Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Control Release ; 369: 734-745, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38604385

RESUMEN

Despite research efforts being made towards preserving (or even regenerating) heart tissue after an ischemic event, there is a lack of resources in current clinical treatment modalities for patients with acute myocardial infarction that specifically address cardiac tissue impairment. Modified messenger RNA (modRNA) presents compelling properties that could allow new therapeutic strategies to tackle the underlying molecular pathways that ultimately lead to development of chronic heart failure. However, clinical application of modRNA for the heart is challenged by the lack of effective and safe delivery systems. Lipid nanoparticles (LNPs) represent a well characterized class of RNA delivery systems, which were recently approved for clinical usage in mRNA-based COVID-19 vaccines. In this study, we evaluated the potential of LNPs for cardiac delivery of modRNA. We tested how variations in C12-200 modRNA-LNP composition affect transfection levels and biodistribution after intramyocardial administration in both healthy and myocardial-infarcted mice, and determined the targeted cardiac cell types. Our data revealed that LNP-mediated modRNA delivery outperforms the current state of the art (modRNA in citrate buffer) upon intramyocardial administration in mice, with only minor differences among the formulations tested. Furthermore, we determined both in vitro and in vivo that the cardiac cells targeted by modRNA-LNPs include fibroblasts, endothelial cells and epicardial cells, suggesting that these cell types could represent targets for therapeutic interference with these LNP formulations. These outcomes may serve as a starting point for LNP development specifically for therapeutic mRNA cardiac delivery applications.


Asunto(s)
Ratones Endogámicos C57BL , Infarto del Miocardio , Miocardio , Nanopartículas , ARN Mensajero , Animales , ARN Mensajero/administración & dosificación , Distribución Tisular , Infarto del Miocardio/terapia , Miocardio/metabolismo , Lípidos/química , Ratones , Humanos , Masculino , Técnicas de Transferencia de Gen , Transfección/métodos , Liposomas
2.
Int J Pharm ; 620: 121741, 2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35421533

RESUMEN

Targeted delivery of nucleic acids is gaining momentum due to improved efficacy, selectivity, increased circulation time and enhanced tissue retention in target cells. Using nucleic acid-based therapies previously undruggable targets have proven now to be amenable for treatment. Currently, several methods for preparing targeted or labelled delivery vehicles for nucleic acids are based on liposomal formulations. Lipid nanoparticles (LNPs) are structurally different from liposomes and these methods should therefore be evaluated before being translated to siRNA LNPs preparation protocols. Here, we describe a robust and facile method for the preparation of targeted or fluorescently labelled siRNA LNPs. Using a copper free strain-promoted azide-alkyne cycloaddition (SPAAC) we demonstrate that post-insertion of ligand-lipid conjugates into preformed LNPs is superior to direct-surface modification because it preserves the physicochemical parameters of the LNPs. We found that the time point of solvent removal by dialysis is critical and affects the hydrodynamic diameter of the LNPs; post-insertion after dialysis shows the smallest increase in hydrodynamic diameter and polydispersity index (PDI). The post-insertion of ligand-lipid conjugates also proceeded with rapid kinetics and high efficacy over a wide temperature range. Using this optimised protocol, we generated siRNA LNPs containing both targeting and fluorescent tracking ligands allowing us to monitor siRNA LNP uptake kinetics in dependence of the targeting ligand. In aggregate, we describe a robust approach for the generation of targeted and labelled siRNA LNPs that allows their controlled and facile decoration with ligand combinations.


Asunto(s)
Liposomas , Nanopartículas , Ligandos , Lípidos , ARN Interferente Pequeño/genética , Diálisis Renal
4.
Gene Ther ; 24(3): 157-166, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28140387

RESUMEN

Nucleic acid-based drugs offer a potentially effective tool for treatment of a variety of diseases, including cancer, cardiovascular diseases, neurological disorders and infectious diseases. However, clinical applications are hindered by instability of RNA molecules in the circulation and lack of efficient vectors that can deliver RNAs to target tissues and into diseased target cells. Synthetic polymer and lipids as well as virus-based vectors are among the most widely explored vehicles for RNA delivery, but clinical progress has been limited as a result of issues related to toxicity, immunogenicity and low efficiency. Most recently, the discovery that extracellular vesicles (EVs) are endogenous RNA carriers, which may display better biocompatibility and higher delivery efficiency as compared with the synthetic systems, has provided a ray of hope in coping with the delivery dilemma, and EV-based gene therapy has already sparked general interest both in academia and industry. In this review, the current knowledge on EV biology and their role in cell-cell communication will be summarized. Promises of EVs as drug carriers and recent technologies on tailoring EVs' biological attributes will be included, and preclinical studies in which EVs have shown promise for therapeutic RNA delivery will be discussed.


Asunto(s)
Vesículas Extracelulares/metabolismo , ARN Interferente Pequeño/administración & dosificación , Tratamiento con ARN de Interferencia/métodos , Animales , Vesículas Extracelulares/trasplante , Humanos
5.
J Control Release ; 224: 77-85, 2016 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-26773767

RESUMEN

Extracellular vesicles (EVs) are increasingly being recognized as candidate drug delivery systems due to their ability to functionally transfer biological cargo between cells. However, the therapeutic applicability of EVs may be limited due to a lack of cell-targeting specificity and rapid clearance of exogenous EVs from the circulation. In order to improve EV characteristics for drug delivery to tumor cells, we have developed a novel method for decorating EVs with targeting ligands conjugated to polyethylene glycol (PEG). Nanobodies specific for the epidermal growth factor receptor (EGFR) were conjugated to phospholipid (DMPE)-PEG derivatives to prepare nanobody-PEG-micelles. When micelles were mixed with EVs derived from Neuro2A cells or platelets, a temperature-dependent transfer of nanobody-PEG-lipids to the EV membranes was observed, indicative of a 'post-insertion' mechanism. This process did not affect EV morphology, size distribution, or protein composition. After introduction of PEG-conjugated control nanobodies to EVs, cellular binding was compromised due to the shielding properties of PEG. However, specific binding to EGFR-overexpressing tumor cells was dramatically increased when EGFR-specific nanobodies were employed. Moreover, whereas unmodified EVs were rapidly cleared from the circulation within 10min after intravenous injection in mice, EVs modified with nanobody-PEG-lipids were still detectable in plasma for longer than 60min post-injection. In conclusion, we propose post-insertion as a novel technique to confer targeting capacity to isolated EVs, circumventing the requirement to modify EV-secreting cells. Importantly, insertion of ligand-conjugated PEG-derivatized phospholipids in EV membranes equips EVs with improved cell specificity and prolonged circulation times, potentially increasing EV accumulation in targeted tissues and improving cargo delivery.


Asunto(s)
Sistemas de Liberación de Medicamentos , Vesículas Extracelulares/química , Polietilenglicoles/química , Administración Intravenosa , Plaquetas/metabolismo , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Receptores ErbB/administración & dosificación , Excipientes , Humanos , Ligandos , Micelas , Nanopartículas , Tamaño de la Partícula , Fosfolípidos/química
6.
Haemophilia ; 22(1): 152-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26189554

RESUMEN

INTRODUCTION AND AIM: Joint bleeding results in blood-induced arthropathy. We investigate whether a joint bleed alters protease-activated receptor (PAR) expression, and whether treatment with small interfering RNA (siRNA) targeted against PAR1-4 attenuates synovitis and cartilage damage. METHODS: Protease-activated receptor expression was evaluated upon a joint bleed in haemophilic mice and in humans. In addition, mice with a joint bleed were randomized between treatment with PAR1-4 siRNA or control and evaluated for the presence of synovitis and cartilage damage. Also, human cartilage was transfected with PAR1-4 siRNA or control, and evaluated for plasmin-induced cartilage damage. RESULTS: Following a joint bleed, we observed an increase in synovial PAR1, -2 and -4 expression, and an increase in chondrocyte PAR2 and -3 expression in mice (all P < 0.05). Also an increase in synovial PAR1 and chondrocyte PAR4 expression in patients was observed (both P < 0.05). Treatment of a joint bleed in haemophilic mice with PAR1-4 siRNA attenuates synovitis and cartilage damage (both P < 0.01). Treatment of human cartilage tissue explants with PAR1-4 siRNA reduced plasmin-induced cartilage damage (P < 0.01). CONCLUSION: This study demonstrates that synovial and chondrocyte PAR expression is altered upon a joint bleed, and that treatment with PAR1-4 siRNA attenuates synovitis and plasmin-induced cartilage damage.


Asunto(s)
Cartílago Articular/patología , Silenciador del Gen , Hemofilia A/complicaciones , Hemorragia/complicaciones , Receptores Proteinasa-Activados/deficiencia , Receptores Proteinasa-Activados/genética , Sinovitis/genética , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Fibrinolisina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hemorragia/genética , Hemorragia/patología , Humanos , Ratones , ARN Interferente Pequeño/genética , Sinovitis/complicaciones , Sinovitis/patología
7.
J Control Release ; 195: 11-20, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25135792

RESUMEN

In the development of potent polymeric gene carriers for gene therapy, a good interaction between the polymer and the nucleotide is indispensable to form small and stable polyplexes. Polymers with relatively high cationic charge density are frequently used to provide these interactions, but high cationic charge is usually associated with severe cytotoxicity. In this study an alternative, nucleotide specific binding interaction based on intercalation was investigated to improve polymer/pDNA complex formation. For this purpose bioreducible poly(amido amine) copolymers (p(CBA-ABOL/Nic)) were synthesized with different degrees of intercalating quaternary nicotinamide (Nic) groups and amide-substituted derivatives in their side chains. The quaternary nicotinamide group was chosen as intercalating moiety because this group is part of the naturally occurring NAD+ coenzyme and is therefore expected to be non-toxic and non-carcinogenic. The presence of the quaternary nicotinamide moieties in the poly(amido amine) copolymers showed to effectively promote self-assembled polyplex formation already at low polymer/DNA ratios and results in decreased polyplex size and increased stability of the polyplexes. Furthermore, in contrast to the primary amine functionalized analogs the quaternary nicotinamide polymers showed to be non-hemolytic, indicating their compatibility with cell membranes. Polymers with 25% of Nic in the side chains induced GFP expressions of about 4-5 times that of linear PEI, which is comparable with p(CBA-ABOL), the parent PAA without Nic, but at a two- to fourfold lower required polymer dose. N-phenylation of the nicotinamide functionality even further reduces the required polymer dose to form stable polyplexes, which is a major improvement for these kinds of cationic polymers.


Asunto(s)
Técnicas de Transferencia de Gen , Sustancias Intercalantes , Niacinamida , Polímeros , Animales , Células COS , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , ADN/administración & dosificación , ADN/química , Eritrocitos/efectos de los fármacos , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Hemólisis/efectos de los fármacos , Humanos , Sustancias Intercalantes/administración & dosificación , Sustancias Intercalantes/química , Niacinamida/administración & dosificación , Niacinamida/química , Plásmidos , Polímeros/administración & dosificación , Polímeros/química
8.
Int J Lab Hematol ; 36(3): 244-53, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24750670

RESUMEN

The majority, if not all, of human cell types secrete extracellular vesicles (EVs) into their environment, at least partly as a means of intercellular communication. These secreted vesicles can be detected in most bodily fluids including blood, urine, and saliva. The number of secreted vesicles and their composition is altered in various pathological conditions, raising opportunities to exploit EVs as diagnostic and/or prognostic biomarkers. For this to become a reality, it is important to reach consensus regarding the standardization of protocols for sample collection, EV isolation, handling, and storage for valid comparison and interpretation of measurements. Depending on the information required, there are several detection options including EV number and size distribution, molecular surface markers, procoagulation activity, and RNA content. For these purposes, different techniques are currently utilized or under development. This review discusses the techniques that have the potential to become standard EV detection methods in a clinical diagnostic setting. In addition to the accuracy of the detection technique, other factors such as high-throughput, cost-effectiveness, time consumption, and required operator skill are important to consider. A combination of increasing fundamental knowledge, technological progress, standardization of sample collection, and processing protocols is required for EVs to become reliable predictors of altered physiology or development of disease suitable for routine clinical diagnostics. Cancer and (cardio)vascular disorders are examples of pathologies where EV detection may be applied in the near future for diagnosis and/or prognosis.


Asunto(s)
Espacio Extracelular/metabolismo , Vesículas Transportadoras/metabolismo , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/metabolismo , Técnicas y Procedimientos Diagnósticos , Enfermedades Hematológicas/diagnóstico , Enfermedades Hematológicas/metabolismo , Humanos , Neoplasias/diagnóstico , Neoplasias/metabolismo
9.
J Control Release ; 163(3): 361-7, 2012 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-23041274

RESUMEN

In the current study, core-crosslinked polymeric micelles (DEX-PMs) loaded with three different DEX derivatives designed to display different drug release kinetics, were evaluated for cancer therapy and compared to another effective nanomedicine formulation (long-circulating liposomes encapsulating dexamethasone, LCL-DEX). Pharmacokinetic studies with both radiolabeled dexamethasone and polymer showed that these polymeric systems have long circulating half-lives and may accumulate at the tumor site to a higher extent than liposomes. The in vitro drug release profiles and circulating drug levels in the blood stream show that DEX-PMs with dexamethasone covalently entrapped via a sulfone ester-containing linker (DMSL2) have prolonged circulation time and intermediate drug release kinetics compared to the other polymeric DEX-releasing systems. Furthermore, as the free dexamethasone circulating levels were similar when administered as DMSL2-PM or LCL-DEX, these systems were evaluated simultaneously for antitumor efficacy in B16F10 melanoma bearing mice. The corticosteroid-targeted systems inhibited tumor growth to a similar extent and both increased survival compared to free drug. Recently antitumor efficacy of targeted formulations has been correlated with a systemic effect: a decrease of white blood cell count. In this study all three polymeric systems, liposomes as well as free drug had similar effects on the number of circulating white blood cells, although white blood cell counts recovered faster in the group receiving free drug. In conclusion, corticosteroid-targeting with a polymeric system or a liposomal system translates in similar therapeutic effects. The proven high versatility of the PM with possible optimization and adjustment of the drug release to that required by the therapeutic application, clearly demonstrates the potential of these systems for the treatment of chronic inflammatory diseases including cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Dexametasona/administración & dosificación , Melanoma Experimental/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Dexametasona/química , Dexametasona/farmacocinética , Liposomas , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Micelas , Polímeros/administración & dosificación , Polímeros/química , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Distribución Tisular , Carga Tumoral/efectos de los fármacos
10.
Curr Top Med Chem ; 12(2): 108-19, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22196278

RESUMEN

RNA interference is a technique to induce sequence-specific gene silencing, but is hampered by inefficient delivery of its mediator, short interfering RNA, into target cells. This review describes recent advances in siRNA delivery using polymeric carrier systems. Structural variations that have been applied to these polymers for optimizing their intracellular trafficking are discussed, as well as strategies for stabilization and targeting to diseased tissues in vivo. Recent findings have highlighted safety issues that need to be taken into account in the design of nanoparticles for clinical application.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , ARN Interferente Pequeño/química , Humanos , Polímeros/química , Interferencia de ARN
11.
J Control Release ; 160(2): 211-6, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21983283

RESUMEN

Angiogenesis is an attractive target for cancer therapy, due to its central position in tumor growth and development. Vascular Endothelial Growth Factor (VEGF) and its receptors (VEGFRs) play a key role in the angiogenic process. A promising strategy for targeting VEGF-mediated angiogenesis is RNA interference (RNAi) using short interfering RNA (siRNA). However, for efficacious RNAi a well-designed siRNA delivery system is crucial. Liposome-Polycation-DNA (LPD) particles form a promising system for siRNA delivery to tumors. In order to target angiogenic endothelial cells, LPD particles may be modified with a targeting ligand, such as a cyclic Arg-Gly-Asp (RGD) peptide that specifically binds to integrins expressed on tumor-associated endothelial cells. In the current study, RGD-targeted PEGylated LPD particles containing VEGFR-2 siRNA were prepared and optimized with respect to their size and charge by varying protamine content, carrier DNA content for stronger complexation, and PEGylation density. The size of the optimized particles was around 200 nm and the ζ-potential was approximately +20 mV. The uptake and silencing efficacy of the RGD-targeted PEGylated LPD particles were evaluated in H5V cells (murine endothelial cells) and Human Umbilical Vein Endothelial cells (HUVECs). When compared to non-targeted LPD particles, enhanced uptake and silencing of VEGFR-2 expression was observed for RGD-targeted PEGylated LPD particles. In conclusion, the RGD-targeted PEGylated LPD particles containing VEGFR-2 siRNA presented here may be a promising approach for targeting VEGF-mediated angiogenesis in cancer therapy.


Asunto(s)
ADN/química , Portadores de Fármacos/química , Células Endoteliales/metabolismo , Neovascularización Patológica/metabolismo , Poliaminas/química , ARN Interferente Pequeño/administración & dosificación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Western Blotting , Técnicas de Cultivo de Célula , Células Endoteliales/patología , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Liposomas , Ratones , Neovascularización Patológica/patología , Tamaño de la Partícula , Polielectrolitos , ARN Interferente Pequeño/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
12.
Angiogenesis ; 14(4): 457-66, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21789714

RESUMEN

Angiogenesis, the sprouting of new blood vessels from the pre-existing vasculature, is a well established target in anti-cancer therapy. It is thought that the Rho GTPase Rac1 is required during vascular endothelial growth factor (VEGF)-mediated angiogenesis. In the present study, we have used a clinically relevant RNA interference approach to silence Rac1 expression. Human umbilical vein endothelial cells were transiently transfected with non-specific control siRNA (siNS) or Rac1 siRNA (siRac1) using electroporation or Lipofectamine 2000. Functional assays with transfected endothelial cells were performed to determine the effect of Rac1 knockdown on angiogenesis in vitro. Silencing of Rac1 inhibited VEGF-mediated tube formation, cell migration, invasion and proliferation. In addition, treatment with Rac1 siRNA inhibited angiogenesis in an in vivo Matrigel plug assay. Intratumoral injections of siRac1 almost completely inhibited the growth of grafted Neuro2a tumors and reduced tumor angiogenesis. Together, these data indicate that Rac1 is an important regulator of VEGF-mediated angiogenesis. Knockdown of Rac1 may represent an attractive approach to inhibit tumor angiogenesis and growth.


Asunto(s)
Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteína de Unión al GTP rac1/metabolismo , Análisis de Varianza , Secuencia de Bases , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno , Combinación de Medicamentos , Electroporación , Humanos , Laminina , Datos de Secuencia Molecular , Proteoglicanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ensayo de Radioinmunoprecipitación , Transfección , Venas Umbilicales/citología , Proteína de Unión al GTP rac1/genética
13.
J Control Release ; 150(2): 177-86, 2011 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-21130817

RESUMEN

By Michael addition polymerization of N,N'-cystaminebisacrylamide (CBA) with variable ratios of 4-amino-1-butanol (ABOL) and ethylene diamine (EDA) or triethylenetetramine (TETA), poly(amido amine) copolymers could be obtained with tunable charge densities. The copolymers were optimized to serve as nonviral vectors in RNA interference (RNAi) to form stable, nanosized polyplexes with siRNA with maximum transfection efficacy. It was observed that at least 20-30% EDA or TETA amino units in the copolymers is necessary to encapsulate siRNA into small and stable polyplexes (< 200 nm). Incorporation of higher amounts of EDA or TETA in the copolymers did not further improve polyplex formation and stability, but the increased cationic charge in these copolymers resulted in increased cytotoxicity and hemolytic activity. Copolymers with 20% EDA showed excellent gene silencing properties in vitro (70% luciferase knockdown in H1299 cells) with negligible cytotoxicity.


Asunto(s)
Técnicas de Transferencia de Gen , Nylons/química , Poliaminas/química , ARN Interferente Pequeño/administración & dosificación , Acrilamidas/química , Amino Alcoholes/química , Disponibilidad Biológica , Tampones (Química) , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ditiotreitol/química , Electroforesis en Gel de Agar , Etilenodiaminas/química , Expresión Génica/genética , Glutatión/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hemólisis/efectos de los fármacos , Humanos , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Espectroscopía de Resonancia Magnética , Peso Molecular , Conformación de Ácido Nucleico , Nylons/síntesis química , Nylons/farmacología , Tamaño de la Partícula , Plásmidos/administración & dosificación , Plásmidos/química , Poliaminas/síntesis química , Poliaminas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/química , ARN Interferente Pequeño/farmacocinética , Electricidad Estática , Transfección , Trientina/química
15.
Curr Cancer Drug Targets ; 9(6): 748-60, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19754359

RESUMEN

Both the epidermal growth factor receptor (EGFR) and the insulin-like growth factor-1 receptor (IGF-1R) can contribute to tumor development and -progression through their effects on cell proliferation, inhibition of apoptosis, angiogenesis, anchorage-independent growth and tumor-associated inflammation. EGFR-targeting monoclonal antibodies and small molecule tyrosine kinase inhibitors are currently in clinical use for the treatment of several types of cancer. However, primary and acquired resistance to these agents often occurs and thereby limits the clinical efficacy of mono-specific targeted therapy. Results from both in vitro and in vivo studies indicate that cross-talk between EGFR and IGF-1R can lead to acquired resistance against EGFR-targeted drugs. This review describes the interface between the EGFR and IGF-1R signaling networks and the implications of the extensive cross-talk between these two receptor systems for cancer therapy. EGFR and IGF-1R interact on multiple levels, either through a direct association between the two receptors, by mediating the availability of each others ligands, or indirectly, via common interaction partners such as G protein coupled receptors (GPCR) or downstream signaling molecules. This multi-layered cross-talk and its involvement in the induction of resistance to targeted therapies provide a clear rationale for dual targeting of EGFR and IGF-1R. We discuss several (potential) strategies to simultaneously inhibit EGFR and IGF-1R signaling as promising novel therapeutic approaches.


Asunto(s)
Receptores ErbB/metabolismo , Receptor Cross-Talk/fisiología , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/fisiología , Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Descubrimiento de Drogas , Activación Enzimática , Receptores ErbB/antagonistas & inhibidores , Humanos , Modelos Biológicos , Receptor Cross-Talk/efectos de los fármacos , Receptor IGF Tipo 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
16.
Curr Pharm Des ; 14(34): 3686-97, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19075744

RESUMEN

The prospect of introducing siRNA in a cell, to induce silencing of the corresponding gene, has encouraged research into RNAi-based therapeutics as treatment for human diseases. At present, the siRNA molecules that are in a more advanced stage of clinical evaluation have a common factor: all are delivered locally at the site of the disease. Thus, the state of the art in delivery of siRNA appears to be the local administration. This can certainly be attributed to the characteristics of siRNA molecules, such as relatively high molecular weight, negative charge, and susceptibility to nuclease degradation, which make systemic application as a drug molecule difficult. When focusing on local administration, the main concerns for siRNA delivery can be restricted to the trafficking of siRNA molecules from the vicinity of the target cells, to the intracellular compartment where RNAi takes place, i.e. the cytoplasm. This contribution is focused on the barriers and challenges in trafficking of siRNA upon local delivery. First, an overview is given on the current state of the art for siRNA delivery in clinical trials. Second, recent successful preclinical studies, involving direct and local administration of siRNA, are reviewed. Third, emphasis is given to the endosomal escape. Some of our recent work is presented: the application of photochemical internalization (PCI) to improve the endosomal escape of siRNA lipoplexes in vivo. Finally, concluding remarks focus on the advantages of employing a technique such as PCI to enhance the endosomal escape of siRNA molecules.


Asunto(s)
Citoplasma/metabolismo , Silenciador del Gen , Fotoquímica , ARN Interferente Pequeño/administración & dosificación , Evaluación Preclínica de Medicamentos , ARN Interferente Pequeño/uso terapéutico
17.
Br J Cancer ; 99(8): 1256-64, 2008 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-18797467

RESUMEN

Vascular disrupting agents (VDAs) are able to affect selectively tumour endothelial cell morphology resulting in vessel occlusion and widespread tumour cell necrosis. However, single-agent antitumour activity of VDAs is typically limited, as tumour regrowth occurs rapidly following drug treatment. To improve the therapeutic effectiveness of VDAs, we investigated liposomal targeting using ZD6126 as a model VDA. ZD6126 is a phosphate-prodrug of the tubulin-binding vascular disrupting agent ZD6126 phenol. ZD6126 was encapsulated into long circulating PEG-liposomes for passive targeting and PEG-liposomes conjugated with peptide ligands containing the RGD-motif for active targeting to alpha(v)-integrins on tumour endothelial cells. ZD6126 could be stably encapsulated, and liposomes displayed minimal leakage in vitro (<10% in 3 weeks). In vivo, upon intravenous injection, free ZD6126 was rapidly converted into ZD6126 phenol, which was cleared from the circulation within minutes. In contrast, ZD6126 encapsulated into either RGD-targeted or PEG liposomes showed prolonged blood circulation times (t(1/2)=10 h), and ZD6126 phenol exposure was also prolonged (t(1/2)=8 h). Both liposomal formulations displayed tumour accumulation plus hepatosplenic uptake by local macrophages. The altered pharmacokinetics and tissue distribution profiles of both liposomal ZD6126 formulations resulted both in single-dose and multiple-dose regimes, in improved therapeutic efficacy in established murine B16.F10 melanomas compared with free ZD6126. The passively and actively targeted liposomes showed equal antitumour efficacy, indicating that delivery of ZD6126 to the tumour tissue may suffice to disrupt tumour blood vessels without the need for specific targeting to the tumour endothelium.


Asunto(s)
Antineoplásicos/administración & dosificación , Melanoma Experimental/tratamiento farmacológico , Compuestos Organofosforados/administración & dosificación , Compuestos Organofosforados/farmacocinética , Animales , Células Endoteliales/efectos de los fármacos , Humanos , Inmunohistoquímica , Liposomas , Macrófagos/efectos de los fármacos , Melanoma Experimental/patología , Ratones , Neovascularización Patológica/tratamiento farmacológico , Distribución Tisular , Venas Umbilicales
18.
Int J Pharm ; 254(1): 55-8, 2003 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-12615409

RESUMEN

Peptides with the RGD amino acid sequence show affinity for the alpha(v)beta(3) integrin, an integrin which is over-expressed on angiogenic endothelium and involved in cell adhesion. A peptide with the sequence ATWLPPR has been demonstrated to show affinity for the vascular endothelial growth factor (VEGF) receptor, a receptor involved in the proliferation of endothelial cells. By coupling these peptides to liposomes, these liposomes can serve as a site-specific drug delivery system to tumor endothelial cells in order to inhibit angiogenesis. In the present study we demonstrate that the coupling of cyclic RGD-peptides or ATWLPPR-peptides to the surface of PEG-liposomes results in binding of these liposomes to endothelial cells in vitro. Subsequent studies with RGD-peptide targeted liposomes in vivo also demonstrate specific binding to the tumor endothelium.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Sistemas de Liberación de Medicamentos , Oligopéptidos/química , Polietilenglicoles/química , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Células Cultivadas , Portadores de Fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Humanos , Técnicas In Vitro , Liposomas/química , Ratones , Microscopía Confocal/instrumentación , Microscopía Confocal/métodos , Neovascularización Patológica/tratamiento farmacológico , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Tiempo , Trasplantes
19.
Pharm Res ; 18(6): 780-7, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11474781

RESUMEN

PURPOSE: To gain insight into the host factors influencing liposome localization at sites of bacterial infection. METHODS: In a unilateral Klebsiella pneumoniae pneumonia rat model, capillary permeability and number of circulating leukocytes was quantified and related to the degree of liposome target localization. RESULTS: Liposome localization was highest in the hemorrhagic zone of infection, a zone characterized by markedly increased capillary permeability and high bacterial numbers. Both liposome localization and capillary permeability correlated positively with severity of infection. Lung instillation of other inflammatory stimuli, such as lipopolysaccharide or 0.1 M HCl inducing increased capillary permeability, also promoted liposome localization. As liposomal target localization in leukopenic rats was similar to that in immunocompetent rats, contribution of circulating leukocytes seems limited. Intrapulmonary distribution of liposomes shows that leukocytes at the target site are involved in liposome uptake after extravasation. CONCLUSIONS: Increased capillary permeability plays a crucial role in liposome localization at the infected site, whereas contribution of leukocytes is limited. These results suggest inflammatory conditions that could benefit from liposomal drug delivery. The involvement of leukocytes in liposome uptake at the target site could be important information in the selection of appropriate drugs.


Asunto(s)
Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/metabolismo , Klebsiella pneumoniae/metabolismo , Liposomas/farmacocinética , Pulmón/metabolismo , Animales , Permeabilidad Capilar , Azul de Evans/farmacocinética , Oro Coloide/farmacocinética , Infecciones por Klebsiella/patología , Liposomas/administración & dosificación , Pulmón/patología , Polietilenglicoles/administración & dosificación , Polietilenglicoles/farmacocinética , Ratas , Solventes/administración & dosificación , Solventes/farmacocinética
20.
J Pharmacol Exp Ther ; 298(1): 369-75, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11408564

RESUMEN

Antimicrobial agents may interact synergistically. But to ensure synergy in vivo, the drugs should both be present at the site of infection at sufficiently high concentrations for an adequate period of time. Coencapsulation of the drugs in a drug carrier may ensure parallel tissue distributions. Since liposomes localize preferentially at sites of infection, this mode of drug delivery could, in addition, increase drug concentrations at the focus of infection. The therapeutic efficacy of gentamicin and ceftazidime coencapsulated into liposomes was examined by monitoring survival in a rat model of an acute unilateral pneumonia caused by antibiotic-susceptible and antibiotic-resistant Klebsiella pneumoniae strains. It is shown that administration of gentamicin in combination with ceftazidime in the free form either as single dose or as 5-day treatment resulted in an additive effect on rat survival in both models. In contrast, targeted delivery of liposome-coencapsulated gentamicin and ceftazidime resulted in a synergistic interaction of the antibiotics in both models. Consequently, liposome coencapsulation of gentamicin and ceftazidime allowed both a shorter course of treatment at lower cumulative doses compared with administration of the antibiotics in the free form to obtain complete survival of rats. Liposomal coencapsulation of synergistic antibiotics may open new perspectives in the treatment of severe infections.


Asunto(s)
Ceftazidima/farmacología , Quimioterapia Combinada/farmacología , Gentamicinas/farmacología , Infecciones por Klebsiella/mortalidad , Klebsiella pneumoniae/efectos de los fármacos , Animales , Cápsulas , Ceftazidima/administración & dosificación , Farmacorresistencia Microbiana , Sinergismo Farmacológico , Quimioterapia Combinada/administración & dosificación , Femenino , Gentamicinas/administración & dosificación , Infecciones por Klebsiella/tratamiento farmacológico , Liposomas , Ratas , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA