Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Front Immunol ; 14: 1272055, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37942313

RESUMEN

Conventional type 1 dendritic cells (cDC1s) are superior in antigen cross-presentation and priming CD8+ T cell anti-tumor immunity and thus, are a target of high interest for cancer immunotherapy. Type I interferon (IFN) is a potent inducer of antigen cross-presentation, but, unfortunately, shows only modest results in the clinic given the short half-life and high toxicity of current type I IFN therapies, which limit IFN exposure in the tumor. CD8+ T cell immunity is dependent on IFN signaling in cDC1s and preclinical studies suggest targeting IFN directly to cDC1s may be sufficient to drive anti-tumor immunity. Here, we engineered an anti-XCR1 antibody (Ab) and IFN mutein (IFNmut) fusion protein (XCR1Ab-IFNmut) to determine whether systemic delivery could drive selective and sustained type I IFN signaling in cDC1s leading to anti-tumor activity and, in parallel, reduced systemic toxicity. We found that the XCR1Ab-IFNmut fusion specifically enhanced cDC1 activation in the tumor and spleen compared to an untargeted control IFN. However, multiple treatments with the XCR1Ab-IFNmut fusion resulted in robust anti-drug antibodies (ADA) and loss of drug exposure. Using other cDC1-targeting Ab-IFNmut fusions, we found that localizing IFN directly to cDC1s activates their ability to promote ADA responses, regardless of the cDC1 targeting antigen. The development of ADA remains a major hurdle in immunotherapy drug development and the cellular and molecular mechanisms governing the development of ADA responses in humans is not well understood. Our results reveal a role of cDC1s in ADA generation and highlight the potential ADA challenges with targeting immunostimulatory agents to this cellular compartment.


Asunto(s)
Interferón Tipo I , Neoplasias , Humanos , Interferón Tipo I/metabolismo , Linfocitos T CD8-positivos , Células Dendríticas , Presentación de Antígeno
3.
Cancer Immunol Immunother ; 72(5): 1327-1335, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36394642

RESUMEN

Type I interferon-mediated activation of immune cells can facilitate the generation of productive tumor antigen-specific T cell responses in solid tumors. The cGAS/STING DNA sensing pathway is a critical upstream mediator of type I interferon production and is an important regulator of anti-tumor immunity. Numerous STING pathway agonists are now being tested in clinical trials, but the effectiveness of this approach is not yet clear and a better understanding of the relative importance of this pathway in various tumor settings is needed. We have evaluated syngeneic tumor models with different baseline inflammatory states to determine the contributions of STING activity in both tumor and non-tumor cellular compartments to anti-tumor immune responses. We find that productive anti-tumor immune responses in the poorly immunogenic B16F10 model show a strong dependence on STING expression in non-tumor cells. In the immunogenic MC38 model, constitutive STING activation in tumor cells can partially bypass the requirement for STING-dependent activity from immune cells. Our findings reveal multiple, context-dependent roles for STING activity in the regulation of anti-tumor immunity and the response to immunotherapy. In preclinical models where STING is basally active, checkpoint inhibition is more likely to have a therapeutic effect and removal of STING signaling from either the tumor or the non-tumor compartment has a minimal effect. Removal of STING signaling in both, however, diminishes the efficacy derived from checkpoint therapy. Further work is needed to understand the heterogeneity of STING signaling in patients, both in tumor cells and the tumor microenvironment, and the best means of harnessing this pathway to generate anti-tumor immunity and improve therapeutic outcomes.


Asunto(s)
Interferón Tipo I , Neoplasias , Humanos , ADN , Inmunidad Innata , Inmunoterapia , Transducción de Señal , Microambiente Tumoral
4.
Sci Transl Med ; 13(608)2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433637

RESUMEN

Therapeutic approaches are needed to promote T cell-mediated destruction of poorly immunogenic, "cold" tumors typically associated with minimal response to immune checkpoint blockade (ICB) therapy. Bispecific T cell engager (BiTE) molecules induce redirected lysis of cancer cells by polyclonal T cells and have demonstrated promising clinical activity against solid tumors in some patients. However, little is understood about the key factors that govern clinical responses to these therapies. Using an immunocompetent mouse model expressing a humanized CD3ε chain (huCD3e mice) and BiTE molecules directed against mouse CD19, mouse CLDN18.2, or human EPCAM antigens, we investigated the pharmacokinetic and pharmacodynamic parameters and immune correlates associated with BiTE efficacy across multiple syngeneic solid-tumor models. These studies demonstrated that pretreatment tumor-associated T cell density is a critical determinant of response to BiTE therapy, identified CD8+ T cells as important targets and mediators of BiTE activity, and revealed an antagonistic role for CD4+ T cells in BiTE efficacy. We also identified therapeutic combinations, including ICB and 4-1BB agonism, that synergized with BiTE treatment in poorly T cell-infiltrated, immunotherapy-refractory tumors. In these models, BiTE efficacy was dependent on local expansion of tumor-associated CD8+ T cells, rather than their recruitment from circulation. Our findings highlight the relative contributions of baseline T cell infiltration, local T cell proliferation, and peripheral T cell trafficking for BiTE molecule-mediated efficacy, identify combination strategies capable of overcoming resistance to BiTE therapy, and have clinical relevance for the development of BiTE and other T cell engager therapies.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Animales , Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD19 , Complejo CD3 , Linfocitos T CD8-positivos , Claudinas , Humanos , Inmunoterapia , Ratones , Neoplasias/tratamiento farmacológico
5.
Cancer Immunol Immunother ; 70(8): 2401-2410, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33511454

RESUMEN

Tumor-associated macrophages (TAMs) are abundant in solid tumors where they exhibit immunosuppressive and pro-tumorigenic functions. Inhibition of TAM proliferation and survival through CSF1R blockade has been widely explored as a cancer immunotherapy. To further define mechanisms regulating CSF1R-targeted therapies, we systematically evaluated the effect of anti-CSF1R treatment on tumor growth and tumor microenvironment (TME) inflammation across multiple murine models. Despite substantial macrophage depletion, anti-CSF1R had minimal effects on the anti-tumor immune response in mice bearing established tumors. In contrast, anti-CSF1R treatment concurrent with tumor implantation resulted in more robust tumor growth inhibition and evidence of enhanced anti-tumor immunity. Our findings suggest only minor contributions of CSF1R-dependent TAMs to the inflammatory state of the TME in established tumors, that immune landscape heterogeneity across different tumor models can influence anti-CSF1R activity, and that alternative treatment schedules and/or TAM depletion strategies may be needed to maximize the clinical benefit of this approach.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Macrófagos Asociados a Tumores/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Inmunoterapia/métodos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología
6.
Cell ; 181(2): 442-459.e29, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32302573

RESUMEN

Single-cell RNA sequencing (scRNA-seq) is a powerful tool for defining cellular diversity in tumors, but its application toward dissecting mechanisms underlying immune-modulating therapies is scarce. We performed scRNA-seq analyses on immune and stromal populations from colorectal cancer patients, identifying specific macrophage and conventional dendritic cell (cDC) subsets as key mediators of cellular cross-talk in the tumor microenvironment. Defining comparable myeloid populations in mouse tumors enabled characterization of their response to myeloid-targeted immunotherapy. Treatment with anti-CSF1R preferentially depleted macrophages with an inflammatory signature but spared macrophage populations that in mouse and human expresses pro-angiogenic/tumorigenic genes. Treatment with a CD40 agonist antibody preferentially activated a cDC population and increased Bhlhe40+ Th1-like cells and CD8+ memory T cells. Our comprehensive analysis of key myeloid subsets in human and mouse identifies critical cellular interactions regulating tumor immunity and defines mechanisms underlying myeloid-targeted immunotherapies currently undergoing clinical testing.


Asunto(s)
Neoplasias del Colon/patología , Células Mieloides/metabolismo , Análisis de la Célula Individual/métodos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Secuencia de Bases/genética , Linfocitos T CD8-positivos/inmunología , China , Neoplasias del Colon/terapia , Neoplasias Colorrectales/patología , Células Dendríticas/inmunología , Femenino , Humanos , Inmunoterapia , Macrófagos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Análisis de Secuencia de ARN/métodos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
7.
Immunity ; 52(1): 36-54, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31940272

RESUMEN

Therapeutics that target the T cell inhibitory checkpoint proteins CTLA-4 and PD(L)1 are efficacious across a broad range of cancers, resulting in reductions in tumor burden and increased long-term survival in subsets of patients. The significant and wide-ranging effects of these immunotherapies have prompted the clinical investigation of additional therapies that modulate anti-tumor immunity through effects on T cells, myeloid cells, and other cell types within the tumor microenvironment. The clinical activity of these newer investigational therapies has been mixed, with some therapeutics showing promise but others not exhibiting appreciable efficacy. In this review, we summarize the results of select recent clinical studies of cancer immunotherapies beyond anti-CTLA-4 and anti-PD(L)1 and discuss how these results are providing new insights into the regulation of human anti-tumor immunity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Linfocitos T/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno CTLA-4/antagonistas & inhibidores , Humanos , Activación de Linfocitos/inmunología , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Microambiente Tumoral/inmunología
9.
JCI Insight ; 52019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31211697

RESUMEN

The lung is a relatively quiescent organ during homeostasis, but has a remarkable capacity for repair after injury. Alveolar epithelial type I cells (AEC1s) line airspaces and mediate gas exchange. After injury, they are regenerated by differentiation from their progenitors - alveolar epithelial type II cells (AEC2s) - which also secrete surfactant to maintain surface tension and alveolar patency. While recent studies showed that the maintenance of AEC2 stemness is Wnt dependent, the molecular mechanisms underlying AEC2-AEC1 differentiation in adult lung repair are still incompletely understood. Here we show that WWTR1 (TAZ) plays a crucial role in AEC differentiation. Using an in vitro organoid culture system, we found that tankyrase inhibition can efficiently block AEC2-AEC1 differentiation, and this effect was due to the inhibition of TAZ. In a bleomycin induced lung injury model, conditional deletion of TAZ in AEC2s dramatically reduced AEC1 regeneration during recovery, leading to exacerbated alveolar lesions and fibrosis. In patients with idiopathic pulmonary fibrosis (IPF), decreased blood levels of RAGE, a biomarker of AEC1 health, were associated with more rapid disease progression. Our findings implicate TAZ as a critical factor involved in AEC2 to AEC1 differentiation, and hence the maintenance of alveolar integrity after injury.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Lesión Pulmonar/metabolismo , Transactivadores/metabolismo , Transactivadores/farmacología , Proteínas Adaptadoras Transductoras de Señales , Animales , Bleomicina/efectos adversos , Modelos Animales de Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis/efectos de los fármacos , Organogénesis/fisiología , Organoides/metabolismo , Regeneración/fisiología , Células Madre/metabolismo , Transactivadores/genética , Transcriptoma , beta Catenina/genética , beta Catenina/metabolismo
10.
J Immunol ; 203(4): 1076-1087, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31253728

RESUMEN

Elicitation of tumor cell killing by CD8+ T cells is an effective therapeutic approach for cancer. In addition to using immune checkpoint blockade to reinvigorate existing but unresponsive tumor-specific T cells, alternative therapeutic approaches have been developed, including stimulation of polyclonal T cell cytolytic activity against tumors using bispecific T cell engager (BiTE) molecules that simultaneously engage the TCR complex and a tumor-associated Ag. BiTE molecules are efficacious against hematologic tumors and are currently being explored as an immunotherapy for solid tumors. To understand mechanisms regulating BiTE molecule--mediated CD8+ T cell activity against solid tumors, we sought to define human CD8+ T cell populations that efficiently respond to BiTE molecule stimulation and identify factors regulating their cytolytic activity. We find that human CD45RA+CCR7- CD8+ T cells are highly responsive to BiTE molecule stimulation, are enriched in genes associated with cytolytic effector function, and express multiple unique inhibitory receptors, including leukocyte Ig-like receptor B1 (LILRB1). LILRB1 and programmed cell death protein 1 (PD1) were found to be expressed by distinct CD8+ T cell populations, suggesting different roles in regulating the antitumor response. Engaging LILRB1 with its ligand HLA-G on tumor cells significantly inhibited BiTE molecule-induced CD8+ T cell activation. Blockades of LILRB1 and PD1 induced greater CD8+ T cell activation than either treatment alone. Together, our data suggest that LILRB1 functions as a negative regulator of human CD8+ effector T cells and that blocking LILRB1 represents a unique strategy to enhance BiTE molecule therapeutic activity against solid tumors.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antígenos CD/inmunología , Inmunoterapia/métodos , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Anticuerpos Biespecíficos/inmunología , Humanos , Receptor Leucocitario Tipo Inmunoglobulina B1/antagonistas & inhibidores , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/inmunología , Células Tumorales Cultivadas
11.
J Leukoc Biol ; 2018 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-29393979

RESUMEN

The generation and regulation of innate immune signals are key determinants of autoimmune pathogenesis. Emerging evidence suggests that parallel processes operating in the setting of solid tumors can similarly determine the balance between tolerance and immunity and ultimately the effectiveness of the antitumor immune response. In both contexts, self-specific responses start with innate immune cell activation that leads to the initial break in self-tolerance, which can be followed by immune response amplification and maturation through innate-adaptive crosstalk, and finally immune-mediated tissue/tumor destruction that can further potentiate inflammation. Of particular importance for these processes is type I IFN, which is induced in response to endogenous ligands, such as self-nucleic acids, and acts on myeloid cells to promote the expansion of autoreactive or tumor-specific T cells and their influx into the target tissue. Evidence from the study of human disease pathophysiology and genetics and mouse models of disease has revealed an extensive and complex network of negative regulatory pathways that has evolved to restrain type I IFN production and activity. Here, we review the overlapping features of self- and tumor-specific immune responses, including the central role that regulators of the type I IFN response and innate immune cell activation play in maintaining tolerance, and discuss how a better understanding of the pathophysiology of autoimmunity can help to identify new approaches to promote immune-mediated tumor destruction.

12.
Methods ; 127: 45-52, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28434998

RESUMEN

Intra-vital two-photon microscopy (2P-IVM) allows for in-situ investigation of tissue organization, cell behavior and the dynamic interactions between different cell types in their natural environment. This methodology has also expanded our understanding of the immune response against pathogens. Leishmania are protozoan intracellular parasites that have adapted to successfully establish infection within the context of an inflammatory response in the skin following transmission by the bite of an infected sand fly. The generation of fluorescent transgenic parasites coupled with the increased availability of different types of fluorescent transgenic reporter mice has facilitated the study of the host-parasite interaction in the skin, significantly impacting our understanding of cutaneous leishmaniasis. In this review we will discuss 2P-IVM in the context of Leishmania infection of the mouse ear skin and describe a simple and minimally invasive procedure that allows long-term imaging of this host-pathogen interaction.


Asunto(s)
Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Leishmania major/fisiología , Leishmaniasis Cutánea/fisiopatología , Microscopía/métodos , Piel/parasitología , Animales , Ratones , Ratones Transgénicos , Microorganismos Modificados Genéticamente , Psychodidae
13.
J Crohns Colitis ; 11(5): 610-620, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28453768

RESUMEN

BACKGROUND AND AIMS: The αEß7 integrin is crucial for retention of T lymphocytes at mucosal surfaces through its interaction with E-cadherin. Pathogenic or protective functions of these cells during human intestinal inflammation, such as ulcerative colitis [UC], have not previously been defined, with understanding largely derived from animal model data. Defining this phenotype in human samples is important for understanding UC pathogenesis and is of translational importance for therapeutic targeting of αEß7-E-cadherin interactions. METHODS: αEß7+ and αEß7- colonic T cell localization, inflammatory cytokine production and expression of regulatory T cell-associated markers were evaluated in cohorts of control subjects and patients with active UC by immunohistochemistry, flow cytometry and real-time PCR of FACS-purified cell populations. RESULTS: CD4+αEß7+ T lymphocytes from both healthy controls and UC patients had lower expression of regulatory T cell-associated genes, including FOXP3, IL-10, CTLA-4 and ICOS in comparison with CD4+αEß7- T lymphocytes. In UC, CD4+αEß7+ lymphocytes expressed higher levels of IFNγ and TNFα in comparison with CD4+αEß7- lymphocytes. Additionally the CD4+αEß7+ subset was enriched for Th17 cells and the recently described Th17/Th1 subset co-expressing both IL-17A and IFNγ, both of which were found at higher frequencies in UC compared to control. CONCLUSION: αEß7 integrin expression on human colonic CD4+ T cells was associated with increased production of pro-inflammatory Th1, Th17 and Th17/Th1 cytokines, with reduced expression of regulatory T cell-associated markers. These data suggest colonic CD4+αEß7+ T cells are pro-inflammatory and may play a role in UC pathobiology.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Colitis Ulcerosa/inmunología , Colon/citología , Integrinas/inmunología , Adulto , Anciano , Estudios de Casos y Controles , Colitis Ulcerosa/metabolismo , Colon/inmunología , Citocinas/metabolismo , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Adulto Joven
14.
Thorax ; 72(9): 780-787, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28250200

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is associated with aberrant expression of developmental pathways, including Hedgehog (Hh). As Hh signalling contributes to multiple pro-fibrotic processes, Hh inhibition may represent a therapeutic option for IPF. However, no non-invasive biomarkers are available to monitor lung Hh activity. METHODS: We assessed gene and protein expression in IPF and control lung biopsies, mouse lung, fibroblasts stimulated in vitro with sonic hedgehog (SHh), and plasma in IPF patients versus controls, and cancer patients before and after treatment with vismodegib, a Hh inhibitor. RESULTS: Lung tissue from IPF patients exhibited significantly greater expression of Hh-related genes versus controls. The gene most significantly upregulated in both IPF lung biopsies and fibroblasts stimulated in vitro with SHh was CXCL14, which encodes a soluble secreted chemokine whose expression is inhibited in vitro by the addition of vismodegib. CXCL14 expression was induced by SHh overexpression in mouse lung. Circulating CXCL14 protein levels were significantly higher in plasma from IPF patients than controls. In cancer patients, circulating CXCL14 levels were significantly reduced upon vismodegib treatment. CONCLUSIONS: CXCL14 is a systemic biomarker that could be used to identify IPF patients with increased Hh pathway activity and monitor the pharmacodynamic effects of Hh antagonist therapy in IPF. TRIAL REGISTRATION NUMBER: Post-results, NCT00968981.


Asunto(s)
Quimiocinas CXC/biosíntesis , Proteínas Hedgehog/fisiología , Fibrosis Pulmonar Idiopática/metabolismo , Anciano , Anilidas/farmacología , Animales , Antineoplásicos/farmacología , Biomarcadores/metabolismo , Células Cultivadas , Quimiocinas CXC/sangre , Quimiocinas CXC/efectos de los fármacos , Quimiocinas CXC/genética , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Fibrosis Pulmonar Idiopática/genética , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/tratamiento farmacológico , Piridinas/farmacología , Transducción de Señal/genética , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
15.
Gastroenterology ; 150(2): 477-87.e9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26522261

RESUMEN

BACKGROUND & AIMS: Etrolizumab is a humanized monoclonal antibody against the ß7 integrin subunit that has shown efficacy vs placebo in patients with moderate to severely active ulcerative colitis (UC). Patients with colon tissues that expressed high levels of the integrin αE gene (ITGAE) appeared to have the best response. We compared differences in colonic expression of ITGAE and other genes between patients who achieved clinical remission with etrolizumab vs those who did. METHODS: We performed a retrospective analysis of data collected from 110 patients with UC who participated in a phase 2 placebo-controlled trial of etrolizumab, as well as from 21 patients with UC or without inflammatory bowel disease (controls) enrolled in an observational study at a separate site. Colon biopsies were collected from patients in both studies and analyzed by immunohistochemistry and gene expression profiling. Mononuclear cells were isolated and analyzed by flow cytometry. We identified biomarkers associated with response to etrolizumab. In the placebo-controlled trial, clinical remission was defined as total Mayo Clinic Score ≤2, with no individual subscore >1, and mucosal healing was defined as endoscopic score ≤1. RESULTS: Colon tissues collected at baseline from patients who had a clinical response to etrolizumab expressed higher levels of T-cell-associated genes than patients who did not respond (P < .05). Colonic CD4(+) integrin αE(+) cells from patients with UC expressed higher levels of granzyme A messenger RNA (GZMA mRNA) than CD4(+) αE(-) cells (P < .0001); granzyme A and integrin αE protein were detected in the same cells. Of patients receiving 100 mg etrolizumab, a higher proportion of those with high levels of GZMA mRNA (41%) or ITGAE mRNA (38%) than those with low levels of GZMA (6%) or ITGAE mRNA (13%) achieved clinical remission (P < .05) and mucosal healing (41% GZMA(high) vs 19% GZMA(low) and 44% ITGAE(high) vs 19% ITGAE(low)). Compared with ITGAE(low) and GZMA(low) patients, patients with ITGAE(high) and GZMA(high) had higher baseline numbers of epithelial crypt-associated integrin αE(+) cells (P < .01 for both), but a smaller number of crypt-associated integrin αE(+) cells after etrolizumab treatment (P < .05 for both). After 10 weeks of etrolizumab treatment, expression of genes associated with T-cell activation and genes encoding inflammatory cytokines decreased by 40%-80% from baseline (P < .05) in patients with colon tissues expressing high levels of GZMA at baseline. CONCLUSIONS: Levels of GZMA and ITGAE mRNAs in colon tissues can identify patients with UC who are most likely to benefit from etrolizumab; expression levels decrease with etrolizumab administration in biomarker(high) patients. Larger, prospective studies of markers are needed to assess their clinical value.


Asunto(s)
Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD/metabolismo , Colitis Ulcerosa/tratamiento farmacológico , Colon/efectos de los fármacos , Fármacos Gastrointestinales/uso terapéutico , Granzimas/metabolismo , Cadenas alfa de Integrinas/metabolismo , Antígenos CD/genética , Biopsia , Ensayos Clínicos Fase II como Asunto , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/genética , Colon/enzimología , Colon/patología , Perfilación de la Expresión Génica/métodos , Granzimas/genética , Humanos , Inmunohistoquímica , Cadenas alfa de Integrinas/genética , Valor Predictivo de las Pruebas , ARN Mensajero/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Inducción de Remisión , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento , Cicatrización de Heridas/efectos de los fármacos
16.
Sci Transl Med ; 7(301): 301ra129, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26290411

RESUMEN

Increasing evidence suggests that asthma is a heterogeneous disorder regulated by distinct molecular mechanisms. In a cross-sectional study of asthmatics of varying severity (n = 51), endobronchial tissue gene expression analysis revealed three major patient clusters: TH2-high, TH17-high, and TH2/17-low. TH2-high and TH17-high patterns were mutually exclusive in individual patient samples, and their gene signatures were inversely correlated and differentially regulated by interleukin-13 (IL-13) and IL-17A. To understand this dichotomous pattern of T helper 2 (TH2) and TH17 signatures, we investigated the potential of type 2 cytokine suppression in promoting TH17 responses in a preclinical model of allergen-induced asthma. Neutralization of IL-4 and/or IL-13 resulted in increased TH17 cells and neutrophilic inflammation in the lung. However, neutralization of IL-13 and IL-17 protected mice from eosinophilia, mucus hyperplasia, and airway hyperreactivity and abolished the neutrophilic inflammation, suggesting that combination therapies targeting both pathways may maximize therapeutic efficacy across a patient population comprising both TH2 and TH17 endotypes.


Asunto(s)
Asma/inmunología , Asma/metabolismo , Células Th17/metabolismo , Células Th2/metabolismo , Animales , Células Cultivadas , Femenino , Humanos , Interleucina-13/metabolismo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos BALB C , Transducción de Señal
17.
Thorax ; 70(1): 48-56, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25217476

RESUMEN

BACKGROUND: There is microscopic spatial and temporal heterogeneity of pathological changes in idiopathic pulmonary fibrosis (IPF) lung tissue, which may relate to heterogeneity in pathophysiological mediators of disease and clinical progression. We assessed relationships between gene expression patterns, pathological features, and systemic biomarkers to identify biomarkers that reflect the aggregate disease burden in patients with IPF. METHODS: Gene expression microarrays (N=40 IPF; 8 controls) and immunohistochemical analyses (N=22 IPF; 8 controls) of lung biopsies. Clinical characterisation and blood biomarker levels of MMP3 and CXCL13 in a separate cohort of patients with IPF (N=80). RESULTS: 2940 genes were significantly differentially expressed between IPF and control samples (|fold change| >1.5, p<0.05). Two clusters of co-regulated genes related to bronchiolar epithelium or lymphoid aggregates exhibited substantial heterogeneity within the IPF population. Gene expression in bronchiolar and lymphoid clusters corresponded to the extent of bronchiolisation and lymphoid aggregates determined by immunohistochemistry in adjacent tissue sections. Elevated serum levels of MMP3, encoded in the bronchiolar cluster, and CXCL13, encoded in the lymphoid cluster, corresponded to disease severity and shortened survival time (p<10(-7) for MMP3 and p<10(-5) for CXCL13; Cox proportional hazards model). CONCLUSIONS: Microscopic pathological heterogeneity in IPF lung tissue corresponds to specific gene expression patterns related to bronchiolisation and lymphoid aggregates. MMP3 and CXCL13 are systemic biomarkers that reflect the aggregate burden of these pathological features across total lung tissue. These biomarkers may have clinical utility as prognostic and/or surrogate biomarkers of disease activity in interventional studies in IPF.


Asunto(s)
Quimiocina CXCL13/genética , Regulación de la Expresión Génica , Fibrosis Pulmonar Idiopática/genética , Pulmón/patología , Metaloproteinasa 3 de la Matriz/genética , Anciano , Anciano de 80 o más Años , Linfocitos B , Quimiocina CXCL13/biosíntesis , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Inmunohistoquímica , Pulmón/metabolismo , Masculino , Metaloproteinasa 3 de la Matriz/biosíntesis , Persona de Mediana Edad , Pronóstico , Índice de Severidad de la Enfermedad
18.
J Immunol ; 193(1): 111-9, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24879793

RESUMEN

IL-13 can bind to two distinct receptors: a heterodimer of IL-13Rα1/IL-4Rα and IL-13Rα2. Whereas IL-13Rα1/IL-4Rα engagement by IL-13 leads to the activation of STAT6, the molecular events triggered by IL-13 binding to IL-13Rα2 remain incompletely understood. IL-4 can bind to and signal through the IL-13Rα1/IL-4Rα complex but does not interact with IL-13Rα2. Idiopathic pulmonary fibrosis is a progressive and generally fatal parenchymal lung disease of unknown etiology with no current pharmacologic treatment options that substantially prolong survival. Preclinical models of fibrotic diseases have implicated IL-13 activity on multiple cell types, including macrophages and fibroblasts, in initiating and perpetuating pathological fibrosis. In this study, we show that IL-13, IL-4, IL-13Rα2, and IL-13-inducible target genes are expressed at significantly elevated levels in lung tissue from patients with idiopathic pulmonary fibrosis compared with control lung tissue. IL-4 and IL-13 induce virtually identical transcriptional responses in human monocytes, macrophages, and lung fibroblasts. IL-13Rα2 expression can be induced in lung fibroblasts by IL-4 or IL-13 via a STAT6-dependent mechanism, or by TNF-α via a STAT6-independent mechanism. Endogenously expressed IL-13Rα2 decreases, but does not abolish, sensitivity of lung fibroblasts to IL-13 and does not affect sensitivity to IL-4. Genome-wide transcriptional analyses of lung fibroblasts stimulated with IL-13 in the presence of Abs that selectively block interactions of IL-13 with IL-13Rα1/IL-4Rα or IL-13Rα2 show that endogenously expressed IL-13Rα2 does not activate any unique IL-13-mediated gene expression patterns, confirming its role as a decoy receptor for IL-13 signaling.


Asunto(s)
Fibroblastos/inmunología , Regulación de la Expresión Génica/inmunología , Fibrosis Pulmonar Idiopática/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Interleucina-13/inmunología , Pulmón/inmunología , Transducción de Señal/inmunología , Femenino , Fibroblastos/patología , Estudio de Asociación del Genoma Completo , Humanos , Fibrosis Pulmonar Idiopática/patología , Subunidad alfa1 del Receptor de Interleucina-13/inmunología , Interleucina-4/inmunología , Subunidad alfa del Receptor de Interleucina-4/inmunología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Monocitos/inmunología , Monocitos/patología , Factor de Transcripción STAT6/inmunología , Factor de Necrosis Tumoral alfa/inmunología
19.
Lancet ; 384(9940): 309-18, 2014 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-24814090

RESUMEN

BACKGROUND: Etrolizumab is a humanised monoclonal antibody that selectively binds the ß7 subunit of the heterodimeric integrins α4ß7 and αEß7. We aimed to assess etrolizumab in patients with moderately-to-severely active ulcerative colitis. METHODS: In this double-blind, placebo-controlled, randomised, phase 2 study, patients with moderately-to-severely active ulcerative colitis who had not responded to conventional therapy were recruited from 40 referral centres in 11 countries. Eligible patients (aged 18-75 years; Mayo Clinic Score [MCS] of 5 of higher [or ≥6 in USA]; and disease extending 25 cm or more from anal verge) were randomised (1:1:1) to one of two dose levels of subcutaneous etrolizumab (100 mg at weeks 0, 4, and 8, with placebo at week 2; or 420 mg loading dose [LD] at week 0 followed by 300 mg at weeks 2, 4, and 8), or matching placebo. The primary endpoint was clinical remission at week 10, defined as MCS of 2 or less (with no individual subscore of >1), analysed in the modified intention-to-treat population (mITT; all randomly assigned patients who had received at least one dose of study drug, had at least one post-baseline disease-activity assessment, and had a centrally read screening endoscopic subscore of ≥2). This study is registered with ClinicalTrials.gov, number NCT01336465. FINDINGS: Between Sept 2, 2011, and July 11, 2012, 124 patients were randomly assigned, of whom five had a endoscopic subscore of 0 or 1 and were excluded from the mITT population, leaving 39 patients in the etrolizumab 100 mg group, 39 in the etrolizumab 300 mg plus LD group, and 41 in the placebo group for the primary analyses. No patients in the placebo group had clinical remission at week 10, compared with eight (21% [95% CI 7-36]) patients in the etrolizumab 100 mg group (p=0·0040) and four (10% [0·2-24]) patients in the 300 mg plus LD group (p=0·048). Adverse events occurred in 25 (61%) of 41 patients in the etrolizumab 100 mg group (five [12%] of which were regarded as serious), 19 (48%) of 40 patients in the etrolizumab 300 mg plus LD group (two [5%] serious), and 31 (72%) of 43 patients in the placebo group (five [12%] serious). INTERPRETATION: Etrolizumab was more likely to lead to clinical remission at week 10 than was placebo. Therefore, blockade of both α4ß7 and αEß7 might provide a unique therapeutic approach for the treatment of ulcerative colitis, and phase 3 studies have been planned. FUNDING: Genentech.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Adulto , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados/administración & dosificación , Método Doble Ciego , Femenino , Humanos , Masculino , Inducción de Remisión/métodos , Factores de Tiempo , Resultado del Tratamiento
20.
J Immunol ; 192(4): 1651-1660, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24431231

RESUMEN

Exposure to pathogens in the periphery elicits effector T cell differentiation in local lymph nodes followed by migration of activated T cells to and within the infected site. However, the relationships among pathogen abundance, Ag display on MHC molecules, effector T cell dynamics, and functional responses at the infected sites are incompletely characterized. In this study, we compared CD4(+) T cell effector dynamics and responses during pulmonary mycobacterial infection versus acute influenza infection. Two-photon imaging together with in situ as well as ex vivo analysis of cytokine production revealed that the proportion of migration-arrested, cytokine-producing effector T cells was dramatically higher in the influenza-infected lungs due to substantial differences in Ag abundance in the two infectious states. Despite the marked inflammatory conditions associated with influenza infection, histocytometric analysis showed that cytokine production was focal, with a restriction to areas of significant Ag burden. Optimal effector function is thus constrained by the availability of TCR ligands, pointing to the value of increasing Ag stimulation rather than effector numbers in harnessing CD4(+) T cells for therapeutic purposes in such conditions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Tuberculosis Pulmonar/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Citocinas/biosíntesis , Inflamación/inmunología , Interferón gamma/metabolismo , Hígado/inmunología , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología , Orthomyxoviridae/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...