Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Pharm Biopharm ; 193: 208-217, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37956784

RESUMEN

Photodynamic therapy (PDT) for deep-seated tumors is still challenging due to the limited penetration of visible light through tissues. To resolve this limitation, systems based on bioluminescence resonance energy transfer (BRET), that do not require an external light source are proposed. Herein, for BRET-activated PDT we developed proteinaceous BRET-pair consisting of luciferase NanoLuc, which acts as energy donor upon addition of luciferase specific substrate furimazine, and phototoxic protein SOPP3 as a photosensitizer. We have shown that hybrid protein NanoLuc-SOPP3 is an excellent BRET pair with BRET ratio of 1.12. Targeted delivery of NanoLuc-SOPP3 BRET pair via tumor-specific small liposomes (∼100 nm) to tumors overexpressing the HER2-receptor (human epidermal growth factor receptor 2) was demonstrated in vitro and in vivo. The proposed BRET-activated system has been shown to significantly suppress tumor growth in a model of subcutaneous and, more importantly, deep-seated tumor model. Taking into account the in vivo efficiency of proposed BRET-activated system, we believe that it has great potential for depth-independent PDT and can significantly broaden the application of PDT in the clinic.


Asunto(s)
Neoplasias , Fotoquimioterapia , Humanos , Liposomas , Luciferasas/genética , Luciferasas/metabolismo , Transferencia de Energía , Neoplasias/tratamiento farmacológico
2.
Cancers (Basel) ; 14(19)2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-36230729

RESUMEN

Peptide nucleic acid (PNA) may be used in various biomedical applications; however, these are currently limited, due to its low solubility in aqueous solutions. In this study, a methodology to overcome this limitation is demonstrated, as well as the effect of PNA on cell viability. We show that extruding a mixture of natural phospholipids and short (6-22 bases), cytosine-rich PNA through a 100 nm pore size membrane under mild acidic conditions resulted in the formation of small (60-90 nm in diameter) multilamellar vesicles (SMVs) comprising several (3-5) concentric lipid membranes. The PNA molecules, being positively charged under acidic conditions (due to protonation of cytosine bases in the sequence), bind electrostatically to negatively charged phospholipid membranes. The large membrane surface area allowed the encapsulation of thousands of PNA molecules in the vesicle. SMVs were conjugated with the designed ankyrin repeat protein (DARPin_9-29), which interacts with human epidermal growth factor receptor 2 (HER2), overexpressed in human breast cancer. The conjugate was shown to enter HER2-overexpressing cells by receptor-mediated endocytosis. PNA molecules, released from lysosomes, aggregate in the cytoplasm into micron-sized particles, which interfere with normal cell functioning, causing cell death. The ability of DARPin-functionalized SMVs to specifically deliver large quantities of PNA to cancer cells opens a new promising avenue for cancer therapy.

3.
Light Sci Appl ; 11(1): 38, 2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35190528

RESUMEN

Photodynamic therapy (PDT) is one of the most appealing photonic modalities for cancer treatment based on anticancer activity of light-induced photosensitizer-mediated reactive oxygen species (ROS), but a limited depth of light penetration into tissues does not make possible the treatment of deep-seated neoplasms and thus complicates its widespread clinical adoption. Here, we introduce the concept of genetically encoded bioluminescence resonance energy transfer (BRET)-activated PDT, which combines an internal light source and a photosensitizer (PS) in a single-genetic construct, which can be delivered to tumors seated at virtually unlimited depth and then triggered by the injection of a substrate to initiate their treatment. To illustrate the concept, we engineered genetic NanoLuc-miniSOG BRET pair, combining NanoLuc luciferase flashlight and phototoxic flavoprotein miniSOG, which generates ROS under luciferase-substrate injection. We prove the concept feasibility in mice bearing NanoLuc-miniSOG expressing tumor, followed by its elimination under the luciferase-substrate administration. Then, we demonstrate a targeted delivery of NanoLuc-miniSOG gene, via tumor-specific lentiviral particles, into a tumor, followed by its successful elimination, with tumor-growth inhibition (TGI) coefficient exceeding 67%, which confirms a great therapeutic potential of the proposed concept. In conclusion, this study provides proof-of-concept for deep-tissue "photodynamic" therapy without external light source that can be considered as an alternative for traditional PDT.

4.
Molecules ; 26(22)2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34833876

RESUMEN

Barnase is an extracellular ribonuclease secreted by Bacillus amyloliquefaciens that was originally studied as a small stable enzyme with robust folding. The identification of barnase intracellular inhibitor barstar led to the discovery of an incredibly strong protein-protein interaction. Together, barnase and barstar provide a fully genetically encoded toxin-antitoxin pair having an extremely low dissociation constant. Moreover, compared to other dimerization systems, the barnase-barstar module provides the exact one-to-one ratio of the complex components and possesses high stability of each component in a complex and high solubility in aqueous solutions without self-aggregation. The unique properties of barnase and barstar allow the application of this pair for the engineering of different variants of targeted anticancer compounds and cytotoxic supramolecular complexes. Using barnase in suicide gene therapy has also found its niche in anticancer therapy. The application of barnase and barstar in contemporary experimental cancer therapy is reflected in the review.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Ribonucleasas/metabolismo , Bacillus/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/fisiología , Humanos , Cinética , Modelos Moleculares , Nanotecnología/métodos , Neoplasias/tratamiento farmacológico , Conformación Proteica/efectos de los fármacos , Ribonucleasas/antagonistas & inhibidores , Ribonucleasas/fisiología
5.
Cancers (Basel) ; 13(20)2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34680384

RESUMEN

Near-infrared phototherapy has great therapeutic potential for cancer treatment. However, for efficient application, in vivo photothermal agents should demonstrate excellent stability in blood and targeted delivery to pathological tissue. Here, we demonstrated that stable bovine serum albumin-coated gold mini nanorods conjugated to a HER2-specific designed ankyrin repeat protein, DARPin_9-29, selectively accumulate in HER2-positive xenograft tumors in mice and lead to a strong reduction in the tumor size when being illuminated with near-infrared light. The results pave the way for the development of novel DARPin-based targeted photothermal therapy of cancer.

6.
Int J Mol Sci ; 22(9)2021 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-34067057

RESUMEN

Cancer cells frequently overexpress specific surface receptors providing tumor growth and survival which can be used for precise therapy. Targeting cancer cell receptors with protein toxins is an attractive approach widely used in contemporary experimental oncology and preclinical studies. Methods of targeted delivery of toxins to cancer cells, different drug carriers based on nanosized materials (liposomes, nanoparticles, polymers), the most promising designed light-activated toxins, as well as mechanisms of the cytotoxic action of the main natural toxins used in modern experimental oncology, are discussed in this review. The prospects of the combined therapy of tumors based on multimodal nanostructures are also discussed.


Asunto(s)
Neoplasias/tratamiento farmacológico , Toxinas Biológicas/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Humanos , Estrés Oxidativo/efectos de los fármacos , Toxinas Biológicas/efectos adversos
7.
ACS Omega ; 6(24): 16000-16008, 2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34179645

RESUMEN

Targeted drug delivery is one of the most intriguing and challenging issues in modern biomedicine. For active targeting, full-size IgG molecules (150 kDa) are usually used. Recent studies have revealed that small artificial polypeptide scaffolds such as DARPins (14 kDa) and affibodies (8 kDa) are much more promising tools for drug delivery due to their small size, artificial nature, low immunogenicity, and many other properties. However, there is no comparative information on the targeting abilities of scaffold polypeptides, which should be taken into account when developing drug delivery systems (DDSs). The present work is the first comprehensive study on the comparison of the effectiveness of different HER2-targeting proteins within the architecture of nanoparticles. Namely, we synthesized trimodal nanoparticles: magnetic, fluorescent, and directed toward HER2 oncomarker on cancer cells. The magnetic particles (MPs) were covalently modified with (i) full-size IgG, 150 kDa, (ii) DARPin_G3, 14 kDa, and (iii) affibody ZHER2:342, 8 kDa. We showed that the number of DARPin_G3 and affibody ZHER2:342 molecules conjugated to the nanoparticle surface are 10 and 40 times higher, respectively, than the corresponding value for trastuzumab. Using the methods of magnetic particle quantification (MPQ)-cytometry and confocal microscopy, we showed that all types of the obtained magnetic conjugates specifically labeled HER2-overexpressing cells. Namely, we demonstrated that particle binding to HER2-positive cells is 1113 ± 39 fg/cell for MP*trastuzumab, 1431 ± 186 fg/cell for MP*ZHER2:342, and 625±21 fg/cell for MP*DARPin_G3, which are 2.77, 2.75, and 2.30 times higher than the corresponding values for control HER2-negative cells. Thus, we showed that the smallest HER2-recognizing polypeptide affibody ZHER2:342 is more effective in terms of specificity and selectivity in nanoparticle-mediated cell labeling.

8.
Cancers (Basel) ; 12(10)2020 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-33081407

RESUMEN

We report here a combined anti-cancer therapy directed toward HER2 and EpCAM, common tumor-associated antigens of breast cancer cells. The combined therapeutic effect is achieved owing to two highly toxic proteins-a low immunogenic variant of Pseudomonas aeruginosa exotoxin A and ribonuclease Barnase from Bacillus amyloliquefaciens. The delivery of toxins to cancer cells was carried out by targeting designed ankyrin repeat proteins (DARPins). We have shown that both target agents efficiently accumulate in the tumor. Simultaneous treatment of breast carcinoma-bearing mice with anti-EpCAM fusion toxin based on LoPE and HER2-specific liposomes loaded with Barnase leads to concurrent elimination of primary tumor and metastases. Monotherapy with anti-HER2- or anti-EpCAM-toxins did not produce a comparable effect on metastases. The proposed approach can be considered as a promising strategy for significant improvement of cancer therapy.

9.
ACS Nano ; 14(10): 12781-12795, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32935975

RESUMEN

When combined with immunotherapy, image-guided targeted delivery of chemotherapeutic agents is a promising direction for combination cancer theranostics, but this approach has so far produced only limited success due to a lack of molecular targets on the cell surface and low therapeutic index of conventional chemotherapy drugs. Here, we demonstrate a synergistic strategy of combination immuno/chemotherapy in conditions of dual regioselective targeting, implying vectoring of two distinct binding sites of a single oncomarker (here, HER2) with theranostic compounds having a different mechanism of action. We use: (i) PLGA nanoformulation, loaded with an imaging diagnostic fluorescent dye (Nile Red) and a chemotherapeutic drug (doxorubicin), and functionalized with affibody ZHER2:342 (8 kDa); (ii) bifunctional genetically engineered DARP-LoPE (42 kDa) immunotoxin comprising of a low-immunogenic modification of therapeutic Pseudomonas exotoxin A (LoPE) and a scaffold targeting protein, DARPin9.29 (14 kDa). According to the proposed strategy, the first chemotherapeutic nanoagent is targeted by the affibody to subdomain III and IV of HER2 with 60-fold specificity compared with nontargeted particles, while the second immunotoxin is effectively targeted by DARPin molecule to subdomain I of HER2. We demonstrate that this dual targeting strategy can enhance anticancer therapy of HER2-positive cells with a very strong synergy, which made possible 1000-fold decrease of effective drug concentration in vitro and a significant enhancement of HER2 cancer therapy compared to monotherapy in vivo. Moreover, this therapeutic combination prevented the appearance of secondary tumor nodes. Thus, the suggested synergistic strategy utilizing dual targeting of the same oncomarker could give rise to efficient methods for aggressive tumors treatment.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Doxorrubicina/farmacología , Inmunoterapia , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Receptor ErbB-2
10.
ACS Appl Mater Interfaces ; 11(38): 34645-34651, 2019 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-31448887

RESUMEN

We have demonstrated that designed ankyrin repeat protein (DARPin) _9-29, which specifically targets human epidermal growth factor receptor 2 (HER2), binds tightly to gold mini nanorods (GNRs). Molecular dynamic simulations showed that a single layer of DARPin_9-29 molecules is formed on the surface of the nanorod and that conjugation with the nanorod does not involve the protein's domain responsible for specific binding to HER2. The nanorod-DARPin (DARPin-GNR) conjugate is specifically bound (in nanomolar concentrations) to human breast adenocarcinoma SK-BR-3 cells overexpressing HER2. Illumination by near-infrared light (850 nm) led to almost complete eradication of the conjugate-treated SK-BR-3 cells; the viability of epithelial human breast cancer cells expressing normal amounts of the receptor was much less affected by the illumination. The results reported here pave the way toward application of DARPin-GNR conjugates in phototherapy of cancer.


Asunto(s)
Adenocarcinoma , Neoplasias de la Mama , Sistemas de Liberación de Medicamentos , Oro , Nanopartículas del Metal , Nanotubos/química , Fototerapia , Receptor ErbB-2/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/terapia , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Femenino , Oro/química , Oro/farmacología , Humanos , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico
11.
Mol Pharm ; 16(3): 995-1008, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30608701

RESUMEN

Designed ankyrin repeat proteins (DARPins) are small engineered scaffold proteins that can be selected for binding to desirable molecular targets. High affinity and small size of DARPins render them promising probes for radionuclide molecular imaging. However, detailed knowledge on many factors influencing their imaging properties is still lacking. We have evaluated two human epidermal growth factor 2 (HER2)-specific DARPins with different size and binding properties. DARPins 9_29-H6 and G3-H6 were radiolabeled with iodine-125 and tricarbonyl technetium-99m and evaluated in vitro. A side-by-side comparison of biodistribution and tumor targeting was performed. HER2-specific tumor accumulation of G3-H6 was demonstrated. A combination of smaller size and higher affinity resulted in a higher tumor uptake of G3-H6 in comparison to 9_29-H6. Technetium-99m labeled G3-H6 demonstrated a better biodistribution profile than 9_29-H6, with several-fold lower uptake in liver. Radioiodinated G3-H6 showed the best tumor-to-organ ratios. The combined effect of affinity, molecular weight, scaffold composition, and nonresidualizing properties of iodine label provided radioiodinated G3-H6 with high clinical potential for imaging of HER2.


Asunto(s)
Repetición de Anquirina , Ancirinas/clasificación , Ancirinas/farmacocinética , Radioisótopos de Yodo/farmacocinética , Neoplasias/diagnóstico por imagen , Receptor ErbB-2/metabolismo , Tecnecio/farmacocinética , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Imagen Molecular , Neoplasias/patología , Unión Proteica , Cintigrafía , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Contrast Media Mol Imaging ; 2018: 6930425, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29977173

RESUMEN

High expression of human epidermal growth factor receptor 2 (HER2) in breast and gastroesophageal carcinomas is a predictive biomarker for treatment using HER2-targeted therapeutics (antibodies trastuzumab and pertuzumab, antibody-drug conjugate trastuzumab DM1, and tyrosine kinase inhibitor lapatinib). Radionuclide molecular imaging of HER2 expression might permit stratification of patients for HER2-targeting therapies. In this study, we evaluated a new HER2-imaging probe based on the designed ankyrin repeat protein (DARPin) 9_29. DARPin 9_29 was labeled with iodine-125 by direct radioiodination and with [99mTc]Tc(CO)3 using the C-terminal hexahistidine tag. DARPin 9_29 preserved high specificity and affinity of binding to HER2-expressing cells after labeling. Uptake of [125I]I-DARPin 9_29 and [99mTc]Tc(CO)3-DARPin 9_29 in HER2-positive SKOV-3 xenografts in mice at 6 h after injection was 3.4 ± 0.7 %ID/g and 2.9 ± 0.7 %ID/g, respectively. This was significantly (p < 0.00005) higher than the uptake of the same probes in HER2-negative Ramos lymphoma xenografts, 0.22 ± 0.09 %ID/g and 0.30 ± 0.05 %ID/g, respectively. Retention of [125I]I-DARPin 9_29 in the lung, liver, spleen, and kidneys was appreciably lower compared with [99mTc]Tc(CO)3-DARPin 9_29, which resulted in significantly (p < 0.05) higher tumor-to-organ ratios. The biodistribution data were confirmed by SPECT/CT imaging. In conclusion, radioiodine is a preferable label for DARPin 9_29.


Asunto(s)
Radioisótopos de Yodo , Imagen Molecular/métodos , Radiofármacos/normas , Receptor ErbB-2/análisis , Tecnecio , Animales , Repetición de Anquirina , Neoplasias de la Mama/química , Neoplasias de la Mama/diagnóstico por imagen , Unión Esofagogástrica/patología , Xenoinjertos , Humanos , Radioisótopos de Yodo/farmacocinética , Ratones , Imagen Molecular/normas , Neoplasias/química , Neoplasias/diagnóstico por imagen , Radiofármacos/farmacocinética , Tecnecio/farmacocinética , Distribución Tisular
13.
Eur J Pharm Biopharm ; 130: 296-305, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29959035

RESUMEN

Since their discovery, liposomes have been widely employed in biomedical research. These nano-size spherical vesicles consisting one or few phospholipid bilayers surrounding an aqueous core are capable of carrying a wide variety of bioactive compounds, including drugs, peptides, nucleic acids, proteins and others. Despite considerable success achieved in synthesis of liposome constructs containing bioactive compounds, preparation of ligand-targeted liposomes comprising large quantities of encapsulated proteins that are capable of affecting pathological cells still remains a big challenge. Here we described a novel method for preparation of small (80-90 nm in diameter) unilamellar liposomes containing very large quantities (thousands of protein molecules per liposome) of heme-containing cytochrome c, highly fluorescent mCherry and highly toxic PE40 (Pseudomonas aeruginosa Exotoxin A domain). Efficient encapsulation of the proteins was achieved through electrostatic interaction between positively charged proteins (at pH lower than pI) and negatively charged liposome membrane. The proteoliposomes containing large quantities of mCherry or PE40 and functionalized with designed ankyrin repeat protein (DARPin)_9-29, which targets human epidermal growth factor receptor 2 (HER2) were shown to specifically stain and kill in sub-nanomolar concentrations HER2-positive cells, overexpressing HER2, respectively. Specific staining and eradication of the receptor-positive cells demonstrated here makes the DARPin-functionalized liposomes carrying large quantities of fluorescent and/or toxic proteins a promising candidate for tumor detection and therapy.


Asunto(s)
ADP Ribosa Transferasas/administración & dosificación , Repetición de Anquirina/genética , Toxinas Bacterianas/administración & dosificación , Citocromos c/administración & dosificación , Exotoxinas/administración & dosificación , Proteínas Luminiscentes/administración & dosificación , Factores de Virulencia/administración & dosificación , ADP Ribosa Transferasas/química , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Toxinas Bacterianas/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Citocromos c/química , Exotoxinas/química , Femenino , Hemo/química , Humanos , Liposomas , Proteínas Luminiscentes/química , Neoplasias Ováricas , Tamaño de la Partícula , Receptor ErbB-2/metabolismo , Factores de Virulencia/química , Proteína Fluorescente Roja , Exotoxina A de Pseudomonas aeruginosa
14.
Bioconjug Chem ; 28(10): 2569-2574, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-28806065

RESUMEN

We demonstrate that the designed ankyrin repeat protein (DARPin)_9-29, which specifically targets human epidermal growth factor receptor 2 (HER 2), binds tightly to gold nanoparticles (GNPs). Binding of the protein strongly increases the colloidal stability of the particles. The results of experimental analysis and molecular dynamics simulations show that approximately 35 DARPin_9-29 molecules are bound to the surface of a 5 nm GNP and that the binding does not involve the receptor-binding domain of the protein. The confocal fluorescent microscopy studies show that the DARPin-coated GNP conjugate specifically interacts with the surface of human cancer cells overexpressing epidermal growth factor receptor 2 (HER2) and enters the cells by endocytosis. The high stability under physiological conditions and high affinity to the receptors overexpressed by cancer cells make conjugates of plasmonic gold nanostructures with DARPin molecules promising candidates for cancer therapy.


Asunto(s)
Repetición de Anquirina , Inmunoconjugados/química , Inmunoconjugados/metabolismo , Nanopartículas del Metal/química , Línea Celular Tumoral , Técnicas de Química Sintética , Humanos , Inmunoconjugados/inmunología , Modelos Moleculares , Receptor ErbB-2/inmunología
15.
J Control Release ; 233: 48-56, 2016 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-27178808

RESUMEN

DARPins fused with other proteins are promising non-immunoglobulin scaffolds for specific binding to target cells. In this study HER2-specific DARPin (DARPin_9-29) was used as a tumor-targeting moiety for the delivery of a cytotoxic agent - the fragment of Pseudomonas aeruginosa exotoxin A. It was determined that DARPin-PE40 possesses a considerable cytotoxic activity and induces apoptosis in HER2-positive cells. Cytotoxic effect of DARPin-PE40 strongly correlates with the HER2 expression level. The effect of intravenous administration of DARPin-PE40 was tested in the xenograft model of breast cancer. It was shown that treatment of animals with DARPin-PE40 caused strong and prolonged suppression of xenograft tumor growth.


Asunto(s)
ADP Ribosa Transferasas/administración & dosificación , Antineoplásicos/administración & dosificación , Toxinas Bacterianas/administración & dosificación , Exotoxinas/administración & dosificación , Inmunotoxinas/administración & dosificación , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Factores de Virulencia/administración & dosificación , ADP Ribosa Transferasas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Toxinas Bacterianas/uso terapéutico , Células CHO , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cricetulus , Exotoxinas/uso terapéutico , Femenino , Humanos , Inmunotoxinas/uso terapéutico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones Desnudos , Proteínas Recombinantes de Fusión/uso terapéutico , Carga Tumoral/efectos de los fármacos , Factores de Virulencia/uso terapéutico , Exotoxina A de Pseudomonas aeruginosa
16.
Theranostics ; 3(11): 831-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24312153

RESUMEN

Tumor-targeted delivery of cytotoxins presents considerable advantages over their passive transport. Chemical conjugation of cytotoxic module to antibody is limited due to insufficient reproducibility of synthesis, and recombinant immunotoxins are aimed to overcome this disadvantage. We obtained genetically encoded immunophotosensitizer 4D5scFv-miniSOG and evaluated its photocytotoxic effect in vitro. A single-chain variable fragment (scFv) of humanized 4D5 antibody was used as a targeting vehicle for selective recognition of the extracellular domain of human epidermal growth factor receptor 2 (HER2/neu) overexpressed in many human carcinomas. As a phototoxic module we used a recently described photoactivated fluorescent flavoprotein miniSOG. We found that recombinant protein 4D5scFv-miniSOG exerts a highly specific photo-induced cytotoxic effect on HER2/neu-positive human breast adenocarcinoma SK-BR-3 cells (IC50= 160 nM). We demonstrated that the 4D5scFv-miniSOG specifically binds to HER2-positive cells and internalizes via receptor-mediated endocytosis. Co-treatment of breast cancer cells with 4D5scFv-miniSOG and Taxol or junction opener protein JO-1 produced remarkable additive effects.


Asunto(s)
Antineoplásicos/farmacología , Flavoproteínas/farmacología , Inmunotoxinas/farmacología , Terapia Molecular Dirigida/métodos , Neoplasias/terapia , Fármacos Fotosensibilizantes/farmacología , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Cadena Única/farmacología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Flavoproteínas/genética , Humanos , Inmunotoxinas/genética , Concentración 50 Inhibidora , Luz , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/metabolismo
17.
Nucleic Acids Res ; 34(13): 3615-24, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16877568

RESUMEN

RNA polymerase III contains seventeen subunits in yeasts (Saccharomyces cerevisiae and Schizosaccharomyces pombe) and in human cells. Twelve of them are akin to the core RNA polymerase I or II. The five other are RNA polymerase III-specific and form the functionally distinct groups Rpc31-Rpc34-Rpc82 and Rpc37-Rpc53. Currently sequenced eukaryotic genomes revealed significant homology to these seventeen subunits in Fungi, Animals, Plants and Amoebozoans. Except for subunit Rpc31, this also extended to the much more distantly related genomes of Alveolates and Excavates, indicating that the complex subunit organization of RNA polymerase III emerged at a very early stage of eukaryotic evolution. The Sch.pombe subunits were expressed in S.cerevisiae null mutants and tested for growth. Ten core subunits showed heterospecific complementation, but the two largest catalytic subunits (Rpc1 and Rpc2) and all five RNA polymerase III-specific subunits (Rpc82, Rpc53, Rpc37, Rpc34 and Rpc31) were non-functional. Three highly conserved RNA polymerase III-specific domains were found in the twelve-subunit core structure. They correspond to the Rpc17-Rpc25 dimer, involved in transcription initiation, to an N-terminal domain of the largest subunit Rpc1 important to anchor Rpc31, Rpc34 and Rpc82, and to a C-terminal domain of Rpc1 that presumably holds Rpc37, Rpc53 and their Rpc11 partner.


Asunto(s)
Evolución Molecular , ARN Polimerasa III/genética , Saccharomyces cerevisiae/enzimología , Schizosaccharomyces/enzimología , Animales , Prueba de Complementación Genética , Humanos , Filogenia , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , ARN Polimerasa III/química , Saccharomyces cerevisiae/genética , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...