Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Control Release ; 336: 443-456, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34186148

RESUMEN

Pyroglutamate-3 amyloid-ß (pGlu3 Aß) is an N-terminally modified, pathogenic form of amyloid-ß that is present in cerebral amyloid plaques and vascular deposits. Here, we used focused ultrasound (FUS) with microbubbles to enhance the intravenous delivery of an Fc-competent anti-pGlu3 Aß monoclonal antibody, 07/2a mAb, across the blood brain barrier (BBB) in an attempt to improve Aß removal and memory in aged APP/PS1dE9 mice, an Alzheimer's disease (AD)-like model of amyloidogenesis. First, we demonstrated that bilateral hippocampal FUS-BBB disruption (FUS-BBBD) led to a 5.5-fold increase of 07/2a mAb delivery to the brains compared to non-sonicated mice 72 h following a single treatment. Then, we determined that three weekly treatments with 07/2a mAb alone improved spatial learning and memory in aged, plaque-rich APP/PS1dE9 mice, and that this improvement occurred faster and in a higher percentage of animals when combined with FUS-BBBD. Mice given the combination treatment had reduced hippocampal plaque burden compared to PBS-treated controls. Furthermore, synaptic protein levels were higher in hippocampal synaptosomes from mice given the combination treatment compared to sham controls, and there were more CA3 synaptic puncta labeled in the APP/PS1dE9 mice given the combination treatment compared to those given mAb alone. Plaque-associated microglia were present in the hippocampi of APP/PS1dE9 mice treated with 07/2a mAb with and without FUS-BBBD. However, we discovered that plaque-associated Ly6G+ monocytes were only present in the hippocampi of APP/PS1dE9 mice that were given FUS-BBBD alone or even more so, the combination treatment. Lastly, FUS-BBBD did not increase the incidence of microhemorrhage in mice with or without 07/2a mAb treatment. Our findings suggest that FUS is a useful tool to enhance delivery and efficacy of an anti-pGlu3 Aß mAb for immunotherapy either via an additive effect or an independent mechanism. We revealed a potential novel mechanism wherein the combination of 07/2a mAb with FUS-BBBD led to greater monocyte infiltration and recruitment to plaques in this AD-like model. Overall, these effects resulted in greater plaque removal, sparing of synapses and improved cognitive function without causing overt damage, suggesting the possibility of FUS-BBBD as a noninvasive method to increase the therapeutic efficacy of drugs or biologics in AD patients.


Asunto(s)
Enfermedad de Alzheimer , Anciano , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Placa Amiloide , Ácido Pirrolidona Carboxílico
2.
Sci Rep ; 10(1): 3294, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32094456

RESUMEN

In clinical trials with early Alzheimer's patients, administration of anti-amyloid antibodies reduced amyloid deposits, suggesting that immunotherapies may be promising disease-modifying interventions against Alzheimer's disease (AD). Specific forms of amyloid beta (Aß) peptides, for example post-translationally modified Aß peptides with a pyroglutamate at the N-terminus (pGlu3, pE3), are attractive antibody targets, due to pGlu3-Aß's neo-epitope character and its propensity to form neurotoxic oligomeric aggregates. We have generated a novel anti-pGlu3-Aß antibody, PBD-C06, which is based on a murine precursor antibody that binds with high specificity to pGlu3-Aß monomers, oligomers and fibrils, including mixed aggregates of unmodified Aß and pGlu3-Aß peptides. PBD-C06 was generated by first grafting the murine antigen binding sequences onto suitable human variable light and heavy chains. Subsequently, the humanized antibody was de-immunized and site-specific mutations were introduced to restore original target binding, to eliminate complement activation and to improve protein stability. PBD-C06 binds with the same specificity and avidity as its murine precursor antibody and elimination of C1q binding did not compromise Fcγ-receptor binding or in vitro phagocytosis. Thus, PBD-C06 was specifically designed to target neurotoxic aggregates and to avoid complement-mediated inflammatory responses, in order to lower the risk for vasogenic edemas in the clinic.


Asunto(s)
Enfermedad de Alzheimer/terapia , Anticuerpos Monoclonales Humanizados/farmacología , Activación de Complemento , Inmunoterapia , Ácido Pirrolidona Carboxílico/química , Alelos , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/química , Animales , Complemento C1q/inmunología , Regiones Determinantes de Complementariedad , Edema/prevención & control , Endocitosis , Epítopos/química , Humanos , Inflamación , Ratones , Mutación , Fagocitosis , Unión Proteica , Procesamiento Proteico-Postraduccional
3.
Alzheimers Res Ther ; 12(1): 12, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31931873

RESUMEN

BACKGROUND: Pyroglutamate-3 Aß (pGlu-3 Aß) is an N-terminally truncated and post-translationally modified Aß species found in Alzheimer's disease (AD) brain. Its increased peptide aggregation propensity and toxicity make it an attractive emerging treatment strategy for AD. We address the question of how the effector function of an anti-pGlu-3 Aß antibody influences the efficacy of immunotherapy in mouse models with AD-like pathology. METHODS: We compared two different immunoglobulin (Ig) isotypes of the same murine anti-pGlu-3 Aß mAb (07/1 IgG1 and 07/2a IgG2a) and a general N-terminal Aß mAb (3A1 IgG1) for their ability to clear Aß and protect cognition in a therapeutic passive immunotherapy study in aged, plaque-rich APPSWE/PS1ΔE9 transgenic (Tg) mice. We also compared the ability of these antibodies and a CDC-mutant form of 07/2a (07/2a-k), engineered to avoid complement activation, to clear Aß in an ex vivo phagocytosis assay and following treatment in APPSLxhQC double Tg mice, and to activate microglia using longitudinal microPET imaging with TSPO-specific 18F-GE180 tracer following a single bolus antibody injection in young and old Tg mice. RESULTS: We demonstrated significant cognitive improvement, better plaque clearance, and more plaque-associated microglia in the absence of microhemorrhage in aged APPSWE/PS1ΔE9 Tg mice treated with 07/2a, but not 07/1 or 3A1, compared to PBS in our first in vivo study. All mAbs cleared plaques in an ex vivo assay, although 07/2a promoted the highest phagocytic activity. Compared with 07/2a, 07/2a-k showed slightly reduced affinity to Fcγ receptors CD32 and CD64, although the two antibodies had similar binding affinities to pGlu-3 Aß. Treatment of APPSLxhQC mice with 07/2a and 07/2a-k mAbs in our second in vivo study showed significant plaque-lowering with both mAbs. Longitudinal 18F-GE180 microPET imaging revealed different temporal patterns of microglial activation for 3A1, 07/1, and 07/2a mAbs and no difference between 07/2a-k and PBS-treated Tg mice. CONCLUSION: Our results suggest that attenuation of behavioral deficits and clearance of amyloid is associated with strong effector function of the anti-pGlu-3 Aß mAb in a therapeutic treatment paradigm. We present evidence that antibody engineering to reduce CDC-mediated complement binding facilitates phagocytosis of plaques without inducing neuroinflammation in vivo. Hence, the results provide implications for tailoring effector function of humanized antibodies for clinical development.


Asunto(s)
Enfermedad de Alzheimer , Vacunas contra el Alzheimer/farmacología , Péptidos beta-Amiloides/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacología , Neuroglía/efectos de los fármacos , Animales , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Inmunización Pasiva/métodos , Inmunoglobulina G , Ratones , Ratones Transgénicos , Procesamiento Proteico-Postraduccional , Ácido Pirrolidona Carboxílico/metabolismo
4.
Cancer Res ; 67(15): 7343-9, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671204

RESUMEN

The potassium channel ether à go-go has been directly linked to cellular proliferation and transformation, although its physiologic role(s) are as of yet unknown. The specific blockade of human Eag1 (hEag1) may not only allow the dissection of the role of the channel in distinct physiologic processes, but because of the implication of hEag1 in tumor biology, it may also offer an opportunity for the treatment of cancer. However, members of the potassium channel superfamily are structurally very similar to one another, and it has been notoriously difficult to obtain specific blockers for any given channel. Here, we describe and validate the first rational design of a monoclonal antibody that selectively inhibits a potassium current in intact cells. Specifically blocking hEag1 function using this antibody inhibits tumor cell growth both in vitro and in vivo. Our data provide a proof of concept that enables the generation of functional antagonistic monoclonal antibodies against ion channels with therapeutic potential. The particular antibody described here, as well as the technique developed to make additional functional antibodies to Eag1, makes it possible to evaluate the potential of the channel as a target for cancer therapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Canales de Potasio Éter-A-Go-Go/inmunología , Neoplasias Mamarias Experimentales/terapia , Neoplasias Pancreáticas/terapia , Bloqueadores de los Canales de Potasio/uso terapéutico , Animales , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Electrofisiología , Ensayo de Inmunoadsorción Enzimática , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Riñón/citología , Riñón/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones SCID , Mutagénesis Sitio-Dirigida , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Trasplante Heterólogo
5.
EBioMedicine ; 2(3): 264-71, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26137564

RESUMEN

BACKGROUND: During early clinical development, prospective identification of a predictive biomarker and validation of an assay method may not always be feasible. Dichotomizing a continuous biomarker measure to classify responders also leads to challenges. We present a case study of a prospective-retrospective approach for a continuous biomarker identified after patient enrollment but defined prospectively before the unblinding of data. An analysis of the strengths and weaknesses of this approach and the challenges encountered in its practical application are also provided. METHODS: HERALD (NCT02134015) was a double-blind, phase 2 study in patients with non-small cell lung cancer (NSCLC) randomized to erlotinib with placebo or with high or low doses of patritumab, a monoclonal antibody targeted against human epidermal growth factor receptor 3 (HER3). While the primary objective was to assess safety and progression-free survival (PFS), a secondary objective was to determine a single predictive biomarker hypothesis to identify subjects most likely to benefit from the addition of patritumab. Although not identified as the primary biomarker in the study protocol, on the basis of preclinical results from 2 independent laboratories, expression levels of the HER3 ligand heregulin (HRG) were prospectively declared the predictive biomarker before data unblinding but after subject enrollment. An assay to measure HRG mRNA was developed and validated. Other biomarkers, such as epidermal growth factor receptor (EGFR) mutation status, were also evaluated in an exploratory fashion. The cutoff value for high vs. low HRG mRNA levels was set at the median delta threshold cycle. A maximum likelihood analysis was performed to evaluate the provisional cutoff. The relationship of HRG values to PFS hazard ratios (HRs) was assessed as a measure of internal validation. Additional NSCLC samples were analyzed to characterize HRG mRNA distribution. RESULTS: The subgroup of patients with high HRG mRNA levels ("HRG-high") demonstrated clinical benefit from patritumab treatment with HRs of 0.37 (P = 0.0283) and 0.29 (P = 0.0027) in the high- and low-dose patritumab arms, respectively. However, only 102 of the 215 randomized patients (47.4%) had sufficient tumor samples for HRG mRNA measurement. Maximum likelihood analysis showed that the provisional cutoff was within the optimal range. In the placebo arm, the HRG-high subgroup demonstrated worse prognosis compared with HRG-low. A continuous relationship was observed between increased HRG mRNA levels and lower HR. Additional NSCLC samples (N = 300) demonstrated a similar unimodal distribution to that observed in this study, suggesting that the defined cutoff may be applicable to future NSCLC studies. CONCLUSIONS: The prospective-retrospective approach was successful in clinically validating a probable predictive biomarker. Post hoc in vitro studies and statistical analyses permitted further testing of the underlying assumptions. However, limitations of this analysis include the incomplete collection of adequate tumor tissue and a lack of stratification. In a phase 3 study, findings are being confirmed, and the HRG cutoff value is being further refined. CLINICALTRIALSGOV NUMBER: NCT02134015.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Neurregulina-1/genética , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Anticuerpos Neutralizantes/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Anticuerpos ampliamente neutralizantes , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Supervivencia sin Enfermedad , Método Doble Ciego , Receptores ErbB/genética , Clorhidrato de Erlotinib/administración & dosificación , Clorhidrato de Erlotinib/uso terapéutico , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Neurregulina-1/sangre , Estudios Prospectivos , Receptor ErbB-3/sangre , Receptor ErbB-3/inmunología , Estudios Retrospectivos , Investigación Biomédica Traslacional , Resultado del Tratamiento
6.
Immunol Lett ; 85(3): 297-300, 2003 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-12663146

RESUMEN

Memory T cells are essential for generating secondary immune responses and so provide long-lived protection from pathogens. The mechanisms that regulate the differentiation and survival of memory T cells are largely unknown. Transgenic mice in which NF-kappaB activity is inhibited by the expression of a dominant-negative form of IkappaB-alpha (mIkappaB-alpha mice) have drastically diminished numbers of CD8(+) memory-phenotype cells. The development of activated mIkappaB-alpha CD8 cells into memory-phenotype CD8 cells was severely impaired after adoptive transfer to lymphopenic hosts. Our findings demonstrate a critical role for NF-kappaB transcription factors in determining the number of memory-phenotype CD8 cells.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/fisiología , Memoria Inmunológica/fisiología , FN-kappa B/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Citometría de Flujo , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Memoria Inmunológica/inmunología , Ratones , Ratones Transgénicos , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores
7.
Nat Commun ; 5: 5237, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25354530

RESUMEN

Protein scaffolds can provide a promising alternative to antibodies for various biomedical and biotechnological applications, including therapeutics. Here we describe the design and development of the Alphabody, a protein scaffold featuring a single-chain antiparallel triple-helix coiled-coil fold. We report affinity-matured Alphabodies with favourable physicochemical properties that can specifically neutralize human interleukin (IL)-23, a pivotal therapeutic target in autoimmune inflammatory diseases such as psoriasis and multiple sclerosis. The crystal structure of human IL-23 in complex with an affinity-matured Alphabody reveals how the variable interhelical groove of the scaffold uniquely targets a large epitope on the p19 subunit of IL-23 to harness fully the hydrophobic and hydrogen-bonding potential of tryptophan and tyrosine residues contributed by p19 and the Alphabody, respectively. Thus, Alphabodies are suitable for targeting protein-protein interfaces of therapeutic importance and can be tailored to interrogate desired design and binding-mode principles via efficient selection and affinity-maturation strategies.


Asunto(s)
Interleucina-23/antagonistas & inhibidores , Péptidos/química , Secuencia de Aminoácidos , Animales , Línea Celular , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Péptidos/uso terapéutico , Psoriasis/prevención & control
8.
Clin Cancer Res ; 19(11): 3078-87, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23591447

RESUMEN

PURPOSE: HER3 is a key dimerization partner for other HER family members, and its expression is associated with poor prognosis. This first-in-human study of U3-1287 (NCT00730470), a fully human anti-HER3 monoclonal antibody, evaluated its safety, tolerability, and pharmacokinetics in patients with advanced solid tumor. EXPERIMENTAL DESIGN: The study was conducted in 2 parts: part 1--sequential cohorts received escalating doses (0.3-20 mg/kg) of U3-1287 every 2 weeks, starting 3 weeks after the first dose; part 2--additional patients received 9, 14, or 20 mg/kg U3-1287 every 2 weeks, based on observed tolerability and pharmacokinetics from part 1. Recommended phase II dose, adverse event rates, pharmacokinetics, and tumor response were determined. RESULTS: Fifty-seven patients (part 1: 26; part 2: 31) received U3-1287. As no dose-limiting toxicities were reported, the maximum-tolerated dose was not reached. The maximum-administered dose was 20 mg/kg every 2 weeks. The most frequent adverse events related to U3-1287 were fatigue (21.1%), diarrhea (12.3%), nausea (10.5%), decreased appetite (7.0%), and dysgeusia (5.3%). No patient developed anti-U3-1287 antibodies. In these heavily pretreated patients, stable disease was maintained 9 weeks or more in 19.2% in part 1 and 10 weeks or more in 25.8% in part 2. CONCLUSION: U3-1287 treatment was well tolerated, and some evidence of disease stabilization was observed. Pharmacokinetic data support U3-1287 dosing of 9 to 20 mg/kg every 2 to 3 weeks. Combination studies of U3-1287 are ongoing.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Anticuerpos Neutralizantes , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Biomarcadores/metabolismo , Anticuerpos ampliamente neutralizantes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/metabolismo , Pronóstico , Receptor ErbB-3/antagonistas & inhibidores , Resultado del Tratamiento
9.
Clin Cancer Res ; 17(11): 3673-85, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531812

RESUMEN

PURPOSE: The goal of the study was to engineer a form of interleukin 2 (IL-2) that, when delivered as a tumor-specific antibody fusion protein, retains the ability to stimulate an antitumor immune response via interaction with the high-affinity IL-2 receptor but has lower toxicity because of the reduced activation of the intermediate-affinity IL-2 receptor. EXPERIMENTAL DESIGN: We investigated changes in the proposed toxin motif of IL-2 by introducing a D20T mutation that has little effect on the activity of free IL-2. We expressed this IL-2 variant as a fusion protein with an antibody (NHS76) that targets the necrotic core of tumors and characterized this molecule (NHS-IL2LT) in vitro and in vivo. RESULTS: NHS-IL2LT was shown to have near normal biological activity in vitro by using T-cell lines expressing the high-affinity IL-2 receptor, but little or no activity by using cell lines expressing only the intermediate IL-2 receptor. Relative to the control antibody fusion protein containing wild-type IL-2, NHS-IL2LT retained antitumor activity against established neuroblastoma and non-small cell lung cancer metastases in syngeneic mouse tumor models but was much better tolerated in immune-competent mice and in cynomolgus monkeys. CONCLUSIONS: The qualities of low toxicity and single-agent efficacy shown suggest that NHS-IL2LT is a good candidate for therapeutic approaches combining standard cytotoxic and immune therapies. In fact, this molecule (also known as Selectikine or EMD 521873) is currently in phase I clinical trial.


Asunto(s)
Interleucina-2/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Región Variable de Inmunoglobulina/metabolismo , Interleucina-2/química , Interleucina-2/genética , Interleucina-2/inmunología , Macaca fascicularis , Ratones , Mieloma Múltiple/inmunología , Mutación , Neuroblastoma/inmunología , Ingeniería de Proteínas , Receptores de Interleucina-2/metabolismo , Proteínas Recombinantes de Fusión/metabolismo
10.
Mol Cell ; 11(3): 619-33, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12667446

RESUMEN

Eukaryotic cells respond to unfolded proteins in their endoplasmic reticulum (ER stress), amino acid starvation, or oxidants by phosphorylating the alpha subunit of translation initiation factor 2 (eIF2alpha). This adaptation inhibits general protein synthesis while promoting translation and expression of the transcription factor ATF4. Atf4(-/-) cells are impaired in expressing genes involved in amino acid import, glutathione biosynthesis, and resistance to oxidative stress. Perk(-/-) cells, lacking an upstream ER stress-activated eIF2alpha kinase that activates Atf4, accumulate endogenous peroxides during ER stress, whereas interference with the ER oxidase ERO1 abrogates such accumulation. A signaling pathway initiated by eIF2alpha phosphorylation protects cells against metabolic consequences of ER oxidation by promoting the linked processes of amino acid sufficiency and resistance to oxidative stress.


Asunto(s)
Aminoácidos/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Estrés Oxidativo , Factor de Transcripción Activador 4 , Animales , Antibacterianos/farmacología , Northern Blotting , Caenorhabditis elegans , División Celular , Separación Celular , Grupo Citocromo c/metabolismo , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/metabolismo , Factor 2 Eucariótico de Iniciación/química , Fibroblastos/metabolismo , Citometría de Flujo , Genotipo , Glutatión/metabolismo , Immunoblotting , Ratones , Microscopía Fluorescente , Mutación , Peroxidasa/metabolismo , Fosforilación , Pruebas de Precipitina , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tunicamicina/farmacología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA