Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Front Immunol ; 12: 785229, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899752

RESUMEN

The complement system has long been recognized as a potential druggable target for a variety of inflammatory conditions. Very few complement inhibitors have been approved for clinical use, but a great number are in clinical development, nearly all of which systemically inhibit complement. There are benefits of targeting complement inhibition to sites of activation/disease in terms of efficacy and safety, and here we describe P-selectin targeted complement inhibitors, with and without a dual function of directly blocking P-selectin-mediated cell-adhesion. The constructs are characterized in vitro and in murine models of hindlimb ischemia/reperfusion injury and hindlimb transplantation. Both constructs specifically targeted to reperfused hindlimb and provided protection in the hindlimb ischemia/reperfusion injury model. The P-selectin blocking construct was the more efficacious, which correlated with less myeloid cell infiltration, but with similarly reduced levels of complement deposition. The blocking construct also improved tissue perfusion and, unlike the nonblocking construct, inhibited coagulation, raising the possibility of differential application of each construct, such as in thrombotic vs. hemorrhagic conditions. Similar outcomes were obtained with the blocking construct following vascularized composite graft transplantation, and treatment also significantly increased graft survival. This is outcome may be particularly pertinent in the context of vascularized composite allograft transplantation, since reduced ischemia reperfusion injury is linked to a less rigorous alloimmune response that may translate to the requirement of a less aggressive immunosuppressive regime for this normally nonlife-threatening procedure. In summary, we describe a new generation of targeted complement inhibitor with multi-functionality that includes targeting to vascular injury, P-selectin blockade, complement inhibition and anti-thrombotic activity. The constructs described also bound to both mouse and human P-selectin which may facilitate potential translation.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Inactivadores del Complemento/administración & dosificación , Miembro Posterior/irrigación sanguínea , Miembro Posterior/trasplante , Selectina-P/antagonistas & inhibidores , Receptores de Complemento 3b/administración & dosificación , Daño por Reperfusión/prevención & control , Anticuerpos de Cadena Única/administración & dosificación , Alotrasplante Compuesto Vascularizado , Animales , Inactivadores del Complemento/farmacocinética , Modelos Animales de Enfermedad , Fibrinolíticos/administración & dosificación , Supervivencia de Injerto/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Selectina-P/metabolismo , Flujo Sanguíneo Regional , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Transducción de Señal , Anticuerpos de Cadena Única/farmacocinética
2.
Biochem Pharmacol ; 192: 114724, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34371003

RESUMEN

The COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has quickly spread around the globe. At present, there is no precise and effective treatment for the patients with COVID-19, so rapid development of drugs is urgently needed in order to contain the highly infectious disease. The virus spike protein (S protein) can recognize the angiotensin-converting enzyme 2 (ACE2) receptor on the host cell membrane and undergo a series of conformational changes, protease cleavage and membrane fusion to complete the virus entry, so S protein is an important target for vaccine and drug development. Here we provide a brief overview of molecular mechanisms of virus entry, as well as some potential antiviral agents that act on S/ACE2 protein-protein interaction. Specifically, we focused on experimentally validated and/or computational prediction identified inhibitors that target SARS-CoV-2 S protein, ACE2 and enzymes associated with viral infection. This review offers valuable information for the discovery and development of potential antiviral agents in combating SARS-CoV-2. In addition, with the deepening understanding of the mechanism of SARS-CoV-2 infection, more targeted prevention and treatment drugs will be explored with the aid of the advanced technology in the future.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/administración & dosificación , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/inmunología , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/prevención & control , Ácido Glicirretínico/administración & dosificación , Humanos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , SARS-CoV-2/inmunología , Anticuerpos de Cadena Única/administración & dosificación , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/inmunología
3.
Front Immunol ; 12: 715719, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34413859

RESUMEN

The interaction of the Fc region of therapeutic antibodies and antibody-drug conjugates with Fcγ receptors (FcγRs) can lead to unpredictable and severe side effects. Over the last decades several strategies have been developed to overcome this drawback, including extensive Fc- and glycoengineering and antibody isotype switching. However, these approaches result in permanently Fc-silenced antibody derivates which partially or completely lack antibody-mediated effector functions. Nevertheless, for a majority of antibody-based drugs, Fc-mediated effector functions, like antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP) as well as complement-dependent cytotoxicity (CDC), represent the most substantial modes of action. We argued that a new strategy combining the beneficial properties of Fc-silencing and controlled activation of effector functions can pave the way to potent antibody therapeutics, reducing the FcγRs-mediated off-target toxicity. We present a novel Fc-tamed antibody format, where the FcγR-binding sites of antibodies are blocked by anti-isotypic masking units, hindering the association of FcγR and complement component 1 (c1q) to the Fc domain. The masking units were genetically fused to trastuzumab, including a protease-addressable peptide-liker. Our Fc-tamed antibodies demonstrated completely abolished interaction to soluble high-affinity Fcγ-Receptor I and c1q. In reporter cell-based ADCC assays, our Fc-tamed antibodies exhibited a 2,700 to 7,100-fold reduction in activation, compared to trastuzumab. Upon demasking by a tumor-associated protease, the Fc-activated antibodies demonstrated restored FcγR-binding, c1q-binding and the ability to induce potent ADCC activation. Furthermore, cell killing assays using donor-derived NK cells were performed to validate the functionality of the Fc-tamed antibody variants. To our knowledge, this approach represents the first non-permanently Fc-silenced antibody, which can be re-activated by a tumor-associated protease, eventually extending the field of novel antibody formats.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Fragmentos Fc de Inmunoglobulinas/inmunología , Péptido Hidrolasas/metabolismo , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/efectos adversos , Biomarcadores , Línea Celular Tumoral , Pollos , Humanos , Hidrólisis , Metaloproteinasa 9 de la Matriz/metabolismo , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/metabolismo , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/efectos adversos , Anticuerpos de Cadena Única/farmacología , Trastuzumab/administración & dosificación , Trastuzumab/efectos adversos , Trastuzumab/farmacología
4.
Theranostics ; 11(13): 6293-6314, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995659

RESUMEN

Radioimmunotherapy (RIT) is FDA-approved for the clinical management of liquid malignancies, however, its use for solid malignancies remains a challenge. The putative benefit of RIT lies in selective targeting of antigens expressed on the tumor surface using monoclonal antibodies, to systemically deliver cytotoxic radionuclides. The past several decades yielded dramatic improvements in the quality, quantity, recent commercial availability of alpha-, beta- and Auger Electron-emitting therapeutic radiometals. Investigators have created new or improved existing bifunctional chelators. These bifunctional chelators bind radiometals and can be coupled to antigen-specific antibodies. In this review, we discuss approaches to develop radiometal-based RITs, including the selection of radiometals, chelators and antibody platforms (i.e. full-length, F(ab')2, Fab, minibodies, diabodies, scFv-Fc and nanobodies). We cite examples of the performance of RIT in the clinic, describe challenges to its implementation, and offer insights to address gaps toward translation.


Asunto(s)
Radioinmunoterapia/métodos , Radiofármacos/uso terapéutico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/metabolismo , Antineoplásicos Inmunológicos/uso terapéutico , Quelantes/administración & dosificación , Quelantes/metabolismo , Química Clic , Ensayos Clínicos como Asunto , Fraccionamiento de la Dosis de Radiación , Sistemas de Liberación de Medicamentos , Predicción , Humanos , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Linfoma no Hodgkin/radioterapia , Ratones , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/radioterapia , Especificidad de Órganos , Medicina de Precisión , Tolerancia a Radiación , Radiofármacos/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/uso terapéutico , Anticuerpos de Dominio Único/administración & dosificación , Anticuerpos de Dominio Único/uso terapéutico , Radioisótopos de Itrio/administración & dosificación , Radioisótopos de Itrio/uso terapéutico
5.
Nat Commun ; 12(1): 444, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33469002

RESUMEN

Glioblastoma multiforme (GBM) is the most common and aggressive form of primary brain cancer, for which effective therapies are urgently needed. Chimeric antigen receptor (CAR)-based immunotherapy represents a promising therapeutic approach, but it is often impeded by highly immunosuppressive tumor microenvironments (TME). Here, in an immunocompetent, orthotopic GBM mouse model, we show that CAR-T cells targeting tumor-specific epidermal growth factor receptor variant III (EGFRvIII) alone fail to control fully established tumors but, when combined with a single, locally delivered dose of IL-12, achieve durable anti-tumor responses. IL-12 not only boosts cytotoxicity of CAR-T cells, but also reshapes the TME, driving increased infiltration of proinflammatory CD4+ T cells, decreased numbers of regulatory T cells (Treg), and activation of the myeloid compartment. Importantly, the immunotherapy-enabling benefits of IL-12 are achieved with minimal systemic effects. Our findings thus show that local delivery of IL-12 may be an effective adjuvant for CAR-T cell therapy for GBM.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Inmunoconjugados/administración & dosificación , Inmunoterapia Adoptiva/métodos , Interleucina-12/administración & dosificación , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/inmunología , Encéfalo/patología , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Receptores ErbB/inmunología , Femenino , Glioblastoma/diagnóstico por imagen , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Inmunoconjugados/inmunología , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/inmunología , Inyecciones Intralesiones/métodos , Interleucina-12/inmunología , Imagen por Resonancia Magnética Intervencional , Ratones , Receptores Quiméricos de Antígenos/inmunología , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología
6.
Nat Biomed Eng ; 5(5): 399-413, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33046866

RESUMEN

Therapies employing chimeric antigen receptor T cells (CAR-T cells) targeting tumour-associated antigens (TAAs) can lead to on-target-off-tumour toxicity and to resistance, owing to TAA expression in normal tissues and to TAA expression loss in tumour cells. These drawbacks can be circumvented by CAR-T cells targeting tumour-specific driver gene mutations, such as the four-nucleotide duplication in the oncogene nucleophosmin (NPM1c), which creates a neoepitope presented by the human leukocyte antigen with the A2 serotype (HLA-A2) that has been observed in about 35% of patients with acute myeloid leukaemia (AML). Here, we report a human single-chain variable fragment (scFv), identified via yeast surface display, that specifically binds to the NPM1c epitope-HLA-A2 complex but not to HLA-A2 or to HLA-A2 loaded with control peptides. In vitro and in mice, CAR-T cells with the scFv exhibit potent cytotoxicity against NPM1c+HLA-A2+ leukaemia cells and primary AML blasts, but not NPM1c-HLA-A2+ leukaemia cells or HLA-A2- tumour cells. Therapies using NPM1c CAR-T cells for the treatment of NPM1c+HLA-A2+ AML may limit on-target-off-tumour toxicity and tumour resistance.


Asunto(s)
Linfocitos T CD8-positivos/trasplante , Leucemia Mieloide Aguda/terapia , Proteínas Nucleares/química , Receptores Quiméricos de Antígenos/metabolismo , Anticuerpos de Cadena Única/administración & dosificación , Animales , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Epítopos/inmunología , Antígeno HLA-A2/inmunología , Humanos , Inmunoterapia Adoptiva , Leucemia Mieloide Aguda/inmunología , Ratones , Proteínas Nucleares/inmunología , Nucleofosmina , Células PC-3 , Prueba de Estudio Conceptual , Anticuerpos de Cadena Única/farmacología
7.
Eur J Pharm Biopharm ; 158: 233-244, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33271301

RESUMEN

The tumour endothelial marker 1 (TEM1/endosialin/CD248) is a receptor overexpressed in several human solid tumours and silenced in normal adult tissues, representing a suitable and potentially safe target for radioimmunotherapy of sarcoma. To develop new tools with improved TEM1 targeting properties, a new panel of antibody fragments was for the first time evaluated preclinically following 125I radiolabelling. The antibody fragment 1C1m-Fc, with the highest human/murine TEM1 binding affinity, was extensively characterized in vitro and in vivo in a Ewing's sarcoma human xenograft mouse model. In silico studies were also performed to elucidate the influence of a single amino acid mutation in the complementarity-determining region (CDR3) of the heavy chain, upon affinity maturation of the parental clone 1C1-Fc. From this study, 1C1m-Fc emerged as a promising candidate for the development of TEM1-targeted radioimmunoconjugates, namely to be further explored for theranostic applications with other suitable medical radionuclides.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Inmunoconjugados/administración & dosificación , Neoplasias/radioterapia , Radioinmunoterapia/métodos , Anticuerpos de Cadena Única/administración & dosificación , Animales , Línea Celular Tumoral , Regiones Determinantes de Complementariedad/genética , Simulación por Computador , Femenino , Humanos , Inmunoconjugados/genética , Inmunoconjugados/farmacocinética , Radioisótopos de Yodo , Ratones , Mutación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/farmacocinética , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Theranostics ; 10(25): 11404-11415, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33052222

RESUMEN

Through protein engineering and a novel pegylation strategy, a diabody specific to tumor-associated glycoprotein 72 (TAG-72) (PEG-AVP0458) has been created to optimize pharmacokinetics and bioavailability to tumor. We report the preclinical and clinical translation of PEG-AVP0458 to a first-in-human clinical trial of a diabody. Methods: Clinical translation followed characterization of PEG-AVP0458 drug product and preclinical biodistribution and imaging assessments of Iodine-124 trace labeled PEG-AVP0458 (124I-PEG-AVP0458). The primary study objective of the first-in-human study was the safety of a single protein dose of 1.0 or 10 mg/m2 124I-PEG-AVP0458 in patients with TAG-72 positive relapsed/ metastatic prostate or ovarian cancer. Secondary study objectives were evaluation of the biodistribution, tumor uptake, pharmacokinetics and immunogenicity. Patients were infused with a single-dose of 124I labeled PEG-AVP0458 (3-5 mCi (111-185 MBq) for positron emission tomography (PET) imaging, performed sequentially over a one-week period. Safety, pharmacokinetics, biodistribution, and immunogenicity were assessed up to 28 days after infusion. Results: PEG-AVP0458 was radiolabeled with 124I and shown to retain high TAG-72 affinity and excellent targeting of TAG-72 positive xenografts by biodistribution analysis and PET imaging. In the first-in-human trial, no adverse events or toxicity attributable to 124I-PEG-AVP0458 were observed. Imaging was evaluable in 5 patients, with rapid and highly specific targeting of tumor and minimal normal organ uptake, leading to high tumor:blood ratios. Serum concentration values of 124I-PEG-AVP0458 showed consistent values between patients, and there was no significant difference in T½α and T½ß between dose levels with mean (± SD) results of T½α = 5.10 ± 4.58 hours, T½ß = 46.19 ± 13.06 hours. Conclusions: These data demonstrates the safety and feasibility of using pegylated diabodies for selective tumor imaging and potential delivery of therapeutic payloads in cancer patients.


Asunto(s)
Anticuerpos Biespecíficos/efectos adversos , Antígenos de Neoplasias/metabolismo , Antineoplásicos/efectos adversos , Neoplasias/tratamiento farmacológico , Radiofármacos/efectos adversos , Adulto , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Antineoplásicos/genética , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Disponibilidad Biológica , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Estudios de Factibilidad , Femenino , Humanos , Infusiones Intravenosas , Radioisótopos de Yodo/administración & dosificación , Radioisótopos de Yodo/efectos adversos , Radioisótopos de Yodo/farmacocinética , Masculino , Ratones , Neoplasias/diagnóstico , Neoplasias/inmunología , Neoplasias/patología , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radiofármacos/administración & dosificación , Radiofármacos/química , Radiofármacos/farmacocinética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/efectos adversos , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/farmacocinética , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Acta Neuropathol Commun ; 8(1): 126, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32762731

RESUMEN

With evidence supporting the prion-like spreading of extracellular tau as a mechanism for the initiation and progression of Alzheimer's disease (AD), immunotherapy has emerged as a potential disease-modifying strategy to target tau. Many studies have proven effective to clear pathological tau species in animal models of AD, and several clinical trials using conventional immunotherapy with anti-tau native antibodies are currently active. We have previously generated a vectorized scFv derived from the conformation-dependent anti-tau antibody MC1, scFvMC1, and demonstrated that its intracranial injection was able to prevent tau pathology in adult tau mice. Here, we show that, in a prevention paradigm and in two different tau transgenic models (JNPL3 and P301S), a one-time intramuscular injection of AAV1-scFvMC1 generated a long-lasting peripheral source of anti-tau scFvMC1 and significantly reduced insoluble and soluble tau species in the brain. Moreover, our data showed that scFvMC1 was internalized by the microglia, in the absence of overt inflammation. This study demonstrates the efficacy of intramuscular delivery of vectorized scFv to target tau, and suggests a new potential application to treat AD and the other tauopathies.


Asunto(s)
Enfermedad de Alzheimer/patología , Inmunoterapia/métodos , Anticuerpos de Cadena Única/administración & dosificación , Proteínas tau/antagonistas & inhibidores , Adenoviridae , Animales , Modelos Animales de Enfermedad , Vectores Genéticos , Humanos , Inyecciones Intramusculares , Ratones , Ratones Transgénicos
10.
FASEB J ; 34(9): 11577-11593, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32738178

RESUMEN

Targeted drug delivery to the endothelium has the potential to generate localized therapeutic effects at the blood-tissue interface. For some therapeutic cargoes, it is essential to maintain contact with the bloodstream to exert protective effects. The pharmacokinetics (PK) of endothelial surface-targeted affinity ligands and biotherapeutic cargo remain a largely unexplored area, despite obvious translational implications for this strategy. To bridge this gap, we site-specifically radiolabeled mono- (scFv) and bivalent (mAb) affinity ligands specific for the endothelial cell adhesion molecules, PECAM-1 (CD31) and ICAM-1 (CD54). Radiotracing revealed similar lung biodistribution at 30 minutes post-injection (79.3% ± 4.2% vs 80.4% ± 10.6% ID/g for αICAM and 58.9% ± 3.6% ID/g vs. 47.7% ± 5.8% ID/g for αPECAM mAb vs. scFv), but marked differences in organ residence time, with antibodies demonstrating an order of magnitude greater area under the lung concentration vs. time curve (AUCinf 1698 ± 352 vs. 53.3 ± 7.9 ID/g*hrs for αICAM and 1023 ± 507 vs. 114 ± 37 ID/g*hrs for αPECAM mAb vs scFv). A physiologically based pharmacokinetic model, fit to and validated using these data, indicated contributions from both superior binding characteristics and prolonged circulation time supporting multiple binding-detachment cycles. We tested the ability of each affinity ligand to deliver a prototypical surface cargo, thrombomodulin (TM), using one-to-one protein conjugates. Bivalent mAb-TM was superior to monovalent scFv-TM in both pulmonary targeting and lung residence time (AUCinf 141 ± 3.2 vs 12.4 ± 4.2 ID/g*hrs for ICAM and 188 ± 90 vs 34.7 ± 19.9 ID/g*hrs for PECAM), despite having similar blood PK, indicating that binding strength is more important parameter than the kinetics of binding. To maximize bivalent target engagement, we synthesized an oriented, end-to-end anti-ICAM mAb-TM conjugate and found that this therapeutic had the best lung residence time (AUCinf 253 ± 18 ID/g*hrs) of all TM modalities. These observations have implications not only for the delivery of TM, but also potentially all therapeutics targeted to the endothelial surface.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Endotelio Vascular/metabolismo , Molécula 1 de Adhesión Intercelular/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Anticuerpos de Cadena Única/administración & dosificación , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Células Endoteliales/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Ligandos , Pulmón/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/farmacocinética , Distribución Tisular
11.
Anim Sci J ; 91(1): e13399, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32512648

RESUMEN

Probiotics have been defined as live microorganisms that are administered in an appropriate amount to provide health benefits to the host animal. In this study, we investigated the effect of L. salivarius DJ-sa-01 secreting the 3D8 single-chain variable fragment (3D8 scFv) on the growth performance, cytokine secretion, and intestinal microbial flora of chickens. The experiment was divided into the control group and L. salivarius expressing 3D8 scFv experimental group. Chicken was fed 109 colony-forming units (CFUs) of wild-type (WT) L. salivarius or 3D8 scFv-secreting L. salivarius daily for 35 days. The administration of L. salivarius expressing 3D8 scFv significantly improved the body weight of chickens compared with the administration of WT L. salivarius. A 16S ribosomal RNA metagenomic analysis showed that Firmicutes, Proteobacteria, Actinobacteria, and Bacteroidetes were the dominant phyla in both experimental groups. At the genus level, Lactobacillus was more abundant (22.82%) in the L. salivarius/3D8 group compared with the WT L. salivarius group. The serum levels of cytokines, such as IL-8, TNF-α, IL-1ß, IFN-γ, IL-4, and IGF1, were significantly reduced in the L. salivarius/3D8-treated chickens. In summary, our results suggest that L. salivarius expressing 3D8 scFv could be considered a feed additive for improving the growth performance, immune function, and disease resistance of poultry.


Asunto(s)
Alimentación Animal , Fenómenos Fisiológicos Nutricionales de los Animales , Pollos/crecimiento & desarrollo , Pollos/inmunología , Pollos/microbiología , Dieta/veterinaria , Suplementos Dietéticos , Microbioma Gastrointestinal , Homeostasis , Ligilactobacillus salivarius , Anticuerpos de Cadena Única/administración & dosificación , Animales , Citocinas/sangre , Femenino
12.
Toxins (Basel) ; 12(4)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32316084

RESUMEN

Envenoming due to Loxosceles spider bites still remains a neglected disease of particular medical concern in the Americas. To date, there is no consensus for the treatment of envenomed patients, yet horse polyclonal antivenoms are usually infused to patients with identified severe medical conditions. It is widely known that venom proteins in the 30-35 kDa range with sphingomyelinase D (SMasesD) activity, reproduce most of the toxic effects observed in loxoscelism. Hence, we believe that monoclonal antibody fragments targeting such toxins might pose an alternative safe and effective treatment. In the present study, starting from the monoclonal antibody LimAb7, previously shown to target SMasesD from the venom of L. intermedia and neutralize its dermonecrotic activity, we designed humanized antibody V-domains, then produced and purified as recombinant single-chain antibody fragments (scFvs). These molecules were characterized in terms of humanness, structural stability, antigen-binding activity, and venom-neutralizing potential. Throughout this process, we identified some blocking points that can impact the Abs antigen-binding activity and neutralizing capacity. In silico analysis of the antigen/antibody amino acid interactions also contributed to a better understanding of the antibody's neutralization mechanism and led to reformatting the humanized antibody fragment which, ultimately, recovered the functional characteristics for efficient in vitro venom neutralization.


Asunto(s)
Anticuerpos Monoclonales , Antivenenos , Anticuerpos de Cadena Única , Venenos de Araña/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos/inmunología , Antivenenos/administración & dosificación , Antivenenos/inmunología , Eritrocitos/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Modelos Moleculares , Pruebas de Neutralización , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/inmunología , Picaduras de Arañas/terapia , Venenos de Araña/efectos adversos , Arañas/inmunología
13.
Int J Nanomedicine ; 14: 8755-8768, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31806970

RESUMEN

PURPOSE: Single-chain variable fragments (scFvs) are one of the smallest antigen-binding units having the invaluable advantage to be expressed by a unique short open reading frame (ORF). Despite their reduced size, spontaneous cell entry of scFvs remains inefficient, hence precluding the possibility to target intracellular antigens. Here, we describe an original strategy to deliver scFvs inside target cells through engineered extracellular vesicles (EVs). This approach relies on the properties of a Human Immunodeficiency Virus (HIV)-1 Nef mutant protein referred to as Nefmut. It is a previously characterized Nef allele lacking basically all functions of wt Nef, yet strongly accumulating in the EV lumen also when fused at its C-terminus with a foreign protein. To gain the proof-of-principle for the efficacy of the proposed strategy, the tumor-promoting Human Papilloma Virus (HPV)16-E7 protein was considered as a scFv-specific intracellular target. The oncogenic effect of HPV16-E7 relies on its binding to the tumor suppressor pRb protein leading to a dysregulated cell duplication. Interfering with this interaction means impairing the HPV16-E7-induced cell proliferation. METHODS: The Nefmut gene was fused in frame at its 3'-terminus with the ORF coding for a previously characterized anti-HPV16-E7 scFv. Interaction between the Nefmut-fused anti-HPV16-E7 scFv and the HPV16-E7 protein was tested by both confocal microscope and co-immunoprecipitation analyses on co-transfected cells. The in cis anti-proliferative effect of the Nefmut/anti-HPV16-E7 scFv was assayed by transfecting HPV16-infected cells. The anti-proliferative effect of EVs engineered with Nefmut/anti-HPV16-E7 scFv on HPV16-E7-expressing cells was evaluated in two ways: i) through challenge with purified EVs by a Real-Time Cell Analysis system and ii) in transwell co-cultures by an MTS-based assay. RESULTS: The Nefmut/anti-HPV16-E7 scFv chimeric product is efficiently uploaded in EVs, binds HPV16-E7, and inhibits the proliferation of HPV16-E7-expressing cells. Most important, challenge with cell-free EVs incorporating the Nefmut/anti-HPV16-E7 scFv led to the inhibition of proliferation of HPV16-E7-expressing cells. The proliferation of these cells was hindered also when they were co-cultured in transwells with cells producing EVs uploading Nefmut/anti-HPV16-E7 scFv. CONCLUSION: Our data represent the proof-of-concept for the possibility to target intracellular antigens through EV-mediated delivery of scFvs. This finding could be relevant to design novel methods of intracellular therapeutic interventions.


Asunto(s)
Vesículas Extracelulares/inmunología , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Anticuerpos de Cadena Única/administración & dosificación , Efecto Espectador , Línea Celular , Proliferación Celular , Técnicas de Cocultivo , Exosomas/inmunología , Exosomas/metabolismo , Vesículas Extracelulares/genética , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 16/patogenicidad , Humanos , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/prevención & control , Anticuerpos de Cadena Única/genética , Transfección , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
14.
Methods Mol Biol ; 2054: 283-294, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31482462

RESUMEN

A series of ligand-targeted nanosystems have been rapidly exploited to selectively deliver drug molecules to desired cell populations. The conjugation of protein ligands to the nanoparticle (NP) surface endows nanovehicles with active targeting properties. However, the nonspecific covalent coupling of protein ligands to nanocarriers may compromise the protein targeting due to the uncontrolled ligand orientation as well as the decline in ligand activity during linkage process. With this regard, biomimetic synthetic strategies are employed for the preparation of genetically engineered nanovesicles (GNV) from cellular plasma membrane with targeting moieties on the surface in a ligand-oriented manner. Herein, we introduce the biomimetic synthetic strategy and procedures for GNV preparation. This chapter may guide readers to design analogous NPs for cell-specific targeting by displaying particular protein probes (e.g., antibody, nanobody, and single-chain antibody) on the surface of GNVs.


Asunto(s)
Antineoplásicos/administración & dosificación , Ingeniería Genética/métodos , Nanopartículas/química , Neoplasias/terapia , Nanomedicina Teranóstica/métodos , Animales , Antineoplásicos/farmacocinética , Materiales Biomiméticos/síntesis química , Línea Celular Tumoral , Membrana Celular/genética , Terapia Combinada/métodos , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Composición de Medicamentos/métodos , Exosomas/genética , Humanos , Hipertermia Inducida/métodos , Liposomas , Ratones , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Técnicas Fotoacústicas , Fotoquimioterapia/métodos , Anticuerpos de Cadena Única/administración & dosificación , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Drug Metab Dispos ; 47(10): 1136-1145, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31387870

RESUMEN

In this study, we evaluated the effect of size on tumor disposition of protein therapeutics, including the plasma and tumor pharmacokinetics (PK) of trastuzumab (∼150 kDa), FcRn-nonbinding trastuzumab (∼150 kDa), F(ab)2 fragment of trastuzumab (∼100 kDa), Fab fragment of trastuzumab (∼50 kDa), and trastuzumab scFv (∼27 kDa) in both antigen (i.e., HER2)-overexpressing (N87) and antigen-nonexpressing (MDA-MB-468) tumor-bearing mice. The observed data were used to develop the maximum tumor uptake versus molecular weight and tumor-to-plasma area under the curve (AUC) ratio versus molecular weight relationships. Comparison of the PK of different sizes of FcRn nonbinding molecules in target-expressing tumor showed that ∼100 kDa is an optimal size to achieve maximum tumor uptake and ∼50 kDa is an optimal size to achieve maximum tumor-to-plasma exposure ratio of protein therapeutics. The PK data were also used to validate a systems PK model for tumor disposition of different-sized protein therapeutics. The PK model was able to predict a priori the PK of all five molecules in both tumor types reasonably well (within 2- to 3-fold). In addition, the model captured the bell-shaped relationships observed between maximum tumor uptake and molecular weight and between tumor-to-plasma AUC ratio and molecular weight. Our results provide an unprecedented insight into the effect of size and target engagement on the tumor PK of protein therapeutics. Our results also provide further validation of the tumor disposition model, which can be used to support discovery, development, and preclinical-to-clinical translation of different sizes of protein therapeutics. SIGNIFICANCE STATEMENT: This article highlights the importance of molecular size and target engagement on the tumor disposition of protein therapeutics. Our results suggest that ∼100 kDa is an optimal size to achieve maximum tumor uptake and ∼50 kDa is an optimal size to achieve maximum tumor-to-plasma exposure ratio for non-FcRn-binding targeted protein therapeutics. We also demonstrate that a systems pharmacokinetics model developed to characterize tumor disposition of protein therapeutics can predict a priori the disposition of different-sized protein therapeutics in target-expressing and target-nonexpressing solid tumors.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Neoplasias/tratamiento farmacológico , Receptor ErbB-2/antagonistas & inhibidores , Receptores Fc/metabolismo , Anticuerpos de Cadena Única/farmacología , Trastuzumab/farmacocinética , Animales , Área Bajo la Curva , Línea Celular Tumoral , Humanos , Masculino , Ratones , Modelos Biológicos , Peso Molecular , Neoplasias/sangre , Neoplasias/patología , Receptor ErbB-2/metabolismo , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/química , Distribución Tisular , Trastuzumab/administración & dosificación , Trastuzumab/química , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Sci ; 110(9): 2722-2733, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31461572

RESUMEN

Mesothelin (MSLN) shows increased expression in various cancer cells. For clinical application of antibodies as a positron emission tomography (PET) imaging reagent, a human shortened antibody is essential both for avoiding redundant immune responses and for providing rapid imaging. Therefore, we cloned a single-chain fragment of variable regions (scFv) from a human-derived gene sequence. This was achieved through the construction of a naïve phage library derived from human tonsil lymphocytes. Using a column with human recombinant MSLN, we carried out bio-panning of phage-variants by colony formation. We first obtained 120 clones that were subjected to selection in an ELISA using human recombinant MSLN as a solid phase antigen, and 15 phage clones of scFv with a different sequence were selected and investigated by flow cytometry (FCM). Then, six variants were selected and the individual scFv gene was synthesized in the VL and VH domains and expressed in Chinese hamster ovary cells. Mammalian cell-derived human-origin scFv clones were analyzed by FCM again, and one MSLN highly specific scFv clone was established. PET imaging by 89 Zr-labeled scFv was done in mice bearing xenografts with MSLN-expressing cancer cells, and tumor legions were successfully visualized. The scFv variant established in the present study may be potentially useful for cancer diagnosis by PET imaging.


Asunto(s)
Proteínas Ligadas a GPI/inmunología , Neoplasias/diagnóstico por imagen , Radiofármacos/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Células CHO , Línea Celular Tumoral , Clonación Molecular , Cricetulus , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/aislamiento & purificación , Proteínas Ligadas a GPI/metabolismo , Humanos , Masculino , Mesotelina , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Imagen Molecular/métodos , Neoplasias/patología , Biblioteca de Péptidos , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Radioisótopos , Radiofármacos/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Anticuerpos de Cadena Única/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Circonio
17.
PLoS One ; 14(8): e0220986, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31430333

RESUMEN

A promising strategy for the enhancement of vaccine-mediated immune responses is by directly targeting protein antigens to immune cells. Targeting of antigens to the dendritic cell (DC) molecule Clec9A has been shown to enhance antibody affinity and titers for model antigens, and influenza and enterovirus antigens, and may be advantageous for immunogens that otherwise fail to elicit antibodies with sufficient titers and breadth for broad protection, such as the envelope protein (Env) of HIV. Previously employed targeting strategies often utilize receptor-specific antibodies, however it is impractical to conjugate a bivalent IgG antibody to oligomeric antigens, including HIV Env trimers. Here we designed single chain variable fragment (scFv) and single chain Fab (scFab) constructs of a Clec9A-targeting antibody, expressed as genetically fused conjugates with the soluble ectodomain of Env, gp140. This conjugation did not affect the presentation of Env neutralising antibody epitopes. The scFab moiety was shown to be more stable than scFv, and in the context of gp140 fusions, was able to mediate better binding to recombinant and cell surface-expressed Clec9A, although the level of binding to cell-surface Clec9A was lower than that of the anti-Clec9A IgG. However, binding to Clec9A on the surface of DCs was not detected. Mouse immunization experiments suggested that the Clec9A-binding activity of the scFab-gp140 conjugate was insufficient to enhance Env-specific antibody responses. This is an important first proof of principle study demonstrating the conjugation of a scFab to an oligomeric protein antigen, and that an scFab displays better antigen binding than the corresponding scFv. Future developments of this technique that increase the scFab affinity will provide a valuable means to target oligomeric proteins to cell surface antigens of interest, improving vaccine-generated immune responses.


Asunto(s)
Vacunas contra el SIDA/inmunología , Infecciones por VIH/terapia , Proteínas Recombinantes de Fusión/inmunología , Anticuerpos de Cadena Única/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Animales , Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos , Antígenos Virales/genética , Antígenos Virales/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos/inmunología , Femenino , Células HEK293 , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Inmunogenicidad Vacunal , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Ratones , Prueba de Estudio Conceptual , Dominios Proteicos/genética , Dominios Proteicos/inmunología , Receptores Mitogénicos/inmunología , Receptores Mitogénicos/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/genética , Vacunación/métodos , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/administración & dosificación , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética
19.
J Alzheimers Dis ; 70(4): 1069-1091, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31306135

RESUMEN

The intracellular deposition of amyloid-ß (Aß) peptides has been described in the brains of both Alzheimer's disease (AD) patients and animal models. A correlation between the intracellular amyloid burden and neurodegeneration has recently been reported in a triple-transgenic AD (3xTg-AD) murine model. In the present study, we assessed the effect of scFv-h3D6, an anti-Aß single-chain variable fragment (scFv) derived from the antibody bapineuzumab, on amyloid pathology in 5-month-old 3xTg-AD female mice, focusing on intracellular Aß clearance, neuronal survival, and functional abilities. We also examined neuroinflammation and the histology of peripheral organ samples to detect any adverse effects. A single intraperitoneal injection of scFv-h3D6 dramatically reduced intracellular Aß burden in the deep layers of the cerebral cortex, pyramidal cells layer of the hippocampus, and basolateral amygdalar nucleus. The treatment prevented neuronal loss in the hippocampus and amygdala, while neither astrogliosis nor microgliosis was induced. Instead, an increase in the size of the white pulp after the treatment indicated that the spleen could be involved in the clearance mechanism. Although the treatment did not ameliorate behavioral and psychological symptoms of dementia-like symptoms, the results of cognitive testing pointed to a noticeable improvement in spatial memory. These findings indicated that the mechanism underlying the therapeutic effect of scFv-h3D6 was the clearance of intracellular Aß, with subsequent prevention of neuronal loss and amelioration of cognitive disabilities. The treatment was safe in terms of neuroinflammation and kidney and liver function, whereas some effects on the spleen were observed.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Anticuerpos Monoclonales Humanizados/administración & dosificación , Líquido Intracelular/metabolismo , Neuronas/metabolismo , Anticuerpos de Cadena Única/administración & dosificación , Memoria Espacial/fisiología , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Líquido Intracelular/efectos de los fármacos , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
20.
J Immunother Cancer ; 7(1): 191, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31324216

RESUMEN

BACKGROUND: Glucocorticoid-induced TNFR-related protein (TNFRSF18, GITR, CD357), expressed by T cells, and its ligand (TNFSF18, GITRL), expressed by myeloid populations, provide co-stimulatory signals that boost T cell activity. Due to the important role that GITR plays in regulating immune functions, agonistic stimulation of GITR is a promising therapeutic concept. Multiple strategies to induce GITR signaling have been investigated. The limited clinical efficacy of antibody-based GITR agonists results from structural and functional characteristics of antibodies that are unsuitable for stimulating the well-defined trimeric members of the TNFRSF. METHODS: To overcome limitations of antibody-based TNFRSF agonists, we have developed HERA-GITRL, a fully human hexavalent TNF receptor agonist (HERA) targeting GITR and mimicking the natural signaling concept. HERA-GITRL is composed of a trivalent but single-chain GITRL-receptor-binding-domain (scGITRL-RBD) unit fused to an IgG1 derived silenced Fc-domain serving as dimerization scaffold. A specific mouse surrogate, mmHERA-GITRL, was also generated to examine in vivo activity in respective mouse tumor models. RESULTS: For functional characterization of HERA-GITRL in vitro, human immune cells were isolated from healthy-donor blood and stimulated with anti-CD3 antibody in the presence of HERA-GITRL. Consistently, HERA-GITRL increased the activity of T cells, including proliferation and differentiation, even in the presence of regulatory T cells. In line with these findings, mmHERA-GITRL enhanced antigen-specific clonal expansion of both CD4+ (OT-II) and CD8+ (OT-I) T cells in vivo while having no effect on non-specific T cells. In addition, mmHERA-GITRL showed single-agent anti-tumor activity in two subcutaneous syngeneic colon cancer models (CT26wt and MC38-CEA). Importantly, this activity is independent of its FcγR-binding functionality, as both mmHERA-GITRL with a functional Fc- and a silenced Fc-domain showed similar tumor growth inhibition. Finally, in a direct in vitro comparison to a bivalent clinical benchmark anti-GITR antibody and a trivalent GITRL, only the hexavalent HERA-GITRL showed full biological activity independent of additional crosslinking. CONCLUSION: In this manuscript, we describe the development of HERA-GITRL, a true GITR agonist with a clearly defined mechanism of action. By clustering six receptor chains in a spatially well-defined manner, HERA-GITRL induces potent agonistic activity without being dependent on additional FcγR-mediated crosslinking.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/agonistas , Anticuerpos de Cadena Única/administración & dosificación , Linfocitos T Reguladores/inmunología , Factores de Necrosis Tumoral/química , Animales , Línea Celular Tumoral , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Activación de Linfocitos , Macaca fascicularis , Ratones , Proteínas Recombinantes de Fusión/inmunología , Transducción de Señal , Anticuerpos de Cadena Única/inmunología , Factores de Necrosis Tumoral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...