Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Lancet Neurol ; 23(1): 37-45, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38101901

RESUMEN

BACKGROUND: Converging lines of evidence suggest that microglia are relevant to Parkinson's disease pathogenesis, justifying exploration of therapeutic agents thought to attenuate pathogenic microglial function. We sought to test the safety and efficacy of NLY01-a brain-penetrant, pegylated, longer-lasting version of exenatide (a glucagon-like peptide-1 receptor agonist) that is believed to be anti-inflammatory via reduction of microglia activation-in Parkinson's disease. METHODS: We report a 36-week, randomised, double-blind, placebo-controlled study of NLY01 in participants with early untreated Parkinson's disease conducted at 58 movement disorder clinics in the USA. Participants meeting UK Brain Bank or Movement Disorder Society research criteria for Parkinson's disease were randomly allocated (1:1:1) to one of two active treatment groups (2·5 mg or 5·0 mg NLY01) or matching placebo, based on a central computer-generated randomisation scheme using permuted block randomisation with varying block sizes. All participants, investigators, coordinators, study staff, and sponsor personnel were masked to treatment assignments throughout the study. The primary efficacy endpoint for the primary analysis population (defined as all randomly assigned participants who received at least one dose of study drug) was change from baseline to week 36 in the sum of Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS) parts II and III. Safety was assessed in the safety population (all randomly allocated participants who received at least one dose of the study drug) with documentation of adverse events, vital signs, electrocardiograms, clinical laboratory assessments, physical examination, and scales for suicidality, sleepiness, impulsivity, and depression. This trial is complete and registered at ClinicalTrials.gov, NCT04154072. FINDINGS: The study took place between Jan 28, 2020, and Feb 16, 2023. 447 individuals were screened, of whom 255 eligible participants were randomly assigned (85 to each study group). One patient assigned to placebo did not receive study treatment and was not included in the primary analysis. At 36 weeks, 2·5 mg and 5·0 mg NLY01 did not differ from placebo with respect to change in sum scores on MDS-UPDRS parts II and III: difference versus placebo -0·39 (95% CI -2·96 to 2·18; p=0·77) for 2·5 mg and 0·36 (-2·28 to 3·00; p=0·79) for 5·0 mg. Treatment-emergent adverse events were similar across groups (reported in 71 [84%] of 85 patients on 2·5 mg NLY01, 79 [93%] of 85 on 5·0 mg, and 73 [87%] of 84 on placebo), with gastrointestinal disorders the most commonly observed class in active groups (52 [61%] for 2·5 mg, 64 [75%] for 5·0 mg, and 30 [36%] for placebo) and nausea the most common event overall (33 [39%] for 2·5 mg, 49 [58%] for 5·0 mg, and 16 [19%] for placebo). No deaths occurred during the study. INTERPRETATION: NLY01 at 2·5 and 5·0 mg was not associated with any improvement in Parkinson's disease motor or non-motor features compared with placebo. A subgroup analysis raised the possibility of motor benefit in younger participants. Further study is needed to determine whether these exploratory observations are replicable. FUNDING: D&D Pharmatech-Neuraly.


Asunto(s)
Exenatida , Agonistas Receptor de Péptidos Similares al Glucagón , Enfermedad de Parkinson , Humanos , Método Doble Ciego , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/complicaciones , Resultado del Tratamiento , Exenatida/análogos & derivados , Exenatida/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Agonistas Receptor de Péptidos Similares al Glucagón/uso terapéutico
2.
Nat Commun ; 10(1): 1128, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850660

RESUMEN

Scleroderma is an autoimmune rheumatic disorder accompanied by severe fibrosis in skin and other internal organs. During scleroderma progression, resident fibroblasts undergo activation and convert to α-smooth muscle actin (α-SMA) expressing myofibroblasts (MFBs) with increased capacity to synthesize collagens and fibrogenic components. Accordingly, MFBs are a major therapeutic target for fibrosis in scleroderma and treatment with blocking MFBs could produce anti-fibrotic effects. TLY012 is an engineered human TNF-related apoptosis-inducing ligand (TRAIL) which induces selective apoptosis in transformed cells expressing its cognate death receptors (DRs). Here we report that TLY012 selectively blocks activation of dermal fibroblasts and induces DR-mediated apoptosis in α-SMA+ MFBs through upregulated DR5 during its activation. In vivo, TLY012 reverses established skin fibrosis to near-normal skin architecture in mouse models of scleroderma. Thus, the TRAIL pathway plays a critical role in tissue remodeling and targeting upregulated DR5 in α-SMA+ MFBs is a viable therapy for fibrosis in scleroderma.


Asunto(s)
Actinas/genética , Dermis/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Esclerodermia Sistémica/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Actinas/metabolismo , Adulto , Anciano , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Diferenciación Celular , Colágeno/genética , Colágeno/metabolismo , Dermis/metabolismo , Dermis/patología , Modelos Animales de Enfermedad , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Persona de Mediana Edad , Terapia Molecular Dirigida , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ingeniería de Proteínas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/patología , Transducción de Señal
3.
Nat Med ; 24(7): 931-938, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29892066

RESUMEN

Activation of microglia by classical inflammatory mediators can convert astrocytes into a neurotoxic A1 phenotype in a variety of neurological diseases1,2. Development of agents that could inhibit the formation of A1 reactive astrocytes could be used to treat these diseases for which there are no disease-modifying therapies. Glucagon-like peptide-1 receptor (GLP1R) agonists have been indicated as potential neuroprotective agents for neurologic disorders such as Alzheimer's disease and Parkinson's disease3-13. The mechanisms by which GLP1R agonists are neuroprotective are not known. Here we show that a potent, brain-penetrant long-acting GLP1R agonist, NLY01, protects against the loss of dopaminergic neurons and behavioral deficits in the α-synuclein preformed fibril (α-syn PFF) mouse model of sporadic Parkinson's disease14,15. NLY01 also prolongs the life and reduces the behavioral deficits and neuropathological abnormalities in the human A53T α-synuclein (hA53T) transgenic mouse model of α-synucleinopathy-induced neurodegeneration16. We found that NLY01 is a potent GLP1R agonist with favorable properties that is neuroprotective through the direct prevention of microglial-mediated conversion of astrocytes to an A1 neurotoxic phenotype. In light of its favorable properties, NLY01 should be evaluated in the treatment of Parkinson's disease and related neurologic disorders characterized by microglial activation.


Asunto(s)
Astrocitos/patología , Microglía/patología , Fármacos Neuroprotectores/metabolismo , Enfermedad de Parkinson/patología , Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , alfa-Sinucleína/metabolismo
4.
MAbs ; 5(2): 208-18, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23575268

RESUMEN

The recognition that few human diseases are thoroughly addressed by mono-specific, monoclonal antibodies (mAbs) continues to drive the development of antibody therapeutics with additional specificities and enhanced activity. Historically, efforts to engineer additional antigen recognition into molecules have relied predominantly on the reformatting of immunoglobulin domains. In this report we describe a series of fully functional mAbs to which additional specificities have been imparted through the recombinant fusion of relatively short polypeptides sequences. The sequences are selected for binding to a particular target from combinatorial libraries that express linear, disulfide-constrained, or domain-based structures. The potential for fusion of peptides to the N- and C- termini of both the heavy and light chains affords the bivalent expression of up to four different peptides. The resulting molecules, called zybodies, can gain up to four additional specificities, while retaining the original functionality and specificity of the scaffold antibody. We explore the use of two clinically significant oncology antibodies, trastuzumab and cetuximab, as zybody scaffolds and demonstrate functional enhancements in each case. The affect of fusion position on both peptide and scaffold function is explored, and penta-specific zybodies are demonstrated to simultaneously engage five targets (ErbB2, EGFR, IGF-1R, Ang2 and integrin αvß3). Bispecific, trastuzumab-based zybodies targeting ErbB2 and Ang2 are shown to exhibit superior efficacy to trastuzumab in an angiogenesis-dependent xenograft tumor model. A cetuximab-based bispecific zybody that targeting EGFR and ErbB3 simultaneously disrupted multiple intracellular signaling pathways; inhibited tumor cell proliferation; and showed efficacy superior to that of cetuximab in a xenograft tumor model.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos , Neoplasias/terapia , Péptidos/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Secuencia de Aminoácidos , Angiopoyetina 2/química , Angiopoyetina 2/genética , Angiopoyetina 2/inmunología , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Proliferación Celular/efectos de los fármacos , Cetuximab , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Datos de Secuencia Molecular , Neovascularización Patológica , Péptidos/genética , Péptidos/inmunología , Péptidos/metabolismo , Ingeniería de Proteínas/métodos , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Transducción de Señal , Trastuzumab , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Addict Biol ; 18(1): 30-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22264200

RESUMEN

Although substantial research effort has focused on developing pharmacological treatments for cocaine abuse, no effective medications have been developed. Recent studies show that enzymes that metabolize cocaine in the periphery, forestalling its entry into the brain, can prevent cocaine toxicity and its behavioral effects in rodents. Here we report on effects of one such enzyme (Albu-CocH) on the pharmacokinetic and behavioral effects of cocaine in squirrel monkeys. Albu-CocH was developed from successive mutations of human butyrylcholinesterase (BChE) and has 1000-fold greater catalytic activity against cocaine than naturally occurring BChE. Pharmacokinetic studies showed that Albu-CocH (5 mg/kg) had a half-life of 56.6 hours in squirrel monkeys. In these studies, plasma levels of cocaine following i.v. 1 mg/kg cocaine were reduced 2 hours after administration of Albu-CocH, whereas plasma levels of the cocaine metabolite ecgonine methyl ester were increased. These effects were still evident 72 hours following Albu-CocH administration. In behavioral experiments in monkeys, pre-treatment with 5 mg/kg Albu-CocH dramatically decreased self-administration of a reinforcing dose of i.v. cocaine (30 µg/kg/injection) for over 24 hours. Pre-treatment with 5 mg/kg Albu-CocH also attenuated the reinstatement of extinguished cocaine self-administration by an i.v. priming injection of cocaine (0.1 or 0.3 mg/kg) and, in separate studies, attenuated the discriminative-stimulus effects of cocaine. The ability of Albu-CocH to attenuate the abuse-related effects of cocaine in squirrel monkeys indicates that further investigation of BChE mutants as potential treatment for cocaine abuse and toxicity is warranted.


Asunto(s)
Albúminas/farmacología , Butirilcolinesterasa/farmacología , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Cocaína/farmacocinética , Inhibidores de Captación de Dopamina/farmacocinética , Albúminas/farmacocinética , Análisis de Varianza , Animales , Formación de Anticuerpos/efectos de los fármacos , Biocatálisis , Butirilcolinesterasa/farmacocinética , Cocaína/administración & dosificación , Cocaína/antagonistas & inhibidores , Aprendizaje Discriminativo/efectos de los fármacos , Inhibidores de Captación de Dopamina/administración & dosificación , Inhibidores de Captación de Dopamina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Semivida , Humanos , Masculino , Refuerzo en Psicología , Saimiri , Autoadministración
6.
MAbs ; 4(5): 600-13, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22864384

RESUMEN

Despite the clinical success of anti-tumor necrosis factor (TNF) therapies in the treatment of inflammatory conditions such as rheumatoid arthritis, Crohn disease and psoriasis, full control of the diseases only occurs in a subset of patients and there is a need for new therapeutics with improved efficacy against broader patient populations. One possible approach is to combine biological therapeutics, but both the cost of the therapeutics and the potential for additional toxicities needs to be considered. In addition to the various mediators of immune and inflammatory pathways, angiogenesis is reported to contribute substantially to the overall pathogenesis of inflammatory diseases. The combination of an anti-angiogenic agent with anti-TNF into one molecule could be more efficacious without the risk of severe immunosuppression. To evaluate this approach with our Zybody technology, we generated bispecific antibodies that contain an Ang2 targeting peptide genetically fused to the anti-TNF antibody adalimumab (Humira®). The bispecific molecules retain the binding and functional characteristics of the anti-TNF antibody, but with additional activity that neutralizes Ang2. In a TNF transgenic mouse model of arthritis, the bispecific anti-TNF-Ang2 molecules showed a dose-dependent reduction in both clinical symptoms and histological scores that were significantly better than that achieved by adalimumab alone.


Asunto(s)
Angiopoyetina 2/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Factor de Necrosis Tumoral alfa/inmunología , Adalimumab , Angiopoyetina 2/genética , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales Humanizados/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/terapia , Línea Celular , Modelos Animales de Enfermedad , Humanos , Inflamación/terapia , Ratones , Ratones Transgénicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Resultado del Tratamiento
7.
J Immunol ; 178(11): 7325-33, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17513783

RESUMEN

TNF-like cytokine (TL1A) is a newly identified member of the TNF superfamily of ligands that is important for T cell costimulation and Th1 polarization. However, despite increasing information about its functions, very little is known about expression of TL1A in normal or pathological states. In this study, we report that mononuclear phagocytes appear to be a major source of TL1A in rheumatoid arthritis (RA), as revealed by their strong TL1A expression in either synovial fluids or synovial tissue of rheumatoid factor (RF)-seropositive RA patients, but not RF-/RA patients. Accordingly, in vitro experiments revealed that human monocytes express and release significant amounts of soluble TL1A when stimulated with insoluble immune complexes (IC), polyethylene glycol precipitates from the serum of RF+/RA patients, or with insoluble ICs purified from RA synovial fluids. Monocyte-derived soluble TL1A was biologically active as determined by its capacity to induce apoptosis of the human erythroleukemic cell line TF-1, as well as to cooperate with IL-12 and IL-18 in inducing the production of IFN-gamma by CD4(+) T cells. Because RA is a chronic inflammatory disease with autoimmune etiology, in which ICs, autoantibodies (including RF), and various cytokines contribute to its pathology, our data suggest that TL1A could be involved in its pathogenesis and contribute to the severity of RA disease that is typical of RF+/RA patients.


Asunto(s)
Complejo Antígeno-Anticuerpo/fisiología , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Complejo Antígeno-Anticuerpo/biosíntesis , Complejo Antígeno-Anticuerpo/aislamiento & purificación , Artritis Reumatoide/patología , Línea Celular Tumoral , Células Cultivadas , Citocinas/fisiología , Humanos , Inmunoprecipitación , Monocitos/química , Monocitos/patología , Polietilenglicoles/metabolismo , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/fisiología , Solubilidad , Líquido Sinovial/química , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Membrana Sinovial/química , Membrana Sinovial/inmunología , Membrana Sinovial/patología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/sangre , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
8.
J Biol Chem ; 280(26): 25111-8, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15851468

RESUMEN

Bone morphogenetic proteins (BMPs), a subset of the transforming growth factor (TGF)-beta superfamily, regulate a diverse array of cellular functions during development and in the adult. BMP-9 (also known as growth and differentiation factor (GDF)-2) potently induces osteogenesis and chondrogenesis, has been implicated in the differentiation of cholinergic neurons, and may help regulate glucose metabolism. We have determined the structure of BMP-9 to 2.3 A and examined the differences between our model and existing crystal structures of other BMPs, both in isolation and in complex with their receptors. TGF-beta ligands are translated as precursors, with pro-regions that generally dissociate after cleavage from the ligand, but in some cases (including GDF-8 and TGF-beta1, -2, and -3), the pro-region remains associated after secretion from the cell and inhibits binding of the ligand to its receptor. Although the proregion of BMP-9 remains tightly associated after secretion, we find, in several cell-based assays, that the activities of BMP-9 and BMP-9.pro-region complex were equivalent. Activin receptor-like kinase 1 (ALK-1), an orphan receptor in the TGF-beta family, was also identified as a potential receptor for BMP-9 based on surface plasmon resonance studies (BIAcore) and the ability of soluble ALK-1 to block the activity of BMP-9.pro-region complex in cell-based assays.


Asunto(s)
Proteínas Morfogenéticas Óseas/química , Cristalografía por Rayos X/métodos , Células 3T3-L1 , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo II , Fosfatasa Alcalina/metabolismo , Secuencia de Aminoácidos , Animales , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 6 , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Condrogénesis , Cromatografía , Electroforesis en Gel de Poliacrilamida , Genes Reporteros , Glucosa/metabolismo , Factor 2 de Diferenciación de Crecimiento , Ligandos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Miostatina , Neuronas/metabolismo , Osteogénesis , Unión Proteica , Ratas , Homología de Secuencia de Aminoácido , Transducción de Señal , Resonancia por Plasmón de Superficie , Factor de Crecimiento Transformador beta/metabolismo
9.
J Mol Recognit ; 18(1): 94-102, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15382264

RESUMEN

B lymphocyte stimulator (BLyS) is a tumor necrosis factor (TNF) family member and a key regulator of B cell responses. We employed a phage display-based approach to identify peptides that bind BLyS with high selectivity and affinity. Sequence analysis of first-generation BLyS-binding peptides revealed two dominant peptide motifs, including one containing a conserved DxLT sequence. Selected linear peptides with this motif were found to bind BLyS with K(D) values of 1-3 microM. In order to improve the binding affinity for BLyS, consensus residues flanking the DxLT sequence were seeded into a second-generation, BLyS affinity maturation library (BAML). BAML phage were subjected to stringent binding competition conditions to select for isolates expressing high-affinity peptide ligands for BLyS. Post-selection analysis of BAML peptide sequences resulted in the identification of a core decapeptide motif (WYDPLTKLWL). Peptides containing this core motif exhibited K(D) values as low as 26 nM, approximately 100-fold lower than that of first-generation peptides. A fluorescence anisotropy assay was developed to monitor the protein-protein interaction between BLyS labeled with a ruthenium chelate, and TACI-Fc, a soluble form of a BLyS receptor. Using this assay it was found that a BAML peptide disrupts this high-affinity protein-protein interaction. This demonstrates the potential of short peptides for disruption of high affinity cytokine-receptor interactions.


Asunto(s)
Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Biblioteca de Péptidos , Péptidos/química , Péptidos/aislamiento & purificación , Receptores del Factor de Necrosis Tumoral/química , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/química , Secuencias de Aminoácidos , Factor Activador de Células B , Bioensayo , Secuencia Conservada , Disulfuros/química , Polarización de Fluorescencia , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Péptidos/metabolismo , Mapeo de Interacción de Proteínas/métodos , Receptores del Factor de Necrosis Tumoral/fisiología , Rutenio/química , Proteína Activadora Transmembrana y Interactiva del CAML , Factor de Necrosis Tumoral alfa/metabolismo
10.
Blood ; 105(2): 830-7, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15358625

RESUMEN

We have recently shown that granulocyte-colony-stimulating factor (G-CSF)- and interferon-gamma (IFN-gamma)-activated human neutrophils accumulate and release remarkable amounts of soluble B-lymphocyte stimulator (BLyS) in vitro. In this study, we provide evidence that neutrophils migrating into skin window exudates (SWEs) developed in healthy volunteers and in patients with rheumatoid arthritis (RA), synthesized, and released BLyS in response to locally produced G-CSF. Accordingly, the concentrations of soluble BLyS in SWEs were significantly more elevated than in serum. Because the levels of SWE BLyS, but not SWE G-CSF, were higher in patients with RA than in healthy subjects, we examined the effect of CXCL8/IL-8, C5a, and other proinflammatory mediators that dramatically accumulate in RA SWEs and in inflamed synovial fluids. We show that CXCL1/GROalpha, CXCL8/IL-8, C5a, immune complexes, tumor necrosis factor-alpha (TNF-alpha), leukotriene B4, N-formyl-methionyl-leucyl-phenylalanine (fMLP), and lipopolysaccharide (LPS), which by themselves do not induce BLyS de novo synthesis, act as potent secretagogues for BLyS, which is mainly stored in Golgi-related compartments within G-CSF-treated neutrophils, as determined by immunogold electron microscopy. This action is pivotal in greatly amplifying neutrophil-dependent BLyS release in SWEs of patients with RA compared with healthy subjects. Collectively, our data uncover a novel mechanism that might dramatically exacerbate the release of BLyS by neutrophils during pathologic inflammatory responses.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Mediadores de Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , Neutrófilos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Anciano , Factor Activador de Células B , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Piel/citología , Piel/inmunología , Piel/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
11.
Arthritis Rheum ; 48(12): 3475-86, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14673998

RESUMEN

OBJECTIVE: To assess the overexpression of B lymphocyte stimulator (BLyS) over time in patients with systemic lupus erythematosus (SLE). METHODS: Sixty-eight SLE patients were followed up longitudinally for a median 369 days. At each physician encounter, disease activity was assessed by the Systemic Lupus Erythematosus Disease Activity Index, and blood was collected for determination of the serum BLyS level, blood BLyS messenger RNA (mRNA) level, and cell surface BLyS expression. Twenty normal control subjects underwent similar laboratory evaluations. RESULTS: In contrast to the uniformly normal serum BLyS and blood BLyS mRNA phenotypes in control subjects, SLE patients displayed marked heterogeneity, with 50% and 61% of patients manifesting persistently or intermittently elevated serum BLyS and blood BLyS mRNA phenotypes, respectively. Surface BLyS expression by SLE peripheral blood mononuclear cells was also often increased. Treatment of patients who had elevated serum BLyS levels with intensive courses of high-dose corticosteroids resulted in marked reductions in serum BLyS levels, and tapering of the corticosteroid dosage often resulted in increases in serum BLyS levels. Serum BLyS levels generally correlated with anti-double-stranded DNA (anti-dsDNA) titers (in those with detectable anti-dsDNA titers), but changes in serum BLyS levels did not correlate with changes in disease activity in individual patients. Serum BLyS phenotype did not associate with specific organ system involvement. CONCLUSION: Dysregulation of BLyS over extended periods of time is common in patients with SLE. Neutralization of BLyS activity with an appropriate BLyS antagonist may be therapeutically beneficial.


Asunto(s)
Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/fisiopatología , Proteínas de la Membrana/genética , Factor de Necrosis Tumoral alfa/genética , Corticoesteroides/uso terapéutico , Adulto , Anciano , Antígenos de Superficie/genética , Autoanticuerpos/sangre , Factor Activador de Células B , ADN/inmunología , Femenino , Expresión Génica/inmunología , Humanos , Estudios Longitudinales , Lupus Eritematoso Sistémico/tratamiento farmacológico , Masculino , Proteínas de la Membrana/sangre , Persona de Mediana Edad , ARN Mensajero/sangre
12.
Arthritis Rheum ; 48(11): 3253-65, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14613291

RESUMEN

OBJECTIVE: To identify and characterize a fully human antibody directed against B lymphocyte stimulator (BLyS), a tumor necrosis factor-related cytokine that plays a critical role in the regulation of B cell maturation and development. Elevated levels of BLyS have been implicated in the pathogenesis of autoimmune diseases. METHODS: A human phage display library was screened for antibodies against human BLyS. A human monoclonal antibody, LymphoStat-B, specific for human BLyS was obtained from the library screening and subsequent affinity optimization mutagenesis. The antibody was tested for inhibition of human BLyS in vitro and in an in vivo murine model. Additionally, the consequences of BLyS inhibition were tested in vivo by administration of LymphoStat-B to cynomolgus monkeys. RESULTS: LymphoStat-B bound with high affinity to human BLyS and inhibited the binding of BLyS to its 3 receptors, TACI, BCMA, and BLyS receptor 3/BAFF-R. LymphoStat-B potently inhibited BLyS-induced proliferation of B cells in vitro, and administration of LymphoStat-B to mice prevented human BLyS-induced increases in splenic B cell numbers and IgA titers. In cynomolgus monkeys, administration of LymphoStat-B resulted in decreased B cell representation in both spleen and mesenteric lymph nodes. CONCLUSION: A fully human monoclonal antibody has been isolated that binds to BLyS with high affinity and neutralizes human BLyS bioactivity in vitro and in vivo. Administration of this antibody to cynomolgus monkeys resulted in B cell depletion in spleen and lymph node. This antibody may prove therapeutically useful in the treatment of autoimmune diseases in humans.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Linfocitos B/metabolismo , Proteínas de la Membrana , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Receptor del Factor Activador de Células B , Antígeno de Maduración de Linfocitos B , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inyecciones Intravenosas , Leucocitos Mononucleares/efectos de los fármacos , Ganglios Linfáticos/citología , Ganglios Linfáticos/efectos de los fármacos , Macaca fascicularis , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Pruebas de Neutralización , Receptores del Factor de Necrosis Tumoral/administración & dosificación , Receptores del Factor de Necrosis Tumoral/inmunología , Bazo/citología , Bazo/efectos de los fármacos , Proteína Activadora Transmembrana y Interactiva del CAML
13.
Arthritis Rheum ; 48(4): 982-92, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12687540

RESUMEN

OBJECTIVE: To determine whether synovial fluid (SF) levels and cell-surface expression of B lymphocyte stimulator (BLyS) protein and SF levels of APRIL are elevated in patients with inflammatory arthritis (IA). METHODS: Same-day blood and SF samples from 89 patients with 103 knee effusions (81 knees with IA and 22 with noninflammatory arthritis [NIA]) were evaluated for BLyS protein and APRIL levels by enzyme-linked immunosorbent assay. Blood and SF mononuclear cells were double-stained for surface BLyS protein and surface CD14 (monocyte marker) and were analyzed by flow cytometry. Complete blood cell counts and SF nucleated cell counts were performed by the clinical hematology laboratory. RESULTS: BLyS protein levels were higher in SF than in corresponding serum samples from both IA and NIA patients. SF BLyS protein levels, but not surface expression of BLyS protein, were disproportionately elevated in IA patients. APRIL levels were higher in SF than in corresponding serum samples from most IA patients but not NIA patients. SF BLyS protein and APRIL levels correlated with each other, and each correlated with SF monocyte, lymphocyte, neutrophil, and total nucleated cell counts. Although SF and serum BLyS protein levels correlated with each other, SF and serum APRIL levels did not, suggesting that SF BLyS protein levels are more dependent upon systemic factors than are SF APRIL levels. Moreover, in 8 patients who underwent sequential arthrocenteses, changes in SF BLyS protein levels did not immutably parallel changes in SF APRIL levels, indicating their differential regulation. CONCLUSION: BLyS protein and APRIL are locally produced in inflamed joints. Their respective SF levels are differentially regulated, suggesting that they serve different functions. Together, their local production may foster survival and/or expansion of B cells that produce pathogenic autoantibodies and/or promote local T cell activation and consequent joint destruction.


Asunto(s)
Artritis Reumatoide/metabolismo , Articulación de la Rodilla/metabolismo , Proteínas de la Membrana/metabolismo , Neuropéptidos/metabolismo , Proteínas Nucleares/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Factor Activador de Células B , Recuento de Células , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/citología , Monocitos/metabolismo , Líquido Sinovial/citología , Líquido Sinovial/metabolismo
14.
Nat Biotechnol ; 21(3): 302-7, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12598909

RESUMEN

A coordinated effort combining bioinformatic tools with high-throughput cell-based screening assays was implemented to identify novel factors involved in T-cell biology. We generated a unique library of cDNAs encoding predicted secreted and transmembrane domain-containing proteins generated by analyzing the Human Genome Sciences cDNA database with a combination of two algorithms that predict signal peptides. Supernatants from mammalian cells transiently transfected with this library were incubated with primary T cells and T-cell lines in several high-throughput assays. Here we describe the discovery of a T cell factor, TIP (T cell immunomodulatory protein), which does not show any homology to proteins with known function. Treatment of primary human and murine T cells with TIP in vitro resulted in the secretion of IFN-gamma, TNF-alpha, and IL-10, whereas in vivo TIP had a protective effect in a mouse acute graft-versus-host disease (GVHD) model. Therefore, combining functional genomics with high-throughput cell-based screening is a valuable and efficient approach to identifying immunomodulatory activities for novel proteins.


Asunto(s)
Enfermedad Injerto contra Huésped/tratamiento farmacológico , Análisis de Secuencia de Proteína/métodos , Factores Supresores Inmunológicos/administración & dosificación , Factores Supresores Inmunológicos/química , Linfocitos T/metabolismo , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica/métodos , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Humanos , Riñón/química , Riñón/embriología , Riñón/inmunología , Ratones/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Factores Supresores Inmunológicos/genética , Factores Supresores Inmunológicos/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Linfocitos T/química , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Transfección/métodos
15.
J Immunol ; 169(8): 4314-21, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12370363

RESUMEN

BLyS and APRIL are two members of the TNF superfamily that are secreted by activated myeloid cells and have costimulatory activity on B cells. BLyS and APRIL share two receptors, TACI and BCMA, whereas a third receptor, BAFF-R, specifically binds BLyS. Both BLyS and APRIL have been described as homotrimeric molecules, a feature common to members of the TNF superfamily. In this study, we show that APRIL and BLyS can form active heterotrimeric molecules when coexpressed and that circulating heterotrimers are present in serum samples from patients with systemic immune-based rheumatic diseases. These findings raise the possibility that active BLyS/APRIL heterotrimers may play a role in rheumatic and other autoimmune diseases and that other members of the TNF ligand superfamily may also form active soluble heterotrimers.


Asunto(s)
Linfocitos B/inmunología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/fisiología , Receptores del Factor de Necrosis Tumoral/biosíntesis , Receptores del Factor de Necrosis Tumoral/fisiología , Enfermedades Reumáticas/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/fisiología , Animales , Artritis Psoriásica/sangre , Artritis Psoriásica/inmunología , Artritis Reactiva/sangre , Artritis Reactiva/inmunología , Artritis Reumatoide/sangre , Artritis Reumatoide/inmunología , Receptor del Factor Activador de Células B , Linfocitos B/metabolismo , Línea Celular , Células Cultivadas , Femenino , Humanos , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Activación de Linfocitos/genética , Proteínas de la Membrana/sangre , Proteínas de la Membrana/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Polimiositis/sangre , Polimiositis/inmunología , Receptores del Factor de Necrosis Tumoral/sangre , Receptores del Factor de Necrosis Tumoral/aislamiento & purificación , Enfermedades Reumáticas/sangre , Espondilitis Anquilosante/sangre , Espondilitis Anquilosante/inmunología , Transfección , Células Tumorales Cultivadas , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/aislamiento & purificación
16.
Cancer Chemother Pharmacol ; 50(3): 202-12, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12203102

RESUMEN

PURPOSE: Repifermin (keratinocyte growth factor-2, KGF-2) is a growth factor that selectively induces epithelial cell proliferation, differentiation and migration. The objective of this study was to assess the effect of repifermin on in vitro tumor cell proliferation and in vivo tumor growth using a variety of human carcinoma cell lines with differing growth rates and levels of KGF receptor (KGFR) expression. METHODS: Potential effects of repifermin on in vitro cell proliferation were evaluated by alamarBlue and/or [(3)H]-thymidine incorporation assays under a range of serum conditions. In vivo tumor growth was evaluated by implanting KGFR(+) carcinomas subcutaneously into nude mice and measuring tumor growth over time in mice injected intravenously (i.v.) or intraperitoneally (i.p.) with repifermin or placebo. RESULTS: In vitro, none of the 30 human carcinoma cell lines tested demonstrated a substantial increase in proliferation in response to repifermin over the concentration range 0.01 to 1000 ng/ml. In vivo results showed no significant tumor growth-promoting activity when single- or multiple-cycle intravenous injections of repifermin (1 mg/kg) were given to athymic nude mice inoculated with human KGFR(+) tumors of the pharynx (Detroit 562, FaDu), colon (Caco-2), salivary gland (A-253) or tongue (SCC-25, CAL 27). In addition, repifermin (0.2 or 2 mg/kg) injected i.p. for 2 weeks had no effect on the growth of eight other human carcinomas including those of the ovary (NIH:OVCAR-3, SK-OV 3, PA-1), bladder (SCaBER), epidermis (A 431), lung (SW 900), breast (MDA-MB-231) and cervix (SiHa). CONCLUSIONS: Repifermin had no in vitro or in vivo proliferative effects on KGFR(+) human epithelial-like tumors. This failure to stimulate tumor cell growth highlights the ability of repifermin to specifically target normal epithelial tissue. This is critical to the safety profile of repifermin, since it is currently in phase II clinical trials for the treatment of cancer patients with mucositis resulting from chemo- or radiotherapy.


Asunto(s)
Carcinoma/patología , División Celular/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/biosíntesis , Animales , Células Epiteliales/efectos de los fármacos , Factor 10 de Crecimiento de Fibroblastos , Humanos , Ratones , Ratones Desnudos , Neoplasias Experimentales , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/efectos de los fármacos , Células Tumorales Cultivadas
17.
Clin Immunol ; 104(2): 115-22, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12165272

RESUMEN

To assess the helper T cell dependence of B lymphocyte stimulator (BLyS) protein-driven autoantibody production in vivo, serum levels of BLyS protein, total IgG, and anti-IgG anti-phospholipid (aPhL) autoantibodies from HIV-infected patients (n = 105) with varying degrees of CD4+ cell depletion and healthy control donors at low risk for HIV (n = 64) were determined. Peripheral blood mononuclear cells from these subjects were stained for surface expression of BLyS protein. Monocyte surface expression and serum levels of BLyS protein were increased in HIV-infected patients as were serum total IgG and IgG aPhL autoantibody levels. No associations were detected between increased serum BLyS protein levels and patient age, sex, disease duration, history of opportunistic infection or malignancy, or serum total IgG levels. However, serum levels of IgG aPhL autoantibodies were greater in patients with high serum BLyS protein levels than in those with normal serum BLyS protein levels. Importantly, this association between serum levels of BLyS protein and IgG aPhL was appreciated only in patients who were not severely CD4+ cell-depleted and not in patients who were severely CD4+ cell-depleted (peripheral blood CD4+ cell counts

Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , VIH-1 , VIH-2 , Leucocitos Mononucleares/inmunología , Proteínas de la Membrana/análisis , Factor de Necrosis Tumoral alfa/análisis , Adulto , Anciano , Autoanticuerpos/sangre , Factor Activador de Células B , Recuento de Linfocito CD4 , Estudios de Cohortes , Femenino , Infecciones por VIH/sangre , Humanos , Inmunoglobulina G/sangre , Masculino , Proteínas de la Membrana/sangre , Persona de Mediana Edad , Fosfolípidos/inmunología
18.
AAPS PharmSci ; 4(2): E8, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12102617

RESUMEN

Repifermin (truncated, recombinant human keratinocyte growth factor-2, KGF-2) was evaluated in cynomolgus monkeys and healthy humans during a phase 1 trial. Monkeys received vehicle or repifermin at 20, 75, or 200 microg/kg IV or 750 microg/kg subcutaneous (SC) daily for 29 days. Clinical observations were made during the entire dosing period. Gross and microscopic changes were assessed at necropsy. Pharmacokinetic parameters and immunogenicity were evaluated in these monkeys and in humans, following a single or 7 daily IV bolus injections of 1, 5, 25, or 50 microg/kg repifermin. In monkeys, repifermin was well tolerated, and histologic evaluation demonstrated dose-dependent, reversible thickening of the mucosa throughout the alimentary tract, except for the stomach. In the alimentary tract tissues, nonepithelial tissues were not affected, indicating a specificity of repifermin for epithelial cells. Pharmacokinetics in both monkeys and humans were dose proportional, showed lack of drug accumulation with repeated daily dosing, and were characterized by high volumes of distribution and clearance rates, indicating substantial tissue binding and metabolism. Repifermin was not markedly immunogenic following multiple daily IV injections in either species. Serum repifermin concentrations in humans were comparable to those attained in monkeys that produced significant pharmacological effects on epithelial cells in the alimentary tract. These findings provide additional support for the ongoing clinical development of repifermin for diseases involving epithelial injury.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos/farmacocinética , Adolescente , Adulto , Anciano , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Femenino , Factor 10 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Humanos , Inyecciones Intravenosas , Inyecciones Subcutáneas , Macaca fascicularis , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...