Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Immunol ; 203(7): 1897-1908, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31484735

RESUMEN

Sepsis is characterized as life-threatening organ dysfunction caused by a dysregulated host immune response to infection. The purpose of this investigation was to determine the differential effect of sepsis on innate versus adaptive immunity, in humans, by examining RNA expression in specific immune cell subsets, including monocytes/macrophages and CD4 and CD8 T cells. A second aim was to determine immunosuppressive mechanisms operative in sepsis that might be amenable to immunotherapy. Finally, we examined RNA expression in peripheral cells from critically ill nonseptic patients and from cancer patients to compare the unique immune response in these disorders with that occurring in sepsis. Monocytes, CD4 T cells, and CD8 T cells from septic patients, critically ill nonseptic patients, patients with metastatic colon cancer, and healthy controls were analyzed by RNA sequencing. Sepsis induced a marked phenotypic shift toward downregulation of multiple immune response pathways in monocytes suggesting that impaired innate immunity may be fundamental to the immunosuppression that characterizes the disorder. In the sepsis cohort, there was a much more pronounced effect on gene transcription in CD4 T cells than in CD8 T cells. Potential mediators of sepsis-induced immunosuppression included Arg-1, SOCS-1, and SOCS-3, which were highly upregulated in multiple cell types. Multiple negative costimulatory molecules, including TIGIT, Lag-3, PD-1, and CTLA-4, were also highly upregulated in sepsis. Although cancer had much more profound effects on gene transcription in CD8 T cells, common immunosuppressive mechanisms were present in all disorders, suggesting that immunoadjuvant therapies that are effective in one disease may also be efficacious in the others.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Monocitos/inmunología , Neoplasias/inmunología , ARN Neoplásico/inmunología , Sepsis/inmunología , Análisis de Secuencia de ARN , Adulto , Anciano , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Enfermedad Crítica , Femenino , Humanos , Tolerancia Inmunológica , Masculino , Persona de Mediana Edad , Monocitos/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias/genética , Neoplasias/patología , Estudios Prospectivos , ARN Neoplásico/genética , Sepsis/genética , Sepsis/patología
2.
J Infect Dis ; 209(7): 1039-44, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24133182

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) disease is associated with aberrant immune activation, and coinfection with hepatitis C virus (HCV) exacerbates hepatic inflammation and fibrosis. However, the role of HIV-1 infection or host immune modulation in liver pathogenesis is not clearly defined. Here, we report that regulatory T (Treg) cells prevent liver immunopathogenesis during HIV-1 infection in a humanized mouse model. In the absence of Treg cells, HIV-1 infection induced liver fibrosis associated with hepatic stellate cell activation, hepatitis, and liver injury. Our findings provide new insight linking Treg cells and liver immunopathogenesis during HIV-1 infection.


Asunto(s)
Coinfección/complicaciones , Infecciones por VIH/complicaciones , Hepatitis C/complicaciones , Cirrosis Hepática/prevención & control , Linfocitos T Reguladores/inmunología , Animales , Coinfección/inmunología , Modelos Animales de Enfermedad , Infecciones por VIH/inmunología , VIH-1/inmunología , VIH-1/aislamiento & purificación , Hepacivirus/inmunología , Hepacivirus/aislamiento & purificación , Hepatitis C/inmunología , Humanos , Ratones , Ratones SCID
3.
Gastroenterology ; 140(4): 1334-44, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21237170

RESUMEN

BACKGROUND & AIMS: Studies of hepatitis C virus (HCV) infection, immunopathogenesis, and resulting liver diseases have been hampered by the lack of a small animal model. We developed humanized mice with human immune system and liver tissues to improve the studies of hepatitis C virus pathogenesis and treatment. METHODS: To promote engraftment of human hepatocytes, we expressed a fusion protein of the FK506 binding protein (FKBP) and caspase 8 under control of the albumin promoter (AFC8), which induces liver cell death, in Balb/C Rag2(-/-) γC-null mice. Cotransplantation of human CD34(+) human hematopoietic stem cells (HSC) and hepatocyte progenitors into the transgenic mice led to efficient engraftment of human leukocytes and hepatocytes. We then infected these humanized mice (AFC8-hu HSC/Hep) with primary HCV isolates and studied HCV-induced immune responses and liver diseases. RESULTS: AFC8-hu HSC/Hep mice supported HCV infection in the liver and generated a human immune T-cell response against HCV. HCV infection induced liver inflammation, hepatitis, and fibrosis, which correlated with activation of stellate cells and expression of human fibrogenic genes. CONCLUSIONS: AFC8-hu HSC/Hep mice are a useful model of HCV infection, the immune response, and liver disease because they contain human immune system and liver cells. These mice become infected with HCV, generate a specific immune response against the virus, and develop liver diseases that include hepatitis and fibrosis. This model might also be used to develop therapeutics for HCV infection.


Asunto(s)
Modelos Animales de Enfermedad , Hepacivirus/inmunología , Hepatitis C Crónica/inmunología , Hepatocitos/trasplante , Ratones Transgénicos , Animales , Caspasa 8/genética , Caspasa 8/inmunología , Proteínas de Unión al ADN/genética , Femenino , Hepatitis C Crónica/genética , Hepatitis C Crónica/patología , Hepatocitos/inmunología , Hepatocitos/patología , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/inmunología , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos BALB C , Células Madre/inmunología , Células Madre/patología , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/inmunología , Trasplante Heterólogo
4.
Clin Transl Sci ; 15(11): 2625-2639, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36097345

RESUMEN

A phase I trial (NCT03447314; 204686) evaluated the safety and efficacy of GSK1795091, a Toll-like receptor 4 (TLR4) agonist, in combination with immunotherapy (GSK3174998 [anti-OX40 monoclonal antibody], GSK3359609 [anti-ICOS monoclonal antibody], or pembrolizumab) in patients with solid tumors. The primary endpoint was safety; other endpoints included efficacy, pharmacokinetics, and pharmacodynamics (PD). Manufacturing of GSK1795091 formulation was modified during the trial to streamline production and administration, resulting in reduced PD (cytokine) activity. Fifty-four patients received GSK1795091 with a combination partner; 32 received only the modified GSK1795091 formulation, 15 received only the original formulation, and seven switched mid-study from the original to the modified formulation. Despite the modified formulation demonstrating higher systemic GSK1795091 exposure compared with the original formulation, the transient, dose-dependent elevations in cytokine and chemokine concentrations were no longer observed (e.g., IP-10, IL10, IL1-RA). Most patients (51/54; 94%) experienced ≥1 treatment-emergent adverse event (TEAE) during the study. Safety profiles were similar between formulations, but a higher incidence of TEAEs associated with immune responses (chills, fatigue, pyrexia, nausea, and vomiting) were observed with the original formulation. No conclusions can be made regarding GSK1795091 anti-tumor activity due to the limited data collected. Manufacturing changes were hypothesized to have caused the change in biological activity in this study. Structural characterization revealed GSK1795091 aggregate size in the modified formulation to be twice that in the original formulation, suggesting a negative correlation between GSK1795091 aggregate size and PD activity. This may have important clinical implications for future development of structurally similar compounds.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias , Humanos , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Citocinas , Lípido A/uso terapéutico , Neoplasias/tratamiento farmacológico , Receptor Toll-Like 4/agonistas , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos
5.
Blood ; 112(7): 2858-68, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18544681

RESUMEN

The role of FoxP3(+)CD4(+) regulatory T (Treg) cells in HIV-1 disease in vivo is poorly understood due to the lack of a robust model. We report here that CD4(+)FoxP3(+) T cells are developed in all lymphoid organs in humanized Rag2(-/-)gammaC(-/-) (DKO-hu HSC) mice and they display both Treg phenotype and Treg function. These FoxP3(+) Treg cells are preferentially infected and depleted by a pathogenic HIV-1 isolate in HIV-infected DKO-hu HSC mice; and depletion of Treg cells is correlated with induction of their apoptosis in vivo. When CD4(+)CD25(+/hi) Treg cells are depleted with the IL-2-toxin fusion protein (denileukin diftitox), HIV-1 infection is significantly impaired. This is demonstrated by reduced levels of productively infected cells in lymphoid organs and lower plasma viremia. Therefore, FoxP3(+) Treg cells are productively infected and play an important role in acute HIV-1 infection in vivo. The DKO-hu HSC mouse will be a valuable model to study human Treg functions and their role in HIV-1 pathogenesis in vivo.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Factores de Transcripción Forkhead/metabolismo , Infecciones por VIH/inmunología , VIH-1/inmunología , Cadenas gamma de Inmunoglobulina/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/virología , Animales , Apoptosis/efectos de los fármacos , Proteínas de Unión al ADN/inmunología , Toxina Diftérica/farmacología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Humanos , Cadenas gamma de Inmunoglobulina/metabolismo , Interleucina-2/farmacología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Depleción Linfocítica , Ratones , Ratones Noqueados , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Replicación Viral/efectos de los fármacos
6.
Clin Ther ; 42(8): 1519-1534.e33, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32739049

RESUMEN

PURPOSE: Interest in Toll-like receptor (TLR) agonists for cancer treatment has been renewed after promising preliminary clinical data in combination with checkpoint inhibitors. This first-in-human study assessed the safety, tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) properties of intravenous GSK1795091, a synthetic TLR4 agonist, in healthy volunteers as a precursor to evaluation in patients with cancer. METHODS: Healthy participants were randomized (1:3; double-blinded manner) to receive placebo or a single intravenous injection of GSK1795091 at doses of 7-100 ng. The primary objective was to evaluate the safety and tolerability of GSK1795091; secondary and exploratory objectives were to characterize GSK1795091 PK and PD properties. FINDINGS: Forty participants received study treatment (10 received placebo and 30 received GSK1795091). Overall, 3 of the 10 participants (30%) who received placebo and 16 of the 30 (53%) who received GSK1795091 experienced ≥1 adverse event (AE). The most common AEs were influenza-like illness, headache, back pain, and increased body temperature. One participant experienced late-occurring AEs (alanine aminotransferase and aspartate aminotransferase increases), considered possibly related to GSK1795091. No serious AEs were reported. GSK1795091 PK properties were characterized by dose proportional increase in exposure. Transient and dose-dependent changes in induced cytokine and chemokine concentrations and immune cell counts were observed 1-4 h after GSK1795091 administration and returned to baseline within 24 h. IMPLICATIONS: Intravenously administered GSK1795091 was acceptably tolerated in healthy volunteers, had favorable PK properties, and stimulated immune cell changes in a dose-dependent manner, providing evidence of target engagement and downstream pharmacology. These results supported the design and initiation of a repeat-dose study of intravenous GSK1795091 in combination with other immunotherapies in patients with advanced cancer. ClinicalTrials.gov identifier: NCT02798978.


Asunto(s)
Antineoplásicos , Glucolípidos , Receptor Toll-Like 4/agonistas , Adulto , Alanina Transaminasa/sangre , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Área Bajo la Curva , Aspartato Aminotransferasas/sangre , Dolor de Espalda/inducido químicamente , Temperatura Corporal/efectos de los fármacos , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Glucolípidos/administración & dosificación , Glucolípidos/efectos adversos , Glucolípidos/farmacocinética , Cefalea/inducido químicamente , Voluntarios Sanos , Humanos , Inmunoterapia , Infusiones Intravenosas , Recuento de Leucocitos , Masculino , Persona de Mediana Edad
7.
MAbs ; 12(1): 1755069, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32343620

RESUMEN

Complex cellular targets such as G protein-coupled receptors (GPCRs), ion channels, and other multi-transmembrane proteins represent a significant challenge for therapeutic antibody discovery, primarily because of poor stability of the target protein upon extraction from cell membranes. To assess whether a limited set of membrane-bound antigen formats could be exploited to identify functional antibodies directed against such targets, we selected a GPCR of therapeutic relevance (CCR1) and identified target binders using an in vitro yeast-based antibody discovery platform (AdimabTM) to expedite hit identification. Initially, we compared two different biotinylated antigen formats overexpressing human CCR1 in a 'scouting' approach using a subset of the antibody library. Binders were isolated using streptavidin-coated beads, expressed as yeast supernatants, and screened using a high-throughput binding assay and flow cytometry on appropriate cell lines. The most suitable antigen was then selected to isolate target binders using the full library diversity. This approach identified a combined total of 183 mAbs with diverse heavy chain sequences. A subset of clones exhibited high potencies in primary cell chemotaxis assays, with IC50 values in the low nM/high pM range. To assess the feasibility of any further affinity enhancement, full-length hCCR1 protein was purified, complementary-determining region diversified libraries were constructed from a high and lower affinity mAb, and improved binders were isolated by fluorescence-activated cell sorting selections. A significant affinity enhancement was observed for the lower affinity parental mAb, but not the high affinity mAb. These data exemplify a methodology to generate potent human mAbs for challenging targets rapidly using whole cells as antigen and define a route to the identification of affinity-matured variants if required.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Afinidad de Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Receptores CCR1/inmunología , Receptores Acoplados a Proteínas G/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Biblioteca de Péptidos , Unión Proteica/efectos de los fármacos , Receptores CCR1/antagonistas & inhibidores , Receptores CCR1/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
8.
Open Forum Infect Dis ; 6(4): ofz106, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31041337

RESUMEN

Mice were infected with influenza and treated with a CXCR2 antagonist in combination with antiviral or antiviral alone starting 4 days postinfection. Neutrophil recruitment to the lung was reduced, and improvements in health outcomes and lung consolidation were observed in combination-treated mice with no evidence of worsening outcome.

9.
Open Forum Infect Dis ; 6(4): ofz163, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31041358

RESUMEN

BACKGROUND: Excessive neutrophil migration has been correlated with influenza symptom severity. Danirixin (GSK1325756), a selective and reversible antagonist of C-X-C chemokine receptor 2, decreases neutrophil activation and transmigration to areas of inflammation. This study evaluated the efficacy and safety of intravenous (IV) danirixin co-administered with oseltamivir for the treatment of adults hospitalized with influenza. METHODS: In this phase 2b, double-blind, 3-arm study (NCT02927431), influenza-positive participants were randomized 2:2:1 to receive danirixin 15mg intravenously (IV) twice daily (bid) + oral oseltamivir 75mg bid (OSV), danirixin 50mg IV bid + OSV, or placebo IV bid + OSV, for up to 5 days. The primary endpoint was time to clinical response (TTCR). RESULTS: In total, 10 participants received study treatment (danirixin 15mg + OSV, n = 4; danirixin 50mg + OSV, n = 4; placebo + OSV, n = 2) before the study was terminated early due to low enrollment. All participants achieved a clinical response. Median (95% confidence interval) TTCR was 4.53 days (2.95, 5.71) for danirixin 15mg + OSV, 4.76 days (2.71, 5.25) for danirixin 50mg + OSV, and 1.33 days (0.71, 1.95) for placebo + OSV. Adverse events (AEs) were generally of mild or moderate intensity; no serious AEs were considered treatment-related. Interleukin-8 levels increased in nasal samples (using synthetic absorptive matrix strips) and decreased serum neutrophil-elastase-mediated degradation of elastin decreased in danirixin-treated participants, suggesting effective target engagement. CONCLUSIONS: Interpretation of efficacy results is restricted by the low participant numbers. The safety and tolerability profile of danirixin was consistent with previous studies. CLINICAL TRIAL INFORMATION: The registration data for the trial are in the ClinicalTrials.gov database, number NCT02927431, and in the EU Clinical Trials Register (https://www.clinicaltrialsregister.eu/) as GSK study 201023, EudraCT 2016-002512-40. Anonymized individual participant data and study documents can be requested for further research from www.clinicalstudydatarequest.com.

10.
Open Forum Infect Dis ; 6(4): ofz072, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31024969

RESUMEN

BACKGROUND: Danirixin (DNX), a selective and reversible CXC chemokine receptor 2 antagonist, inhibits neutrophil transmigration and activation. This study assessed the safety, tolerability, and clinical effect of DNX with and without oseltamivir (OSV) in adults with acute, uncomplicated influenza. METHODS: This was a placebo-controlled, double-blind, Phase IIa study. Participants (18-64 years) with influenza-like symptoms (onset ≤48 hours) and positive influenza rapid antigen test were randomized 2:1:2:1 to DNX, placebo, DNX+OSV, or OSV (75 mg each, administered twice daily for 5 days) and followed for 28 days. Primary endpoints included frequency of adverse events (AEs) and serious AEs (SAEs). The effect of DNX on virologic response and clinical effect on influenza symptoms were secondary endpoints. RESULTS: A total of 45 participants were enrolled, 35 of whom were confirmed influenza positive by polymerase chain reaction analysis. The highest incidence of AEs was in the placebo group (4 of 7, 57%), followed by the DNX+OSV (7 of 16, 44%), DNX (3 of 15, 20%), and OSV (0 of 7, 0%) groups. One SAE (T-wave abnormality) was reported in the DNX group (unrelated to treatment). No differences in viral load assessments were observed among treatment groups. CONCLUSIONS: Danirixin treatment was well tolerated and did not impede viral clearance.

11.
Nat Protoc ; 7(9): 1608-17, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22899330

RESUMEN

Establishing a small animal model that accurately recapitulates hepatotropic pathogens, including hepatitis C virus (HCV) infection and immunopathogenesis, is essential for the study of hepatitis virus-induced liver disease and for therapeutics development. This protocol describes our recently developed humanized mouse model for studying HCV and other hepatotropic infections, human immune response and hepatitis and liver fibrosis. The first 5-h stage is the isolation of human liver progenitor and hematopoietic stem cells from fetal liver. Next, AFC8 immunodeficient mice are transplanted with the isolated progenitor/stem cells. This generally takes 2 h. The transplanted mice are then treated for a month with the mouse liver apoptosis-inducing AFC8 dimerizer and left for an additional 2-month period to permit human liver and immune cell growth as well as system reconstitution and development before inoculation with HCV clinical isolates. HCV infection, human immune response and liver disease are observed with high incidence from approximately 2 months after inoculation.


Asunto(s)
Quimera/inmunología , Células Madre Hematopoyéticas/citología , Hepatitis C/inmunología , Hígado/fisiopatología , Modelos Animales , Animales , Proteínas de Unión al ADN/genética , Dimerización , Trasplante de Células Madre Hematopoyéticas , Humanos , Hígado/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Tacrolimus/análogos & derivados
12.
PLoS One ; 5(5): e10585, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20498840

RESUMEN

BACKGROUND: Infection with adeno-associated virus (AAV) vector with liver tropism leads to persistent expression of foreign antigens in the mouse liver, with no significant liver inflammation or pathology. This provides a model to investigate antigen persistence in the liver and strategies to modulate host immunity to reduce or clear the foreign antigen expressed from AAV vector in the liver. METHODS/PRINCIPAL FINDINGS: We showed that expressing LIGHT with an adenovirus vector (Ad) in mice with established AAV in the liver led to clearance of the AAV. Ad-LIGHT enhanced CD8 effector T cells in the liver, correlated with liver inflammation. LTbetaR-Ig proteins blocked Ad-LIGHT in clearing AAV. Interestingly, in LTbetaR-null mice, Ad-LIGHT still cleared AAV but caused no significant liver inflammation. CONCLUSIONS/SIGNIFICANCE: Our data suggest that LIGHT interaction with the LTbetaR plays a critical role in liver inflammation but is not required for LIGHT-mediated AAV clearance. These findings will shed light on developing novel immuno-therapeutics in treating people chronically infected with hepato-tropic viruses.


Asunto(s)
Antígenos Virales/inmunología , Dependovirus/inmunología , Inflamación/virología , Hígado/patología , Hígado/virología , Transducción de Señal/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Dependovirus/genética , Genoma Viral/genética , Inflamación/patología , Receptor beta de Linfotoxina/deficiencia , Ratones , Infecciones por Parvoviridae/patología , Infecciones por Parvoviridae/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA