Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nanotechnology ; 33(24)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35255487

ABSTRACT

It is still highly desired to develop efficient, resource-abundant and inexpensive electrocatalysts to improve the sluggish kinetics of oxygen evolution reaction (OER) in electrochemical water splitting systems. In this work, the large-area ultrathin (2.52 nm thick) Ce-doped La2O3nanofilms were developed via a facile and reliable ionic layer epitaxy method with different Ce content. The ultrathin Ce-doped La2O3nanofilm with optimum composition of La1.22Ce0.78O3exhibited an excellent OER performance with a very low overpotential of 221 mV at 10 mA cm-2and a small Tafel slope of 33.7 mV dec-1. A remarkable high mass activity of 6263.2 A g-1was also obtained from ultrathin La1.22Ce0.78O3nanofilm at the overpotential of 221 mV. Such a high mass activity was three orders of magnitude higher than state-of-the-art commercial IrO2powders (3.8 A g-1) and more than 30 times higher than La2O3nanofilm (196.7 A g-1) without Ce doping at the same overpotential. This high mass activity was even significantly higher than other recently reported typical OER catalysts. The substantial OER performance gain by the Ce doping was attributed to the improved conductivity and electrochemical active surface areas of nanofilms as a result of favorable tuning on the charge transfer and electronic structures. This work provides a promising approach to develop high-performance two-dimensional (2D) electrocatalysts by effective heteroatom doping strategy.

2.
Pharmaceutics ; 10(3)2018 Aug 02.
Article in English | MEDLINE | ID: mdl-30072675

ABSTRACT

Enhancing the dissolution of insoluble active ingredients comprises one of the most important issues in the pharmaceutical and biomaterial fields. Here, a third generation solid dispersion (3rd SD) of ferulic acid was designed and fabricated by a modified coaxial electrospinning process. A traditional second generation SD (2nd SD) was also prepared by common one-fluid blending electrospinning and was used as a control. With poly(vinyl alcohol) as the fiber matrix and polyvinylpyrrolidone K10 as an additive in the 3rd SDs, the two electrospinning processes were investigated. The prepared 2nd and 3rd SDs were subjected to a series of characterizations, including X-ray diffraction (XRD), scanning electron microscope (SEM), hydrophilicity and in vitro drug dissolving experiments. The results demonstrate that both SDs were monolithic nanocomposites and that the drugs were amorphously distributed within the matrix. However, the 3rd SDs had better morphology with smaller size, narrower size distribution, and smaller water contact angles than the 2nd SDs. Dissolution tests verified that the 3rd SDs could release their loaded cargoes within 60 s, which was over three times faster than the 2nd SDs. Therefore, a combined strategy based on the modified coaxial electrospinning and the logical selections of drug carriers is demonstrated for creating advanced biomaterials.

3.
J Immunother Cancer ; 3: 37, 2015.
Article in English | MEDLINE | ID: mdl-26500773

ABSTRACT

BACKGROUND: Clinical targeting of TNFR family of receptors (CD40, CD134 and CD137) with immunostimulatory monoclonal antibodies has been successful in cancer immunotherapy. However, targeting of CD27 with a mAb is a relatively new approach to provide costimulation of immune cells undergoing activation. Thus, activation of human CD27 (TNFRSF7) with a monoclonal antibody (varlilumab) has previously been demonstrated to result in T cell activation and anti-tumor activity in preclinical models, and is currently in early phase clinical trials in patients with advanced malignancies. In this study we used an in vitro system using human peripheral blood T cells to characterize the varlilumab-mediated costimulatory effects in combination with TCR stimulation in terms of phenotypic, transcriptional and functionality changes. METHODS: T cells were isolated from normal volunteer PBMCs using magnetic bead isolation kits and stimulated in vitro with plate bound anti-CD3 Ab (OKT3) and varlilumab or control Ab for 72 h. Activation profiles were monitored by ELISA or Luminex-based testing cytokine/chemokine releases, cell surface phenotyping for costimulatory and coinhibitory markers and CFSE dye dilution by proliferating T cells and Tregs. Changes in gene expression and transcriptome analysis of varlilumab-stimulated T cells was carried on Agilent Human whole genome microarray datasets using a suite of statistical and bioinformatic software tools. RESULTS: Costimulation of T cells with varlilumab required continuous TCR signaling as pre-activated T cells were unable to produce cytokines with CD27 signaling alone. Analysis of T cell subsets further revealed that memory CD4+ and CD8+ T cells were specifically activated with a bias toward CD8+ T lymphocyte proliferation. Activation was accompanied by upregulated cell surface expression of costimulatory [4-1BB, OX40, GITR and ICOS] and coinhibitory [PD-1] molecules. Importantly, varlilumab costimulation did not activate purified Tregs as measured by cytokine production, proliferation and suppression of dividing non-Treg T cells. Analysis of changes in gene expression during varlilumab stimulation of T cells revealed modulation of pro-inflammatory signatures consistent with cellular activation and proliferation, with the IL-2 pathway showing the highest frequency of gene modulation. CONCLUSIONS: Altogether, the data reveal the requirements and T cell subtype-specific effects of CD27 costimulation, and helps select relevant biomarkers for studying the effects of varlilumab in patients.

4.
Sci Transl Med ; 6(232): 232ra51, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24739759

ABSTRACT

Immune-based therapies for cancer are generating substantial interest because of the success of immune checkpoint inhibitors. This study aimed to enhance anticancer immunity by exploiting the capacity of dendritic cells (DCs) to initiate T cell immunity by efficient uptake and presentation of endocytosed material. Delivery of tumor-associated antigens to DCs using receptor-specific monoclonal antibodies (mAbs) in the presence of DC-activating agents elicits robust antigen-specific immune responses in preclinical models. DEC-205 (CD205), a molecule expressed on DCs, has been extensively studied for its role in antigen processing and presentation. CDX-1401 is a vaccine composed of a human mAb specific for DEC-205 fused to the full-length tumor antigen NY-ESO-1. This phase 1 trial assessed the safety, immunogenicity, and clinical activity of escalating doses of CDX-1401 with the Toll-like receptor (TLR) agonists resiquimod (TLR7/8) and Hiltonol (poly-ICLC, TLR3) in 45 patients with advanced malignancies refractory to available therapies. Treatment induced humoral and cellular immunity to NY-ESO-1 in patients with confirmed NY-ESO-1-expressing tumors across various dose levels and adjuvant combinations. No dose-limiting or grade 3 toxicities were reported. Thirteen patients experienced stabilization of disease, with a median duration of 6.7 months (range, 2.4+ to 13.4 months). Two patients had tumor regression (~20% shrinkage in target lesions). Six of eight patients who received immune-checkpoint inhibitors within 3 months after CDX-1401 administration had objective tumor regression. This first-in-human study of a protein vaccine targeting DCs demonstrates its feasibility, safety, and biological activity and provides rationale for combination immunotherapy strategies including immune checkpoint blockade.


Subject(s)
Antigens, CD/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Epitopes/immunology , Immunity, Humoral/immunology , Lectins, C-Type/immunology , Membrane Proteins/immunology , Receptors, Cell Surface/immunology , Adult , Aged , Aged, 80 and over , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Cancer Vaccines/pharmacokinetics , Cytokines/metabolism , Dose-Response Relationship, Immunologic , Female , Humans , Immunity, Cellular/immunology , Immunoglobulin G/immunology , Interferon-gamma/biosynthesis , Lymphocyte Subsets/metabolism , Male , Middle Aged , Minor Histocompatibility Antigens , T-Lymphocytes/immunology , Vaccination
5.
Clin Cancer Res ; 17(14): 4844-53, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21632857

ABSTRACT

PURPOSE: The use of tumor-derived proteins as cancer vaccines is complicated by tolerance to these self-antigens. Tolerance may be broken by immunization with activated, autologous, ex vivo generated and antigen-loaded, antigen-presenting cells (APC); however, targeting tumor antigen directly to APC in vivo would be a less complicated strategy. We wished to test whether targeted delivery of an otherwise poorly immunogenic, soluble antigen to APC through their mannose receptors (MR) would induce clinically relevant immunity. EXPERIMENTAL DESIGN: Two phase I studies were conducted with CDX-1307, a vaccine composed of human chorionic gonadotropin beta-chain (hCG-ß) fused to an MR-specific monoclonal antibody, administered either locally (intradermally) or systemically (intravenously) in patients with advanced epithelial malignancies. An initial dose escalation of single-agent CDX-1307 was followed by additional cohorts of CDX-1307 combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) and the Toll-like receptor (TLR) 3 agonist polyinosinic-polycytidylic acid (poly-ICLC) and TLR7/8 agonist resiquimod to activate the APC. RESULTS: CDX-1307 induced consistent humoral and T-cell responses to hCG-ß when coadministered with TLR agonists. Greater immune responses and clinical benefit, including the longest duration of stable disease, were observed with immunization combined with local TLR agonists. Immune responses were induced equally efficiently in patients with elevated and nonelevated levels of serum hCG-ß. Antibodies within the serum of vaccinated participants had tumor suppressive function in vitro. Toxicity consisted chiefly of mild injection site reactions. CONCLUSIONS: APC targeting and activation induce adaptive immunity against poorly immunogenic self-antigens which has implications for enhancing the efficacy of cancer immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , Autoantigens/immunology , Cancer Vaccines/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Toll-Like Receptors/agonists , Antigen-Presenting Cells/metabolism , Autoantigens/metabolism , Cancer Vaccines/pharmacokinetics , Cancer Vaccines/toxicity , Cell Line, Tumor , Chorionic Gonadotropin, beta Subunit, Human/blood , Chorionic Gonadotropin, beta Subunit, Human/immunology , Female , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Male , Neoplasm Staging , Neoplasms/pathology , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/toxicity , Skin/immunology , Skin/metabolism , Skin/pathology , Toll-Like Receptors/metabolism , Treatment Outcome
6.
PLoS One ; 6(5): e20068, 2011.
Article in English | MEDLINE | ID: mdl-21637763

ABSTRACT

Treatment of invasive adenovirus (Ad) disease in hematopoietic stem cell transplant (SCT) recipients with capsid protein hexon-specific donor T cells is under investigation. We propose that cytotoxic T cells (CTLs) targeted to the late protein hexon may be inefficient in vivo because the early Ad protein E3-19K downregulates HLA class I antigens in infected cells. In this study, CD8+ T cells targeted to highly conserved HLA A2-restricted epitopes from the early regulatory protein DNA polymerase (P-977) and late protein hexon (H-892) were compared in peripheral blood (PB) and tonsils of naturally infected adults. In tonsils, epitope-specific pentamers detected a significantly higher frequency of P-977+CD8+ T cells compared to H-892+CD8+ T cells; this trend was reversed in PB. Tonsil epitope-specific CD8+ T cells expressed IFN-γ and IL-2 but not perforin or TNF-α, whereas PB T cells were positive for IFN-γ, TNF-α, and perforin. Tonsil epitope-specific T cells expressed lymphoid homing marker CCR7 and exhibited lower levels of the activation marker CD25 but higher proliferative potential than PB T cells. Finally, in parallel with the kinetics of mRNA expression, P-977-specific CTLs lysed targets as early as 8 hrs post infection. In contrast, H-892-specific CTLs did not kill unless infected fibroblasts were pretreated with IFN-γ to up regulate HLA class I antigens, and cytotoxicity was delayed until 16-24 hours. These data show that, in contrast to hexon CTLs, central memory type DNA polymerase CTLs dominate the lymphoid compartment and kill fibroblasts earlier after infection without requiring exogenous IFN-γ. Thus, use of CTLs targeted to both early and late Ad proteins may improve the efficacy of immunotherapy for life-threatening Ad disease in SCT recipients.


Subject(s)
Adenovirus Early Proteins/blood , Adenovirus Early Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/blood , Capsid Proteins/immunology , Immunologic Memory/immunology , Palatine Tonsil/metabolism , Adult , Biomarkers , DNA-Directed DNA Polymerase/blood , DNA-Directed DNA Polymerase/immunology , Epitopes/immunology , Humans , Kinetics , Phenotype , T-Lymphocytes, Cytotoxic/metabolism
7.
Virology ; 352(2): 338-44, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16780916

ABSTRACT

In adenovirus-12 tumorigenic cells, the viral E1A-12 protein mediates transcriptional down-regulation of the major histocompatibility complex (MHC) class I genes by targeting the class I enhancer. Here, we demonstrate by a combination of antisense and chromatin immunoprecipitation (ChIP) analysis that E1A-12 is a physical component of the class I enhancer repression complex, known to comprise COUP-TFII and histone deacetylase 1 (HDAC1). Significantly, E1A antisense was shown to co-eliminate E1A-12 as well as HDAC1 and HDAC8, but not HDAC3, from the enhancer repression complex. Consistent with elimination of HDAC1 and HDAC8, E1A antisense also resulted in a dramatic increase in histone acetylation, a hallmark of transcriptionally active chromatin. Importantly, MHC class I antigen expression was restored on the surface of E1A antisense-transfected cells. These results demonstrate that E1A-12 is associated with the MHC class I complex and apparently mediates class I transcriptional down-regulation by enacting chromatin repression through HDAC1 and HDAC8.


Subject(s)
Adenoviridae/genetics , Adenoviridae/pathogenicity , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/physiology , Genes, MHC Class I , Histone Deacetylases/genetics , Adenoviridae/immunology , Adenoviridae/physiology , Adenovirus E1A Proteins/immunology , Animals , Base Sequence , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Histocompatibility Antigens Class I/metabolism , Mice , Oligodeoxyribonucleotides, Antisense/genetics
8.
Virology ; 306(1): 68-76, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12620799

ABSTRACT

In adenovirus type 12 transformed cells, the down-regulation of MHC class I transcription contributes to the tumorigenic phenotype and is solely mediated by Ad12 E1A. Previous in vitro studies with class I enhancer sequences have indicated that there is an increased binding of repressor COUP-TFII and its associated HDAC and a decreased binding of activator NF-kappaB. In this study, we used chromatin immunoprecipitation (ChIP) assay in order to determine in vivo whether these proteins regulate class I transcription by affecting chromatin. The ChIP assay revealed that there is lack of chromatin histone acetylation in the region of the class I enhancer in Ad12-transformed cells. This is regulated by histone deacetylation as it was further demonstrated in vivo that COUP-TFII and HDAC are associated with the class I enhancer chromatin. In agreement with in vitro studies, NF-kappaB could be recruited to the class I enhancer following induction by TNF-alpha. However, this enhancer-bound NF-kappaB failed to up-regulate class I expression because the class I enhancer chromatin remained repressed as a result of histone deacetylation by HDAC in association with COUP-TFII. Thus, we have demonstrated for the first time that repression of chromatin through histone deacetylation is a major mechanism in down-regulating class I transcription in Ad12-transformed cells. Finally, Ad12 E1A, a non-DNA binding protein, was shown to be present in the natural protein complex bound to the class I enhancer.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Histocompatibility Antigens Class I/metabolism , Histone Deacetylases/metabolism , NF-kappa B/metabolism , Receptors, Steroid , Transcription Factors/metabolism , Transcription, Genetic , Adenoviridae , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Base Sequence , COUP Transcription Factor II , COUP Transcription Factors , Cell Transformation, Viral , Enhancer Elements, Genetic/genetics , Histocompatibility Antigens Class I/genetics , Mice , Molecular Sequence Data , Rats , Transcriptional Activation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...