Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Methods Mol Biol ; 2702: 347-372, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37679629

RESUMEN

Antibody libraries came into existence 30 years ago when the accumulating sequence data of immunoglobulin genes and the advent of PCR technology made it possible to clone antibody gene repertoires. Phage display (most common) and additional display and screening technologies were applied to pan out desired binding specificities from antibody libraries. As other antibody discovery tools, phage display is not an off-the-shelf technology and not offered as a kit but rather requires experience and expertise for making it indeed very useful.Next-generation sequencing (NGS) coupled with bioinformatics is a powerful tool for analyzing large amount of DNA sequence output of the panning. Here, we demonstrate how NGS analysis of phage biopanning (phage-Seq) of complex antibody libraries can facilitate the antibody discovery process and provide insights regarding the biopanning process (see Fig. 1).


Asunto(s)
Bacteriófagos , Anticuerpos de Cadena Única , Humanos , Anticuerpos de Cadena Única/genética , Genes de Inmunoglobulinas , Secuenciación de Nucleótidos de Alto Rendimiento , Bioprospección
2.
Theranostics ; 13(11): 3497-3508, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37441597

RESUMEN

Background and rationale: Cancer therapy have evolved remarkably over the past decade, providing new strategies to inhibit cancer cell growth using immune modulation, with or without gene therapy. Specifically, suicide gene therapies and immunotoxins have been investigated for the treatment of tumors by direct cancer cell cytotoxicity. Recent advances in mRNA delivery also demonstrated the potential of mRNA-based vaccines and immune-modulators for cancer therapeutics by utilizing nanocarriers for mRNA delivery. Methods: We designed a bacterial toxin-encoding modified mRNA, delivered by lipid nanoparticles into a B16-melanoma mouse model. Results: We showed that local administration of LNPs entrapping a modified mRNA that encodes for a bacterial toxin, induced significant anti-tumor effects and improved overall survival of treated mice. Conclusions: We propose mmRNA-loaded LNPs as a new class of anti-tumoral, toxin-based therapy.


Asunto(s)
Toxinas Bacterianas , Nanopartículas , Neoplasias , Ratones , Animales , ARN Mensajero/genética , Liposomas , Terapia Genética , Neoplasias/terapia , Toxinas Bacterianas/genética
3.
Nat Cancer ; 4(3): 401-418, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36797502

RESUMEN

Brain metastasis still encompass very grim prognosis and therefore understanding the underlying mechanisms is an urgent need toward developing better therapeutic strategies. We uncover the intricate interactions between recruited innate immune cells and resident astrocytes in the brain metastatic niche that facilitate metastasis of melanoma and breast cancer. We show that granulocyte-derived lipocalin-2 (LCN2) induces inflammatory activation of astrocytes, leading to myeloid cell recruitment to the brain. LCN2 is central to inducing neuroinflammation as its genetic targeting or bone-marrow transplantation from LCN2-/- mice was sufficient to attenuate neuroinflammation and inhibit brain metastasis. Moreover, high LCN2 levels in patient blood and brain metastases in multiple cancer types were strongly associated with disease progression and poor survival. Our findings uncover a previously unknown mechanism, establishing a central role for the reciprocal interactions between granulocytes and astrocytes in promoting brain metastasis and implicate LCN2 as a prognostic marker and potential therapeutic target.


Asunto(s)
Astrocitos , Neoplasias Encefálicas , Ratones , Animales , Lipocalina 2/genética , Lipocalina 2/metabolismo , Astrocitos/metabolismo , Enfermedades Neuroinflamatorias , Neoplasias Encefálicas/genética , Inmunidad Innata
4.
Allergy ; 78(2): 464-476, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36070083

RESUMEN

BACKGROUND: Eosinophilic esophagitis (EoE) is a chronic, food-driven allergic disease, characterized by eosinophil-rich inflammation in the esophagus. The histopathological and clinical features of EoE have been attributed to overproduction of the type 2 cytokines IL-4 and IL-13, which mediate profound alterations in the esophageal epithelium and neutralizing of their shared receptor component (IL-4Rα) with a human antibody drug (dupilumab) demonstrates clinical efficacy. Yet, the relative contribution of IL-4 and IL-13 and whether the type II IL-4 receptor (comprised of the IL-4Rα chain in association with IL-13Rα1) mediates this effect has not been determined. METHODS: Experimental EoE was induced in WT, Il13ra1-/- , and Krt14Cre /Il13ra1fl/fl mice by skin-sensitized using 4-ethoxymethylene-2-phenyl-2-oxazolin (OXA) followed by intraesophageal challenges. Esophageal histopathology was determined histologically. RNA was extracted and sequenced for transcriptome analysis and compared with human EoE RNAseq data. RESULTS: Induction of experimental EoE in mice lacking Il13ra1 and in vivo IL-13 antibody-based neutralization experiments blocked antigen-induced esophageal epithelial and lamina propria thickening, basal cell proliferation, eosinophilia, and tissue remodeling. In vivo targeted deletion of Il13ra1 in esophageal epithelial cells rendered mice protected from experimental EoE. Single-cell RNA sequencing analysis of human EoE biopsies revealed predominant expression of IL-13Rα1 in epithelial cells and that EoE signature genes correlated with IL-13 expression compared with IL-4. CONCLUSIONS: We demonstrate a definitive role for IL-13 signaling via IL-13Rα1 in EoE. These data provide mechanistic insights into the mode of action of current therapies in EoE and highlight the type II IL-4R as a future therapeutic target.


Asunto(s)
Esofagitis Eosinofílica , Humanos , Ratones , Animales , Esofagitis Eosinofílica/patología , Subunidad alfa1 del Receptor de Interleucina-13/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Interleucina-13/metabolismo , Células Epiteliales/metabolismo
5.
Front Immunol ; 13: 835774, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35350789

RESUMEN

Diabetes is a metabolic disease that may lead to different life-threatening complications. While insulin constitutes a beneficial treatment, its use may be limited due to increased degradation and an increase in side effects such as weight gain and hypoglycemia. Small molecule inhibitors to insulin-degrading enzyme (IDE) have been previously suggested as a potential treatment for diabetes through their ability to reduce insulin degradation and thus increase insulin activity. Nevertheless, their tendency to bind to the zinc ion in the catalytic site of IDE may affect other important metalloproteases and limit their clinical use. Here, we describe the isolation of an IDE-specific antibody that specifically inhibits insulin degradation by IDE. Using phage display, we generated a human IDE-specific antibody that binds human and mouse IDE with high affinity and specificity and can differentiate between active IDE to a mutated IDE with reduced catalytic activity in the range of 30 nM. We further assessed the ability of that IDE-inhibiting antibody to improve insulin activity in vivo in an STZ-induced diabetes mouse model. Since human antibodies may stimulate the mouse immune response to generate anti-human antibodies, we reformatted our inhibitory antibody to a "reverse chimeric" antibody that maintained the ability to inhibit IDE in vitro, but consisted of mouse constant regions, for reduced immunogenicity. We discovered that one intraperitoneal (IP) administration of the IDE-specific antibody in STZ-induced diabetic mice improved insulin activity in an insulin tolerance test (ITT) assay and reduced blood glucose levels. Our results suggest that antibody-mediated inhibition of IDE may be beneficial on improving insulin activity in a diabetic environment.


Asunto(s)
Diabetes Mellitus Experimental , Insulisina , Animales , Anticuerpos , Dominio Catalítico , Diabetes Mellitus Experimental/tratamiento farmacológico , Modelos Animales de Enfermedad , Insulina/metabolismo , Insulisina/metabolismo , Ratones
6.
Transl Vis Sci Technol ; 10(10): 16, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34817617

RESUMEN

Purpose: Semaphorin 3A (Sema-3A) is a secreted protein that deflects axons from inappropriate regions and induces neuronal cell death. Intravitreal application of polyclonal antibodies against Sema-3A prevents loss of retinal ganglion cells ensuing from axotomy of optic nerves. This suggested a therapeutic approach for neuroprotection via inhibition of the Sema-3A pathway. Methods: To develop potent and specific Sema-3A antagonists, we isolated monoclonal anti-Sema-3A antibodies from a human antibody phage display library and optimized low-molecular weight Sema-3A signaling inhibitors. The best inhibitors were identified using in vitro scratch assays and semiquantitative repulsion assays. Results: A therapeutic approach for neuroprotection must have a long duration of action. Therefore, antibodies and low-molecular weight inhibitors were formulated in extruded implants to allow controlled and prolonged release. Following release from the implants, Sema-3A inhibitors antagonized Sema-3A effects in scratch and repulsion assays and protected retinal ganglion cells in animal models of optic nerve injury, retinal ischemia, and glaucoma. Conclusions and Translational Relevance: Collectively, our findings indicate that the identified Sema-3A inhibitors should be further evaluated as therapeutic candidates for the treatment of Sema-3A-driven central nervous system degenerative processes.


Asunto(s)
Células Ganglionares de la Retina , Semaforina-3A , Animales , Axones , Axotomía , Movimiento Celular , Humanos
7.
Lupus ; 30(5): 775-784, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33554716

RESUMEN

INTRODUCTION: Antiphospholipid syndrome (APS) is an autoimmune disorder manifested by thromboembolic events, recurrent spontaneous abortions and elevated titers of circulating antiphospholipid antibodies. In addition, the presence of antiphospholipid antibodies seems to confer a fivefold higher risk for stroke or transient ischemic attack. Although the major antigen of APS is ß2 glycoprotein I, it is now well established that antiphospholipid antibodies are heterogeneous and bind to various targets. Recently, antibodies to Annexin A2 (ANXA2) have been reported in APS. This is of special interest since data indicated ANXA2 as a key player in fibrinolysis. Therefore, in the present study we assessed whether anti-ANXA2 antibodies play a pathological role in thrombosis associated disease. MATERIALS AND METHODS: Mice were induced to produce anti-ANXA2 antibodies by immunization with ANXA2 (iANXA2) and control mice were immunized with adjuvant only. A middle cerebral artery occlusion stroke model was applied to the mice. The outcome of stroke severity was assessed and compared between the two groups. RESULTS: Our results indicate that antibodies to ANXA2 lead to a more severe stroke as demonstrated by a significant larger stroke infarct volume (iANXA2 133.9 ± 3.3 mm3 and control 113.7 ± 7.4 mm3; p = 0.017) and a more severe neurological outcome (iANXA2 2.2 ± 0.2, and control 1.5 ± 0.18; p = 0.03). CONCLUSIONS: This study supports the hypothesis that auto-antibodies to ANXA2 are an independent risk factor for cerebral thrombosis. Consequently, we propose screening for anti-ANXA2 antibodies should be more widely used and patients that exhibit the manifestations of APS should be closely monitored by physicians.


Asunto(s)
Anexina A2/inmunología , Síndrome Antifosfolípido/inmunología , Autoanticuerpos/inmunología , Trombosis Intracraneal/metabolismo , Adulto , Animales , Anexina A2/administración & dosificación , Anexina A2/metabolismo , Anticuerpos Antifosfolípidos/sangre , Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/metabolismo , Autoanticuerpos/metabolismo , Autoinmunidad/inmunología , Modelos Animales de Enfermedad , Femenino , Fibrinólisis/inmunología , Humanos , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/fisiopatología , Inyecciones Subcutáneas , Trombosis Intracraneal/etiología , Ataque Isquémico Transitorio/inmunología , Ratones , Ratones Endogámicos BALB C/inmunología , Persona de Mediana Edad , Factores de Riesgo , Índice de Severidad de la Enfermedad , Accidente Cerebrovascular/inmunología , beta 2 Glicoproteína I/metabolismo
8.
Nat Commun ; 11(1): 5851, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33203857

RESUMEN

HIV viremia can be controlled by chronic antiretroviral therapy. As a potentially single-shot alternative, B cells engineered by CRISPR/Cas9 to express anti-HIV broadly neutralizing antibodies (bNAbs) are capable of secreting high antibody titers. Here, we show that, upon immunization of mice, adoptively transferred engineered B cells home to germinal centers (GC) where they predominate over the endogenous response and differentiate into memory and plasma cells while undergoing class switch recombination (CSR). Immunization with a high affinity antigen increases accumulation in GCs and CSR rates. Boost immunization increases the rate of engineered B cells in GCs and antibody secretion, indicating memory retention. Finally, antibody sequences of engineered B cells in the spleen show patterns of clonal selection. Therefore, B cells can be engineered into what could be a living and evolving drug.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Monoclonales/genética , Linfocitos B/inmunología , Anticuerpos ampliamente neutralizantes/genética , Anticuerpos Anti-VIH/genética , Memoria Inmunológica/genética , Vacunas contra el SIDA/genética , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Linfocitos B/fisiología , Linfocitos B/trasplante , Anticuerpos ampliamente neutralizantes/sangre , Anticuerpos ampliamente neutralizantes/inmunología , Ingeniería Genética/métodos , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/inmunología , Inmunización , Isotipos de Inmunoglobulinas/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación
9.
Cancer Immunol Immunother ; 69(7): 1337-1352, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32219500

RESUMEN

Cleavage of the MUC1 glycoprotein yields two subunits, an extracellular alpha-subunit bound to a smaller transmembrane beta-subunit. Monoclonal antibodies (mAbs) directed against the MUC1 alpha-beta junction comprising the SEA domain, a stable cell-surface moiety, were generated. Sequencing of all seven anti-SEA domain mAbs showed that they clustered into four groups and sequences of all groups are presented here. mAb DMB5F3 with picomolar affinity for the MUC1 SEA target was selected for further evaluation. Immunohistochemical staining of a series of malignancies with DMB5F3 including lung, prostate, breast, colon, and pancreatic carcinomas revealed qualitative and qualitative differences between MUC1 expression on normal versus malignant cells: DMB5F3 strongly stained malignant cells in a near-circumferential pattern, whereas MUC1 in normal pancreatic and breast tissue showed only weak apical positivity of ductal/acinar cells. Humanized chimeric DMB5F3 linked to ZZ-PE38 (ZZ IgG-binding protein fused to Pseudomonas exotoxin) induced vigorous cytotoxicity of MUC1+ malignant cells in vitro. The intensity of cell killing correlated with the level of MUC1 expression by the target cell, suggesting a MUC1 expression threshold for cell killing. MUC1+ Colo357 pancreatic cancer cells xenotransplanted into nude and SCID mice models were treated with the chDMB5F3:ZZ-PE38 immunocomplex. In both transplant models, chDMB5F3:ZZ-PE38 exhibited significant in vivo anti-tumor activity, suppressing up to 90% of tumor volume in the SCID model compared with concomitant controls. The efficacy of chDMB5F3:ZZ-PE38 immunotoxin in mediating tumor killing both in vitro and in vivo strongly suggests a clinical role for anti-MUC1 SEA antibody in the treatment of MUC1-expressing malignancies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Inmunotoxinas/inmunología , Mucina-1/química , Mucina-1/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Dominios Proteicos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Sci Immunol ; 5(44)2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-32060143

RESUMEN

IL-13 and IL-4 are potent mediators of type 2-associated inflammation such as those found in atopic dermatitis (AD). IL-4 shares overlapping biological functions with IL-13, a finding that is mainly explained by their ability to signal via the type 2 IL-4 receptor (R), which is composed of IL-4Rα in association with IL-13Rα1. Nonetheless, the role of the type 2 IL-4R in AD remains to be clearly defined. Induction of two distinct models of experimental AD in Il13ra1 -/- mice, which lack the type 2 IL-4R, revealed that dermatitis, including ear and epidermal thickening, was dependent on type 2 IL-4R signaling. Expression of TNF-α was dependent on the type 2 IL-4R, whereas induction of IL-4, IgE, CCL24, and skin eosinophilia was dependent on the type 1 IL-4R. Neutralization of IL-4, IL-13, and TNF-α as well as studies in bone marrow-chimeric mice revealed that dermatitis, TNF-α, CXCL1, and CCL11 expression were exclusively mediated by IL-13 signaling via the type 2 IL-4R expressed by nonhematopoietic cells. Conversely, induction of IL-4, CCL24, and eosinophilia was dependent on IL-4 signaling via the type 1 IL-4R expressed by hematopoietic cells. Last, we pharmacologically targeted IL-13Rα1 and established a proof of concept for therapeutic targeting of this pathway in AD. Our data provide mechanistic insight into the differential roles of IL-4, IL-13, and their receptor components in allergic skin and highlight type 2 IL-4R as a potential therapeutic target in AD and other allergic diseases such as asthma and eosinophilic esophagitis.


Asunto(s)
Dermatitis Atópica/inmunología , Interleucina-13/inmunología , Receptores Tipo II de Interleucina-4/inmunología , Transducción de Señal/inmunología , Animales , Dermatitis Atópica/inducido químicamente , Dinitrofluorobenceno , Femenino , Interleucina-13/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxazolona
11.
Environ Microbiol ; 22(1): 32-44, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31602783

RESUMEN

Horizontal gene transfer via plasmids plays a pivotal role in microbial evolution. The forces that shape plasmidomes functionality and distribution in natural environments are insufficiently understood. Here, we present a comparative study of plasmidomes across adjacent microbial environments present in different individual rumen microbiomes. Our findings show that the rumen plasmidome displays enormous unknown functional potential currently unannotated in available databases. Nevertheless, this unknown functionality is conserved and shared with published rat gut plasmidome data. Moreover, the rumen plasmidome is highly diverse compared with the microbiome that hosts these plasmids, across both similar and different rumen habitats. Our analysis demonstrates that its structure is shaped more by stochasticity than selection. Nevertheless, the plasmidome is an active partner in its intricate relationship with the host microbiome with both interacting with and responding to their environment.


Asunto(s)
Bacterias/genética , Microbiota/genética , Plásmidos/genética , Rumen/microbiología , Animales , Transferencia de Gen Horizontal
12.
J Control Release ; 313: 33-41, 2019 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-31634546

RESUMEN

Interferon regulatory factor 8 (IRF8) protein plays a critical role in the differentiation, polarization, and activation of mononuclear phagocytic cells. In light of previous studies, we explored the therapeutic potential of IRF8 inhibition as immunomodulatory therapy for inflammatory bowel disease (IBD). To this end, we utilized siRNA-loaded lipid-based nanoparticles (siLNPs) and demonstrated a ∼90% reduction of IRF8 mRNA levels in vitro (PV < 0.0001), alongside a notable reduction in IRF8 protein. Moreover, silencing IRF8 ex vivo in splenocytes lead to a profound downregulation of IRF8 protein, followed by an immunomodulatory effect, as represented by a decrease in the secretion of TNFα, IL6 and IL12/IL23 (IL12p40) proinflammatory cytokines (PV = 0.0045, 0.0330, <0.0001, respectively). In order to silence IRF8 in vivo, selectively in inflammatory leukocytes, we used siLNPs that were coated with anti-Ly6C antibodies via our recently published ASSET targeting approach. Through this strategy, we have demonstrated a selective binding of the targeted-LNPs (T-LNPs) to Ly6C + inflammatory leukocytes. Finally, an immunomodulatory effect was demonstrated in vivo in an IBD mouse model with a profound decrease of TNFα, IL6, IL12/IL23, and IL1ß pro-inflammatory cytokines (n = 5, PV < 0.0001, <0.0001, <0.0001, 0.02, respectively) and an improvement of colon-morphology as assessed by colon-length measurements and colonoscopy (PV < 0.0001). Overall, using antibody-targeted siLNPs, we showed a notable reduction of IRF8 mRNA and protein and demonstrated a targeted immunomodulation therapeutic effect ex vivo and in vivo, in the DSS colitis model. We claim that a selective silencing of IRF8 in inflammatory leukocytes (such as Ly6C+) may serve as a therapeutic approach for treating inflammatory disorders.


Asunto(s)
Antiinflamatorios/metabolismo , Enfermedades Inflamatorias del Intestino/terapia , Factores Reguladores del Interferón/genética , Leucocitos/metabolismo , Lípidos/química , Nanopartículas/química , ARN Interferente Pequeño/metabolismo , Animales , Anticuerpos/química , Anticuerpos/metabolismo , Colesterol/química , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Humanos , Inmunomodulación , Factores Reguladores del Interferón/metabolismo , Interleucinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Células RAW 264.7 , Propiedades de Superficie , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
13.
Drug Resist Updat ; 45: 13-29, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31382144

RESUMEN

Immune checkpoint inhibitors (ICI) have emerged as a remarkable treatment option for diverse cancer types. Currently, ICIs are approved for an expanding array of cancer indications. However, the majority of patients still do not demonstrate a durable long-term response following ICI therapy. In addition, many patients receiving ICI therapy develop immune-related adverse events (irAEs) affecting a wide variety of organs. To increase the percentage of patients who benefit from ICI therapy and to reduce the occurrence of irAEs, there is an ongoing effort to combine current ICIs with novel checkpoints inhibitors or other therapeutic approaches to achieve a synergistic effect which is larger than the sum of its parts. In this review we highlight the essential factors for more effective ICI combinations. We describe how the design of these strategies should be driven by the tumor's immunological context. We analyze current combination strategies and describe how they can be improved to unleash the immune system's full anti-cancer potential as well as convert immunologically "cold" tumors into "hot" ones. We examine the efforts to combine current ICIs (PD-1 and CTLA-4) with novel checkpoints (TIM-3, LAG-3, VISTA, TIGIT and others), immunotherapies (CAR-T cells and Cancer Vaccines) and delivery strategies (bispecific antibodies and other delivery platforms). Importantly, we outline how can one optimally combine ICIs with traditional pillars of cancer therapy such as radiation therapy (RT) and chemotherapy. We discuss the considerations regarding successful combination with RT and chemotherapy; these include fractionation schemes and selection of chemotherapeutics which can both directly eradicate cancer cells as well as increase the infiltration of immune cells into tumors. Finally, we critically assess these approaches and attempt to establish their strengths and weaknesses based on pre-clinical and clinical data.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Terapia Combinada/métodos , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Animales , Humanos , Factores Inmunológicos/inmunología , Neoplasias/inmunología
14.
Nano Lett ; 19(3): 1838-1843, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30817160

RESUMEN

The supply of growth factors to engineered tissues is essential for many physiological processes. These processes include the proper organization of the cells into functioning tissues, maintenance of their viability, vasculogenesis, proliferation, and differentiation. Systems to efficiently control the release of growth factors were previously incorporated into tissue engineering scaffolds to affect cells. However, because the initial concentration of the factors in these systems is finite, their ability to provide a long-term physiological effect is limited. Here, we report on a new reloadable system in which 3D fibrous scaffolds conjugated with an anti His-tag antibody enable the retention and controlled release of any His-tag-modified proteinaceous growth factor. The scaffolds can be reloaded in vitro or in vivo with any His-tagged biomolecule at any time according to the physiological need. We show the ability of the scaffolds to release angiogenic factors in a static cell culture or under flow in a microfluidics device and effect on endothelial cells. We also demonstrate the potential of the system to be sequentially reloaded in vivo with various factors, and as a proof of concept, we provide evidence for the efficient in vivo vascularization of scaffolds after reloading with tagged VEGF.


Asunto(s)
Inmunoconjugados/química , Ingeniería de Tejidos , Andamios del Tejido/química , Factor A de Crecimiento Endotelial Vascular/química , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Humanos , Inmunoconjugados/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología
15.
J Mech Behav Biomed Mater ; 91: 345-354, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30654195

RESUMEN

Detecting the presence of circulating tumor cells (CTCs) in peripheral blood can be useful for monitoring treatment in patients, metastasis prognosis, and even early detection. The epidermal growth factor receptor (EGFR) is overexpressed in carcinoma, e.g. in colorectal cancer. Here, we use atomic force microscopy (AFM) force spectroscopy to study the mechanical properties of A431 cells, which simulate EGFR-overexpressing epithelial CTCs and were magnetically isolated by Bio-Ferrography (BF). BF is found useful in isolating individual cancerous cells for mechanical testing, thus avoiding cell-cell interactions. Different stages in the pre-isolation sample preparation steps (namely, cell fixation, PLL coating of the glass substrate, and immunomagnetic labeling) are found to affect the estimated Young's modulus. The BF magnetic isolation step itself does not change the elasticity of the captured cells in comparison to the pre-isolated microbeads-bound cells. The reported increase in the estimated Young's modulus between BF-isolated target cells and fixed cells that are not bound to magnetic microbeads can be used as a quantitative mechanical indicator for objective detection of CTCs. Furthermore, we report a 2.8-fold increase in the adhesion force between the AFM tip and the BF-isolated cells compared to the pre-isolated magnetic microbead-bound A431 fixed cells. This adhesion force correlation could potentially serve as an additional quantitative mechanical indicator for distinguishing between the target and background cells, without the use of cell staining assay and subjective analysis by an expert pathologist. This study demonstrates the powerful combination of the highly sensitive cell isolation by BF and the subsequent analysis of mechanical properties of individual captured cancerous cells by AFM. This combination has potential use in cancer research.


Asunto(s)
Fenómenos Mecánicos , Microscopía de Fuerza Atómica , Fenómenos Biomecánicos , Línea Celular Tumoral , Módulo de Elasticidad , Humanos , Metástasis de la Neoplasia
16.
Front Immunol ; 10: 2921, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921180

RESUMEN

Drugs formulated from monoclonal antibodies (mAbs) are clinically effective in various diseases. Repeated administration of mAbs, however, elicits an immune response in the form of anti-drug-antibodies (ADA), thereby reducing the drug's efficacy. Notwithstanding their importance, the molecular landscape of ADA and the mechanisms involved in their formation are not fully understood. Using a newly developed quantitative bio-immunoassay, we found that ADA concentrations specific to TNFα antagonists can exceed extreme concentrations of 1 mg/ml with a wide range of neutralization capacity. Our data further suggest a preferential use of the λ light chain in a subset of neutralizing ADA. Moreover, we show that administration of TNFα antagonists result in a vaccine-like response whereby ADA formation is governed by the extrafollicular T cell-independent immune response. Our bio-immunoassay coupled with insights on the nature of the immune response can be leveraged to improve mAb immunogenicity assessment and facilitate improvement in therapeutic intervention strategies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Humanos , Inmunoensayo , Factor de Necrosis Tumoral alfa/inmunología
17.
Methods Mol Biol ; 1904: 431-454, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30539485

RESUMEN

Bispecific antibodies (bsAbs) are antibodies with two binding sites directed at different antigens, enabling therapeutic strategies not possible with conventional monoclonal antibodies (mAbs). Since bispecific antibodies are regarded as promising therapeutic agents, many different bispecific design modalities have been evaluated. Many of these are based on antibody fragments or on inclusion of non-antibody components. For some therapeutic applications, full-size, native IgG-like bsAbs may be the optimal format.To prepare bsAbs in IgG format, two challenges should be met. One is that each heavy chain will only pair with the heavy chain of the second specificity and that heavy chain homodimerization will be prevented. The second is that each heavy chain will only pair with the light chain of its own specificity and that pairing with the light chain of the second specificity will be prevented. The first solution to the first criterion (known as knobs into holes, KIH) was presented in 1996 by Genentech and additional solutions were presented more recently. However, until recently, out of >120 published formats, only a handful of solutions for the second criterion that make it possible to produce a bispecific IgG by a single expressing cell were suggested.Here, we present a protocol for preparing bsAbs in IgG format in transfected mammalian cells. For heavy chain dimerization we use KIH while as a solution for the second challenge-correct pairing of heavy and light chains of bispecific IgGs we present our "BIClonals" technology; an engineered (artificial) disulfide bond between the antibodies' variable domains that asymmetrically replaces the natural disulfide bond between CH1 and CL.During our studies of bsAbs we found that H-L chain pairing seems to be driven by VH-VL interfacial interactions that differ between different antibodies; hence, there is no single optimal solution for effective and precise assembly of bispecific IgGs that suits every antibody sequence, making it necessary to carefully evaluate the optimal solution for each new antibody.


Asunto(s)
Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Biespecíficos/inmunología , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/genética , Línea Celular , Expresión Génica , Vectores Genéticos/genética , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/inmunología , Ingeniería de Proteínas , Transfección
18.
Methods Mol Biol ; 1904: 455-480, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30539486

RESUMEN

Monoclonal antibodies (mAbs) are currently the fastest growing class of therapeutic proteins. Parallel to full-length IgG format the development of recombinant technologies provided the production of smaller recombinant antibody variants. The single-chain variable fragment (scFv) antibody is a minimal form of functional antibody comprised of the variable domains of immunoglobulin light and heavy chains connected by a flexible linker. In most cases, scFvs are expressed in the periplasm bacterium E. coli. The production of soluble scFvs is more effective in quantity, however, under the reducing conditions of the E. coli bacterial cytoplasm it is inefficient because of the inability of the disulfide bonds to form. Hence, scFvs are either secreted to the periplasm as soluble proteins or expressed in the cytoplasm as insoluble inclusion bodies and recovered by refolding. The cytoplasmic expression of scFvs as a C-terminal fusion to maltose-binding protein (MBP) provided the high-level production of stable, soluble, and functional fusion protein. The below protocol provides the detailed description of MBP-scFv production in E. coli utilizing two expression systems: pMALc-TNN and pMALc-NHNN. Although the MBP tag does not disrupt the most of antibody activities, the MBP-TNN-scFv product can be cleaved by Tobacco Etch Virus (TEV) protease in order to obtain untagged scFv.The second protocol is for efficient production of Fab antibody fragments as MBP fusion proteins secreted by transiently transfected mammalian cells. While transient transfection is a fast and effective way of obtaining several mgs of antibody for initial screening and validation of antibodies, some antibody sequences express poorly or not at all. For such antibodies, fusion to MBP provides an effective approach for solving the expression problem.


Asunto(s)
Citoplasma/metabolismo , Escherichia coli/crecimiento & desarrollo , Anticuerpos de Cadena Única/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Células HEK293 , Humanos , Ingeniería de Proteínas , Replegamiento Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética
19.
J Control Release ; 291: 80-89, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30342077

RESUMEN

BACKGROUND: Trastuzumab is a monoclonal antibody which demonstrates efficacy for HER2 positive breast cancer patients. Recently, an increased incidence of brain metastasis in trastuzumab-treated patients has been reported. The reason for this may be the effectiveness of systemic trastuzumab allowing patients to survive longer thus providing time for brain metastases to develop, along with the lack of penetration of systemic therapies through the blood brain barrier. In recent years, several administration routes to the brain have been evaluated. Albeit advances in the field, there is still a need for improved delivery of therapeutic antibodies to the brain. To address this challenge, we have developed two gene therapy-based methods enabling continuous secretion of active trastuzumab in the brain. METHODS: We have developed two gene therapy approaches for the delivery of the therapeutic anti-HER2 monoclonal antibody, trastuzumab, to the brain. We utilized the helper dependent adenovirus vector, containing trastuzumab light and heavy chains coding sequences (HDAd-trastuzumab). In the first approach, we used the Transduced Autologous Restorative Gene Therapy (TARGT) platform, in which dermal fibroblasts of human and mouse origin, are ex-vivo transduced with HDAd-trastuzumab vector, rendering continuous secretion of active trastuzumab from the cells locally. These genetically engineered cells were subsequently implanted intracranially to mice, contralateral to HER2 positive breast carcinoma cells inoculation site, enabling continuous secretion of trastuzumab in the brain. In the second approach, we used the same HDAd-trastuzumab viral vector, directly injected intracranially, contralateral to the HER2 positive breast carcinoma cells inoculation site. Both methods enabled therapeutic concentrations of local in-vivo production of active trastuzumab in a mouse model of brain metastatic breast cancer. RESULTS: Trastuzumab secreted from the TARGT platform demonstrated in-vitro affinity and immune recruitment activity (ADCC) similar to recombinant trastuzumab (Herceptin, Genentech). When implanted in the brain of HER2 positive tumor-bearing mice, both the TARGT platform of dermal fibroblasts engineered to secrete trastuzumab and direct injection of HDAd-trastuzumab demonstrated remarkable intracranial tumor growth inhibitory effect. CONCLUSIONS: This work presents two gene therapy approaches for the administration of therapeutic antibodies to the brain. The TARGT platform of dermal fibroblasts engineered to secrete active trastuzumab, and the direct injection of HDAd-trastuzumab viral vector, both rendered continuous in-vivo secretion of active trastuzumab in the brain and demonstrated high efficacy. These two approaches present a proof of concept for promising gene therapy based administration methods for intracranial tumors as well as other brain diseases.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/terapia , Neoplasias de la Mama/patología , Técnicas de Transferencia de Gen , Trastuzumab/uso terapéutico , Adenoviridae/genética , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/metabolismo , Neoplasias Encefálicas/patología , Neoplasias de la Mama/terapia , Células Cultivadas , Sistemas de Liberación de Medicamentos/métodos , Femenino , Fibroblastos/metabolismo , Fibroblastos/trasplante , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Transducción Genética , Trastuzumab/administración & dosificación , Trastuzumab/genética
20.
J Virol Methods ; 260: 62-69, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30003926

RESUMEN

Environmental surveillance (EnvS)2 is an important tool for monitoring the presence of poliovirus in endemic and poliovirus free regions. Unlike acute flaccid paralysis (AFP)3 surveillance, EnvS can monitor large populations using small numbers of samples and detect the introduction of poliovirus even before the appearance of AFP cases. Early detection and timely response can prevent the onset of poliovirus associated AFP, as was demonstrated by silent poliovirus transmission in Israel in 2013. Although EnvS is currently recommended as supplementary to AFP surveillance, it is limited to laboratories with equipment for poliovirus concentration and to regions where samples can be easily transported under temperature controlled conditions to such facilities. However the highest risk of poliovirus re-emergence is in developing countries where such conditions do not exist. We developed and evaluated an affinity purification method using antibody or poliovirus receptor (CD155) presenting bacteriophage covered magnetic beads for poliovirus concentration. This method requires only simple, inexpensive and portable equipment. Though tested only on Sabin 1 spiked sewage samples it provided better recovery than our current polyethylene glycol (PEG)4/NaCl- based concentration method. On site use of this method might facilitate EnvS in currently inaccessible remote regions by significantly reducing the volume of sample that needs to be transported back to the laboratory under temperature-controlled conditions5.


Asunto(s)
Monitoreo del Ambiente/métodos , Poliomielitis/prevención & control , Poliovirus/aislamiento & purificación , Anticuerpos Monoclonales/química , Anticuerpos Antivirales/química , Proteínas de Unión al GTP/química , Humanos , Imanes , Microesferas , Poliomielitis/virología , ARN Viral/química , Receptores Virales/química , Factores de Tiempo , Acoplamiento Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...