Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 14(2)2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35215976

RESUMO

The evolution of the SARS-CoV-2 virus during the COVID-19 pandemic was accompanied by the emergence of new heavily mutated viral variants with increased infectivity and/or resistance to detection by the human immune system. To respond to the urgent need for advanced methods and materials to empower a better understanding of the mechanisms of virus's adaptation to human host cells and to the immuno-resistant human population, we suggested using recombinant filamentous bacteriophages, displaying on their surface foreign peptides termed "mimotopes", which mimic the structure of viral receptor-binding sites on the viral spike protein and can serve as molecular probes in the evaluation of molecular mechanisms of virus infectivity. In opposition to spike-binding antibodies that are commonly used in studying the interaction of the ACE2 receptor with SARS-CoV-2 variants in vitro, phage spike mimotopes targeted to other cellular receptors would allow discovery of their role in viral infection in vivo using cell culture, tissue, organs, or the whole organism. Phage mimotopes of the SARS-CoV-2 Spike S1 protein have been developed using a combination of phage display and molecular mimicry concepts, termed here "phage mimicry", supported by bioinformatics methods. The key elements of the phage mimicry concept include: (1) preparation of a collection of p8-type (landscape) phages, which interact with authentic active receptors of live human cells, presumably mimicking the binding interactions of human coronaviruses such as SARS-CoV-2 and its variants; (2) discovery of closely related amino acid clusters with similar 3D structural motifs on the surface of natural ligands (FGF1 and NRP1), of the model receptor of interest FGFR and the S1 spike protein; and (3) an ELISA analysis of the interaction between candidate phage mimotopes with FGFR3 (a potential alternative receptor) in comparison with ACE2 (the authentic receptor).


Assuntos
Bacteriófagos/genética , Técnicas de Visualização da Superfície Celular/métodos , Mimetismo Molecular , Receptores de Superfície Celular/genética , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Bacteriófagos/metabolismo , Sítios de Ligação , Humanos , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Ligação Viral
2.
Viruses ; 11(11)2019 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-31717800

RESUMO

Peptide-displayed phage libraries are billion-clone collections of diverse chimeric bacteriophage particles, decorated by genetically fused peptides built from a random combination of natural amino acids. Studying the molecular evolution of peptide-displayed libraries in mammalian model systems, using in vivo phage display techniques, can provide invaluable knowledge about the underlying physiology of the vasculature system, allow recognition of organ- and tissue-specific networks of protein-protein interactions, and provide ligands for targeted diagnostics and therapeutics. Recently, we discovered that landscape phage libraries, a specific type of multivalent peptide phage display library, expose on their surface comprehensive collections of elementary binding units (EBUs), which can form short linear motifs (SLiMs) that interact with functional domains of physiologically relevant proteins. Because of their unique structural and functional features, landscape phages can use an alternative mechanism of directed molecular evolution, i.e., combinatorial avidity selection. These discoveries fueled our interest in revisiting the in vivo evolution of phage displayed libraries using another format of display, i.e., landscape phages. In this study, we monitored the evolution of a landscape phage library in a mouse model with and without an implanted human breast cancer tumor xenograft. As expected, the multivalent architecture of landscape phage displayed proteins provided strong tissue selectivity and resulted in a huge diversity of tissue penetrating, chimeric phage particles. We identified several types of EBU interactions that evolved during the course of tissue distribution, which included interactions of EBUs with all tissue types, those EBUs that interacted selectively with specific organs or tissues with shared gene expression profiles or functionalities, and other EBUs that interacted in a tissue-selective manner. We demonstrated that landscape phage libraries are a rich collection of unique nanobioparticles that can be used to identify functional organ and tissue-binding elements after the evolution of a phage display library in vivo.


Assuntos
Bacteriófagos/genética , Neoplasias da Mama/genética , Técnicas de Visualização da Superfície Celular/métodos , Biblioteca Gênica , Animais , Evolução Molecular , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Nus
3.
Viruses ; 11(9)2019 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-31454976

RESUMO

Low performance of actively targeted nanomedicines required revision of the traditional drug targeting paradigm and stimulated the development of novel phage-programmed, self-navigating drug delivery vehicles. In the proposed smart vehicles, targeting peptides, selected from phage libraries using traditional principles of affinity selection, are substituted for phage proteins discovered through combinatorial avidity selection. Here, we substantiate the potential of combinatorial avidity selection using landscape phage in the discovery of Short Linear Motifs (SLiMs) and their partner domains. We proved an algorithm for analysis of phage populations evolved through multistage screening of landscape phage libraries against the MDA-MB-231 breast cancer cell line. The suggested combinatorial avidity selection model proposes a multistage accumulation of Elementary Binding Units (EBU), or Core Motifs (CorMs), in landscape phage fusion peptides, serving as evolutionary initiators for formation of SLiMs. Combinatorial selection has the potential to harness directed molecular evolution to create novel smart materials with diverse novel, emergent properties.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Técnicas de Química Combinatória/métodos , Evolução Molecular Direcionada , Sistemas de Liberação de Medicamentos/métodos , Antineoplásicos/análise , Bacteriófagos/metabolismo , Linhagem Celular Tumoral , Humanos , Nanomedicina , Biblioteca de Peptídeos , Peptídeos/química
4.
J Drug Target ; 26(5-6): 505-515, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29132246

RESUMO

Pancreatic cancer has been a life-threatening illness associated with high incidence and mortality rates. Paclitaxel (PCT) that causes mitotic arrest in cancer cells disrupting microtubule function is used for pancreatic cancer treatment. Nausea, anorexia and abdominal pain are some of the typical dose-limiting toxicity associated gastrointestinal side effects of the drug. Here, we present the use of polymeric mixed micelles to enable a targeted delivery of PCT and to provide additional advantages such as enhanced drug solubility, bioavailability and minimal dose-limiting toxicity. Also, these micelles self-assemble with pancreatic cancer cells-specific phage proteins P38, L1 and with the hydrophobic drug PCT resolving the issue of complex chemistry efforts normally needed for any conjugation. Our cytotoxicity and binding experiment results in vitro in 2 D and 3 D models suggested that the phage protein-targeted drug-loaded micelles bind and exhibit higher cell killing over the non-targeted ones.


Assuntos
Bacteriófagos/química , Sistemas de Liberação de Medicamentos , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Portadores de Fármacos/química , Humanos , Micelas , Paclitaxel/farmacologia , Neoplasias Pancreáticas/patologia , Polímeros/química
5.
Expert Opin Drug Deliv ; 14(3): 373-384, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27466706

RESUMO

INTRODUCTION: New phage-directed nanomedicines have emerged recently as a result of the in-depth study of the genetics and structure of filamentous phage and evolution of phage display and phage nanobiotechnology. This review focuses on the progress made in the development of the cancer-targeted nanomaterials and discusses the trends in using phage as a bioselectable molecular navigation system. Areas covered: The merging of phage display technologies with nanotechnology in recent years has proved promising in different areas of medicine and technology, such as medical diagnostics, molecular imaging, vaccine development and targeted drug/gene delivery, which is the focus of this review. The authors used data obtained from their research group and sourced using Science Citation Index (Web of Science) and NCBI PubMed search resources. Expert opinion: First attempts of adapting traditional concepts of direct targeting of tumor using phage-targeted nanomedicines has shown minimal improvements. With discovery and study of biological and technical barriers that prevent anti-tumor drug delivery, a paradigm shift from traditional drug targeting to nanomedicine navigation systems is required. The advanced bacteriophage-driven self-navigation systems are thought to overcome those barriers using more precise, localized phage selection methods, multi-targeting 'promiscuous' ligands and advanced multifunctional nanomedicine platforms.


Assuntos
Antineoplásicos/administração & dosagem , Bacteriófagos , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Técnicas de Transferência de Genes , Humanos , Ligantes , Nanomedicina/métodos , Nanoestruturas
6.
Comb Chem High Throughput Screen ; 19(5): 412-22, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27095536

RESUMO

Cancer cell-specific diagnostic or therapeutic tools are commonly believed to significantly increase the success rate of cancer diagnosis and targeted therapies. To extend the repertoire of available cancer cell-specific phage fusion proteins and study their efficacy as navigating moieties, we used two landscape phage display libraries f8/8 and f8/9 displaying an 8- or 9-mer random peptide fusion to identify a panel of novel peptide families that are specific to Calu-3 cells. Using a phage capture assay, we showed that two of the selected phage clones, ANGRPSMT and VNGRAEAP (phage and their recombinant proteins are named by the sequence of the fusion peptide), are selective for the Calu-3 cell line in comparison to phenotypically normal lung epithelial cells and distribute into unique subcellular fractions.


Assuntos
Neoplasias Pulmonares/tratamento farmacológico , Terapia de Alvo Molecular/métodos , Biblioteca de Peptídeos , Sequência de Aminoácidos , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Neoplasias Pulmonares/patologia , Proteínas de Fusão de Membrana/metabolismo , Peptídeos , Frações Subcelulares/metabolismo
7.
Protein Eng Des Sel ; 29(3): 93-103, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26764410

RESUMO

Cancer cell-specific targeting ligands against numerous cancer cell lines have been selected previously and used as ligands for cell-specific delivery of chemotherapies and various nanomedicines. However, tumor heterogeneity is one recognized problem hampering clinical translation of targeted anti-cancer medicines. Therefore, a novel class of targeting ligands is required that recognize receptors expressed between a variety of cancer phenotypes, identified here as 'promiscuous' ligands. In this work, promiscuous phage fusion proteins were first identified by a novel selection scheme to enrich for pan-cancer cell binding abilities, as indicated by conserved structural motifs identified previously in other cancer types. Additionally, peptide sequences containing a combination of motifs were identified to modulate binding. A panel of phage fusion proteins was studied for their specificity and selectivity for lung and pancreatic cancer cells. Phage displaying the fusion peptides GSLEEVSTL or GEFDELMTM, the two predominate clones with greatest binding ability, were used to modify preformed, doxorubicin-loaded, liposomes. These modified liposomes increased cytotoxicity up to 8.1-fold in several cancer cell lines when compared with unmodified liposomal doxorubicin. Taken together, these data indicate that promiscuous phage proteins, selected against different cancer cell lines, can be used as targeting ligands for treatment of heterogeneous tumor populations.


Assuntos
Terapia de Alvo Molecular , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular Tumoral , Clonagem Molecular , Doxorrubicina/administração & dosagem , Doxorrubicina/uso terapêutico , Humanos , Espaço Intracelular/metabolismo , Ligantes , Lipossomos , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Especificidade por Substrato
8.
Front Microbiol ; 6: 628, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26157433

RESUMO

Active tumor targeting of nanomedicines has recently shown significant improvements in the therapeutic activity of currently existing drug delivery systems, such as liposomal doxorubicin (Doxil/Caelyx/Lipodox). Previously, we have shown that isolated pVIII major coat proteins of the fd-tet filamentous phage vector, containing cancer cell-specific peptide fusions at their N-terminus, can be used as active targeting ligands in a liposomal doxorubicin delivery system in vitro and in vivo. Here, we show a novel major coat protein isolation procedure in 2-propanol that allows spontaneous incorporation of the hydrophobic protein core into preformed liposomal doxorubicin with minimal damage or drug loss while still retaining the targeting ligand exposed for cell-specific targeting. Using a panel of 12 structurally unique ligands with specificity toward breast, lung, and/or pancreatic cancer, we showed the feasibility of pVIII major coat proteins to significantly increase the throughput of targeting ligand screening in a common nanomedicine core. Phage protein-modified Lipodox samples showed an average doxorubicin recovery of 82.8% across all samples with 100% of protein incorporation in the correct orientation (N-terminus exposed). Following cytotoxicity screening in a doxorubicin-sensitive breast cancer line (MCF-7), three major groups of ligands were identified. Ligands showing the most improved cytotoxicity included: DMPGTVLP, ANGRPSMT, VNGRAEAP, and ANDVYLD showing a 25-fold improvement (p < 0.05) in toxicity. Similarly DGQYLGSQ, ETYNQPYL, and GSSEQLYL ligands with specificity toward a doxorubicin-insensitive pancreatic cancer line (PANC-1) showed significant increases in toxicity (2-fold; p < 0.05). Thus, we demonstrated proof-of-concept that pVIII major coat proteins can be screened in significantly higher throughput to identify novel ligands displaying improved therapeutic activity in a desired cancer phenotype.

9.
Mol Cancer Ther ; 13(12): 2864-75, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25239936

RESUMO

In an effort to improve the therapeutic index of cancer chemotherapy, we developed an advanced nanopreparation based on the combination of landscape phage display to obtain new targeting ligands with micellar nanoparticles for tumor targeting of water-insoluble neoplastic agents. With paclitaxel as a drug, this self-assembled nanopreparation composed of MCF-7-specific phage protein and polyethylene glycol-phosphatidylethanolamine (PEG-PE) micelles showed selective toxicity to target cancer cells rather than nontarget, non cancer cells in vitro. In vivo, the targeted phage micelles triggered a dramatic tumor reduction and extensive necrosis as a result of improved tumor delivery of paclitaxel. The enhanced anticancer effect was also verified by an enhanced apoptosis and reduced tumor cell proliferation following the treatment with the targeted micellar paclitaxel both in vitro and in vivo. The absence of hepatotoxicity and pathologic changes in tissue sections of vital organs, together with maintenance of overall health of mice following the treatment, further support its translational potential as an effective and safe chemotherapy for improved breast cancer treatment.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Micelas , Nanopartículas , Neoplasias/metabolismo , Neoplasias/patologia , Paclitaxel/administração & dosagem , Fosfatidiletanolaminas , Polietilenoglicóis , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Protein Eng Des Sel ; 27(7): 235-43, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24899628

RESUMO

It is hypothesized that the use of targeted drug delivery systems can significantly improve the therapeutic index of small molecule chemotherapies by enhancing accumulation of the drugs at the site of disease. Phage display offers a high-throughput approach for selection of the targeting ligands. We have successfully isolated phage fusion proteins selective and specific for PANC-1 pancreatic cancer cells. Doxorubicin liposomes (Lipodox) modified with tumor-specific phage fusion proteins enhanced doxorubicin uptake specifically in PANC-1 cells as compared with unmodified Lipodox and also compared with normal breast epithelial cells. Phage protein-targeted Lipodox substantially increased the concentration of doxorubicin in the nuclei of PANC-1 cells in spite of P-glycoprotein-mediated drug efflux. The in vitro cytotoxic activity obtained with pancreatic cell-targeted Lipodox was greater than that of unmodified Lipodox. We present a novel and straightforward method for preparing pancreatic tumor-targeted nanomedicines by anchoring pancreatic cancer-specific phage proteins within the liposome bilayer.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Nanomedicina/métodos , Neoplasias Pancreáticas/patologia , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Sequência de Aminoácidos , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Doxorrubicina/análogos & derivados , Doxorrubicina/metabolismo , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Polietilenoglicóis/metabolismo , Polietilenoglicóis/farmacologia , Transporte Proteico , Proteínas Recombinantes de Fusão/química , Especificidade por Substrato
11.
Nanomedicine ; 10(2): 421-30, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24028893

RESUMO

A novel strategy to improve the therapeutic index of chemotherapy has been developed by the integration of nanotechnology with phage technique. The objective of this study was to combine phage display, identifying tumor-targeting ligands, with a liposomal nanocarrier for targeted delivery of doxorubicin. Following the proof of concept in cell-based experiments, this study focused on in vivo assessment of antitumor activity and potential side-effects of phage fusion protein-modified liposomal doxorubicin. MCF-7-targeted phage-Doxil treatments led to greater tumor remission and faster onset of antitumor activity than the treatments with non-targeted formulations. The enhanced anticancer effect induced by the targeted phage-Doxil correlated with an improved tumor accumulation of doxorubicin. Tumor sections consistently revealed enhanced apoptosis, reduced proliferation activity and extensive necrosis. Phage-Doxil-treated mice did not show any sign of hepatotoxicity and maintained overall health. Therefore, MCF-7-targeted phage-Doxil seems to be an active and tolerable chemotherapy for breast cancer treatment. FROM THE CLINICAL EDITOR: The authors of this study successfully combined phage display with a liposomal nanocarrier for targeted delivery of doxorubicin using MCF-7-targeted phage-Doxil nanocarriers in a rodent model. The method demonstrated improved efficiency and reduced hepatotoxicity, paving the way to future clinical trials addressing breast cancer.


Assuntos
Antineoplásicos/química , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Doxorrubicina/análogos & derivados , Neoplasias/tratamento farmacológico , Alanina Transaminase/metabolismo , Animais , Apoptose , Aspartato Aminotransferases/metabolismo , Bacteriófagos/metabolismo , Doxorrubicina/administração & dosagem , Portadores de Fármacos , Feminino , Humanos , Antígeno Ki-67/metabolismo , Lipossomos/química , Células MCF-7 , Camundongos , Camundongos Nus , Nanomedicina , Necrose , Polietilenoglicóis/administração & dosagem , Proteínas Recombinantes de Fusão/química , Resultado do Tratamento
12.
Mol Pharm ; 10(2): 551-9, 2013 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-23215008

RESUMO

Nucleic acids, including antisense oligonucleotides, small interfering RNA (siRNA), aptamers, and rybozymes, emerged as versatile therapeutics due to their ability to interfere in a well-planned manner with the flow of genetic information from DNA to protein. However, a systemic use of NAs is hindered by their instability in physiological liquids and inability of intracellular accumulation in the site of action. We first evaluated the potential of cancer specific phage fusion proteins as targeting ligands that provide encapsulation, protection, and navigation of siRNA to the target cell. The tumor-specific proteins were isolated from phages that were affinity selected from a landscape phage library against target breast cancer cells. It was found that fusion phage coat protein fpVIII displaying cancer-targeting peptides can effectively encapsulate siRNAs and deliver them into the cells leading to specific silencing of the model gene GAPDH. Complexes of siRNA and phage protein form nanoparticles (nanophages), which were characterized by atomic force microscopy and ELISA, and their stability was demonstrated by resistance of encapsulated siRNA to degradation by serum nucleases. The phage protein/siRNA complexes can make a new type of highly selective, stable, active, and physiologically acceptable cancer nanomedicine.


Assuntos
Neoplasias da Mama/metabolismo , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Proteínas Virais/administração & dosagem , Bacteriófagos/genética , Bacteriófagos/metabolismo , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/química , Células Hep G2 , Humanos , Microscopia de Força Atômica , Microscopia de Fluorescência , Nanopartículas/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Proteínas Virais/química
13.
Nanomedicine ; 7(3): 315-23, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21050894

RESUMO

Efficacy of siRNAs as potential anticancer therapeutics can be increased by their targeted delivery into cancer cells via tumor-specific ligands. Phage display offers a unique approach to identify highly specific and selective ligands that can deliver nanocarriers to the site of disease. In this study, we proved a novel approach for intracellular delivery of siRNAs into breast cancer cells through their encapsulation into liposomes targeted to the tumor cells with preselected intact phage proteins. The targeted siRNA liposomes were obtained by a fusion of two parental liposomes containing spontaneously inserted siRNA and fusion phage proteins. The presence of pVIII coat protein fused to a MCF-7 cell-targeting peptide DMPGTVLP in the liposomes was confirmed by Western blotting. The novel phage-targeted siRNA-nanopharmaceuticals demonstrate significant down-regulation of PRDM14 gene expression and PRDM14 protein synthesis in the target MCF-7 cells. This approach offers the potential for development of new anticancer siRNA-based targeted nanomedicines. FROM THE CLINICAL EDITOR: In this study, the authors report a novel approach for targeted intracellular delivery of siRNAs into breast cancer cells through encapsulation into liposomes targeted to the tumor cells with preselected intact phage proteins.


Assuntos
Bacteriófagos/metabolismo , Neoplasias da Mama/metabolismo , Técnicas de Transferência de Genes , Lipossomos/química , Oligopeptídeos/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Virais de Fusão/metabolismo , Neoplasias da Mama/virologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Feminino , Inativação Gênica , Humanos , Especificidade de Órgãos , Tamanho da Partícula , Transporte Proteico , Proteínas de Ligação a RNA , Proteínas Repressoras/metabolismo , Eletricidade Estática , Fatores de Transcrição , Transcrição Gênica
14.
Nanomedicine ; 6(4): 538-46, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20138246

RESUMO

Tumor-specific cytotoxicity of drugs can be enhanced by targeting them to tumor receptors using tumor-specific ligands. Phage display offers a high-throughput approach to screen for the targeting ligands. We have successfully isolated phage fusion peptides selective and specific for PC3 prostate cancer cells. Also, we have demonstrated a novel approach of targeting liposomes through tumor-specific phage fusion coat proteins, exploiting the intrinsic properties of the phage coat protein as an integral membrane protein. Here we describe the production of Rhodamine-labeled liposomes as well as doxorubicin-loaded long-circulating liposomes targeted to PC3 prostate tumor cells via PC-specific phage peptides, as an extension of our previous studies. Targeting of labeled liposomes was demonstrated using fluorescence microscopy as well as flow cytometry. Targeting of doxorubicin-loaded liposomes enhanced their cytotoxic effect against PC3 cells in vitro, indicating a possible therapeutic advantage. The simplicity of the approach for generating targeted liposomes coupled with the ability to rapidly obtain tumor-specific phage fusion proteins via phage display may contribute to a combinatorial system for the production of targeted liposomal therapeutics for advanced stages of prostate tumor. From the clinical editor: This paper demonstrates targeting cytotoxic agents to tumor receptors using tumor-specific ligands. The authors describe the production of Rhodamine-labeled liposomes as well as doxorubicin loaded long circulating liposomes targeted to PC3 prostate tumor cells via PC-specific phage peptides. This approach may be especially relevant for advanced prostate tumors.


Assuntos
Antineoplásicos/administração & dosagem , Proteínas do Capsídeo/química , Lipossomos/química , Biblioteca de Peptídeos , Neoplasias da Próstata/tratamento farmacológico , Antineoplásicos/química , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Doxorrubicina/toxicidade , Humanos , Lipossomos/metabolismo , Masculino , Nanomedicina/métodos , Peptídeos/química , Peptídeos/metabolismo , Transporte Proteico , Rodaminas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...