Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Immunol Cell Biol ; 96(3): 284-297, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29356071

RESUMEN

Inflammation plays pivotal roles in different stages of tumor development. Screening for predisposing genetic abnormalities and understanding the roles these genes play in the crosstalk between immune and cancer cells will provide new targets for cancer therapy and prevention. The interferon inducible transmembrane (IFITM) genes are involved in pathogenesis of the gastro-intestinal tract. We aimed at delineating the role of IFITM3 in colonic epithelial homeostasis, inflammation and colitis-associated tumorigenesis using IFITM3-deficient mice. Chemical induction of colitis in IFITM3-deficient mice results in significantly increased clinical signs of inflammation and induction of invasive tumorigenesis. Bone marrow transplantation showed that cells of the hematopoietic system are responsible for colitis deterioration. In these mice, impaired cytokine expression skewed inflammatory response toward pathogenic Th17 with reduced expression of the anti-inflammatory cytokine IL10 during the recovery phase. Intriguingly, mice lacking the entire IFITM locus developed spontaneous chronic colitis from the age of 14 weeks. Sequencing the 16S rRNA of naïve mice lacking IFITM3 gene, or the entire locus containing five IFITM genes, revealed these mice had significant bacterial differences from their wild-type littermates. Our novel results provide strong evidence for the essential role of IFITM genes in ameliorating colitis and colitis-associated tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Colitis/inmunología , Colitis/microbiología , Inmunidad , Inflamación/genética , Proteínas de la Membrana/genética , Microbiota , Animales , Colitis/genética , Colitis/patología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Hematopoyesis , Inmunidad/genética , Proteínas de la Membrana/deficiencia , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/genética , Células Mieloides/patología
2.
J Immunol ; 193(11): 5733-43, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25362181

RESUMEN

Adoptive transfer of Ag-specific T lymphocytes is an attractive form of immunotherapy for cancers. However, acquiring sufficient numbers of host-derived tumor-specific T lymphocytes by selection and expansion is challenging, as these cells may be rare or anergic. Using engineered T cells can overcome this difficulty. Such engineered cells can be generated using a chimeric Ag receptor based on common formats composed from Ag-recognition elements such as αß-TCR genes with the desired specificity, or Ab variable domain fragments fused with T cell-signaling moieties. Combining these recognition elements are Abs that recognize peptide-MHC. Such TCR-like Abs mimic the fine specificity of TCRs and exhibit both the binding properties and kinetics of high-affinity Abs. In this study, we compared the functional properties of engineered T cells expressing a native low affinity αß-TCR chains or high affinity TCR-like Ab-based CAR targeting the same specificity. We isolated high-affinity TCR-like Abs recognizing HLA-A2-WT1Db126 complexes and constructed CAR that was transduced into T cells. Comparative analysis revealed major differences in function and specificity of such CAR-T cells or native TCR toward the same antigenic complex. Whereas the native low-affinity αß-TCR maintained potent cytotoxic activity and specificity, the high-affinity TCR-like Ab CAR exhibited reduced activity and loss of specificity. These results suggest an upper affinity threshold for TCR-based recognition to mediate effective functional outcomes of engineered T cells. The rational design of TCRs and TCR-based constructs may need to be optimized up to a given affinity threshold to achieve optimal T cell function.


Asunto(s)
Anticuerpos/inmunología , Vacunas contra el Cáncer , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/fisiología , Afinidad de Anticuerpos , Citotoxicidad Inmunológica , Ingeniería Genética , Antígeno HLA-A2/metabolismo , Humanos , Células Jurkat , Neoplasias/inmunología , Unión Proteica , Transducción de Señal , Especificidad del Receptor de Antígeno de Linfocitos T
3.
Mol Ther ; 22(5): 1018-28, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24686242

RESUMEN

The adoptive transfer of regulatory T cells (Tregs) offers a promising strategy to combat pathologies that are characterized by aberrant immune activation, including graft rejection and autoinflammatory diseases. Expression of a chimeric antigen receptor (CAR) gene in Tregs redirects them to the site of autoimmune activity, thereby increasing their suppressive efficiency while avoiding systemic immunosuppression. Since carcinoembryonic antigen (CEA) has been shown to be overexpressed in both human colitis and colorectal cancer, we treated CEA-transgenic mice that were induced to develop colitis with CEA-specific CAR Tregs. Two disease models were employed: T-cell-transfer colitis as well as the azoxymethane-dextran sodium sulfate model for colitis-associated colorectal cancer. Systemically administered CEA-specific (but not control) CAR Tregs accumulated in the colons of diseased mice. In both model systems, CEA-specific CAR Tregs suppressed the severity of colitis compared to control Tregs. Moreover, in the azoxymethane-dextran sodium sulfate model, CEA-specific CAR Tregs significantly decreased the subsequent colorectal tumor burden. Our data demonstrate that CEA-specific CAR Tregs exhibit a promising potential in ameliorating ulcerative colitis and in hindering colorectal cancer development. Collectively, this study provides a proof of concept for the therapeutic potential of CAR Tregs in colitis patients as well as in other autoimmune inflammatory disorders.


Asunto(s)
Antígeno Carcinoembrionario/biosíntesis , Colitis/terapia , Neoplasias Colorrectales/terapia , Linfocitos T Reguladores/trasplante , Animales , Colitis/complicaciones , Colitis/genética , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/genética , Sulfato de Dextran/administración & dosificación , Modelos Animales de Enfermedad , Humanos , Inmunoterapia Adoptiva/métodos , Ratones , Linfocitos T Reguladores/metabolismo
4.
Cancer Immunol Res ; 12(6): 687-703, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38592331

RESUMEN

Recombinant cytokines have limited anticancer efficacy mostly due to a narrow therapeutic window and systemic adverse effects. IL18 is an inflammasome-induced proinflammatory cytokine, which enhances T- and NK-cell activity and stimulates IFNγ production. The activity of IL18 is naturally blocked by a high-affinity endogenous binding protein (IL18BP). IL18BP is induced in the tumor microenvironment (TME) in response to IFNγ upregulation in a negative feedback mechanism. In this study, we found that IL18 is upregulated in the TME compared with the periphery across multiple human tumors and most of it is bound to IL18BP. Bound IL18 levels were largely above the amount required for T-cell activation in vitro, implying that releasing IL18 in the TME could lead to potent T-cell activation. To restore the activity of endogenous IL18, we generated COM503, a high-affinity anti-IL18BP that blocks the IL18BP:IL18 interaction and displaces precomplexed IL18, thereby enhancing T- and NK-cell activation. In vivo, administration of a surrogate anti-IL18BP, either alone or in combination with anti-PD-L1, resulted in significant tumor growth inhibition and increased survival across multiple mouse tumor models. Moreover, the anti-IL18BP induced pronounced TME-localized immune modulation including an increase in polyfunctional nonexhausted T- and NK-cell numbers and activation. In contrast, no increase in inflammatory cytokines and lymphocyte numbers or activation state was observed in serum and spleen. Taken together, blocking IL18BP using an Ab is a promising approach to harness cytokine biology for the treatment of cancer.


Asunto(s)
Interleucina-18 , Microambiente Tumoral , Animales , Humanos , Interleucina-18/metabolismo , Ratones , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Línea Celular Tumoral , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de los fármacos , Femenino , Ratones Endogámicos C57BL , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
5.
Nano Lett ; 11(11): 4997-5001, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-21985491

RESUMEN

Seamless embedment of electronic devices in biological systems is expected to add the outstanding computing power, memory, and speed of electronics to the biochemical toolbox of nature. Such amalgamation requires transduction of electronic signals into biochemical cues that affect cells. Inspired by biology, where pathways are directed by molecular recognition, we propose and demonstrate a generic electrical-to-biological transducer comprising a two-state electronic antigen and a chimeric cell receptor engineered to bind the antigen exclusively in its "on" state. T-cells expressing these receptors remain inactivated with the antigen in its "off" state. Switching the antigen to its "on" state by an electrical signal leads to its recognition by the T-cells and correspondingly to cell activation.


Asunto(s)
Receptores de Antígenos de Linfocitos T/efectos de la radiación , Anticuerpos de Cadena Única/efectos de la radiación , Linfocitos T/efectos de la radiación , Células Cultivadas , Campos Electromagnéticos , Humanos , Receptores de Antígenos de Linfocitos T/química , Anticuerpos de Cadena Única/química , Linfocitos T/química
6.
Bioorg Med Chem ; 19(2): 798-806, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21194956

RESUMEN

Cystine disulfide bond is a common feature in numerous biologically active peptides and proteins and accordingly its replacement by various surrogates presents a potential route to obtain analogs with improved pharmacokinetic characteristics. The purpose of the present study was to assess whether an azo-bridge can serve as such a surrogate. In view of the marked clinical significance of somatostatin and the brain natriuretic peptide (BNP) we choose these peptides as a model. Three cyclic-azo somatostatin analogs and three cyclic-azo BNP analogs were effectively prepared in solution through azo bond formation between p-amino phenylalanine and His or Tyr residues that were positioned in the peptide sequences in place of the native Cys residues. The peptides binding affinities to the sst2 and ANP-receptor (NPR-A) expressed on rat acinar pancreating carcinoma AR4-2J cell membranes and HeLa cells, respectively, were examined. The somatostatin analogs displayed good to moderate affinities to the rat sst2 in the nM range with best results obtained with peptide 2, that is, IC50 = 8.1 nM. Molecular dynamics simulations on these peptides suggests on a correlation between the observed binding potencies and the degree of conformational space overlapping with that of somatostatin. The BNP analogs exhibited binding affinities to the NPR-A in the nM range with best results obtained with BNP-1, that is, IC50 = 60 nM.


Asunto(s)
Compuestos Azo/química , Cistina/química , Disulfuros/química , Péptido Natriurético Encefálico/análogos & derivados , Somatostatina/análogos & derivados , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Humanos , Concentración de Iones de Hidrógeno , Simulación de Dinámica Molecular , Péptido Natriurético Encefálico/síntesis química , Péptido Natriurético Encefálico/metabolismo , Unión Proteica , Ratas , Somatostatina/síntesis química , Somatostatina/metabolismo
7.
J Med Chem ; 51(1): 126-34, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-18078312

RESUMEN

Affecting an estimated 5% of adults over 65 years of age, Parkinson's disease and Alzheimer's disease are the most common neurodegenerative disorders. Accumulating evidence suggests that oxidative stress induced by the breakdown of iron homeostasis is a major contributor to the neuronal loss observed in neurodegeneration. Thus, brain-permeable iron chelators may present potential therapeutic benefits. In the present study, iron-chelating hydroxamate groups were introduced into the NAP (NAPVSIPQ) peptide, whose neuroprotective qualities have been widely demonstrated. Our experiments revealed that the novel dihydroxamate peptide 3 is capable of inhibiting iron-catalyzed hydroxyl radical formation and lipid peroxidation, abilities that are not part of the repertoire of its parent peptide. In addition, peptide 3 was superior to native NAP in protecting human neuroblastoma cell cultures against the toxicity of hydrogen peroxide. These results suggest that NAP-based iron chelators deserve further investigation in the search for drug candidates for neurodegeneration.


Asunto(s)
Antioxidantes/síntesis química , Ácidos Hidroxámicos/síntesis química , Quelantes del Hierro/síntesis química , Fármacos Neuroprotectores/síntesis química , Oligopéptidos/síntesis química , Animales , Antioxidantes/química , Antioxidantes/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Compuestos Férricos/química , Compuestos Ferrosos/química , Humanos , Peróxido de Hidrógeno/farmacología , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Radical Hidroxilo/química , Técnicas In Vitro , Quelantes del Hierro/química , Quelantes del Hierro/farmacología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Oligopéptidos/química , Oligopéptidos/farmacología , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley
8.
PLoS One ; 13(10): e0205491, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30321203

RESUMEN

Robust preclinical testing is essential to predict clinical safety and efficacy and provide data to determine safe dose for first-in-man studies. There are a growing number of examples where the preclinical development of drugs failed to adequately predict clinical adverse events in part due to their assessment with inappropriate preclinical models. Preclinical investigations of T cell receptor (TCR)-based immunotherapies prove particularly challenging as these biologics are human-specific and thus the conventional testing in animal models is inadequate. As these molecules harness the full force of the immune system, and demonstrate tremendous potency, we set out to design a preclinical package that would ensure adequate evaluation of these therapeutics. Immune Mobilising Monoclonal TCR Against Cancer (ImmTAC) molecules are bi-specific biologics formed of an affinity-enhanced TCR fused to an anti-CD3 effector function. ImmTAC molecules are designed to activate human T lymphocytes and target peptides within the context of a human leukocyte antigen (HLA), thus require an intact human immune system and peptidome for suitable preclinical screening. Here we draw upon the preclinical testing of four ImmTAC molecules, including IMCgp100, the first ImmTAC molecule to reach the clinic, to present our comprehensive, informative and robust approach to in vitro preclinical efficacy and safety screening. This package comprises a broad range of cellular and molecular assays using human tissues and cultured cells to test efficacy, safety and specificity, and hence predict human responses in clinical trials. We propose that this entirely in vitro package offers a potential model to be applied to screening other TCR-based biologics.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Proteínas/farmacología , Anticuerpos de Cadena Única/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Técnicas In Vitro , Flujo de Trabajo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA