Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 14(636): eabg8402, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35294258

RESUMEN

To uncover underlying mechanisms associated with failure of indoleamine 2,3-dioxygenase 1 (IDO1) blockade in clinical trials, we conducted a pilot, window-of-opportunity clinical study in 17 patients with newly diagnosed advanced high-grade serous ovarian cancer before their standard tumor debulking surgery. Patients were treated with the IDO1 inhibitor epacadostat, and immunologic, transcriptomic, and metabolomic characterization of the tumor microenvironment was undertaken in baseline and posttreatment tumor biopsies. IDO1 inhibition resulted in efficient blockade of the kynurenine pathway of tryptophan degradation and was accompanied by a metabolic adaptation that shunted tryptophan catabolism toward the serotonin pathway. This resulted in elevated nicotinamide adenine dinucleotide (NAD+), which reduced T cell proliferation and function. Because NAD+ metabolites could be ligands for purinergic receptors, we investigated the impact of blocking purinergic receptors in the presence or absence of NAD+ on T cell proliferation and function in our mouse model. We demonstrated that A2a and A2b purinergic receptor antagonists, SCH58261 or PSB1115, respectively, rescued NAD+-mediated suppression of T cell proliferation and function. Combining IDO1 inhibition and A2a/A2b receptor blockade improved survival and boosted the antitumor immune signature in mice with IDO1 overexpressing ovarian cancer. These findings elucidate the downstream adaptive metabolic consequences of IDO1 blockade in ovarian cancers that may undermine antitumor T cell responses in the tumor microenvironment.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa , Neoplasias Ováricas , Animales , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Activación de Linfocitos , Ratones , NAD , Neoplasias Ováricas/tratamiento farmacológico , Triptófano/metabolismo , Microambiente Tumoral
2.
Front Immunol ; 12: 678999, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025677

RESUMEN

The immunoregulatory enzyme, indoleamine 2,3-dioxygenase (IDO1) and the PD-1/PD-L1 axis are potent mechanisms that impede effective anti-tumor immunity in ovarian cancer. However, whether the IDO pathway regulates PD-1 expression in T cells is currently unknown. Here we show that tumoral IDO1 expression led to profound changes in tryptophan, nicotinate/nicotinamide, and purine metabolic pathways in the ovarian tumor microenvironment, and to an increased frequency of PD-1+CD8+ tumor infiltrating T cells. We determined that activation of the aryl hydrocarbon receptor (AHR) by kynurenine induced PD-1 expression, and this effect was significantly abrogated by the AHR antagonist CH223191. Mechanistically, kynurenine alters chromatin accessibility in regulatory regions of T cell inhibitory receptors, allowing AHR to bind to consensus XRE motifs in the promoter region of PD-1. These results enable the design of strategies to target the IDO1 and AHR pathways for enhancing anti-tumor immunity in ovarian cancer.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Neoplasias Ováricas/etiología , Neoplasias Ováricas/metabolismo , Receptor de Muerte Celular Programada 1/genética , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Sitios de Unión , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Masculino , Ratones , Neoplasias Ováricas/patología , Receptor de Muerte Celular Programada 1/química , Receptor de Muerte Celular Programada 1/metabolismo , Unión Proteica , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
3.
Am J Reprod Immunol ; 85(3): e13343, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32905653

RESUMEN

PROBLEM: Previous studies identified circulating CD14+ HLA-DRlo/- monocytic cells as an immune suppressive subset in solid malignancies, such as prostate, renal cell carcinoma, and pancreatic cancer. Such monocytic cells have been implicated not only in tumour progression but also as a potential barrier for immunotherapy. This study examined the relationship between the frequency of circulating monocytic cells and epithelial ovarian cancer (EOC) progression pre- and post-frontline chemotherapy, defined by disease stage, which is a leading prognostic factor for this malignancy. METHOD OF STUDY: Incident cases of 236 women with EOC were recruited and comprehensive flow cytometry was utilized to assess the frequency of peripheral blood CD33+ CD11b+ HLA-DR-/low CD14+ CD15- monocytic cells, henceforth termed CD14+ HLA-DRlo/- monocytic cells, prior to and after completion of frontline chemotherapy. Multivariable odds ratios (OR) were used to estimate the association between CD14+ HLA-DRlo/- monocytic cell percentages and disease stage. Wilcoxon signed-rank tests evaluated changes in these monocytic cell levels pre- and post-chemotherapy in a patient subset (n = 70). RESULTS: Patients with elevated frequencies of circulating CD14+ HLA-DRlo/- monocytic cells at diagnosis were at 3.33-fold greater odds of having advanced stage (III/IV) EOC (CI: 1.04-10.64), with a significant trend in increasing CD14+ HLA-DRlo/- monocytic cell levels (P = .04). There was a 2.02% median decrease of these monocytic cells post-chemotherapy among a subset of patients with advanced stage disease (P < .0001). CONCLUSION: These findings support the potential clinical relevance of CD14+ HLA-DRlo/- monocytic cells in EOC for prognosis and may indicate a non-invasive biomarker to measure disease progression.


Asunto(s)
Células Epiteliales/patología , Imidas/inmunología , Neoplasias Ováricas/inmunología , Polifosfatos/inmunología , Anciano , Biomarcadores , Carcinogénesis , Progresión de la Enfermedad , Femenino , Antígenos HLA-DR/metabolismo , Humanos , Tolerancia Inmunológica , Receptores de Lipopolisacáridos/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/diagnóstico , Pronóstico
4.
Oncotarget ; 11(27): 2669-2683, 2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32676168

RESUMEN

CD8+ tumor-infiltrating lymphocytes (TILs) are not all specific for tumor antigens, but can include bystander TILs that are specific for cancer-irrelevant epitopes, and it is unknown whether the T-cell repertoire affects prognosis. To delineate the complexity of anti-tumor T-cell responses, we utilized a computational method for de novo assembly of sequences from CDR3 regions of 369 high-grade serous ovarian cancers from TCGA, and then applied deep TCR-sequencing for analyses of paired tumor and peripheral blood specimens from an independent cohort of 99 ovarian cancer patients. Strongly monoclonal T-cell repertoires were associated with favorable prognosis (PFS, HR = 0.65, 0.50-0.84, p = 0.003; OS, HR = 0.61, 0.44-0.83, p = 0.006) in TCGA cohort. In the validation cohort, we discovered that patients with low T-cell infiltration but low diversity or focused repertoires had clinical outcomes almost indistinguishable from highly-infiltrated tumors (median 21.0 months versus 15.9 months, log-rank p = 0.945). We also found that the degree of divergence of the peripheral repertoire from the TIL repertoire, and the presence of detectable spontaneous anti-tumor immune responses are important determinants of clinical outcome. We conclude that the prognostic significance of TILs in ovarian cancer is dictated by T-cell clonality, degree of overlap with peripheral repertoire, and the presence of detectable spontaneous anti-tumor immune response in the patients. These immunological phenotypes defined by the TCR repertoire may provide useful insights for identifying "TIL-low" ovarian cancer patients that may respond to immunotherapy.

5.
Clin Obstet Gynecol ; 63(1): 48-63, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31833846

RESUMEN

The role of the immune system in the development of cancer has been a subject of ongoing clinical investigation in recent years. Emerging data demonstrate that tumorigenesis resulting in ovarian, uterine, and cervical cancers is a consequence of impaired host immune responses to cancerous cells. Leveraging the immune system through the use of immune checkpoint inhibitors, therapeutic vaccine therapy, and adoptive cell transfer presents a profound opportunity to revolutionize cancer treatment. This review will encompass the role of the immune system in development of gynecologic cancers and highlight recent data regarding immunotherapy applications in ovarian, uterine, and cervical cancers.


Asunto(s)
Neoplasias de los Genitales Femeninos/terapia , Inmunoterapia/tendencias , Femenino , Predicción , Humanos
6.
J Immunother Cancer ; 7(1): 189, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31315674

RESUMEN

BACKGROUND: Cancer immunotherapies are emerging as promising treatment strategies for ovarian cancer patients that experience disease relapse following first line therapy. As such, identifying strategies to bolster anti-tumor immunity and limit immune suppression, while recognizing diverse patterns of tumor response to immunotherapy is critical to selecting treatment combinations that lead to durable therapeutic benefit. METHODS: Using a pre-clinical mouse model, we evaluated a heterologous prime/boost vaccine in combination with checkpoint blockade to treat metastatic intraperitoneal ovarian cancer. Vaccine-elicited CD8+ T cell responses and changes in the tumor microenvironment following treatment were analyzed and compared to treatment outcome. Kinetics of intraperitoneal tumor growth were assessed using non-invasive magnetic resonance imaging (MRI). RESULTS: Vaccine priming followed by antigen-armed oncolytic Maraba virus boosting elicited robust tumor-specific CD8+ T cell responses that improved tumor control and led to unique immunological changes in the tumor, including a signature that correlated with improved clinical outcome of ovarian cancer patients. However, this treatment was not curative and T cells in the tumor microenvironment (TME) were functionally suppressed. Combination PD-1 blockade partially overcame the adaptive resistance in the tumor observed in response to prime/boost vaccination, restoring CD8+ T cell function in the TME and enhancing the therapeutic response. Non-invasive MRI of tumors during the course of combination treatment revealed heterogeneous radiologic response patterns following treatment, including pseudo-progression, which was associated with improved tumor control prior to relapse. CONCLUSIONS: Our findings point to a key hierarchical role for PD-1 signaling and adaptive immune resistance in the ovarian TME in determining the functional fate of tumor-specific CD8+ T cells, even in the context of robust therapy mediated anti-tumor immunity, as well as the ability of multiple unique patterns of therapeutic response to result in durable tumor control.


Asunto(s)
Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/administración & dosificación , Oxidorreductasas Intramoleculares/genética , Ovalbúmina/genética , Neoplasias Ováricas/terapia , Vesiculovirus/fisiología , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Oxidorreductasas Intramoleculares/inmunología , Ratones , Metástasis de la Neoplasia , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Ovalbúmina/inmunología , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/inmunología , Resultado del Tratamiento , Microambiente Tumoral , Vesiculovirus/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Immunother Cancer ; 7(1): 156, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221207

RESUMEN

BACKGROUND: Efficient identification of neoantigen-specific T-cell responses in epithelial ovarian cancer (EOC) remains a challenge. Existing investigations of spontaneous T-cell response to tumor neoepitope in EOC have taken the approach of comprehensive screening all neoantigen candidates, with a validation rate of 0.5-2%. METHODS: Whole-exome and transcriptome sequencing analysis of treatment-naive EOC patients were performed to identify neoantigen candidates, and the immunogenicity of prioritized neoantigens was evaluated by analyzing spontaneous neoantigen-specfic CD4+ and CD8+ T-cell responses in the tumor and/or peripheral blood. The biological relevance of neoantigen-specific T-cell lines and clones were analyzed by evaluating the capacity of autologous ovarian tumor recognition. Genetic transfer of T-cell receptor (TCR) from these neoantigen-specific T-cell clones into peripheral blood T-cells was conducted to generate neoepitope-specific T-cells. The molecular signature associated with positive neoantigen T-cell responses was investigated, and the impacts of expression level and lymphocyte source on neoantigen identification were explored. RESULTS: Using a small subset of prioritized neoantigen candidates, we were able to detect spontaneous CD4+ and/or CD8+ T-cell responses against neoepitopes from autologous lymphocytes in half of treatment-naïve EOC patients, with a significantly improved validation rate of 19%. Tumors from patients exhibiting neoantigen-specific T-cell responses exhibited a signature of upregulated antigen processing and presentation machinery, which was also associated with favorable patient survival in the TCGA ovarian cohort. T-cells specific against two mutated cancer-associated genes, NUP214 and JAK1, recognized autologous tumors. Gene-engineering with TCR from these neoantigen-specific T-cell clones conferred neoantigen-reactivity to peripheral T-cells. CONCLUSIONS: Our study demonstrated the feasibility of efficiently identifying both CD4+ and CD8+ neoantigen-specific T-cells in EOC. Autologous lymphocytes genetically engineered with tumor antigen-specific TCR can be used to generate cells for use in the personalized adoptive T-cell transfer immunotherapy.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Carcinoma Epitelial de Ovario/inmunología , Inmunoterapia/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Femenino , Humanos
8.
Front Immunol ; 10: 48, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30778346

RESUMEN

Inappropriate inflammation exacerbates a vast array of chronic and acute conditions with severe health risks. In certain situations, such as acute sepsis, traditional therapies may be inadequate in preventing severe organ damage or death. We have previously shown cell surface glycan modification by the circulating sialyltransferase ST6Gal-1 regulates de novo inflammatory cell production via a novel extrinsic glycosylation pathway. Here, we show that therapeutic administration of recombinant, bioactive ST6Gal-1 (rST6G) mitigates acute inflammation in a murine model mimicking acute exacerbations experienced by patients with chronic obstructive pulmonary disease (COPD). In addition to suppressing proximal neutrophil recruitment at onset of infection-mediated inflammation, rST6G also muted local cytokine production. Histologically, exposure with NTHI, a bacterium associated with COPD exacerbations, in rST6G-treated animals revealed consistent and pronounced reduction of pulmonary inflammation, characterized by smaller inflammatory cuffs around bronchovascular bundles, and fewer inflammatory cells within alveolar walls, alveolar spaces, and on pleural surfaces. Taken together, the data advance the idea that manipulating circulatory ST6Gal-1 levels has potential in managing inflammatory conditions by leveraging the combined approaches of controlling new inflammatory cell production and dampening the inflammation mediator cascade.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Neumonía/tratamiento farmacológico , Neumonía/etiología , Proteínas Recombinantes/administración & dosificación , Sialiltransferasas/administración & dosificación , Enfermedad Aguda , Animales , Biomarcadores , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Infusiones Intravenosas , Ratones , Ratones Transgénicos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neumonía/diagnóstico , Neumonía/metabolismo , Pronóstico , Índice de Severidad de la Enfermedad , Sialiltransferasas/sangre , Resultado del Tratamiento , beta-D-Galactósido alfa 2-6-Sialiltransferasa
9.
Hematol Oncol Clin North Am ; 32(6): 1025-1039, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30390758

RESUMEN

Clinical progress in cancer immunotherapy has been slow; however, within the last 5 years, breakthrough successes have brought immunotherapy to the forefront in cancer therapy. Promising results have been observed in solid tumors and hematologic malignancies. Most treatment modalities have shown limited efficacy as monotherapy. The complex nature of cancer and the immunosuppressive microenvironment emphasizes the need to personalize immunotherapy by manipulating the patient's own immune system. For successful and long-lasting cure of cancer, a multimodal approach is essential, combining antitumor cell therapy with manipulation of multiple pathways in the tumor microenvironment to ameliorate tumor-induced immunosuppression.


Asunto(s)
Carcinoma Epitelial de Ovario , Inmunoterapia/métodos , Inmunoterapia/tendencias , Neoplasias Ováricas , Microambiente Tumoral/inmunología , Carcinoma Epitelial de Ovario/inmunología , Carcinoma Epitelial de Ovario/patología , Carcinoma Epitelial de Ovario/terapia , Femenino , Humanos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia
10.
Immunology ; 155(2): 202-210, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29772069

RESUMEN

Major progress in the analysis of human immune responses to cancer has been made through the molecular characterization of human tumour antigens. The development of therapeutic strategies for eliciting immune-mediated rejection of tumours has accelerated due to the elucidation of the molecular basis for tumour cell recognition and destruction by immune cells. Of the various human tumour antigens defined to date in ovarian cancer, the cancer-testis (CT) family of antigens have been studied extensively preclinically and clinically because of their testis-restricted expression in normal tissues and ability to elicit robust immune responses. Recent developments in cancer sequencing technologies offer a unique opportunity to identify tumour mutations with the highest likelihood of being expressed and recognized by the immune system. Such mutations, or neoantigens, could potentially serve as specific immune targets for T-cell-mediated destruction of cancer cells. This review will highlight current work in selecting tumour rejection antigens in ovarian cancer for improving the efficacy of immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Ováricas/etiología , Neoplasias Ováricas/metabolismo , Escape del Tumor/inmunología , Animales , Antígenos de Neoplasias/genética , Ensayos Clínicos como Asunto , Femenino , Humanos , Inmunomodulación , Inmunoterapia , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Escape del Tumor/genética
11.
Am J Clin Oncol ; 41(4): 332-338, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-27014931

RESUMEN

OBJECTIVES: To evaluate the safety/efficacy and explore biomarkers for a rationally designed combination of sunitinib and transarterial chemoembolization (TACE) in a prospective phase 2 study of advanced hepatocellular carcinoma (HCC). METHODS: Inoperable HCC patients with Child-Pugh A disease received 37.5 mg sunitinib from days 1 to 7 followed by TACE on day 8. Sunitinib was resumed from days 15 to 36 followed by 2 weeks off. Patients received subsequent sunitinib cycles of 4 weeks on and 2 weeks off. Dynamic contrast-enhanced magnetic resonance imaging and circulating soluble biomarkers were assessed at baseline, day 8, day 10, and day 36. RESULTS: Sixteen patients with liver only (n=10) and extrahepatic disease (n=6) were enrolled. After a median follow-up of 12.8 months, 2 partial responses, 11 stable disease, and 3 clinical deteriorations were seen for a clinical benefit rate of 81%. Median progression-free survival (PFS) was 8 months (95% CI, 4.3-9.3) and overall survival was 14.9 months (95% CI, 6.3-27.1). Eleven of 16 patients (69%) had grade 3/4 toxicities attributable to sunitinib, the most frequent being thrombocytopenia, amylase/lipase elevations, lymphopenia, and fatigue. Mean K (volume transfer constant) and viable tumor percent in consented patients decreased by 27% and 14.8%, respectively, with combination therapy. Soluble vascular endothelial growth factor receptor-2 (sVEGFR2) levels, cytokines (interleukin-8, interleukin-21), and monocytes decreased with combination therapy. Estimated sunitinib IC50 values of 15 and 10 ng/mL modulated K and AUC90. sVEGFR2 levels decreased with K and AUC90. CONCLUSIONS: Encouraging progression-free survival and overall survival were seen with acceptable toxicity in our study of sunitinib and TACE combination in advanced HCC. Potential imaging and serum biomarkers showed increased benefit with combination therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/análisis , Carcinoma Hepatocelular/terapia , Quimioembolización Terapéutica , Neoplasias Hepáticas/terapia , Sunitinib/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Hepatocelular/secundario , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia
12.
Adv Exp Med Biol ; 1036: 129-144, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29275469

RESUMEN

Over the last decade, tryptophan catabolism has been firmly established as a powerful mechanism of innate and adaptive immune tolerance. The catabolism of tryptophan is a central pathway maintaining homeostasis by preventing autoimmunity or immunopathology that would result from uncontrolled and overreacting immune responses. This is driven by the key and rate-limiting enzymes indoleamine-2,3-dioxygenase 1 (IDO1) and tryptophan-2,3-dioxygenase 2 (TDO), resulting in local depletion of tryptophan, while tryptophan catabolites accumulate, including kynurenine and its derivatives, depending on the presence of downstream enzymes in the kynurenine pathway. These metabolic modifications result in a local microenvironment that is profoundly immunosuppressive, as a result of various mechanisms whose respective role remains incompletely characterized. Drugs targeting this pathway, specifically IDO1, are already in clinical trials with the aim at reverting cancer-induced immunosuppression. Recent studies have demonstrated favorable pharmacokinetics profiles for first-generation (Indoximod NLG8189) and second-generation IDO1 inhibitors (INCB024360 and NLG919). Targeting tryptophan catabolism in combination with additional methods of therapy may improve efficacy of cancer immunotherapy. These methods include, but are not limited to vaccination, adoptive cellular therapy, checkpoint inhibitor blockade, and cyclooxygenase-2 (COX2) inhibition. Over the last decade, there has been a considerable increase in our understanding of the regulation and downstream mediators of tryptophan metabolism. This detailed understanding will expand opportunities to interfere with the pathway therapeutically on multiple levels. The object of this chapter is to highlight current and past key findings that implicate tryptophan catabolism as an important mediator of cancer immunity and discuss the development of multiple therapeutic targets.


Asunto(s)
Tolerancia Inmunológica , Inmunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenasa , Proteínas de Neoplasias , Neoplasias , Triptófano , Animales , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Triptófano/inmunología , Triptófano/metabolismo
13.
Gynecol Oncol ; 145(3): 420-425, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28392127

RESUMEN

OBJECTIVES: NY-ESO-1 is a cancer testis antigen and a promising target for immunotherapy. The purpose of this study was to determine the expression frequency, immunogenicity, and clinical impact of NY-ESO-1 in ovarian cancer. METHODS: Immunohistochemistry (IHC), reverse-transcription polymerase chain reaction (RT-PCR), and quantitative-PCR (qRT-PCR) were utilized in an ovarian cancer (including Fallopian tube and primary peritoneal cancers) patient cohort; humoral responses against NY-ESO-1 were determined by ELISA. Clinicopathologic outcomes including progression-free (PFS) and overall (OS) survival were evaluated based on NY-ESO-1 expression. Cohen's kappa (κ) tested agreement between expression tests. RESULTS: NY-ESO-1 expression was detected by any method in 40.7% of 1002 patients' tumors (NY-ESO-1+) and baseline humoral response was identified in 19.0% of 689 tested patients. NY-ESO-1+ patients were older (p<0.001), higher stage (85% stage III/IV vs. 76.4%, p=0.015), less likely to have a complete response to initial therapy (53.9% vs. 68.9%, p=0.002), had more serous histotype (74.5% vs. 66.9%, p=0.011), and had more grade 3 tumors (83.7% vs. 70.8%, p<0.001). There was a trend towards shorter PFS (22.2 vs. 25.0months, p=0.07) and significantly shorter OS (42.9 vs. 50.0months, p=0.003) among NY-ESO-1+ patients. A subset analysis of NY-ESO-1+ patients that received immunotherapy demonstrated improved OS by >2years (52.6 vs. 27.2months, p<0.001). CONCLUSIONS: This study is the first demonstration of an association between NY-ESO-1 expression and an aggressive cancer phenotype. The relatively high expression frequency of NY-ESO-1 in ovarian cancer patients coupled with the poor clinical outcomes in NY-ESO-1+ patients reveals an underappreciated need for targeted therapy against this antigen. In support, our study reveals that NY-ESO-1+ patients enrolled on immunotherapy trials targeting the antigen exhibited an improvement in OS.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/inmunología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos como Asunto , Femenino , Humanos , Inmunohistoquímica , Inmunoterapia , Persona de Mediana Edad , Terapia Molecular Dirigida , Clasificación del Tumor , Neoplasias Ováricas/patología , Fenotipo , Adulto Joven
14.
J Immunol ; 192(11): 5226-35, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24752444

RESUMEN

The detrimental impact of tobacco on human health is clearly recognized, and despite aggressive efforts to prevent smoking, close to one billion individuals worldwide continue to smoke. People with chronic obstructive pulmonary disease are susceptible to recurrent respiratory infections with pathogens, including nontypeable Haemophilus influenzae (NTHI), yet the reasons for this increased susceptibility are poorly understood. Because mortality rapidly increases with multiple exacerbations, development of protective immunity is critical to improving patient survival. Acute NTHI infection has been studied in the context of cigarette smoke exposure, but this is the first study, to our knowledge, to investigate chronic infection and the generation of adaptive immune responses to NTHI after chronic smoke exposure. After chronic NTHI infection, mice that had previously been exposed to cigarette smoke developed increased lung inflammation and compromised adaptive immunity relative to air-exposed controls. Importantly, NTHI-specific T cells from mice exposed to cigarette smoke produced lower levels of IFN-γ and IL-4, and B cells produced reduced levels of Abs against outer-membrane lipoprotein P6, with impaired IgG1, IgG2a, and IgA class switching. However, production of IL-17, which is associated with neutrophilic inflammation, was enhanced. Interestingly, cigarette smoke-exposed mice exhibited a similar defect in the generation of adaptive immunity after immunization with P6. Our study has conclusively demonstrated that cigarette smoke exposure has a profound suppressive effect on the generation of adaptive immune responses to NTHI and suggests the mechanism by which prior cigarette smoke exposure predisposes chronic obstructive pulmonary disease patients to recurrent infections, leading to exacerbations and contributing to mortality.


Asunto(s)
Infecciones por Haemophilus/inmunología , Haemophilus influenzae/inmunología , Neumonía Bacteriana/inmunología , Fumar/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/inmunología , Animales , Linfocitos B/inmunología , Proteínas de la Membrana Bacteriana Externa/farmacología , Enfermedad Crónica , Femenino , Infecciones por Haemophilus/patología , Infecciones por Haemophilus/prevención & control , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Interferón gamma/inmunología , Interleucina-4/inmunología , Ratones , Neumonía Bacteriana/inducido químicamente , Neumonía Bacteriana/prevención & control , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/prevención & control , Fumar/inmunología , Fumar/patología
15.
Oncoimmunology ; 2(7): e24679, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24073364

RESUMEN

The accumulation of immunosuppressive cells and exhausted effector T cells highlight an important immune dysfunction in advanced stage hepatocellular carcinoma (HCC) patients. These cells significantly hamper the efficacy immunotherapies and facilitate HCC progression. We have recently demonstrated that the multipronged depletion of immunosuppressive cells potentially restores effector T-cell function in HCC.

16.
Nanomedicine ; 9(7): 923-34, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23542018

RESUMEN

Chitosan nanoparticles were evaluated as a vaccine delivery system for hepatitis B surface antigen (HBsAg) in the absence of adjuvant. Nano-encapsulated HBsAg (HBsAg chitosan-NP) was endocytosed more rapidly and efficiently by dendritic cells compared to soluble HBsAg. FRET analysis demonstrated that intact nanoparticles were taken up by DCs. To determine the immunogenicity of adjuvant-free nano-encapsulated HBsAg, mice were immunized with a single dose of non-encapsulated HBsAg, HBsAg chitosan-NP, or HBsAg alum. Mice immunized with adjuvant-free nanoparticle elicited anti-HBs antibodies at significantly higher titers compared to mice immunized with HBsAg alum. Elevated numbers of BAFF-R(+) B cells and CD138+ plasma cells account for the heightened anti-HBs response in nanoparticle immunized mice. Increases in Tfh cells provide a mechanism for the accumulation of anti-HBs secreting cells. Thus, chitosan nanoparticle vaccines represent a promising un-adjuvanted platform to generate robust and durable immunity to HBsAg and other subunit antigens following a single low-dose administration. FROM THE CLINICAL EDITOR: In this study, chitosan nanoparticle vaccines are demonstrated as a promising un-adjuvanted platform to generate robust and durable immunity to HBsAg and other subunit antigens following a single low-dose administration in a murine model. The authors also demonstrated superior antibody response induction compared with non-encapsulated HBs antigen and HBsAg aluminum.


Asunto(s)
Adyuvantes Inmunológicos/química , Antígenos de Superficie de la Hepatitis B/inmunología , Vacunas contra Hepatitis B/inmunología , Inmunidad/inmunología , Nanopartículas/química , Animales , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Diferenciación Celular , Quitosano/síntesis química , Quitosano/química , Células Dendríticas/metabolismo , Relación Dosis-Respuesta Inmunológica , Femenino , Hepatitis B/sangre , Hepatitis B/inmunología , Hepatitis B/prevención & control , Hepatitis B/virología , Anticuerpos contra la Hepatitis B/sangre , Inmunización , Recuento de Linfocitos , Ratones , Ratones Endogámicos BALB C , Nanopartículas/ultraestructura , Fenotipo
17.
Cancer Res ; 73(8): 2435-44, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23423978

RESUMEN

The extent to which T-cell-mediated immune surveillance is impaired in human cancer remains a question of major importance, given its potential impact on the development of generalized treatments of advanced disease where the highest degree of heterogeneity exists. Here, we report the first global analysis of immune dysfunction in patients with advanced hepatocellular carcinoma (HCC). Using multi-parameter fluorescence-activated cell sorting analysis, we quantified the cumulative frequency of regulatory T cells (Treg), exhausted CD4(+) helper T cells, and myeloid-derived suppressor cells (MDSC) to gain concurrent views on the overall level of immune dysfunction in these inoperable patients. We documented augmented numbers of Tregs, MDSC, PD-1(+)-exhausted T cells, and increased levels of immunosuppressive cytokines in patients with HCC, compared with normal controls, revealing a network of potential mechanisms of immune dysregulation in patients with HCC. In dampening T-cell-mediated antitumor immunity, we hypothesized that these processes may facilitate HCC progression and thwart the efficacy of immunotherapeutic interventions. In testing this hypothesis, we showed that combined regimens to deplete Tregs, MDSC, and PD-1(+) T cells in patients with advanced HCC restored production of granzyme B by CD8(+) T cells, reaching levels observed in normal controls and also modestly increased the number of IFN-γ producing CD4(+) T cells. These clinical findings encourage efforts to restore T-cell function in patients with advanced stage disease by highlighting combined approaches to deplete endogenous suppressor cell populations that can also expand effector T-cell populations.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Neoplasias Hepáticas/inmunología , Células Mieloides/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígeno CTLA-4/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Inmunofenotipificación , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Activación de Linfocitos , Depleción Linfocítica , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Células Mieloides/metabolismo , Fenotipo , Receptor de Muerte Celular Programada 1/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo
18.
J Biol Chem ; 287(19): 15365-70, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22427662

RESUMEN

The anti-inflammatory properties associated with intravenous immunoglobulin therapy require the sialic acid modification of the N-glycan of the Fc domain of IgG. Sialylation of the Fc fragment is mediated by ß-galactoside α2,6-sialyltransferase 1 (ST6Gal-1), acting on the Gal(ß4)GlcNAc terminal structure of the biantennary N-glycans on the Fc domain. However, little is known regarding the in vivo regulation of Fc sialylation and its role in the progression of inflammatory processes. Here, we report that decreased Fc sialylation of circulatory IgG accompanies the acute phase response elicited by turpentine exposure or upon acute exposure to either nontypeable Haemophilus influenzae or ovalbumin. However, Fc sialylation was increased 3-fold from the base line upon transition to chronic inflammation by repeated exposure to challenge. The P1 promoter of the ST6Gal-1 gene is critical for Fc sialylation, but P1 does not drive ST6Gal-1 expression in B cells. The Siat1ΔP1 mouse, with a dysfunctional P1 promoter, was unable to produce sialylated Fc in the systemic circulation, despite the presence of Gal(ß4)GlcNAc termini on the Fc glycans. The major contribution of P1 action is to synthesize ST6Gal-1 enzymes that are deposited into the systemic circulation. The data strongly indicate that this pool of extracellular ST6Gal-1 in the blood impacts the sialylation of IgG Fc and that defective Fc sialylation is likely a major contributing mechanism for the proinflammatory tendencies previously noted in Siat1ΔP1 animals.


Asunto(s)
Antiinflamatorios/metabolismo , Inmunoglobulina G/metabolismo , Regiones Promotoras Genéticas/genética , Sialiltransferasas/metabolismo , Reacción de Fase Aguda/inducido químicamente , Reacción de Fase Aguda/inmunología , Reacción de Fase Aguda/metabolismo , Animales , Antiinflamatorios/inmunología , Western Blotting , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/metabolismo , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido N-Acetilneuramínico/metabolismo , Ovalbúmina/inmunología , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/microbiología , Polisacáridos/metabolismo , Sialiltransferasas/sangre , Sialiltransferasas/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Trementina , beta-D-Galactósido alfa 2-6-Sialiltransferasa
19.
Adv Exp Med Biol ; 780: 125-41, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21842370

RESUMEN

The respiratory mucosa is exposed to the external environment each time we breathe and therefore requires a robust and sophisticated immune defense system. As with other mucosal sites, the respiratory mucosal immune system must balance its response to pathogens while also regulating inflammatory immune cell-mediated tissue damage. In the airways, a failure to tightly control immune responses to a pathogen can result in chronic inflammation and tissue destruction with an overzealous response being deleterious for the host. Chronic obstructive pulmonary disease (COPD) is the fourth most common cause of death in the US and both the prevalence of and mortality rate of this disease is increasing annually. COPD is characterized by intermittent disease exacerbation. The causal contribution of bacterial infections to exacerbations of COPD is now widely accepted, accounting for at least 50% of all exacerbations. Non-typeable Haemophilus influenzae and Moraxella catarrhalis (both gram-negative bacteria) along with Streptococcus pneumoniae (a gram-positive bacterium) are the three most common bacterial pathogens that cause respiratory tract infections in COPD patients. The colonization of bacteria in the lower airways is similar to a low-grade smoldering infection that induces chronic airway inflammation. Chronic low-grade infection can induce a persistent inflammatory response in the airways and parenchyma. Inefficient removal of bacteria from the lower respiratory tract is characteristic of chronic bronchitis. Inflammation is believed to be central to the pathogenesis of exacerbations, but a clear understanding of the inflammatory changes during an exacerbation of COPD has yet to emerge. As bacterial colonization of the lung in COPD patients is a chronic inflammatory condition highlighted by frequent bouts of exacerbation and clearance, we sought to reproduce this chronic pathogen-mediated inflammation in a murine model by repeatedly delivering the intact, whole, live bacteria intra-tracheally to the lungs.


Asunto(s)
Infecciones Bacterianas/inmunología , Bronquios/inmunología , Citocinas/biosíntesis , Inflamación/inmunología , Intubación Intratraqueal/métodos , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Mucosa Respiratoria/inmunología , Animales , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Bronquios/microbiología , Bronquios/patología , Citocinas/análisis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Haemophilus influenzae/fisiología , Humanos , Inflamación/complicaciones , Inflamación/microbiología , Inflamación/patología , Ratones , Moraxella catarrhalis/fisiología , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/microbiología , Índice de Severidad de la Enfermedad , Fumar/efectos adversos , Streptococcus pneumoniae/fisiología , Contaminación por Humo de Tabaco/efectos adversos
20.
Adv Otorhinolaryngol ; 72: 170-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21865721

RESUMEN

In both infants and adults, infections with non-typeable Haemophilus influenzae (NTHI) results in morbidity and mortality. NTHI strains are the leading cause of bacterial otitis media infections (both acute and recurrent) in young children and are also responsible for chronic obstructive pulmonary disease (COPD) exacerbations in current and former smokers. The realization that NTHI causes serious infections in humans has generated interest in the study of the pathogenesis associated with this bacterium and also stimulated considerable efforts towards the evaluation of candidate vaccines that will elicit protective immunity. As NTHI is exclusively a human pathogen and has not been associated with any diseases in other mammals, special efforts have been necessary to establish animal models of NTHI infection to generate useful data on the pathogenesis of infection and efficacy of potential vaccines. This article provides a brief summary of the role of NTHI in disease and the work that has been accomplished by us and several other investigators.


Asunto(s)
Infecciones por Haemophilus , Haemophilus influenzae/inmunología , Otitis Media , Enfermedad Pulmonar Obstructiva Crónica , Animales , Infecciones por Haemophilus/epidemiología , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/microbiología , Humanos , Morbilidad , Otitis Media/epidemiología , Otitis Media/inmunología , Otitis Media/microbiología , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Tasa de Supervivencia/tendencias , Estados Unidos/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA