Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Anal Bioanal Chem ; 411(3): 777-786, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30470915

RESUMEN

High- and low-density lipoproteins (HDL and LDL) are attractive targets for biomarker discovery. However, ultracentrifugation (UC), the current methodology of choice for isolating HDL and LDL, is tedious, requires large sample volume, results in sample loss, and does not readily provide information on particle size. In this work, human plasma HDL and LDL are separated and collected using semi-preparative asymmetrical flow field-flow fractionation (SP-AF4) and UC. The SP-AF4 and UC separation conditions, sample throughput, and liquid chromatography/mass spectrometry (LC/MS) lipidomic results are compared. Over 600 µg of total proteins is recovered in a single SP-AF4 run, and Western blot results confirm apoA1 pure and apoB100 pure fractions, consistent with HDL and LDL, respectively. The SP-AF4 separation requires ~ 60 min per sample, thus providing a marked improvement over UC which can span hours to days. Lipidome analysis of SP-AF4-prepared HDL and LDL fractions is compared to UC-prepared HDL and LDL samples. Over 270 lipids in positive MS mode and over 140 lipids in negative MS mode are identified by both sample preparation techniques with over 98% overlap between the lipidome. Additionally, lipoprotein size distributions are determined using analytical scale AF4 coupled with multiangle light scattering (MALS) and dynamic light scattering (DLS) detectors. These developments position SP-AF4 as a sample preparation method of choice for lipoprotein biomarker characterization and identification. Graphical abstract ᅟ.


Asunto(s)
Fraccionamiento de Campo-Flujo/métodos , Lipoproteínas HDL/sangre , Lipoproteínas LDL/sangre , Western Blotting , Cromatografía Liquida/métodos , Dispersión Dinámica de Luz/métodos , Humanos , Lipoproteínas HDL/aislamiento & purificación , Lipoproteínas LDL/aislamiento & purificación , Tamaño de la Partícula , Manejo de Especímenes , Espectrometría de Masas en Tándem/métodos , Ultracentrifugación
2.
Biomacromolecules ; 15(3): 915-23, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24392665

RESUMEN

One of the important criteria for achieving efficient nanoparticle-based targeted drug delivery is that the drug is not prematurely released at off-target sites. Here we report the preclinical evaluation of a serum-stable dendrimer-based drug conjugate capable of actively targeting into prostate cancer (PC) cells, delivered through the prostate-specific membrane antigen (PSMA). Multiple molecules of PSMA-binding small molecule glutamate urea (GLA; targeting agent) and the drug methotrexate (MTX) were conjugated to generation 5 PAMAM dendrimer (G5) through Cu-free "click" chemistry. The GLA was conjugated through a stable amide bond, and the MTX was conjugated either through ester (Es)- or amide (Am)-coupling, to generate G5-GLA(m)-(Es)MTX(n) and G5-GLA(m)-(Am)MTX(n), respectively. In serum-containing medium, free MTX was slowly released from "G5-GLA(m)-(Es)MTX(n)", with ~8% MTX released from the dendrimer in 72 h, whereas the MTX on G5-GLA(m)-(Am)MTX(n) was completely stable. The G5-GLA(m)-(Am)MTX(n) bound and internalized into PSMA-expressing LNCaP cells, but not into PSMA-negative PC3 cells. The conjugate-inhibited recombinant dihydrofolate reductase and induced potent cytotoxicity in the LNCaP cells, but not in the PC3 cells. Similar to the action of free GLA, stable amide-linked dendrimer-GLA was capable of inhibiting the enzyme N-acetylated α-linked acidic dipeptidase (NAALADase) activity of PSMA. The G5-GLA(m)-MTX(n) may serve as a serum-stable nanoparticle conjugate to specifically and effectively target and treat PSMA-overexpressing prostate tumors.


Asunto(s)
Antígenos de Superficie/metabolismo , Sistemas de Liberación de Medicamentos , Glutamato Carboxipeptidasa II/metabolismo , Nanopartículas/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Antígenos de Superficie/química , Química Clic , Dendrímeros/administración & dosificación , Dendrímeros/química , Glutamato Carboxipeptidasa II/química , Ácido Glutámico/química , Humanos , Masculino , Metotrexato/administración & dosificación , Metotrexato/química , Nanopartículas/química , Urea/química
3.
Pharm Res ; 30(1): 247-56, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23054086

RESUMEN

PURPOSE: Design and evaluate the in vitro and in vivo efficacy of two extended release morphine formulations developed for IV administration by complexing esterase activated morphine prodrugs to surface-modified, generation 5 (G5) poly(amidoamine) (PAMAM) dendrimer. METHODS: Prodrugs were synthesized, complexed with PAMAM dendrimer, characterized via ultra performance liquid chromatography (UPLC), nuclear magnatic resonance (NMR), and tested in vitro using rat plasma vs. saline control and in an in vivo rat and guinea pig pain model (modified Randall and Selitto test). RESULTS: We demonstrated that complexation with dendrimer allowed the solubilization of the prodrugs for in vivo applications without the need for salt, and that the structural design of the morphine prodrugs allowed the controlled release of morphine which extended the action of morphine-induced analgesia in an animal pain model from 2 h (control) to 6 h (Morphine Prodrug A). CONCLUSION: The concept of complexing/solubilizing appropriately designed esterase-sensitive prodrugs with dendrimer to enhance the sustained release of these drugs may be a useful pharmacokinetic strategy for a range of therapeutics.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Preparaciones de Acción Retardada/química , Dendrímeros/química , Morfina/uso terapéutico , Dolor/tratamiento farmacológico , Profármacos/uso terapéutico , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/química , Animales , Cobayas , Masculino , Morfina/administración & dosificación , Morfina/química , Profármacos/administración & dosificación , Profármacos/química , Ratas , Ratas Sprague-Dawley , Solubilidad
4.
Cancer ; 118(8): 2148-56, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22488668

RESUMEN

BACKGROUND: Growth factor receptors such as epidermal growth factor receptor 1 and human epidermal growth receptor 2 (HER2) are overexpressed in certain cancer cells. Antibodies against these receptors (eg. cetuximab and transtuzumab [Herceptin]) have shown therapeutic value in cancer treatment. The existing methods for the quantification of these receptors in tumors involve immunohistochemistry or DNA quantification, both in extracted tissue samples. The goal of the study was to evaluate whether an optical fiber-based technique can be used to quantify the expression of multiple growth factor receptors simultaneously. METHODS: The authors examined HER2 expression using the monoclonal antibody trastuzumab as a targeting ligand to test their system. They conjugated trastuzumab to 2 different Alexa Fluor dyes with different excitation and emission wavelengths. Two of the dye conjugates were subsequently injected intravenously into mice bearing HER2-expressing subcutaneous tumors. An optical fiber was then inserted into the tumor through a 30-gauge needle, and using a single laser beam as the excitation source, the fluorescence emitted by the 2 conjugates was identified and quantified by 2-photon optical fiber fluorescence. RESULTS: The 2 conjugates bound to the HER2-expressing tumor competitively in a receptor-specific fashion, but they failed to bind to a similar cell tumor that did not express HER2. The concentration of the conjugate present in the tumor as determined by 2-photon optical fiber fluorescence was shown to serve as an index of the HER2 expression levels. CONCLUSIONS: These studies offer a minimally invasive technique for the quantification of tumor receptors simultaneously.


Asunto(s)
Fibras Ópticas , Receptores de Factores de Crecimiento/análisis , Animales , Anticuerpos Monoclonales Humanizados , Carbocianinas , Línea Celular Tumoral , Receptores ErbB/análisis , Femenino , Colorantes Fluorescentes , Ratones , Ratones Desnudos , Neoplasias/metabolismo , Receptor ErbB-2/análisis , Trastuzumab
5.
Mol Pharm ; 9(9): 2669-2676, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22827500

RESUMEN

Our previous studies have demonstrated that a generation 5 dendrimer (G5) conjugated with both folic acid (FA) and methotrexate (MTX) has a higher chemotherapeutic index than MTX alone. Despite this, batch-to-batch inconsistencies in the number of FA and MTX molecules linked to each dendrimer led to conjugate batches with varying biological activity, especially when scaleup synthesis was attempted. Since the MTX is conjugated through an ester linkage, there were concerns that biological inconsistency could also result from serum esterase activity and differential bioavailability of the targeted conjugate. In order to resolve these problems, we undertook a novel approach to synthesize a polyvalent G5-MTX(n) conjugate through click chemistry, attaching the MTX to the dendrimer through an esterase-stable amide linkage. Surface plasmon resonance binding studies show that a G5-MTX(10) conjugate synthesized in this manner binds to the FA receptor (FR) through polyvalent interaction showing 4300-fold higher affinity than free MTX. The conjugate inhibits dihydrofolate reductase, and induces cytotoxicity in FR-expressing KB cells through FR-specific cellular internalization. Thus, the polyvalent MTX on the dendrimer serves the dual role as a targeting molecule as well as a chemotherapeutic drug. The newly synthesized G5-MTX(n) conjugate may serve as a FR-targeted chemotherapeutic with potential for cancer therapy.


Asunto(s)
Dendrímeros/química , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/metabolismo , Metotrexato/química , Disponibilidad Biológica , Línea Celular Tumoral , Dendrímeros/administración & dosificación , Esterasas/sangre , Humanos , Células KB , Metotrexato/administración & dosificación , Terapia Molecular Dirigida/métodos , Neoplasias/sangre , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo
6.
Biomacromolecules ; 13(4): 982-91, 2012 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-22360561

RESUMEN

Our group previously developed a multifunctional, targeted cancer therapeutic based on Generation 5 (G5) polyamidoamine (PAMAM) dendrimers. In those studies we conjugated the targeting molecule folic acid (FA) and the chemotherapeutic drug methotrexate (MTX) sequentially. This complex macromolecule was shown to selectively bind and kill KB tumor cells that overexpress folate receptor (FR) in vitro and in vivo. However, the multistep conjugation strategy employed in the synthesis of the molecule resulted in heterogeneous populations having differing numbers and ratios of the functionally antagonistic FA and MTX. This led to inconsistent and sometimes biologically inactive batches of molecules, especially during large-scale synthesis. We here resolved this issue by using a novel triazine scaffold approach that reduces the number of dendrimer conjugation steps required and allows for the synthesis of G5 conjugates with defined ratios of FA and MTX. Although an unoccupied γ-glutamyl carboxylate of FA has been previously suggested to be nonessential for FR binding, the functional requirement of an open α-carboxylate still remains unclear. In an attempt to also address this question, we have synthesized isomeric FA dendrimer conjugates (α-carboxyl or γ-carboxyl linked). Competitive binding studies revealed that both linkages have virtually identical affinity toward FR on KB cells. Our studies show that a novel bifunctional triazine-based conjugate G5-Triazine-γMTX-αFA with identical numbers of FA and MTX binds to FR through a polyvalent interaction and induces cytotoxicity in KB cells through FR-mediated cellular internalization, inducing higher toxicity as compared to conjugates synthesized by the multistep strategy. This work serves as a proof of concept for the development of bifunctional dendrimer conjugates that require a defined ratio of two functional molecules.


Asunto(s)
Antineoplásicos/farmacología , Dendrímeros/química , Dendrímeros/farmacología , Ácido Fólico/farmacología , Metotrexato/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Ácido Fólico/química , Humanos , Células KB , Sustancias Macromoleculares/síntesis química , Sustancias Macromoleculares/química , Sustancias Macromoleculares/farmacología , Metotrexato/química , Estructura Molecular , Relación Estructura-Actividad , Células Tumorales Cultivadas
7.
Photochem Photobiol Sci ; 11(4): 653-60, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22234658

RESUMEN

Nanoparticle (NP)-based targeted drug delivery involves cell-specific targeting followed by a subsequent therapeutic action from the therapeutic carried by the NP system. NPs conjugated with methotrexate (MTX), a potent inhibitor of dihydrofolate reductase (DHFR) localized in cytosol, have been under investigation as a delivery system to target cancer cells to enhance the therapeutic index of methotrexate, which is otherwise non-selectively cytotoxic. Despite improved therapeutic activity from MTX-conjugated NPs in vitro and in vivo, the therapeutic action of these conjugates following cellular entry is poorly understood; in particular it is unclear whether the therapeutic activity requires release of the MTX. This study investigates whether MTX must be released from a nanoparticle in order to achieve the therapeutic activity. We report herein light-controlled release of methotrexate from a dendrimer-based conjugate and provide evidence suggesting that MTX still attached to the nanoconjugate system is fully able to inhibit the activity of its enzyme target and the growth of cancer cells.


Asunto(s)
Dendrímeros/química , Antagonistas del Ácido Fólico/toxicidad , Metotrexato/toxicidad , Nanoconjugados/química , Tetrahidrofolato Deshidrogenasa/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/química , Humanos , Nanoconjugados/toxicidad , Neoplasias/tratamiento farmacológico , Fotólisis , Espectrofotometría Ultravioleta , Tetrahidrofolato Deshidrogenasa/metabolismo , Rayos Ultravioleta
8.
Arthritis Rheum ; 63(9): 2671-80, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21618461

RESUMEN

OBJECTIVE: To investigate the uptake of a poly(amidoamine) dendrimer (generation 5 [G5]) nanoparticle covalently conjugated to polyvalent folic acid (FA) as the targeting ligand into macrophages, and to investigate the activity of an FA- and methotrexate (MTX)-conjugated dendrimer (G5-FA-MTX) as a therapeutic for the inflammatory disease of arthritis. METHODS: In vitro studies were performed in macrophage cell lines and in isolated mouse macrophages to check the cellular uptake of fluorescence-tagged G5-FA nanoparticles, using flow cytometry and confocal microscopy. In vivo studies were conducted in a rat model of collagen-induced arthritis to evaluate the therapeutic potential of G5-FA-MTX. RESULTS: Folate-targeted dendrimer bound and internalized in a receptor-specific manner into both folate receptor ß-expressing macrophage cell lines and primary mouse macrophages. The conjugate G5-FA-MTX acted as a potent antiinflammatory agent and reduced arthritis-induced parameters of inflammation such as ankle swelling, paw volume, cartilage damage, bone resorption, and body weight decrease. CONCLUSION: The use of folate-targeted nanoparticles to specifically target MTX into macrophages may provide an effective clinical approach for antiinflammatory therapy in rheumatoid arthritis.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Portadores de Fármacos , Ácido Fólico/uso terapéutico , Macrófagos/efectos de los fármacos , Metotrexato/uso terapéutico , Nanopartículas/uso terapéutico , Animales , Artritis Experimental/inducido químicamente , Línea Celular , Células Cultivadas , Ácido Fólico/administración & dosificación , Metotrexato/administración & dosificación , Ratones , Nanopartículas/administración & dosificación
9.
Bioconjug Chem ; 22(4): 679-89, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21425790

RESUMEN

A modular dendrimer-based drug delivery platform was designed to improve upon existing limitations in single dendrimer systems. Using this modular strategy, a biologically active platform containing receptor mediated targeting and fluorescence imaging modules was synthesized by coupling a folic acid (FA) conjugated dendrimer with a fluorescein isothiocyanate (FITC) conjugated dendrimer. The two different dendrimer modules were coupled via the 1,3-dipolar cycloaddition reaction ("click" chemistry) between an alkyne moiety on the surface of the first dendrimer and an azide moiety on the second dendrimer. Two simplified model systems were also synthesized to develop appropriate "click" reaction conditions and aid in spectroscopic assignments. Conjugates were characterized by (1)H NMR spectroscopy and NOESY. The FA-FITC modular platform was evaluated in vitro with a human epithelial cancer cell line (KB) and found to specifically target the overexpressed folic acid receptor.


Asunto(s)
Dendrímeros/metabolismo , Portadores de Fármacos/metabolismo , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Receptores de Folato Anclados a GPI/análisis , Ácido Fólico/metabolismo , Química Clic , Dendrímeros/síntesis química , Dendrímeros/química , Portadores de Fármacos/síntesis química , Portadores de Fármacos/química , Colorantes Fluorescentes/química , Receptores de Folato Anclados a GPI/biosíntesis , Ácido Fólico/química , Humanos , Isotiocianatos/química , Células KB , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Estructura Molecular , Propiedades de Superficie , Células Tumorales Cultivadas
10.
Bioorg Med Chem ; 19(8): 2557-64, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21459000

RESUMEN

A saccharide-terminated generation 3 (G3) polyamidoamine (PAMAM) dendrimer was synthesized as a drug carrier. Utilizing this dendritic platform, we have successfully synthesized polyvalent conjugates (G3-MTX) containing the drug methotrexate (MTX). Surface Plasmon Resonance (SPR) results showed that G3-MTX presented three orders of magnitude enhancement in binding avidity to folate-binding protein (FBP) as compared to the free folic acid (FA). Flow cytometric and confocal microscopic analysis showed that conjugate (G3-MTX-FI) containing imaging agent fluorescein-5(6)-carboxamidohexanoic acid (FI) was internalized into folate receptor (FR)-expressing KB cells in dose-dependent and receptor-mediated fashion. The G3-MTX induced a dose-dependent cytotoxicity in the KB cells. Therefore, the polyvalent G3-MTX may have potential as an anticancer nanodevice for the specific targeting and killing of FR-expressing tumor cells.


Asunto(s)
Dendrímeros/química , Portadores de Fármacos/síntesis química , Metotrexato/administración & dosificación , Antimetabolitos Antineoplásicos/administración & dosificación , Carbohidratos , Línea Celular Tumoral , Dendrímeros/farmacocinética , Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos , Receptor 1 de Folato , Humanos , Poliaminas , Unión Proteica/efectos de los fármacos
11.
Anal Chem ; 82(12): 5211-8, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20486666

RESUMEN

Real-time measurement of specific biomolecular interactions is critical to many areas of biological research. A number of label-free techniques for directly monitoring biomolecular binding have been developed, but it is still challenging to measure the binding kinetics of very small molecules, to detect low concentrations of analyte molecules, or to detect low affinity interactions. In this study, we report the development of a highly sensitive photonic crystal biosensor for label-free, real-time biomolecular binding analysis. We characterize the performance of this biosensor using a standard streptavidin-biotin binding system. Optimization of the surface functionalization methods for streptavidin immobilization on the silica sensing surface is presented, and the specific binding of biotinylated analyte molecules ranging over 3 orders of magnitude in molecular weight, including very small molecules (<250 Da), DNA oligonucleotides, proteins, and antibodies (>150 000 Da), are detected in real time with a high signal-to-noise ratio. Finally, we document the sensor efficiency for low mass adsorption, as well as multilayered molecular interactions. By all important metrics for sensitivity, we anticipate this photonic crystal biosensor will provide new capabilities for highly sensitive measurements of biomolecular binding.


Asunto(s)
Técnicas Biosensibles/métodos , Biotina/metabolismo , Estreptavidina/metabolismo , Anticuerpos/metabolismo , Sitios de Unión , Biotinilación , Oligonucleótidos/metabolismo , Fotones , Unión Proteica , Proteínas/metabolismo , Sensibilidad y Especificidad
12.
Bioconjug Chem ; 21(3): 489-95, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20128612

RESUMEN

A targeted dendrimeric anticancer prodrug, a conjugate of generation 5 (G5) polyamidoamine (PAMAM) dendrimer, folic acid (FA), and methotrexate (MTX), has been successfully synthesized by using a novel "one pot" approach which is simple, reproducible, and feasible for large-scale synthesis. All dendrimer products have been characterized by (1)H NMR, MALDI-TOF, GPC, and HPLC. With this new method, the ratio of FA versus MTX attached to the dendrimer can be easily tuned to achieve the desired therapeutic effect. A new analytical approach for calculating the numbers of FA and MTX attached to the dendrimer has been established. In vitro studies performed on FA receptor-expressing KB cells show that the new conjugate has a similar affinity and cytotoxic potency to G5-FA-MTX synthesized using the traditional multiple-step approach.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/síntesis química , Dendrímeros/química , Sistemas de Liberación de Medicamentos , Ácido Fólico/química , Metotrexato/farmacología , Poliaminas/química , Profármacos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Dendrímeros/síntesis química , Relación Dosis-Respuesta a Droga , Humanos , Células KB , Metotrexato/química , Estructura Molecular , Profármacos/administración & dosificación , Profármacos/química , Profármacos/farmacología , Relación Estructura-Actividad
13.
Bioorg Med Chem Lett ; 20(2): 700-3, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19962894

RESUMEN

Fibroblast Growth Factor Receptor (FGFR) is overexpressed in a wide variety of tumors, and therefore is an attractive target for drug delivery. Recombinant FGF-1 was purified and attached to a fifth-generation (G5) polyamidoamine dendrimer. The specific binding and internalization of this conjugate labeled with FITC was demonstrated by flow cytometry as well as by confocal microscopic analysis in cell lines expressing FGFR. The binding and uptake of FGF-conjugated dendrimers was completely blocked by excess nonconjugated FGF-1. Confocal microscopic analysis showed cytosolic as well as nuclear localization. Multivalent G5-FGF nanoparticles may serve as a platform for cytosolic as well as nuclear drug delivery in tumor cells, and as an FGF delivery agent for angiogenesis and wound healing. Our study shows for the first time the applicability of a dendrimer nanodevice for tumor cell targeting through FGFR.


Asunto(s)
Antineoplásicos/administración & dosificación , Dendrímeros/química , Factor 1 de Crecimiento de Fibroblastos/administración & dosificación , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Núcleo Celular/metabolismo , Sistemas de Liberación de Medicamentos , Factor 1 de Crecimiento de Fibroblastos/química , Factor 1 de Crecimiento de Fibroblastos/genética , Citometría de Flujo , Fluoresceína-5-Isotiocianato/química , Humanos , Neoplasias/tratamiento farmacológico , Poliaminas/química , Receptores de Factores de Crecimiento de Fibroblastos/química , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
14.
Bioorg Med Chem Lett ; 20(17): 5191-4, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20659800

RESUMEN

This communication describes the synthesis and in vitro biological evaluation of novel generation 5 PAMAM dendrimers conjugated with riboflavin as a targeting ligand. Cell-based experiments demonstrated that a dendrimer conjugated with riboflavin is able to undergo cellular binding and uptake in KB cells, and when the dendrimer is also conjugated with methotrexate, the riboflavin dendrimer conjugate can potently inhibit cell growth.


Asunto(s)
Dendrímeros/farmacología , Nanotecnología , Neoplasias/tratamiento farmacológico , Riboflavina/química , División Celular/efectos de los fármacos , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Células HeLa , Humanos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
15.
Bioorg Med Chem Lett ; 20(21): 6250-3, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20833544

RESUMEN

Two morphine prodrugs ('PDA' and 'PDB') were synthesized and the kinetics of esterase-mediated morphine release from these prodrugs were determined when incubated with plasma from different animal species. Morphine was rapidly released from PDA by all species plasma with the maximum reached within 5-10min; the released morphine was biologically active as determined by an in vitro cAMP assay. The morphine was released from PDB at a slower and species-dependent rate (mouse>rat>guinea pig>human). Morphine's release from PDB appeared to be mediated by carboxyl esterases as the release was inhibited by the carboxyl esterase inhibitor benzil. PDA nor PDB induce cytotoxicity in the neuronal cell lines SK-NSH and SH-SY5Y. The carboxyl and amino functional moieties present on the linker portions of PDA and PDB, respectively, may facilitate their conjugation to nanoparticles to tailor morphine pharmacokinetics and specific targeting. These studies suggest the potential clinical utility of these prodrugs for morphine release at desired rates by administration of their mixture at selected ratios.


Asunto(s)
Analgésicos Opioides/sangre , Analgésicos Opioides/farmacocinética , Morfina/sangre , Morfina/farmacocinética , Profármacos/síntesis química , Profármacos/farmacocinética , Animales , Hidrolasas de Éster Carboxílico/metabolismo , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Cobayas , Humanos , Hidrólisis , Indicadores y Reactivos , Espectroscopía de Resonancia Magnética , Ratones , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/metabolismo , Estándares de Referencia
16.
Biomacromolecules ; 10(12): 3207-14, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-19924846

RESUMEN

Poly(amidoamine) (PAMAM) dendrimers carrying different amounts of surface amino groups were synthesized and tested for their effects on cellular cytotoxicity, lysosomal pH, and mitochondria-dependent apoptosis. In KB cells, the PAMAM dendrimers were taken up into the lysosomal compartment, and they increased the lysosomal pH and cytotoxicity as a function of the number of surface amino groups on the dendrimer. PAMAM dendrimers that were surface-neutralized by acetylation of >80% of the surface amino groups failed to show any cytotoxicity. The positively charged, amine-terminated PAMAM dendrimer induced cellular apoptosis, as demonstrated by mitochondrial membrane potential changes and caspase activity measurements. These results suggest that PAMAM dendrimers are endocytosed into the KB cells through a lysosomal pathway, leading to lysosomal alkalinization and induction of mitochondria-mediated apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Lisosomas/efectos de los fármacos , Poliaminas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Dendrímeros , Humanos , Concentración de Iones de Hidrógeno , Mitocondrias/efectos de los fármacos , Poliaminas/síntesis química , Poliaminas/química
17.
Bioorg Med Chem Lett ; 19(17): 5016-20, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19632838

RESUMEN

Hypoxia is known to occur in tissues in response to narcotic analgesic therapy using as a result of respiratory depression. The aim of this study was to synthesize a narcotic antagonist pro-drug that can be activated by tissue hypoxia to prevent the damage associated with respiratory depression. We synthesized three different pro-drugs of the narcotic antagonist naloxone utilizing indolequinone as the hypoxia-sensitive moiety. The indolequinone structure in the pro-drugs was designed to have an open reactive point at the N-1 position offering the possibility of further conjugation with macromolecules to modify the bio-availability of these pro-drugs in vivo. A pro-drug (labeled 1) where naloxone and the indolequinone moiety were linked through a carbonate bond was rapidly hydrolyzed in phosphate buffered saline. However, two additional pro-drugs (labeled 2 and 3) having carbamate linkers were stable in phosphate buffered saline for 24h. The reductive release of naloxone from the pro-drugs was achieved in the presence of the bio-reductive enzyme DT-Diaphorase, with about 80% release occurring from the two pro-drugs in 24h. More than 99% of naloxone was released from pro-drug 2 in 30% human plasma, however the release only occurred under hypoxic conditions. This system provides a potential means for feedback control to counter critical respiratory depression induced by narcotic analgesics.


Asunto(s)
Indolquinonas/química , Naloxona/análogos & derivados , Antagonistas de Narcóticos/síntesis química , Profármacos/síntesis química , Células Sanguíneas/efectos de los fármacos , Hipoxia de la Célula , Humanos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naloxona/síntesis química , Naloxona/farmacología , Antagonistas de Narcóticos/química , Antagonistas de Narcóticos/farmacología , Profármacos/química , Profármacos/farmacología
18.
Biomacromolecules ; 9(2): 603-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18193839

RESUMEN

Binding of ligands on to epidermal growth factor receptor (EGFR) can stimulate cell growth; therefore, any application employing EGF as a targeting ligand for a "drug carrier" must evaluate the effect of the conjugate on cell growth. We report the synthesis and in vitro biological activity of EGF molecules coupled to a fluorescein-labeled polyamidoamine dendrimer. The conjugate bound and internalized into several EGFR-expressing cell lines in a receptor-specific fashion. The conjugate effectively induced EGFR phosphorylation and acted as a superagonist by stimulating cell growth to a greater degree than free EGF. Concomitant administration of the chemotherapeutic drug methotrexate completely inhibited cell growth to a degree similar to its effect in the absence of the conjugate. Thus, dendrimer-EGF conjugates serve as EGFR superagonists, but this activity can be overcome by chemotherapeutic drugs. The agonist activity of these materials must be taken into consideration when using EGF conjugates for imaging applications.


Asunto(s)
Dendrímeros/química , Factor de Crecimiento Epidérmico/análogos & derivados , Receptores ErbB/agonistas , Animales , Línea Celular , Línea Celular Tumoral , Dendrímeros/metabolismo , Dendrímeros/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Humanos , Ratones , Unión Proteica/fisiología
19.
J Biomed Opt ; 13(1): 014024, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18315382

RESUMEN

Fluorescence quantification in tissues using conventional techniques can be difficult due to the absorption and scattering of light in tissues. Our previous studies have shown that a single-mode optical fiber (SMF)-based, two-photon optical fiber fluorescence (TPOFF) probe could be effective as a minimally invasive, real-time technique for quantifying fluorescence in solid tumors. We report improved results with this technique using a solid, double-clad optical fiber (DCF). The DCF can maintain a high excitation rate by propagating ultrashort laser pulses down an inner single-mode core, while demonstrating improved collection efficiency by using a high-numerical aperture multimode outer core confined with a second clad. We have compared the TPOFF detection efficiency of the DCF versus the SMF with standard solutions of the generation 5 poly(amidoamine) dendrimer (G5) nanoparticles G5-6TAMRA (G5-6T) and G5-6TAMRA-folic acid (G5-6T-FA). The DCF probe showed three- to five-fold increases in the detection efficiency of these conjugates, in comparison to the SMF. We also demonstrate the applicability of the DCF to quantify the targeted uptake of G5-6T-FA in mouse tumors expressing the FA receptor. These results indicate that the TPOFF technique using the DCF probe is an appropriate tool to quantify low nanomolar concentrations of targeted fluorescent probes from deep tissue.


Asunto(s)
Tecnología de Fibra Óptica/instrumentación , Colorantes Fluorescentes/farmacocinética , Microscopía Fluorescente/instrumentación , Nanopartículas/ultraestructura , Espectrometría de Fluorescencia/instrumentación , Transductores , Animales , Dendrímeros , Sistemas de Liberación de Medicamentos/métodos , Diseño de Equipo , Análisis de Falla de Equipo , Tecnología de Fibra Óptica/métodos , Humanos , Células KB , Ratones , Ratones SCID , Microscopía Fluorescente/métodos , Fibras Ópticas , Espectrometría de Fluorescencia/métodos
20.
Cancer Res ; 65(12): 5317-24, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15958579

RESUMEN

Prior studies suggested that nanoparticle drug delivery might improve the therapeutic response to anticancer drugs and allow the simultaneous monitoring of drug uptake by tumors. We employed modified PAMAM dendritic polymers <5 nm in diameter as carriers. Acetylated dendrimers were conjugated to folic acid as a targeting agent and then coupled to either methotrexate or tritium and either fluorescein or 6-carboxytetramethylrhodamine. These conjugates were injected i.v. into immunodeficient mice bearing human KB tumors that overexpress the folic acid receptor. In contrast to nontargeted polymer, folate-conjugated nanoparticles concentrated in the tumor and liver tissue over 4 days after administration. The tumor tissue localization of the folate-targeted polymer could be attenuated by prior i.v. injection of free folic acid. Confocal microscopy confirmed the internalization of the drug conjugates into the tumor cells. Targeting methotrexate increased its antitumor activity and markedly decreased its toxicity, allowing therapeutic responses not possible with a free drug.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Colorantes Fluorescentes/administración & dosificación , Metotrexato/administración & dosificación , Nanoestructuras , Poliaminas/administración & dosificación , Radiofármacos/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Proteínas Portadoras/metabolismo , Dendrímeros , Modelos Animales de Enfermedad , Portadores de Fármacos/farmacocinética , Femenino , Colorantes Fluorescentes/farmacocinética , Receptores de Folato Anclados a GPI , Humanos , Células KB , Metotrexato/farmacocinética , Ratones , Ratones Desnudos , Ratones SCID , Poliaminas/farmacocinética , Radiofármacos/farmacocinética , Receptores de Superficie Celular/metabolismo , Distribución Tisular , Tritio/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA