content and their hydrophobicity. Therefore, the addition of two I residues at C-terminal enhanced the anticancer activity of the peptides by increasing their hydrophobicity and their helical content. These two variants also exhibited strong anticancer activity against colorectal cancer multicellular tumour spheroids (MCTS). The higher toxicity of the peptides in cancer cells compared to normal cells was the result of higher penetration into the negatively charged cancer cell membranes, leading to higher cellular uptake, and their cytotoxic effect was mainly exerted by damaging the mitochondrial membranes leading to apoptosis. The results from this study provide a basis for rational design of new α-helical ACPs with enhanced anticancer activity and selectivity.


Asunto(s)
Péptidos , Cationes , Dicroismo Circular , Interacciones Hidrofóbicas e Hidrofílicas , Péptidos/farmacología , Conformación Proteica en Hélice alfa , Estructura Secundaria de Proteína
18.
Toxins (Basel) ; 13(11)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34822576

RESUMEN

A chimeric, bispecific Janus lectin has recently been engineered with different, rationally oriented recognition sites. It can bind simultaneously to sialylated and fucosylated glycoconjugates. Because of its multivalent architecture, this lectin reaches nanomolar avidities for sialic acid and fucose. The lectin was designed to detect hypersialylation-a dysregulation in physiological glycosylation patterns, which promotes the tumor growth and progression of several cancer types. In this study, the characteristic properties of this bispecific Janus lectin were investigated on human cells by flow cytometry and confocal microscopy in order to understand the fundamentals of its interactions. We evaluated its potential in targeted drug delivery, precisely leading to the cellular uptake of liposomal content in human epithelial cancer cells. We successfully demonstrated that Janus lectin mediates crosslinking of glyco-decorated giant unilamellar vesicles (GUVs) and H1299 lung epithelial cells. Strikingly, the Janus lectin induced the internalization of liposomal lipids and also of complete GUVs. Our findings serve as a solid proof of concept for lectin-mediated targeted drug delivery using glyco-decorated liposomes as possible drug carriers to cells of interest. The use of Janus lectin for tumor recognition certainly broadens the possibilities for engineering diverse tailor-made lectin constructs, specifically targeting extracellular structures of high significance in pathological conditions.


Asunto(s)
Lectinas/metabolismo , Liposomas/metabolismo , Humanos , Lectinas/química , Células Tumorales Cultivadas
19.
Nanomaterials (Basel) ; 11(9)2021 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-34578538

RESUMEN

Mesoporous silica nanoparticles have been widely applied as carriers for cancer treatment. Among the different types of stimuli-responsive drug delivery systems, those sensitive to redox stimuli have attracted much attention. Their relevance arises from the high concentration of reductive species that are found within the cells, compared to bloodstream, which leads to the drug release taking place only inside cells. This review is intended to provide a comprehensive overview of the most recent trends in the design of redox-responsive mesoporous silica nanoparticles. First, a general description of the biological rationale of this stimulus is presented. Then, the different types of gatekeepers that are able to open the pore entrances only upon application of reductive conditions will be introduced. In this sense, we will distinguish among those targeted and those non-targeted toward cancer cells. Finally, a new family of bridged silica nanoparticles able to degrade their structure upon application of this type of stimulus will be presented.

20.
Artículo en Inglés | MEDLINE | ID: mdl-32195232

RESUMEN

Gold nanoparticles are elective candidate for cancer therapy. Current efforts are devoted to developing innovative methods for their synthesis. Besides, understanding their interaction with cells have become increasingly important for their clinical application. This work aims to describe a simple approach for the synthesis of extra-small gold nanoparticles for breast cancer therapy. In brief, a biocompatible and biodegradable polyamidoamine (named AGMA1-SH), bearing 20%, on a molar basis, thiol-functionalized repeat units, is employed to stabilize and coat extra-small gold nanospheres of different sizes (2.5, 3.5, and 5 nm in gold core), and to generate a nanoplatform for the link with Trastuzumab monoclonal antibody for HER2-positive breast cancer targeting. Dynamic light scattering, transmission electron microscopy, ultraviolet visible spectroscopy, X-ray powder diffraction, circular dichroism, protein quantification assays are used for the characterization. The targeting properties of the nanosystems are explored to achieve enhanced and selective uptake of AGMA1-SH-gold nanoparticles by in vitro studies against HER-2 overexpressing cells, SKBR-3 and compared to HER-2 low expressing cells, MCF-7, and normal fibroblast cell line, NIH-3T3. In vitro physicochemical characterization demonstrates that gold nanoparticles modified with AGMA1-SH are more stable in aqueous solution than the unmodified ones. Additionally, the greater gold nanoparticles size (5-nm) is associated with a higher stability and conjugation efficiency with Trastuzumab, which retains its folding and anticancer activity after the conjugation. In particular, the larger Trastuzumab functionalized nanoparticles displays the highest efficacy (via the pro-apoptotic protein increase, anti-apoptotic components decrease, survival-proliferation pathways downregulation) and internalization (via the activation of the classical clathrin-mediated endocytosis) in HER-2 overexpressing SKBR-3 cells, without eliciting significant effects on the other cell lines. The use of biocompatible AGMA1-SH for producing covalently stabilized gold nanoparticles to achieve selective targeting, cytotoxicity and uptake is completely novel, offering an important advancement for developing new anticancer conjugated-gold nanoparticles.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA