Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioengineered ; 14(1): 2252667, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37661761

RESUMEN

Disadvantages of using murine monoclonal antibodies (mAb) in human therapy, such as immunogenicity response, led to the development of technologies to transform murine antibodies into human antibodies. The murine anti-FGF2 3F12E7 mAb was proposed as a promising agent to treat metastatic melanoma tumors; once it blocks the FGF2, responsible for playing a role in tumor growth, angiogenesis, and metastasis. Considering the therapeutic potential of anti-FGF2 3F12E7 mAb and its limited use in humans due to its origin, we used this antibody as the template for a guided selection humanization technique to obtain human anti-FGF2 mAbs. Three Fab libraries (murine, hybrid, and human) were constructed for humanization. The libraries were phage-displayed, and the panning was performed against recombinant human FGF2 (rFGF2). The selected human variable light and heavy chains were cloned into AbVec vectors for full-length IgG expression into HEK293-F cells. Surface plasmon resonance analyses showed binding to rFGF2 of seven mAbs out of 20 expressed. Assays performed with these mAbs resulted in two that showed proliferation reduction and cell migration attenuation of HUVEC and SK-Mel-28 melanoma cells. In-silico analyses predicted that these two human anti-FGF2 mAbs interact with FGF2 at a similar patch of residues than the chimeric anti-FGF2 antibody, comprehending a region within the heparin-binding domains of FGF2, essential for its function. These results are comparable to those achieved by the murine anti-FGF2 3F12E7 mAb and showed success in the humanization process and selection of two human mAbs with the potential to inhibit undesirable FGF2 roles.


The guided selection humanization process enabled the production of 20 human mAbs anti-FGF2;Seven human anti-FGF2 mAbs showed binding to the rFGF2 antigen in the SPR binding assay;Two human anti-FGF2 mAbs inhibited the proliferation and migration of HUVEC and SK-Mel-28 cells and were predicted to contact the FGF2 at a similar patch of residues than the original mAb.


Asunto(s)
Anticuerpos Monoclonales , Melanoma , Humanos , Animales , Ratones , Hibridomas , Células HEK293 , Proliferación Celular
2.
Sci Rep ; 11(1): 1432, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33446839

RESUMEN

Single-chain variable fragments (scFvs) are small-sized artificial constructs composed of the immunoglobulin heavy and light chain variable regions connected by a peptide linker. We have previously described an anti-fibroblast growth factor 2 (FGF2) immunoglobulin G (IgG) monoclonal antibody (mAb), named 3F12E7, with notable antitumor potential revealed by preclinical assays. FGF2 is a known angiogenesis-associated molecule implicated in tumor progression. In this report, we describe a recombinant scFv format for the 3F12E7 mAb. The results demonstrate that the generated 3F12E7 scFv, although prone to aggregation, comprises an active anti-FGF2 product that contains monomers and small oligomers. Functionally, the 3F12E7 scFv preparations specifically recognize FGF2 and inhibit tumor growth similar to the corresponding full-length IgG counterpart in an experimental model. In silico molecular analysis provided insights into the aggregation propensity and the antigen-recognition by scFv units. Antigen-binding determinants were predicted outside the most aggregation-prone hotspots. Overall, our experimental and prediction dataset describes an scFv scaffold for the 3F12E7 mAb and also provides insights to further engineer non-aggregated anti-FGF2 scFv-based tools for therapeutic and research purposes.


Asunto(s)
Inhibidores de la Angiogénesis/química , Antineoplásicos Inmunológicos/química , Factor 2 de Crecimiento de Fibroblastos/química , Proteínas de Neoplasias/química , Anticuerpos de Cadena Única , Humanos , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética
3.
Front Immunol ; 10: 1023, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31156623

RESUMEN

Several studies report the key role of the vascular endothelial growth factor (VEGF) signaling on angiogenesis and on tumor growth. This has led to the development of a number of VEGF-targeted agents to treat cancer patients by disrupting the tumor blood vessel supply. Of them, bevacizumab, an FDA-approved humanized monoclonal antibody against VEGF, is the most promising. Although the use of antibodies targeting the VEGF pathway has shown clinical benefits associated with a reduction in the tumor blood vessel density, the inhibition of VEGF-driven vascular effects is only part of the functional mechanism of these therapeutic agents in the tumor ecosystem. Compelling reports have demonstrated that VEGF confers, in addition to the activation of angiogenesis-related processes, immunosuppressive properties in tumors. It is also known that structural remodeling of the tumor blood vessel bed by anti-VEGF approaches affect the influx and activation of immune cells into tumors, which might influence the therapeutic results. Besides that, part of the therapeutic effects of antiangiogenic antibodies, including their role in the tumor vascular network, might be triggered by Fc receptors in an antigen-independent manner. In this mini-review, we explore the role of VEGF inhibitors in the tumor microenvironment with focus on the immune system, discussing around the functional contribution of both bevacizumab's Fab and Fc domains to the therapeutic results and the combination of bevacizumab therapy with other immune-stimulatory settings, including adjuvant-based vaccine approaches.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Inmunológicos/farmacología , Susceptibilidad a Enfermedades/inmunología , Neoplasias/etiología , Neoplasias/metabolismo , Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Epítopos/inmunología , Humanos , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Factores Inmunológicos/administración & dosificación , Inmunomodulación/efectos de los fármacos , Modelos Biológicos , Terapia Molecular Dirigida/métodos , Neoplasias/patología , Neoplasias/terapia , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/etiología , Neovascularización Patológica/metabolismo
4.
Biochimie ; 166: 173-183, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30981871

RESUMEN

Formation of new blood vessels from preexisting ones, a process known as angiogenesis, is one of the limiting steps for success in treatment of ischemic disorders. Therefore, efforts to understanding and characterize new agents capable to stimulate neovascularization are a worldwide need. Crataeva tapia bark lectin (CrataBL) has been shown to have chemoattractant properties for endothelial cells through the stimulation of migration and invasiveness of human umbilical vein endothelial cells (HUVEC) because it is a positively charged protein with high affinity to glycosaminoglycan. In addition, CrataBL increased the production of chondroitin and heparan sulfate in endothelial cells. These findings orchestrated specific adhesion on collagen I and phosphorylation of tyrosine kinase receptors, represented by vascular endothelial growth factor receptor-2 (VEGFR-2) and fibroblast growth factor receptor (FGFR), whose downstream pathways trigger the angiogenic cascade increasing cell viability, cytoskeleton rearrangement, cell motility, and tube formation. Moreover, CrataBL inhibited the activity of matrix metalloproteases type 2 (MMP-2), a protein related to tissue remodeling. Likewise, CrataBL improved wound healing and increased the number of follicular structures in lesioned areas produced in the dorsum-cervical region of C57BL/6 mice. These outcomes altogether indicate that CrataBL is a pro-angiogenic and healing agent.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Condroitín/metabolismo , Heparitina Sulfato/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Lectinas de Plantas/farmacología , Animales , Capparaceae/metabolismo , Movimiento Celular/efectos de los fármacos , Factores Quimiotácticos/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Cicatrización de Heridas/efectos de los fármacos
5.
Cancer Sci ; 107(4): 551-5, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27079440

RESUMEN

Tumors require blood supply and, to overcome this restriction, induce angiogenesis. Vascular endothelial growth factor (VEGF) plays an important role in this process, which explains the great number of antiangiogenic therapies targeting VEGF. The research and development of targeted therapy has led to the approval of bevacizumab, a humanized anti-VEGF monoclonal antibody (mAb), in clinical settings. However, side effects have been reported, usually as a consequence of bolus-dose administration of the antibody. This limitation could be circumvented through the use of anti-idiotype (Id) antibodies. In the present study, we evaluated the efficacy of an active VEGF-binding immune response generated by an anti-bevacizumab idiotype mAb, 10.D7. The 10.D7 anti-Id mAb vaccination led to detectable levels of VEGF-binding anti-anti-Id antibodies. In order to examine whether this humoral immune response could have implications for tumor development, 10.D7-immunized mice were challenged with B16-F10 tumor cells. Mice immunized with 10.D7 anti-Id mAb revealed reduced tumor growth when compared to control groups. Histological analyses of tumor sections from 10.D7-immunized mice showed increased necrotic areas, decreased CD31-positive vascular density and reduced CD68-positive cell infiltration. Our results encourage further therapeutic studies, particularly if one considers that the anti-Id therapeutic vaccination maintains stable levels of VEGF-binding antibodies, which might be useful in the control of tumor relapse.


Asunto(s)
Anticuerpos Antiidiotipos/administración & dosificación , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/inmunología , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/inmunología , Animales , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Bevacizumab/administración & dosificación , Bevacizumab/efectos adversos , Línea Celular Tumoral , Humanos , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Cancer Lett ; 371(2): 151-60, 2016 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-26655277

RESUMEN

Compelling evidence suggests that fibroblast growth factor 2 (FGF2), overexpressed in melanomas, plays an important role in tumor growth, angiogenesis and metastasis. In this study, we evaluated the therapeutic use of a new anti-FGF2 monoclonal antibody (mAb), 3F12E7, using for that the B16-F10 melanoma model. The FGF2 neutralizing effect of this antibody was certified by in vitro assays, which allowed the further track of its possible in vivo application. 3F12E7 mAb could be retained in B16-F10 tumors, as shown by antibody low-pH elution and nuclear medicine studies, and also led to reduction in number and size of metastatic foci in lungs, when treatment starts one day after intravenous injection of B16-F10 cells. Such data were accompanied by decreased CD34(+) tumor vascular density and impaired subcutaneous tumor outgrowth. Treatments starting one week after melanoma cell intravenous injection did not reduce tumor burden, remaining the therapeutic effectiveness restricted to early-adopted regimens. Altogether, the presented anti-FGF2 3F12E7 mAb stands as a promising agent to treat metastatic melanoma tumors in adjuvant settings.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Neoplasias Pulmonares/prevención & control , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de la Angiogénesis/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Antígenos CD34/metabolismo , Línea Celular Tumoral , Factor 2 de Crecimiento de Fibroblastos/inmunología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inmunohistoquímica , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Masculino , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/metabolismo , Melanoma Experimental/secundario , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Carga Tumoral
7.
J Gene Med ; 12(3): 310-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20077434

RESUMEN

BACKGROUND: Vascular endothelial growth factor (VEGF) has mostly been tested to treat ischemic diseases, although the outcomes obtained are not satisfactory. Our hypothesis is that the local transient expression of VEGF and stem cell mobilizer granulocyte colony-stimulating factor (G-CSF) genes in ischemic limbs can complement their activities and be more efficient for limb recovery. METHODS: Limb ischemia was surgically induced in mice and 50 microg of VEGF and/or G-CSF genes were locally transferred by electroporation. After 3-4 weeks, evidence of necrosis by visual inspection, capillary density, muscle mass, muscle force and hematopoietic cell mobilization were evaluated. RESULTS: After 4 weeks, 70% and 90% of the animals of the ischemic group (IG) and VEGF-treated group (VG), respectively, presented limb necrosis, in contrast to only 10% observed in the group of mice treated with both VEGF and G-CSF genes (VGG). Recovery of muscle mass and muscle force was higher than 60% in the VGG compared to the non-ischemic group. The mobilization of Sca1+ cells and neutrophils was also higher in the VGG, which may explain the lower level of necrosis observed in this group (22%, in contrast to 70% in the IG). Capillary density and degree of fibrosis were determined in weeks 3 and 4, and also showed a clear benefit as a result of the use of the G-CSF and VEGF genes together. CONCLUSIONS: Gene therapy using VEGF and G-CSF demonstrated a synergistic effect promoting vessel and tissue repair in mouse hind limb ischemia.


Asunto(s)
Extremidades/irrigación sanguínea , Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos/genética , Isquemia/terapia , Enfermedades Vasculares Periféricas/terapia , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Isquemia/sangre , Isquemia/etiología , Masculino , Ratones , Ratones Endogámicos BALB C , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/fisiología , Neovascularización Fisiológica/genética , Enfermedades Vasculares Periféricas/complicaciones , Regeneración/genética
8.
J Gene Med ; 11(4): 345-53, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19194978

RESUMEN

BACKGROUND: Granulocyte-colony-stimulating factor (GM-CSF) is a pleiotropic factor for hematopoiesis that stimulates myeloblasts, monoblasts and mobilization of bone marrow stem cells. Therefore, the GM-CSF gene is a potential candidate for vessel formation and tissue remodeling in the treatment of ischemic diseases. METHODS: A new mouse limb ischemia was established by surgery and gene transfer was performed by injection of 100 microg of a plasmid carrying GM-CSF. Muscle force and weight, histology, capillary density, circulating stem cells and monocytes were determined after 3-4 weeks. RESULTS: More than 60% of nontreated ischemic animals showed gangrene below the heel after 4 weeks, whereas the GM-CSF gene-treated animals showed only darkening of nails or toes. These animals demonstrated a full recovery of the affected muscles in terms of weight, force and muscle fiber structure, but the muscles of nontreated ischemic animals lost approximately 50% weight, 86% force and their regular structure. When the GM-CSF gene was injected into the contralateral limb, only partial loss was observed, demonstrating a distant effect of GM-CSF. The capillary density in the GM-CSF-treated group was 52% higher in relation to the nontreated group. Blood analysis by flow cytometry showed that the GM-CSF-treated group had 10-20% higher levels of circulating monocytes and Sca-1(+). CONCLUSIONS: We conclude that the direct administration of GM-CSF gene in limb ischemia had a strong therapeutic effect because it promoted the recovery of muscle mass, force and structure by mobilizing therapeutic cells and augmenting the number of vessels.


Asunto(s)
Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Isquemia/terapia , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Extremidades/patología , Hematopoyesis/efectos de los fármacos , Ratones , Músculo Esquelético/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Plásmidos/administración & dosificación , Resultado del Tratamiento
9.
Vaccine ; 26(31): 3812-7, 2008 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-18556095

RESUMEN

Aiming to evaluate some parameters to influence the immune response to DNA vaccination, we compare three protocols of DNA immunization (i.m. injections, i.m. injections followed by electroporation, and the effect of i.p. injection of stably antigen-transfected cells before DNA administration), using three different antigens. Statistical analyses showed that electroporation after intramuscular injections provided an immune response comparable to that obtained by pre-treatment with antigen-transfected cells and similar to that obtained by protein immunization. The results allowed us selecting a protocol that worked well for all three antigens and reinforced the idea that high level of gene expression is essential to get good immunization.


Asunto(s)
Electroporación , Vacunación/métodos , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Traslado Adoptivo , Animales , Femenino , Inyecciones Intramusculares , Ratones , Ratones Endogámicos BALB C
10.
J Biotechnol ; 127(4): 615-25, 2007 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16989916

RESUMEN

Since carcinoembryonic antigen (CEA) is expressed during embryonic life, it is not immunogenic in humans. The use of anti-idiotypic (Id) antibodies as a surrogate of antigen in the immunization has been considered a promising strategy for breaking tolerance to some tumor associated antigens. We have described an anti-Id monoclonal antibody (MAb), designated 6.C4, which is able to mimic CEA functionally. The anti-Id MAb 6.C4 was shown to elicit antibodies that recognized CEA in vitro and in vivo. In the present study, we sought to verify whether a single chain (scFv) antibody obtained, the scFv 6.C4, would retain the ability to mimic CEA. Two scFv containing the variable heavy and light chain domains of 6.C4 were constructed with a 15-amino acid linker: one with and another without signal peptide. DNA immunization of mice with both forms of scFv individually elicited antibodies able to recognize CEA.


Asunto(s)
Adenocarcinoma/inmunología , Anticuerpos Antiidiotipos/genética , Antígeno Carcinoembrionario/inmunología , Neoplasias del Colon/inmunología , Fragmentos de Inmunoglobulinas/genética , Región Variable de Inmunoglobulina/genética , Imitación Molecular/inmunología , Adenocarcinoma/patología , Animales , Anticuerpos Antiidiotipos/administración & dosificación , Anticuerpos Antiidiotipos/inmunología , Especificidad de Anticuerpos , Neoplasias del Colon/patología , Femenino , Humanos , Hibridomas/inmunología , Inmunización , Fragmentos de Inmunoglobulinas/administración & dosificación , Fragmentos de Inmunoglobulinas/inmunología , Región Variable de Inmunoglobulina/administración & dosificación , Región Variable de Inmunoglobulina/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA