Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(6): 1020-1032, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38831106

RESUMEN

The efficacy of T cell-based immunotherapies is limited by immunosuppressive pressures in the tumor microenvironment. Here we show a predominant role for the interaction between BTLA on effector T cells and HVEM (TNFRSF14) on immunosuppressive tumor microenvironment cells, namely regulatory T cells. High BTLA expression in chimeric antigen receptor (CAR) T cells correlated with poor clinical response to treatment. Therefore, we deleted BTLA in CAR T cells and show improved tumor control and persistence in models of lymphoma and solid malignancies. Mechanistically, BTLA inhibits CAR T cells via recruitment of tyrosine phosphatases SHP-1 and SHP-2, upon trans engagement with HVEM. BTLA knockout thus promotes CAR signaling and subsequently enhances effector function. Overall, these data indicate that the BTLA-HVEM axis is a crucial immune checkpoint in CAR T cell immunotherapy and warrants the use of strategies to overcome this barrier.


Asunto(s)
Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Receptores Inmunológicos , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Microambiente Tumoral , Animales , Humanos , Inmunoterapia Adoptiva/métodos , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Ratones , Microambiente Tumoral/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Linfocitos T Reguladores/inmunología , Transducción de Señal , Línea Celular Tumoral , Neoplasias/inmunología , Neoplasias/terapia , Ratones Noqueados
2.
Radiology ; 310(2): e231406, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38411517

RESUMEN

Background Chimeric antigen receptor (CAR) T cells are a promising cancer therapy; however, reliable and repeatable methods for tracking and monitoring CAR T cells in vivo remain underexplored. Purpose To investigate direct and indirect imaging strategies for tracking the biodistribution of CAR T cells and monitoring their therapeutic effect in target tumors. Materials and Methods CAR T cells co-expressing a tumor-targeting gene (anti-CD19 CAR) and a human somatostatin receptor subtype 2 (hSSTr2) reporter gene were generated from human peripheral blood mononuclear cells. After direct labeling with zirconium 89 (89Zr)-p-isothiocyanatobenzyl-desferrioxamine (DFO), CAR T cells were intravenously injected into immunodeficient mice with a CD19-positive and CD19-negative human tumor xenograft on the left and right flank, respectively. PET/MRI was used for direct in vivo imaging of 89Zr-DFO-labeled CAR T cells on days 0, 1, 3, and 7 and for indirect cell imaging with the radiolabeled somatostatin receptor-targeted ligand gallium 68 (68Ga)-DOTA-Tyr3-octreotide (DOTATOC) on days 6, 9, and 13. On day 13, mice were euthanized, and tissues and tumors were excised. Results The 89Zr-DFO-labeled CAR T cells were observed on PET/MRI scans in the liver and lungs of mice (n = 4) at all time points assessed. However, they were not visualized in CD19-positive or CD19-negative tumors, even on day 7. Serial 68Ga-DOTATOC PET/MRI showed CAR T cell accumulation in CD19-positive tumors but not in CD19-negative tumors from days 6 to 13. Notably, 68Ga-DOTATOC accumulation in CD19-positive tumors was highest on day 9 (mean percentage injected dose [%ID], 3.7% ± 1.0 [SD]) and decreased on day 13 (mean %ID, 2.6% ± 0.7) in parallel with a decrease in tumor volume (day 9: mean, 195 mm3 ± 27; day 13: mean, 127 mm3 ± 43) in the group with tumor growth inhibition. Enhanced immunohistochemistry staining of cluster of differentiation 3 (CD3) and hSSTr2 was also observed in excised CD19-positive tumor tissues. Conclusion Direct and indirect cell imaging with PET/MRI enabled in vivo tracking and monitoring of CAR T cells in an animal model. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Bulte in this issue.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , Xenoinjertos , Radioisótopos de Galio , Receptores de Somatostatina , Leucocitos Mononucleares , Distribución Tisular , Tomografía de Emisión de Positrones , Imagen por Resonancia Magnética , Modelos Animales de Enfermedad , Linfocitos T
3.
Mol Cancer ; 22(1): 200, 2023 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-38066564

RESUMEN

BACKGROUND: Commercial anti-CD19 chimeric antigen receptor T-cell therapies (CART19) are efficacious against advanced B-cell non-Hodgkin lymphoma (NHL); however, most patients ultimately relapse. Several mechanisms contribute to this failure, including CD19-negative escape and CAR T dysfunction. All four commercial CART19 products utilize the FMC63 single-chain variable fragment (scFv) specific to a CD19 membrane-distal epitope and characterized by slow association (on) and dissociation (off) rates. We hypothesized that a novel anti-CD19 scFv that engages an alternative CD19 membrane-proximal epitope independent of FMC63 and that is characterized by faster on- and off-rates could mitigate CART19 failure and improve clinical efficacy. METHODS: We developed an autologous CART19 product with 4-1BB co-stimulation using a novel humanized chicken antibody (h1218). This antibody is specific to a membrane-proximal CD19 epitope and harbors faster on/off rates compared to FMC63. We tested h1218-CART19 in vitro and in vivo using FMC63-CART19-resistant models. We conducted a first-in-human multi-center phase I clinical trial to test AT101 (clinical-grade h1218-CART19) in patients with relapsed or refractory (r/r) NHL. RESULTS: Preclinically, h1218- but not FMC63-CART19 were able to effectively eradicate lymphomas expressing CD19 point mutations (L174V and R163L) or co-expressing FMC63-CAR19 as found in patients relapsing after FMC63-CART19. Furthermore, h1218-CART19 exhibited enhanced killing of B-cell malignancies in vitro and in vivo compared with FMC63-CART19. Mechanistically, we found that h1218-CART19 had reduced activation-induced cell death (AICD) and enhanced expansion compared to FMC63-CART19 owing to faster on- and off-rates. Based on these preclinical results, we performed a phase I dose-escalation trial, testing three dose levels (DL) of AT101 (the GMP version of h1218) using a 3 + 3 design. In 12 treated patients (7 DLBCL, 3 FL, 1 MCL, and 1 MZL), AT101 showed a promising safety profile with 8.3% grade 3 CRS (n = 1) and 8.3% grade 4 ICANS (n = 1). In the whole cohort, the overall response rate was 91.7%, with a complete response rate of 75.0%, which improved to 100% in DL-2 and -3. AT101 expansion correlates with CR and B-cell aplasia. CONCLUSIONS: We developed a novel, safe, and potent CART19 product that recognizes a membrane-proximal domain of CD19 with fast on- and off-rates and showed significant efficacy and promising safety in patients with relapsed B-cell NHL. TRIAL REGISTRATION: NCT05338931; Date: 2022-04-01.


Asunto(s)
Linfoma no Hodgkin , Receptores de Antígenos de Linfocitos T , Receptores Quiméricos de Antígenos , Humanos , Anticuerpos , Antígenos CD19 , Epítopos/metabolismo , Inmunoterapia Adoptiva/efectos adversos , Linfoma no Hodgkin/terapia , Linfoma no Hodgkin/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores
4.
Biochem Biophys Res Commun ; 545: 33-39, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33535104

RESUMEN

Zika virus (ZIKV) infection in both infants and adults is associated with neurological complications including, but not limited to, microcephaly and Guillain-Barre syndrome. Antibody therapy can be effective against virus infection. We isolated ZIKV envelope domain III-specific neutralizing antibodies (nAbs) from two convalescent patients with ZIKV infection. One antibody, 2F-8, exhibited potent in vitro neutralizing activity against Asian and American strains of ZIKV. To prevent FcγR-mediated antibody-dependent enhancement, we prepared IgG1 with LALA variation. A single dose of 2F-8 in the context of IgG1 or IgG1-LALA prior to or post lethal ZIKV challenge conferred complete protection in mice.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Proteínas del Envoltorio Viral/inmunología , Virus Zika/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Antivirales/administración & dosificación , Especificidad de Anticuerpos , Modelos Animales de Enfermedad , Femenino , Humanos , Técnicas In Vitro , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Pruebas de Neutralización , Embarazo , Dominios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Proteínas del Envoltorio Viral/química , Virus Zika/química , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/prevención & control
5.
PLoS Pathog ; 15(2): e1007375, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30707748

RESUMEN

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease localized to China, Japan, and Korea that is characterized by severe hemorrhage and a high fatality rate. Currently, no specific vaccine or treatment has been approved for this disease. To develop a therapeutic agent for SFTS, we isolated antibodies from a phage-displayed antibody library that was constructed from a patient who recovered from SFTS virus (SFTSV) infection. One antibody, designated as Ab10, was reactive to the Gn envelope glycoprotein of SFTSV and protected host cells and A129 mice from infection in both in vitro and in vivo experiments. Notably, Ab10 protected 80% of mice, even when injected 5 days after inoculation with a lethal dose of SFTSV. Using cross-linker assisted mass spectrometry and alanine scanning, we located the non-linear epitope of Ab10 on the Gn glycoprotein domain II and an unstructured stem region, suggesting that Ab10 may inhibit a conformational alteration that is critical for cell membrane fusion between the virus and host cell. Ab10 reacted to recombinant Gn glycoprotein in Gangwon/Korea/2012, HB28, and SD4 strains. Additionally, based on its epitope, we predict that Ab10 binds the Gn glycoprotein in 247 of 272 SFTSV isolates previously reported. Together, these data suggest that Ab10 has potential to be developed into a therapeutic agent that could protect against more than 90% of reported SFTSV isolates.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Phlebovirus/inmunología , Adulto , Animales , Anticuerpos Neutralizantes/fisiología , Anticuerpos Antivirales/metabolismo , Infecciones por Bunyaviridae/terapia , Epítopos/inmunología , Femenino , Fiebre , Glutamina/inmunología , Glutamina/metabolismo , Glicoproteínas/inmunología , Células HEK293 , Humanos , Leucopenia , Masculino , Ratones , Ratones Noqueados , Pruebas de Neutralización , Phlebovirus/patogenicidad , República de Corea , Trombocitopenia/inmunología , Proteínas del Envoltorio Viral/inmunología
6.
J Cell Mol Med ; 24(2): 1738-1749, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31863639

RESUMEN

Glioblastoma is an aggressive brain tumour found in adults, and the therapeutic approaches available have not significantly increased patient survival. Recently, we discovered that ELTD1, an angiogenic biomarker, is highly expressed in human gliomas. Polyclonal anti-ELTD1 treatments were effective in glioma pre-clinical models, however, pAb binding is potentially promiscuous. Therefore, the aim of this study was to determine the effects of an optimized monoclonal anti-ELTD1 treatment in G55 xenograft glioma models. MRI was used to assess the effects of the treatments on animal survival, tumour volumes, perfusion rates and binding specificity. Immunohistochemistry and histology were conducted to confirm and characterize microvessel density and Notch1 levels, and to locate the molecular probes. RNA-sequencing was used to analyse the effects of the mAb treatment. Our monoclonal anti-ELTD1 treatment significantly increased animal survival, reduced tumour volumes, normalized the vasculature and showed higher binding specificity within the tumour compared with both control- and polyclonal-treated mice. Notch1 positivity staining and RNA-seq results suggested that ELTD1 has the ability to interact with and interrupt Notch1 signalling. Although little is known about ELTD1, particularly about its ligand and pathways, our data suggest that our monoclonal anti-ELTD1 antibody is a promising anti-angiogenic therapeutic in glioblastomas.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Receptores Acoplados a Proteínas G/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Pollos , Glioblastoma/patología , Humanos , Ratones , Microvasos/efectos de los fármacos , Microvasos/patología , Receptores Notch/metabolismo , Carga Tumoral/efectos de los fármacos
7.
Methods ; 154: 125-135, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30292795

RESUMEN

Antibody selection for antibody-drug conjugates (ADCs) has traditionally depended on its internalization into the target cell, although ADC efficacy also relies on recycling of the receptor-ADC complex, endo-lysosomal trafficking, and subsequent linker/antibody proteolysis. In this study, we observed that a bispecific anti-murine platelet-derived growth factor receptor beta (mPDGFRß) x cotinine single-chain variable fragment (scFv)-kappa constant region (Cκ)-scFv fusion protein and cotinine-duocarmycin can form an ADC-like complex to induce cytotoxicity against mPDGFRß expressing cells. Multiple anti-mPDGFRß antibody candidates can be produced in this bispecific scFv-Cκ-scFv fusion protein format and tested for their ability to deliver cotinine-conjugated cytotoxic drugs, thus providing an improved approach for antibody selection in ADC development.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Inmunoconjugados/uso terapéutico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Cotinina , Humanos , Inmunoconjugados/farmacología , Ratones , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/inmunología
9.
Mol Pharm ; 16(1): 165-172, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30521347

RESUMEN

A number of cancer-targeting peptide-drug conjugates (PDCs) have been explored as alternatives to antibody-drug conjugates (ADCs) for targeted cancer therapy. However, the much shorter circulation half-life of PDCs compared with ADCs in vivo has limited their therapeutic value and thus their translation into the clinic, highlighting the need to develop new approaches for extending the half-life of PDCs. Here, we report a new strategy for targeted cancer therapy of a PDC based on a molecular hybrid between an antihapten antibody and a hapten-labeled PDC. An anticotinine antibody (Abcot) was used as a model antihapten antibody. The anticancer drug SN38 was linked to a cotinine-labeled aptide specific to extra domain B of fibronectin (cot-APTEDB), yielding the model PDC, cot-APTEDB-SN38. The cotinine-labeled PDC showed specific binding to and cytotoxicity toward an EDB-overexpressing human glioblastoma cell line (U87MG) and also formed a hybrid complex (HC) with Abcot in situ, designated HC[cot-APTEDB-SN38/Abcot]. In glioblastoma-bearing mice, in situ HC[cot-APTEDB-SN38/Abcot] significantly extended the circulation half-life of cot-APTEDB-SN38 in blood, and it enhanced accumulation and penetration within the tumor and, ultimately, inhibition of tumor growth. These findings suggest that the present platform holds promise as a new, targeted delivery strategy for PDCs in anticancer therapy.


Asunto(s)
Anticuerpos/química , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Péptidos/química , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Femenino , Glioma/tratamiento farmacológico , Humanos , Inmunoconjugados/química , Etiquetado Corte-Fin in Situ , Irinotecán/química , Irinotecán/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos
11.
Int J Mol Sci ; 20(2)2019 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-30669409

RESUMEN

: YYB-101 is a humanized rabbit anti-human hepatocyte growth factor (HGF)-neutralizing antibody currently in clinical trial. To test the effect of HGF neutralization with antibody on anti-cancer T cell immunity, we generated surrogate antibodies that are reactive to the mouse homologue of the epitope targeted by YYB-101. First, we immunized a chicken with human HGF and monitored changes in the B cell repertoire by next-generation sequencing (NGS). We then extracted the VH gene repertoire from the NGS data, clustered it into components by sequence homology, and classified the components by the change in the number of unique VH sequences and the frequencies of the VH sequences within each component following immunization. Those changes should accompany the preferential proliferation and somatic hypermutation or gene conversion of B cells encoding HGF-reactive antibodies. One component showed significant increases in the number and frequencies of unique VH sequences and harbored genes encoding antibodies that were reactive to human HGF and competitive with YYB-101 for HGF binding. Some of the antibodies also reacted to mouse HGF. The selected VH sequences shared 98.3% identity and 98.9% amino acid similarity. It is therefore likely that the antibodies encoded by them all react to the epitope targeted by YYB-101.


Asunto(s)
Anticuerpos/genética , Anticuerpos/inmunología , Mapeo Epitopo , Epítopos de Linfocito B/inmunología , Factor de Crecimiento de Hepatocito/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Secuencia de Aminoácidos , Animales , Anticuerpos/química , Linfocitos B/inmunología , Linfocitos B/metabolismo , Pollos , Mapeo Epitopo/métodos , Biblioteca de Genes , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Ratones
12.
Int J Mol Sci ; 20(20)2019 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-31614869

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) induces severe aggravating respiratory failure in infected patients, frequently resulting in mechanical ventilation. As limited therapeutic antibody is accumulated in lung tissue following systemic administration, inhalation is newly recognized as an alternative, possibly better, route of therapeutic antibody for pulmonary diseases. The nebulization process, however, generates diverse physiological stresses, and thus, the therapeutic antibody must be resistant to these stresses, remain stable, and form minimal aggregates. We first isolated a MERS-CoV neutralizing antibody that is reactive to the receptor-binding domain (RBD) of spike (S) glycoprotein. To increase stability, we introduced mutations into the complementarity-determining regions (CDRs) of the antibody. In the HCDRs (excluding HCDR3) in this clone, two hydrophobic residues were replaced with Glu, two residues were replaced with Asp, and four residues were replaced with positively charged amino acids. In LCDRs, only two Leu residues were replaced with Val. These modifications successfully generated a clone with significantly greater stability and equivalent reactivity and neutralizing activity following nebulization compared to the original clone. In summary, we generated a MERS-CoV neutralizing human antibody that is reactive to recombinant MERS-CoV S RBD protein for delivery via a pulmonary route by introducing stabilizing mutations into five CDRs.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Regiones Determinantes de Complementariedad/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Administración por Inhalación , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/genética , Chlorocebus aethiops , Regiones Determinantes de Complementariedad/genética , Humanos , Células Vero
13.
Angew Chem Int Ed Engl ; 58(7): 2005-2010, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30600870

RESUMEN

Peptide-based therapeutics have suffered from a short plasma half-life. On the other hand, antibodies suffer from poor penetration into solid tumors owing to their large size. Herein, we present a new molecular form, namely a hybrid complex between a hapten-labeled bispecific peptide and an anti-hapten antibody ("HyPEP-body"), that may be able to overcome the aforementioned limitation. The bispecific peptide containing a cotinine tag was synthesized by linking a peptide specific to fibronectin extra domain B (EDB) and a peptide able to bind and inhibit vascular endothelial growth factor (VEGF), yielding cot-biPEPEDB-VEGF . Simple mixing of cot-biPEPEDB-VEGF and anti-cotinine antibody (Abcot ) yielded the hybrid complex, HyPEPEDB-VEGF . HyPEPEDB-VEGF retained the characteristics of the included peptides, and showed improved pharmacokinetic behavior. Moreover, HyPEPEDB-VEGF showed tumor growth inhibition with excellent tumor accumulation and penetration. These findings suggest that the hybrid platform described here offers a solution for most peptide therapeutics that suffer from a short circulation half-life in blood.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Péptidos/farmacología , Animales , Anticuerpos Biespecíficos/química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Células PC-3 , Péptidos/química , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Biochem Biophys Res Commun ; 496(3): 846-851, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29374509

RESUMEN

Viral pathogens have evolved a wide range of tactics to evade host immune responses and thus propagate effectively. One efficient tactic is to divert host immune responses toward an immunodominant decoy epitope and to induce non-neutralizing antibodies toward this epitope. Therefore, it is expected that the amount of decoy epitope in a subunit vaccine can affect the level of neutralizing antibody in an immunized animal. In this study, we tested this hypothesis by generating an antibody specific to the decoy epitope on the capsid protein of porcine circovirus type 2 (PCV2). Using this antibody, we found that two commercial vaccines contained statistically different amounts of the decoy epitope. The vaccine with lower levels of decoy epitope induced a significantly higher level of neutralizing antibody after immunization. This antibody can be used as an analytical tool to monitor the quality of a vaccine from batch to batch.


Asunto(s)
Vacunas contra el Adenovirus/administración & dosificación , Anticuerpos Neutralizantes/inmunología , Infecciones por Circoviridae/inmunología , Infecciones por Circoviridae/prevención & control , Circovirus/inmunología , Vacunas Virales/inmunología , Vacunas Virales/toxicidad , Animales , Anticuerpos Neutralizantes/sangre , Circovirus/efectos de los fármacos , Epítopos/inmunología , Cobayas , Resultado del Tratamiento , Vacunación/métodos
15.
PLoS Biol ; 13(5): e1002152, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25992628

RESUMEN

Epidermal growth factor receptor variant III (EGFRvIII) has been associated with glioma stemness, but the direct molecular mechanism linking the two is largely unknown. Here, we show that EGFRvIII induces the expression and secretion of pigment epithelium-derived factor (PEDF) via activation of signal transducer and activator of transcription 3 (STAT3), thereby promoting self-renewal and tumor progression of glioma stem cells (GSCs). Mechanistically, PEDF sustained GSC self-renewal by Notch1 cleavage, and the generated intracellular domain of Notch1 (NICD) induced the expression of Sox2 through interaction with its promoter region. Furthermore, a subpopulation with high levels of PEDF was capable of infiltration along corpus callosum. Inhibition of PEDF diminished GSC self-renewal and increased survival of orthotopic tumor-bearing mice. Together, these data indicate the novel role of PEDF as a key regulator of GSC and suggest clinical implications.


Asunto(s)
Receptores ErbB/metabolismo , Proteínas del Ojo/metabolismo , Glioma/etiología , Células Madre Neoplásicas/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Serpinas/metabolismo , Animales , Comunicación Autocrina , Progresión de la Enfermedad , Femenino , Glioma/metabolismo , Glioma/mortalidad , Células HEK293 , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Receptores Notch/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción STAT3/metabolismo
16.
Biochem Biophys Res Commun ; 493(1): 325-331, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28888985

RESUMEN

The proto-oncogene tyrosine kinase ROS1 plays a key role in carcinogenesis through gene rearrangement to form a fusion protein with other genes, in which the C-terminal intracellular region of ROS1 participates. The possibility of wild type ROS1 overexpression through epigenetic regulation has been proposed. Here, we generated an antibody, 3B20, reactive to the N-terminal region of ROS1 to use it for the detection of wild type ROS1 in cancerous tissues. Using immunoblot and immunoprecipitation analyses, we found that 3B20 also reacted with heat shock proteins (Hsp)70s. Using homology searching, ROS1 and Hsp70s were found to share an identical amino acid sequence: DLGT. Using alanine mutagenesis of ROS1, the epitope was found to harbor this sequence. To modify the idiotope with the aim of selecting more specific antibodies, we introduced random mutations into the heavy chain complementarity-determining region 3 and successfully generated an antibody clone, 3B20-G1K, with a point mutation that only reacted with ROS1 in enzyme-linked immunosorbent assays, and in immunoblot and immunoprecipitation analysis. In immunohistochemical analysis using 3B20-G1K, ROS1 was found to be absent in normal lung tissues and was overexpressed in a case of lung adenocarcinoma.


Asunto(s)
Adenocarcinoma/inmunología , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Antineoplásicos/inmunología , Proteínas de Arabidopsis/genética , Neoplasias Pulmonares/inmunología , Proteínas Nucleares/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos/administración & dosificación , Proteínas de Arabidopsis/inmunología , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/inmunología , Diseño de Fármacos , Sinergismo Farmacológico , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mutagénesis Sitio-Dirigida/métodos , Proteínas Nucleares/inmunología , Mutación Puntual/genética , Proto-Oncogenes Mas , Proto-Oncogenes , Distribución Tisular , Células Tumorales Cultivadas
17.
Biotechnol Appl Biochem ; 64(3): 327-336, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-26790760

RESUMEN

A truncated precursor form of prostate-specific antigen (PSA), [-2]proPSA, is a well-known biomarker for prostate cancer. To develop a biomarker assay, highly purified [-2]proPSA is required as a standard reference and for generation of a specific antibody. In this study, we generated an efficient mammalian expression system for producing a recombinant [-2]proPSA-human kappa constant domain (Cκ ) fusion protein. N-terminal amino acid sequencing using Edman degradation demonstrated that over 95% of the recombinant protein produced is [-2]proPSA, thereby showing for the first time that recombinant [-2]proPSA can be produced as a major fraction. We also generated a recombinant chicken antibody specific to [-2]proPSA but not cross-reactive to recombinant [-7]proPSA-Cκ , [-5]proPSA-Cκ , and PSA purified from human seminal fluid in enzyme-linked immunosorbent assay (ELISA) and immunoblot analysis. Also, the recombinant chicken antibody reacted to recombinant [-2]proPSA protein bound to an anti-PSA antibody coated on the micrometer plate in a sandwich ELISA. All of these results suggest that the N-terminus of the [-2]proPSA-Cκ fusion protein resides on the exterior of the protein, thus allowing exposure to the antibody.


Asunto(s)
Anticuerpos Monoclonales/química , Proteínas Aviares/química , Cadenas kappa de Inmunoglobulina/biosíntesis , Calicreínas/biosíntesis , Antígeno Prostático Específico/biosíntesis , Señales de Clasificación de Proteína , Proteínas Recombinantes de Fusión/biosíntesis , Animales , Anticuerpos Monoclonales/inmunología , Proteínas Aviares/inmunología , Pollos , Células HEK293 , Humanos , Cadenas kappa de Inmunoglobulina/química , Cadenas kappa de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/inmunología , Calicreínas/análisis , Calicreínas/genética , Calicreínas/inmunología , Antígeno Prostático Específico/análisis , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/inmunología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
18.
Proc Natl Acad Sci U S A ; 111(17): 6269-74, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24733900

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is a multifunctional protein that participates in signaling pathways initiated by various growth factors and cytokines. It exists in multiple forms including those phosphorylated on Tyr(705) (pYSTAT3) or Ser(727) (pSSTAT3) as well as the unphosphorylated protein (USTAT3). In addition to the canonical transcriptional regulatory role of pYSTAT3, both USTAT3 and pSSTAT3 function as transcriptional regulators by binding to distinct promoter sites and play signaling roles in the cytosol or mitochondria. The roles of each STAT3 species in different biological processes have not been readily amenable to investigation, however. We have now prepared an intrabody that binds specifically and with high affinity to the tyrosine-phosphorylated site of pYSTAT3. Adenovirus-mediated expression of the intrabody in HepG2 cells as well as mouse liver blocked both the accumulation of pYSTAT3 in the nucleus and the production of acute phase response proteins induced by interleukin-6. Intrabody expression did not affect the overall accumulation of pSSTAT3 induced by interleukin-6 or phorbol 12-myristate 13-acetate (PMA), the PMA-induced expression of the c-Fos gene, or the PMA-induced accumulation of pSSTAT3 specifically in mitochondria. In addition, it had no effect on interleukin-6-induced expression of the gene for IFN regulatory factor 1, a downstream target of STAT1. Our results suggest that the engineered intrabody is able to block specifically the downstream effects of pYSTAT3 without influencing those of pSSTAT3, demonstrating the potential of intrabodies as tools to dissect the cellular functions of specific modified forms of proteins that exist as multiple species.


Asunto(s)
Anticuerpos Fosfo-Específicos/farmacología , Fosfotirosina/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Especificidad de Anticuerpos/efectos de los fármacos , Especificidad de Anticuerpos/inmunología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interleucina-6/farmacología , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor de Transcripción STAT3/metabolismo , Anticuerpos de Cadena Única/metabolismo , Acetato de Tetradecanoilforbol/farmacología
19.
Int J Mol Sci ; 18(3)2017 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-28272300

RESUMEN

Vascular cell adhesion molecule-1 (VCAM-1) is closely associated with tumor progression and metastasis. However, the relevance and role of VCAM-1 in lung cancer have not been clearly elucidated. In this study, we found that VCAM-1 was highly overexpressed in lung cancer tissue compared with that of normal lung tissue, and high VCAM-1 expression correlated with poor survival in lung cancer patients. VCAM-1 knockdown reduced migration of A549 human lung cancer cells into Matrigel, and competitive blocking experiments targeting the Ig-like domain 6 of VCAM-1 (VCAM-1-D6) demonstrated that the VCAM-1-D6 domain was critical for VCAM-1 mediated A549 cell migration into Matrigel. Next, we developed a human monoclonal antibody specific to human and mouse VCAM-1-D6 (VCAM-1-D6 huMab), which was isolated from a human synthetic antibody library using phage display technology. Finally, we showed that VCAM-1-D6 huMab had a nanomolar affinity for VCAM-1-D6 and that it potently suppressed the migration of A549 and NCI-H1299 lung cancer cell lines into Matrigel. Taken together, these results suggest that VCAM-1-D6 is a key domain for regulating VCAM-1-mediated lung cancer invasion and that our newly developed VCAM-1-D6 huMab will be a useful tool for inhibiting VCAM-1-expressing lung cancer cell invasion.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Movimiento Celular/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , Molécula 1 de Adhesión Celular Vascular/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Sitios de Unión , Línea Celular Tumoral , Humanos , Ratones , Molécula 1 de Adhesión Celular Vascular/química
20.
Biomacromolecules ; 17(9): 3085-93, 2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27517529

RESUMEN

The liver is the most frequent site of metastasis with a 5-year survival rate of only 20-40%. In this work, hyaluronate (HA)-death receptor 5 antibody (DR5 Ab) conjugate was synthesized as a dual targeting therapeutic agent to treat liver metastasis. Dual targeting was achieved by DR5 Ab, a humanized agonistic monoclonal antibody binding to DR5 frequently overexpressed in many kinds of cancer cells, and by HA, a natural polysaccharide binding to HA receptors highly expressed in both the liver and cancer cells. Thiol end-modified HA was site-specifically conjugated to N-glycan on Fc region of oxidized DR5 Ab using a heterobifunctional linker of 3-(2-pyridyldithio)propionyl hydrazide (PDPH). The successful synthesis of HA-DR5 Ab conjugate was confirmed by (1)H NMR, purpald assay, dynamic light scattering (DLS), and high-performance liquid chromatography (HPLC). In vitro analysis of HA-DR5 Ab conjugate revealed that the conjugation of HA to DR5 Ab did not affect the binding affinity and anticancer efficacy of DR5 Ab. Remarkably, according to in vivo bioimaging study, HA-DR5 Ab conjugate appeared to be highly accumulated in the liver and dramatically effective in inhibiting the tumor growth in liver metastasis model mice.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Ácido Hialurónico/química , Inmunoconjugados/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/química , Animales , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Humanos , Neoplasias Hepáticas/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA