Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Allergy Clin Immunol ; 151(4): 966-975, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36592703

RESUMO

BACKGROUND: Type 2 endotype asthma is driven by IL-4 and IL-13 signaling via IL-4Ra, which is highly expressed on airway epithelium, airway smooth muscle, and immunocytes in the respiratory mucosa, suggesting potential advantages of an inhalable antagonist. Lipocalin 1 (Lcn1), a 16 kDa protein abundant in human periciliary fluid, has a robust drug-like structure well suited to protein engineering, but it has never been used to make an inhaled Anticalin protein therapeutic. OBJECTIVES: We sought to reengineer Lcn1 into an inhalable IL-4Ra antagonist and assess its pharmacodynamic/kinetic profile. METHODS: Lcn1 was systematically modified by directed protein mutagenesis yielding a high-affinity, slowly dissociating, long-acting full antagonist of IL-4Ra designated PRS-060 with properties analogous to dupilumab, competitively antagonizing IL-4Ra-dependent cell proliferation, mucus induction, and eotaxin expression in vitro. Because PRS-060 displayed exquisite specificity for human IL-4Ra, with no cross-reactivity to rodents or higher primates, we created a new triple-humanized mouse model substituting human IL-4Ra, IL-4, and IL-13 at their correct syntenic murine loci to model clinical dosing. RESULTS: Inhaled PRS-060 strongly suppressed acute allergic inflammation indexes in triple-humanized mice with a duration of action longer than its bulk clearance, suggesting that it may act locally in the lung. CONCLUSION: Lcn1 can be reengineered into the Anticalin antagonist PRS-060 (elarekibep), exemplifying a new class of inhaled topical, long-acting therapeutic drugs with the potential to treat type 2 endotype asthma.


Assuntos
Asma , Interleucina-13 , Animais , Humanos , Camundongos , Asma/tratamento farmacológico , Modelos Animais de Doenças , Interleucina-4/genética , Pulmão , Proteínas , Nebulizadores e Vaporizadores , Receptores de Interleucina-4/imunologia
2.
Toxicol Pathol ; 49(2): 235-260, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33455525

RESUMO

The inhalation route is a relatively novel drug delivery route for biotherapeutics and, as a result, there is a paucity of published data and experience within the toxicology/pathology community. In recent years, findings arising in toxicology studies with inhaled biologics have provoked concern and regulatory challenges due, in part, to the lack of understanding of the expected pathology, mechanisms, and adversity induced by this mode of delivery. In this manuscript, the authors describe 12 case studies, comprising 18 toxicology studies, using a range of inhaled biotherapeutics (monoclonal antibodies, fragment antigen-binding antibodies, domain antibodies, therapeutic proteins/peptides, and an oligonucleotide) in rodents, nonhuman primates (NHPs), and the rabbit in subacute (1 week) to chronic (26 weeks) toxicology studies. Analysis of the data revealed that many of these molecules were associated with a characteristic pattern of toxicity with high levels of immunogenicity. Microscopic changes in the airways consisted of a predominantly lymphoid perivascular/peribronchiolar (PV/PB) mononuclear inflammatory cell (MIC) infiltrate, whereas changes in the terminal airways/alveoli were characterized by simple ("uncomplicated") increases in macrophages or inflammatory cell infiltrates ranging from mixed inflammatory cell infiltration to inflammation. The PV/PB MIC changes were considered most likely secondary to immunogenicity, whereas simple increases in alveolar macrophages were most likely secondary to clearance mechanisms. Alveolar inflammatory cell infiltrates and inflammation were likely induced by immune modulation or stimulation through pharmacologic effects on target biology or type III hypersensitivity (immune complex disease). Finally, a group of experts provide introductory thoughts regarding the adversity of inhaled biotherapeutics and the basis for reasonable differences of opinion that might arise between toxicologists, pathologists, and regulators.


Assuntos
Produtos Biológicos , Hipersensibilidade , Administração por Inalação , Animais , Produtos Biológicos/efeitos adversos , Líquido da Lavagem Broncoalveolar , Inflamação , Pulmão , Macrófagos Alveolares , Coelhos
3.
Br J Pharmacol ; 175(7): 1054-1065, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29329501

RESUMO

BACKGROUND AND PURPOSE: Anaemia of chronic disease (ACD) has been linked to iron-restricted erythropoiesis imposed by high circulating levels of hepcidin, a 25 amino acid hepatocyte-derived peptide that controls systemic iron homeostasis. Here, we report the engineering of the human lipocalin-derived, small protein-based anticalin PRS-080 hepcidin antagonist with high affinity and selectivity. EXPERIMENTAL APPROACH: Anticalin- and hepcidin-specific pharmacokinetic (PK)/pharmacodynamic modelling (PD) was used to design and select the suitable drug candidate based on t1/2 extension and duration of hepcidin suppression. The development of a novel free hepcidin assay enabled accurate analysis of bioactive hepcidin suppression and elucidation of the observed plasma iron levels after PRS-080-PEG30 administration in vivo. KEY RESULTS: PRS-080 had a hepcidin-binding affinity of 0.07 nM and, after coupling to 30 kD PEG (PRS-080-PEG30), a t1/2 of 43 h in cynomolgus monkeys. Dose-dependent iron mobilization and hepcidin suppression were observed after a single i.v. dose of PRS-080-PEG30 in cynomolgus monkeys. Importantly, in these animals, suppression of free hepcidin and subsequent plasma iron elevation were sustained during repeated s.c. dosing. After repeated dosing and followed by a treatment-free interval, all iron parameters returned to pre-dose values. CONCLUSIONS AND IMPLICATIONS: In conclusion, we developed a dose-dependent and safe approach for the direct suppression of hepcidin, resulting in prolonged iron mobilization to alleviate iron-restricted erythropoiesis that can address the root cause of ACD. PRS-080-PEG30 is currently in early clinical development.


Assuntos
Hepcidinas/antagonistas & inibidores , Hepcidinas/sangue , Ferro/sangue , Animais , Feminino , Macaca fascicularis , Masculino , Modelos Biológicos
4.
Angiogenesis ; 19(1): 79-94, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26650228

RESUMO

Human tear lipocalin (Tlc) was utilized as a protein scaffold to engineer an Anticalin that specifically binds and functionally blocks vascular endothelial growth factor A (VEGF-A), a pivotal inducer of physiological angiogenesis that also plays a crucial role in several neovascular diseases. Starting from a naive combinatorial library where residues that form the natural ligand-binding site of Tlc were randomized, followed by affinity maturation, the final Anticalin PRS-050 was selected to bind all major splice forms of VEGF-A with picomolar affinity. Moreover, this Anticalin cross-reacts with the murine ortholog. PRS-050 efficiently antagonizes the interaction between VEGF-A and its cellular receptors, and it inhibits VEGF-induced mitogenic signaling as well as proliferation of primary human endothelial cells with subnanomolar IC50 values. Intravitreal administration of the Anticalin suppressed VEGF-induced blood-retinal barrier breakdown in a rabbit model. To allow lasting systemic neutralization of VEGF-A in vivo, the plasma half-life of the Anticalin was extended by site-directed PEGylation. The modified Anticalin efficiently blocked VEGF-mediated vascular permeability as well as growth of tumor xenografts in nude mice, concomitantly with reduction in microvessel density. In contrast to bevacizumab, the Anticalin did not trigger platelet aggregation and thrombosis in human FcγRIIa transgenic mice, thus suggesting an improved safety profile. Since neutralization of VEGF-A activity is well known to exert beneficial effects in cancer and other neovascular diseases, including wet age-related macular degeneration, this Anticalin offers a novel potent small protein antagonist for differentiated therapeutic intervention in oncology and ophthalmology.


Assuntos
Lipocalinas/farmacologia , Terapia de Alvo Molecular , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Complexo Antígeno-Anticorpo/metabolismo , Bevacizumab/farmacologia , Bevacizumab/uso terapêutico , Barreira Hematorretiniana/patologia , Permeabilidade Capilar , Proliferação de Células/efeitos dos fármacos , Feminino , Meia-Vida , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lipocalinas/farmacocinética , Lipocalinas/uso terapêutico , Camundongos Nus , Camundongos Transgênicos , Mitógenos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Polietilenoglicóis/química , Engenharia de Proteínas , Coelhos , Receptores de IgG/metabolismo , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Trombose/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
PLoS One ; 9(5): e94972, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24801709

RESUMO

Anticalins are a novel class of targeted protein therapeutics. The PEGylated Anticalin Angiocal (PRS-050-PEG40) is directed against VEGF-A. The purpose of our study was to compare the performance of diffusion weighted imaging (DWI), dynamic contrast enhanced magnetic resonance imaging (DCE)-MRI and positron emission tomography with the tracer [18F]fluorodeoxyglucose (FDG-PET) for monitoring early response to antiangiogenic therapy with PRS-050-PEG40. 31 mice were implanted subcutaneously with A673 rhabdomyosarcoma xenografts and underwent DWI, DCE-MRI and FDG-PET before and 2 days after i.p. injection of PRS-050-PEG40 (n = 13), Avastin (n = 6) or PBS (n = 12). Tumor size was measured manually with a caliper. Imaging results were correlated with histopathology. In the results, the tumor size was not significantly different in the treatment groups when compared to the control group on day 2 after therapy onset (P = 0.09). In contrast the imaging modalities DWI, DCE-MRI and FDG-PET showed significant differences between the therapeutic compared to the control group as early as 2 days after therapy onset (P<0.001). There was a strong correlation of the early changes in DWI, DCE-MRI and FDG-PET at day 2 after therapy onset and the change in tumor size at the end of therapy (r = -0.58, 0.71 and 0.67 respectively). The imaging results were confirmed by histopathology, showing early necrosis and necroptosis in the tumors. Thus multimodality multiparametric imaging was able to predict therapeutic success of PRS-050-PEG40 and Avastin as early as 2 days after onset of therapy and thus promising for monitoring early response of antiangiogenic therapy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Lipocalinas/uso terapêutico , Imagem Multimodal , Neovascularização Patológica/tratamento farmacológico , Animais , Bevacizumab , Feminino , Fluordesoxiglucose F18 , Imageamento por Ressonância Magnética , Camundongos , Neovascularização Patológica/diagnóstico , Neovascularização Patológica/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos
7.
PLoS One ; 8(10): e75518, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24124495

RESUMO

Mass spectrometry (MS)-based assays for the quantification of the iron regulatory hormone hepcidin are pivotal to discriminate between the bioactive 25-amino acid form that can effectively block the sole iron transporter ferroportin and other naturally occurring smaller isoforms without a known role in iron metabolism. Here we describe the design, validation and use of a novel stable hepcidin-25(+40) isotope as internal standard for quantification. Importantly, the relative large mass shift of 40 Da makes this isotope also suitable for easy-to-use medium resolution linear time-of-flight (TOF) platforms. As expected, implementation of hepcidin-25(+40) as internal standard in our weak cation exchange (WCX) TOF MS method yielded very low inter/intra run coefficients of variation. Surprisingly, however, in samples from kidney disease patients, we detected a novel peak (m/z 2673.9) with low intensity that could be identified as hepcidin-24 and had previously remained unnoticed due to peak interference with the formerly used internal standard. Using a cell-based bioassay it was shown that synthetic hepcidin-24 was, like the -22 and -20 isoforms, a significantly less potent inducer of ferroportin degradation than hepcidin-25. During prolonged storage of plasma at room temperature, we observed that a decrease in plasma hepcidin-25 was paralleled by an increase in the levels of the hepcidin-24, -22 and -20 isoforms. This provides first evidence that all determinants for the conversion of hepcidin-25 to smaller inactive isoforms are present in the circulation, which may contribute to the functional suppression of hepcidin-25, that is significantly elevated in patients with renal impairment. The present update of our hepcidin TOF MS assay together with improved insights in the source and preparation of the internal standard, and sample stability will further improve our understanding of circulating hepcidin and pave the way towards further optimization and standardization of plasma hepcidin assays.


Assuntos
Hepcidinas/sangue , Espectrometria de Massas/métodos , Isoformas de Proteínas/sangue , Humanos , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
8.
Mol Cancer Ther ; 12(11): 2459-71, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24002935

RESUMO

Activation of the MET oncogenic pathway has been implicated in the development of aggressive cancers that are difficult to treat with current chemotherapies. This has led to an increased interest in developing novel therapies that target the MET pathway. However, most existing drug modalities are confounded by their inability to specifically target and/or antagonize this pathway. Anticalins, a novel class of monovalent small biologics, are hypothesized to be "fit for purpose" for developing highly specific and potent antagonists of cancer pathways. Here, we describe a monovalent full MET antagonist, PRS-110, displaying efficacy in both ligand-dependent and ligand-independent cancer models. PRS-110 specifically binds to MET with high affinity and blocks hepatocyte growth factor (HGF) interaction. Phosphorylation assays show that PRS-110 efficiently inhibits HGF-mediated signaling of MET receptor and has no agonistic activity. Confocal microscopy shows that PRS-110 results in the trafficking of MET to late endosomal/lysosomal compartments in the absence of HGF. In vivo administration of PRS-110 resulted in significant, dose-dependent tumor growth inhibition in ligand-dependent (U87-MG) and ligand-independent (Caki-1) xenograft models. Analysis of MET protein levels on xenograft biopsy samples show a significant reduction in total MET following therapy with PRS-110 supporting its ligand-independent mechanism of action. Taken together, these data indicate that the MET inhibitor PRS-110 has potentially broad anticancer activity that warrants evaluation in patients.


Assuntos
Lipocalinas/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sítios de Ligação/efeitos dos fármacos , Células CHO , Linhagem Celular Tumoral , Cricetulus , Relação Dose-Resposta a Droga , Mapeamento de Epitopos , Feminino , Células HT29 , Fator de Crescimento de Hepatócito/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligantes , Lipocalinas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas/uso terapêutico , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
PLoS One ; 6(3): e17659, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21479271

RESUMO

Glaucoma, the most frequent optic neuropathy, is a leading cause of blindness worldwide. Death of retinal ganglion cells (RGCs) occurs in all forms of glaucoma and accounts for the loss of vision, however the molecular mechanisms that cause RGC loss remain unclear. The pro-apoptotic molecule, Fas ligand, is a transmembrane protein that can be cleaved from the cell surface by metalloproteinases to release a soluble protein with antagonistic activity. Previous studies documented that constitutive ocular expression of FasL maintained immune privilege and prevented neoangeogenesis. We now show that FasL also plays a major role in retinal neurotoxicity. Importantly, in both TNFα triggered RGC death and a spontaneous model of glaucoma, gene-targeted mice that express only full-length FasL exhibit accelerated RGC death. By contrast, FasL-deficiency, or administration of soluble FasL, protected RGCs from cell death. These data identify membrane-bound FasL as a critical effector molecule and potential therapeutic target in glaucoma.


Assuntos
Membrana Celular/metabolismo , Proteína Ligante Fas/metabolismo , Glaucoma/metabolismo , Glaucoma/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Animais , Morte Celular , Membrana Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Modelos Animais de Doenças , Proteína Ligante Fas/farmacologia , Glaucoma/complicações , Injeções , Camundongos , Camundongos Mutantes , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/metabolismo , Fibras Nervosas/patologia , Ligação Proteica/efeitos dos fármacos , Degeneração Retiniana/complicações , Degeneração Retiniana/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Solubilidade/efeitos dos fármacos , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/metabolismo
10.
Expert Rev Clin Immunol ; 3(4): 491-501, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20477155

RESUMO

Anticalins are engineered ligand-binding proteins based on the human lipocalin scaffold. Their architecture is characterized by a rigid beta-barrel that supports four structurally hypervariable loops. Similar to antibodies, these loops form the natural ligand-binding site, usually for vitamins, hormones or secondary metabolites. Anticalins with novel specificities can be engineered by reshaping this loop region, using targeted random mutagenesis in combination with functional display and guided selection. Several drug candidates with specificities for exogenous low-molecular-weight substances, peptides and even protein targets (e.g., several disease-related cell surface receptors) have been obtained in this way. Owing to their exquisite specificity and high affinity, Anticalins are particularly attractive as antagonists for the manipulation of immune mechanisms, leading to either inhibitory or stimulatory effects. Compared with antibodies, Anticalins offer several practical advantages as they are much smaller, consist of a single polypeptide chain and can be produced easily in microbial expression systems.

11.
J Immunol ; 172(11): 6598-606, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15153474

RESUMO

A number of studies have documented a critical role for tumor-specific CD4(+) cells in the augmentation of immunotherapeutic effector mechanisms. However, in the context of an extensive tumor burden, chronic stimulation of such CD4(+) T cells often leads to the up-regulation of both Fas and Fas ligand, and coexpression of these molecules can potentially result in activation-induced cell death and the subsequent loss of effector activity. To evaluate the importance of T cell persistence in an experimental model of immunotherapy, we used DO11 Th1 cells from wild-type, Fas-deficient, and Fas ligand-deficient mice as effector populations specific for a model tumor Ag consisting of an OVA-derived transmembrane fusion protein. We found that the prolonged survival of Fas-deficient DO11 Th1 cells led to a more sustained tumor-specific response both in vitro and in vivo. Importantly, both Fas- and Fas ligand-deficient Th1 cells delayed tumor growth and cause regression of established tumors more effectively than wild-type Th1 cells, indicating that resistance to activation-induced cell death significantly enhances T cell effector activity.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/imunologia , Ativação Linfocitária , Neoplasias Experimentais/imunologia , Animais , Linhagem Celular , Proteína Ligante Fas , Feminino , Masculino , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Células Th1/imunologia , Receptor fas/fisiologia
12.
Clin Immunol ; 103(1): 1-6, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11987979

RESUMO

Experimental and physiological expression of the pro-apoptotic molecule Fas-ligand can induce inflammation under certain conditions. Discussed here are the experimental situations, possible mechanisms, and pathways that mediate this response.


Assuntos
Glicoproteínas de Membrana/fisiologia , Animais , Apoptose , Caspase 8 , Caspase 9 , Caspases/fisiologia , Proteína Ligante Fas , Sobrevivência de Enxerto , Humanos , Inflamação/etiologia
13.
Nature ; 416(6881): 603-7, 2002 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-11948342

RESUMO

Autoreactive B cells are present in the lymphoid tissues of healthy individuals, but typically remain quiescent. When this homeostasis is perturbed, the formation of self-reactive antibodies can have serious pathological consequences. B cells expressing an antigen receptor specific for self-immunoglobulin-gamma (IgG) make a class of autoantibodies known as rheumatoid factor (RF). Here we show that effective activation of RF+ B cells is mediated by IgG2a-chromatin immune complexes and requires the synergistic engagement of the antigen receptor and a member of the MyD88-dependent Toll-like receptor (TLR) family. Inhibitor studies implicate TLR9. These data establish a critical link between the innate and adaptive immune systems in the development of systemic autoimmune disease and explain the preponderance of autoantibodies reactive with nucleic acid-protein particles. The unique features of this dual-engagement pathway should facilitate the development of therapies that specifically target autoreactive B cells.


Assuntos
Linfócitos B/imunologia , Cromatina/imunologia , Proteínas de Drosophila , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Fator Reumatoide/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Complexo Antígeno-Anticorpo , Antígenos de Diferenciação/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Desoxirribonuclease I/metabolismo , Camundongos , Fator 88 de Diferenciação Mieloide , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Complemento 3b/deficiência , Receptores de Complemento 3b/imunologia , Receptores de Complemento 3d/deficiência , Receptores de Complemento 3d/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais , Receptor Toll-Like 9 , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...