Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Allergy ; 79(2): 456-470, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38010254

RESUMEN

BACKGROUND: New treatment options with improved safety and novel mechanisms of actions are needed for patients with peanut allergy. OBJECTIVES: To evaluate the safety, tolerability, and immunogenicity of ASP0892, a peanut DNA vaccine, after intradermal (id) or intramuscular (im) administration in adult or adolescent patients with peanut allergy in two phase 1 studies. METHODS: ASP0892 or placebo was administered every 2 weeks for a total of 4 doses. The doses were 1 mg or 4 mg id or 4 mg im for adults, and 1 mg or 4 mg id for adolescents. Immunologic parameters were assessed longitudinally. RESULTS: Thirty-one adults (mean age 24.3 years, 17 males) received ASP0892 (9, 8, 8 patients for 1 mg id, 4 mg id or 4 mg im, respectively) or placebo (2 patients/group). Twenty adolescents (mean age 14.2 years, 11 males) received ASP0892 (8 patients/group) or placebo (2 patients/group). In both studies, the most common treatment-emergent adverse event (TEAE) was injection site pruritus. No deaths or treatment withdrawal were related to TEAEs. No serious TEAEs related to treatment were observed in adult or adolescent patients. ASP0892 treatment led to modest increases in allergen-specific IgG and/or IgG4 in adults (1 mg id, 4 mg im) and adolescents (1 mg id, 4 mg id). No improvements in clinical outcomes, including double-blind placebo-controlled food challenge, were found after ASP0892 treatment. CONCLUSIONS: In two phase 1 studies, ASP0892 was well tolerated with modest but not clinically relevant changes in immune responses. GOV IDENTIFIERS: NCT02851277, NCT03755713.


Asunto(s)
Hipersensibilidad al Cacahuete , Adolescente , Adulto , Humanos , Masculino , Adulto Joven , Arachis , Desensibilización Inmunológica/efectos adversos , Método Doble Ciego , Hipersensibilidad al Cacahuete/diagnóstico , Hipersensibilidad al Cacahuete/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto
2.
Allergy ; 79(2): 485-498, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38112286

RESUMEN

BACKGROUND: Food allergy is a leading cause of anaphylaxis worldwide. Allergen-specific immunotherapy is the only treatment shown to modify the natural history of allergic disease, but application to food allergy has been hindered by risk of severe allergic reactions and short-lived efficacy. Allergen-derived peptides could provide a solution. PVX108 comprises seven short peptides representing immunodominant T-cell epitopes of major peanut allergens for treatment of peanut allergy. METHODS: Pre-clinical safety of PVX108 was assessed using ex vivo basophil activation tests (n = 185). Clinical safety and tolerability of single and repeat PVX108 doses were evaluated in a first-in-human, randomized, double-blind, placebo-controlled trial in peanut-allergic adults (46 active, 21 placebo). The repeat-dose cohort received six doses over 16 weeks with safety monitored to 21 weeks. Exploratory immunological analyses were performed at pre-dose, Week 21 and Month 18 after treatment. RESULTS: PVX108 induced negligible activation of peanut-sensitised basophils. PVX108 was safe and well tolerated in peanut-allergic adults. There were no treatment-related hypersensitivity events or AEs of clinical concern. The only events occurring more frequently in active than placebo were mild injection site reactions. Exploratory immunological analyses revealed a decrease in the ratio of ST2+ Th2A:CCR6+ Th17-like cells within the peanut-reactive Th pool which strengthened following treatment. CONCLUSION: This study supports the concept that PVX108 could provide a safe alternative to whole peanut immunotherapies and provides evidence of durable peanut-specific T-cell modulation. Translation of these findings to clinical efficacy in ongoing Phase 2 trials would provide important proof-of-concept for using peptides to treat food allergy.


Asunto(s)
Anafilaxia , Hipersensibilidad al Cacahuete , Adulto , Humanos , Desensibilización Inmunológica/efectos adversos , Anafilaxia/etiología , Basófilos , Arachis/efectos adversos , Alérgenos , Administración Oral
3.
bioRxiv ; 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-38106196

RESUMEN

Interleukin-2 (IL-2) variants with increased CD25 dependence that selectively expand Foxp3+ regulatory T (TR) cells are in clinical trials for treating inflammatory diseases. Using an Fc-fused IL-2 mutein (Fc.IL-2 mutein) we developed that prevents diabetes in non-obese diabetic (NOD) mice, we show that Fc.IL-2 mutein induced an activated TR population with elevated proliferation, a transcriptional program associated with Stat5- and TCR-dependent gene modules, and high IL-10 and CTLA-4 expression. Increased IL-10 signaling limited surface MHC class II upregulation during conventional dendritic cell (cDC) maturation, while increased CTLA-4-dependent transendocytosis led to the transfer of CD80 and CD86 costimulatory ligands from maturing cDCs to TR cells. In NOD mice, Fc.IL-2 mutein treatment promoted the suppression of cDCs in the inflamed pancreas and pancreatic lymph nodes resulting in T cell anergy. Thus, IL-2 mutein-expanded TR cells have enhanced functional properties and restrict cDC function, offering promise for targeted immunotherapy use in autoimmune disease.

4.
Nat Commun ; 14(1): 7443, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37978185

RESUMEN

The transcriptional and phenotypic characteristics that define alveolar monocyte and macrophage subsets in acute hypoxemic respiratory failure (AHRF) are poorly understood. Here, we apply CITE-seq (single-cell RNA-sequencing and cell-surface protein quantification) to bronchoalveolar lavage and blood specimens longitudinally collected from participants with AHRF to identify alveolar myeloid subsets, and then validate their identity in an external cohort using flow cytometry. We identify alveolar myeloid subsets with transcriptional profiles that differ from other lung diseases as well as several subsets with similar transcriptional profiles as reported in healthy participants (Metallothionein) or patients with COVID-19 (CD163/LGMN). We use information from CITE-seq to determine cell-surface proteins that distinguish transcriptional subsets (CD14, CD163, CD123, CD71, CD48, CD86 and CD44). In the external cohort, we find a higher proportion of CD163/LGMN alveolar macrophages are associated with mortality in AHRF. We report a parsimonious set of cell-surface proteins that distinguish alveolar myeloid subsets using scalable approaches that can be applied to clinical cohorts.


Asunto(s)
Enfermedades Pulmonares , Insuficiencia Respiratoria , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Enfermedades Pulmonares/metabolismo , Insuficiencia Respiratoria/genética
5.
JCI Insight ; 8(21)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37751304

RESUMEN

Variation in the preservation of ß cell function in clinical trials in type 1 diabetes (T1D) has emphasized the need to define biomarkers to predict treatment response. The T1DAL trial targeted T cells with alefacept (LFA-3-Ig) and demonstrated C-peptide preservation in approximately 30% of new-onset T1D individuals. We analyzed islet antigen-reactive (IAR) CD4+ T cells in PBMC samples collected prior to treatment from alefacept- and placebo-treated individuals using flow cytometry and single-cell RNA sequencing. IAR CD4+ T cells at baseline had heterogeneous phenotypes. Transcript profiles formed phenotypic clusters of cells along a trajectory based on increasing maturation and activation, and T cell receptor (TCR) chains showed clonal expansion. Notably, the frequency of IAR CD4+ T cells with a memory phenotype and a unique transcript profile (cluster 3) were inversely correlated with C-peptide preservation in alefacept-treated, but not placebo-treated, individuals. Cluster 3 cells had a proinflammatory phenotype characterized by expression of the transcription factor BHLHE40 and the cytokines GM-CSF and TNF-α, and shared TCR chains with effector memory-like clusters. Our results suggest IAR CD4+ T cells as a potential baseline biomarker of response to therapies targeting the CD2 pathway and warrant investigation for other T cell-related therapies.


Asunto(s)
Diabetes Mellitus Tipo 1 , Humanos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Alefacept/uso terapéutico , Péptido C , Leucocitos Mononucleares/metabolismo , Biomarcadores , Receptores de Antígenos de Linfocitos T/uso terapéutico
6.
J Allergy Clin Immunol ; 152(1): 155-166.e9, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37003475

RESUMEN

BACKGROUND: Despite similar clinical symptoms, peanut-allergic (PA) individuals may respond quite differently to the same therapeutic interventions. OBJECTIVE: This study aimed to determine whether inherent qualities of cell response at baseline could influence response to peanut oral immunotherapy (PnOIT). METHODS: We first performed ex vivo T-cell profiling on peanut-reactive CD154+CD137+ T (pTeff) cells from 90 challenge-confirmed PA individuals. We developed a gating strategy for unbiased assessment of the phenotypic distribution of rare pTeff cells across different memory CD4+ T-cell subsets to define patient immunotype. In longitudinal samples of 29 PA participants enrolled onto the IMPACT trial of PnOIT, we determined whether patient immunotype at baseline could influence response to PnOIT. RESULTS: Our data emphasize the heterogeneity of pTeff cell responses in PA participants with 2 mutually exclusive phenotypic entities (CCR6-CRTH2+ and CCR6+CRTH2-). Our findings lead us to propose that peanut allergy can be classified broadly into at least 2 discrete subtypes, termed immunotypes, with distinct immunologic and clinical characteristics that are based on the proportion of TH2A pTeff cells. PnOIT induced elimination of TH2A pTeff cells in the context of the IMPACT clinical trial. Only 1 PA patient with a low level of TH2A pTeff cells at baseline experienced long-lasting benefit of remission after PnOIT discontinuation. CONCLUSION: Dividing PA patients according to their individual peanut-specific T-cell profile may facilitate patient stratification in clinical settings by identifying which immunotypes might respond best to different therapies.


Asunto(s)
Arachis , Hipersensibilidad al Cacahuete , Humanos , Antígenos , Subgrupos de Linfocitos T , Inmunoterapia , Administración Oral , Alérgenos , Desensibilización Inmunológica
7.
Nat Genet ; 54(5): 603-612, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35513721

RESUMEN

Genome-wide association studies (GWASs) have uncovered hundreds of autoimmune disease-associated loci; however, the causal genetic variants within each locus are mostly unknown. Here, we perform high-throughput allele-specific reporter assays to prioritize disease-associated variants for five autoimmune diseases. By examining variants that both promote allele-specific reporter expression and are located in accessible chromatin, we identify 60 putatively causal variants that enrich for statistically fine-mapped variants by up to 57.8-fold. We introduced the risk allele of a prioritized variant (rs72928038) into a human T cell line and deleted the orthologous sequence in mice, both resulting in reduced BACH2 expression. Naive CD8 T cells from mice containing the deletion had reduced expression of genes that suppress activation and maintain stemness and, upon acute viral infection, displayed greater propensity to become effector T cells. Our results represent an example of an effective approach for prioritizing variants and studying their physiologically relevant effects.


Asunto(s)
Enfermedades Autoinmunes , Estudio de Asociación del Genoma Completo , Alelos , Animales , Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo/métodos , Ratones , Polimorfismo de Nucleótido Simple/genética , Secuencias Reguladoras de Ácidos Nucleicos , Linfocitos T
8.
Allergy ; 77(8): 2534-2548, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35266148

RESUMEN

BACKGROUND: The PALISADE study, an international, phase 3 trial of peanut oral immunotherapy (POIT) with AR101, resulted in desensitization in children and adolescents who were highly allergic to peanut. An improved understanding of the immune mechanism induced in response to food allergen immunotherapy would enable more informed and effective therapeutic strategies. Our main purpose was to examine the immunological changes in blood samples from a subset of peanut-allergic individuals undergoing oral desensitization immunotherapy with AR101. METHODS: Blood samples obtained as part of enrollment screening and at multiple time points during PALISADE study were used to assess basophil and CD4+ T-cell reactivity to peanut. RESULTS: The absence of clinical reactivity to the entry double-blinded placebo-controlled peanut challenge (DBPCFC) was accompanied by a significantly lower basophil sensitivity and T-cell reactivity to peanut compared with DBPCFC reactors. At baseline, peanut-reactive TH2A cells were observed in many but not all peanut-allergic patients and their level in peripheral blood correlates with T-cell reactivity to peanut and with serum peanut-specific IgE and IgG4 levels. POIT reshaped circulating peanut-reactive T-cell responses in a subset-dependent manner. Changes in basophil and T-cell responses to peanut closely paralleled clinical benefits to AR101 therapy and resemble responses in those with lower clinical sensitivity to peanut. However, no difference in peanut-reactive Treg cell frequency was observed between groups. CONCLUSION: Oral desensitization therapy with AR101 leads to decreased basophil sensitivity to peanut and reshapes peanut-reactive T effector cell responses supporting its potential as an immunomodulatory therapy.


Asunto(s)
Hipersensibilidad al Cacahuete , Administración Oral , Adolescente , Alérgenos , Arachis , Niño , Desensibilización Inmunológica/métodos , Humanos , Inmunidad , Hipersensibilidad al Cacahuete/terapia
9.
Immunohorizons ; 6(3): 211-223, 2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35273097

RESUMEN

Successful treatment of inflammatory bowel disease (IBD) with the anti-integrin α4ß7 mAb vedolizumab suggests that interaction of this integrin with addressin mucosal addressin cell adhesion molecule-1 (MAdCAM-1) is central to IBD pathogenesis. Although this was presumed to be due to an inhibition of lymphocyte trafficking to the gut, as has been observed in animal models, we report no depletion of CD4 T cells from the colonic mucosa as a consequence of vedolizumab treatment in humans, regardless of efficacy. Likewise, no upregulation of alternative trafficking mechanisms was observed as a consequence of therapy to suggest that this homeostasis is maintained in patients by a mechanistic escape from inhibition. Instead, we explore a role for MAdCAM-integrin interaction as a gut-specific costimulatory signal, demonstrating that it can replace CD28 ligation to activate human T cells in vitro. This activation through integrin α4ß7 is mediated through the gut-restricted molecule MAdCAM-1, and it cannot be replicated by matrix molecules or proteins that bind other integrins. A detailed analysis of mRNA expression by human T cell subsets following suboptimal TCR stimulation in the presence or absence of CD28 versus MAdCAM-1 costimulation reveals marked similarity in the effect that these two signals have upon T cells, with temporal or quantitative differences detected in the expression of cytokines associated with Th17 cells or pyogenic inflammation. Thus, we describe an alternative costimulatory pathway for T cells in the intestine, through ligation of integrin α4ß7 by MAdCAM-1, which may explain the therapeutic efficacy of vedolizumab and have implications concerning the treatment of IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Integrinas , Animales , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Expresión Génica , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Integrinas/metabolismo
10.
JCI Insight ; 6(22)2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34806648

RESUMEN

Human islet antigen reactive CD4+ memory T cells (IAR T cells) play a key role in the pathogenesis of autoimmune type 1 diabetes (T1D). Using single-cell RNA sequencing (scRNA-Seq) to identify T cell receptors (TCRs) in IAR T cells, we have identified a class of TCRs that share TCRα chains between individuals ("public" chains). We isolated IAR T cells from blood of healthy, new-onset T1D and established T1D donors using multiplexed CD154 enrichment and identified paired TCRαß sequences from 2767 individual cells. More than a quarter of cells shared TCR junctions between 2 or more cells ("expanded"), and 29/47 (~62%) of expanded TCRs tested showed specificity for islet antigen epitopes. Public TCRs sharing TCRα junctions were most prominent in new-onset T1D. Public TCR sequences were more germline like than expanded unique, or "private," TCRs, and had shorter junction sequences, suggestive of fewer random nucleotide insertions. Public TCRα junctions were often paired with mismatched TCRß junctions in TCRs; remarkably, a subset of these TCRs exhibited cross-reactivity toward distinct islet antigen peptides. Our findings demonstrate a prevalent population of IAR T cells with diverse specificities determined by TCRs with restricted TCRα junctions and germline-constrained antigen recognition properties. Since these "innate-like" TCRs differ from previously described immunodominant TCRß chains in autoimmunity, they have implications for fundamental studies of disease mechanisms. Self-reactive restricted TCRα chains and their associated epitopes should be considered in fundamental and translational investigations of TCRs in T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Células Germinativas/metabolismo , Cadenas alfa de Inmunoglobulina/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Adolescente , Adulto , Femenino , Humanos , Masculino , Adulto Joven
11.
Clin Exp Allergy ; 51(12): 1624-1633, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34599624

RESUMEN

BACKGROUND: Characterising the clinical and immunological impact of daily cat exposure in cat-allergic subjects with asthma who live with cats (WC) and those who do not (WoC) may provide understanding of the drivers of the allergic response. METHODS: Clinical and immunological characteristics (skin prick test, spirometry, symptom assessments, immunological markers) were compared between asthmatic subjects WC (n = 10) and WoC (n = 9). RESULTS: WC subjects had greater use of long-acting beta agonists (p < .05) and high-potency corticosteroids. No differences were observed in lung function, nasal and ocular symptoms, or asthma control between the groups. Cat dander- and Fel d 1-specific IgG4 concentrations were higher in WC than WoC subjects (both p < .05). Total IgE and cat dander-, Fel d 1- and Fel d 7-specific IgE concentrations were similar, but Fel d 4-sIgE was higher in WC subjects (p < .05) versus WoC. Basophil sensitivity to cat dander extract and Fel d 1 was lower in WC versus WoC subjects (p < .05) and correlated with higher IgG4 concentrations (r = 0.63; p = .009). Fel d 1-specific CD4+ T-cell responses polarised toward Th2A responses in WC versus WoC subjects; Fel d 1-specific IgE correlated with surface expression of CRTH2 and CD200R (both p ≤ .05). CONCLUSION: Immunological differences observed in WC versus WoC did not reflect clinical tolerance with natural cat exposure. The ability to live with a cat despite allergy could be driven by higher preventative medication use. This study may support design of novel therapeutics for allergy management.


Asunto(s)
Asma , Hipersensibilidad , Alérgenos , Asma/diagnóstico , Glicoproteínas , Humanos , Inmunoglobulina E
12.
Urol Oncol ; 39(4): 240.e1-240.e8, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33602622

RESUMEN

PURPOSE: Surveillance is now the preferred treatment strategy for patients with stage 1A/1B seminoma as reflected by the National Comprehensive Cancer Network guidelines. In this study, we aimed to describe trends in adjuvant management strategy for stage 1A/B seminoma from 2004 to 2016 using the National Cancer Database. MATERIALS AND METHODS: The database was queried for patients diagnosed with stage 1A/1B seminoma between 2004 and 2016. Staging was determined using the American Joint Committee on Cancer guidelines. Surveillance was defined as no treatment with chemotherapy or radiation within 60 days of diagnosis. Proportions of cancer patients utilizing surveillance, radiation, and single-agent chemotherapy were summarized annually. Kaplan-Meier survival analysis was used to compare overall survival between groups. RESULTS: 8,686 patients with stage 1A/1B seminoma met inclusion criteria over the course of the study period. Overall, 3,004 (34.6%) patients began adjuvant chemotherapy or radiation within 60 days. Utilization of surveillance increased from 39.8% in 2004 to 86.8% in 2016 while utilization of radiation decreased from 59.7% to 4.6%. High-volume centers adopted surveillance earlier than low-volume centers. CONCLUSION: This study describes trends in utilization of surveillance, chemotherapy, and radiotherapy for stage 1A/1B seminoma over 12 years. A major shift from utilization of adjuvant treatment to surveillance in patients with stage 1A/B seminoma is observed in this large national cancer database; a minority of patients now receive adjuvant treatment and risk-related toxicities. Survival analysis reveals similar survival at a median 5-year follow-up. The results provide insight into the time needed for clinical practice to adopt the preferred approach of surveillance over the time period studied.


Asunto(s)
Seminoma/terapia , Neoplasias Testiculares/terapia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Estudios Retrospectivos , Seminoma/patología , Neoplasias Testiculares/patología , Terapéutica/tendencias , Adulto Joven
13.
Sci Rep ; 10(1): 15686, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32973217

RESUMEN

Monitoring the frequency of circulatory CXCR5+ (cCXCR5+) CD4+ T cells in periphery blood provides a potential biomarker to draw inferences about T follicular helper (TFH) activity within germinal center. However, cCXCR5+ T cells are highly heterogeneous in their expression of ICOS, PD1 and CD38 and the relationship between different cCXCR5 subsets as delineated by these markers remains unclear. We applied class II tetramer reagents and mass cytometry to investigate the ontogeny of different subsets of cCXCR5+ T cell following yellow fever immunization. Through unsupervised analyses of mass cytometry data, we show yellow fever virus-specific cCXCR5 T cells elicited by vaccination were initially CD38+ICOS+PD1+, but then transitioned to become CD38+ICOS-PD1+ and CD38-ICOS-PD1+ before coming to rest as a CD38-ICOS-PD1- subset. These results imply that most antigen-specific cCXCR5+ T cells, including the CD38-ICOS-PD1- CXCR5+ T cells are derived from the CXCR5+CD38+ICOS+PD1+ subset, the subset that most resembles preTFH/TFH in the germinal center.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunación , Vacuna contra la Fiebre Amarilla/inmunología , Fiebre Amarilla/prevención & control , Linfocitos T CD4-Positivos/metabolismo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Humanos , Receptores CXCR5/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo
14.
Urol Oncol ; 38(7): 641.e1-641.e8, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32307328

RESUMEN

INTRODUCTION: The standard of care (SOC) for primary testicular lymphoma (PTL) is orchiectomy, chemotherapy (CHT), and radiotherapy (RT). We hypothesized that men may not receive SOC and may have worse outcomes. To assess this, we queried the National Cancer Database (NCDB) to analyze treatment patterns and survival in PTL patients. METHODS: The NCDB was queried (2006-2016) for men diagnosed with extranodal lymphoma with primary site testis. Patients were placed in 2 treatment groups (1) orchiectomy with chemotherapy plus radiotherapy (CHT + RT), named the SOC group; and 2) CHT + orchiectomy, or RT + orchiectomy, or orchiectomy alone, grouped as non-SOC. Propensity score matching and Kaplan-Meier analysis were used to investigate 5-year overall survival (OS). RESULTS: Two thousand two hundred thirty-two men with PTL underwent orchiectomy. After exclusions, 891 men were included in the SOC group and 1,006 men were included in the non-SOC group. KM analysis showed 5-year OS was significantly higher in the SOC group vs. non-SOC for all stages (hazard ratio = 0.54, with 95% confidence interval 0.45 to 0.65, P < 0.0001). CONCLUSIONS: This study represents one of the largest PTL cohort reported to date reflecting current treatments and shows men receiving standard of care treatment have significantly improved survival. Additionally, analysis reveals that most men included in the NCDB do not receive the standard of care.


Asunto(s)
Linfoma/tratamiento farmacológico , Linfoma/radioterapia , Neoplasias Testiculares/tratamiento farmacológico , Neoplasias Testiculares/radioterapia , Adolescente , Adulto , Estudios de Cohortes , Humanos , Linfoma/mortalidad , Masculino , Persona de Mediana Edad , Análisis de Supervivencia , Neoplasias Testiculares/mortalidad , Adulto Joven
15.
Immunohorizons ; 4(1): 14-22, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974109

RESUMEN

Mucosal-associated invariant T (MAIT) cells acquire effector function in response to proinflammatory signals, which synergize with TCR-mediated signals. We asked if cell-intrinsic regulatory mechanisms exist to curtail MAIT cell effector function akin to the activation-induced expression of inhibitory receptors by conventional T cells. We examined human MAIT cells from blood and oral mucosal tissues by RNA sequencing and found differential expression of immunoregulatory genes, including CTLA-4, by MAIT cells isolated from tissue. Using an ex vivo experimental setup, we demonstrate that inflammatory cytokines were sufficient to induce CTLA-4 expression on the MAIT cell surface in the absence of TCR signals. Even brief exposure to the cytokines IL-12, IL-15, and IL-18 was sufficient for sustained CTLA-4 expression by MAIT cells. These data suggest that control of CTLA-4 expression is fundamentally different between MAIT cells and conventional T cells. We propose that this mechanism serves to limit MAIT cell-mediated tissue damage.


Asunto(s)
Antígenos de Superficie/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/inmunología , Citocinas/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Sangre/inmunología , Femenino , Expresión Génica/inmunología , Humanos , Inflamación/genética , Masculino , Persona de Mediana Edad , Membrana Mucosa/inmunología , Receptores de Antígenos de Linfocitos T/inmunología
16.
Nat Commun ; 11(1): 219, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31924795

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy has produced remarkable anti-tumor responses in patients with B-cell malignancies. However, clonal kinetics and transcriptional programs that regulate the fate of CAR-T cells after infusion remain poorly understood. Here we perform TCRB sequencing, integration site analysis, and single-cell RNA sequencing (scRNA-seq) to profile CD8+ CAR-T cells from infusion products (IPs) and blood of patients undergoing CD19 CAR-T immunotherapy. TCRB sequencing shows that clonal diversity of CAR-T cells is highest in the IPs and declines following infusion. We observe clones that display distinct patterns of clonal kinetics, making variable contributions to the CAR-T cell pool after infusion. Although integration site does not appear to be a key driver of clonal kinetics, scRNA-seq demonstrates that clones that expand after infusion mainly originate from infused clusters with higher expression of cytotoxicity and proliferation genes. Thus, we uncover transcriptional programs associated with CAR-T cell behavior after infusion.


Asunto(s)
Antígenos CD19/inmunología , Inmunoterapia Adoptiva , Inmunoterapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Selección Clonal Mediada por Antígenos/inmunología , Humanos , Cinética , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Análisis de Secuencia de ARN , Linfocitos T Citotóxicos/inmunología , Transcriptoma
17.
Front Oncol ; 10: 570752, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33520695

RESUMEN

Checkpoint inhibitors (CPIs) increase antitumor activity by unblocking regulators of the immune response. This action can provoke a wide range of immunologic and inflammatory side effects, some of which can be fatal. Recent studies suggest that CPI-induced immune-related adverse events (irAEs) may predict survival and response. However, little is known about the mechanisms of this association. This study was undertaken to evaluate the influence of tumor diagnosis and preexisting clinical factors on the types of irAEs experienced by cancer patients treated with CPIs. The correlation between irAEs and overall survival (OS) was also assessed. All cancer patients treated with atezolizumab (ATEZO), ipilimumab (IPI), nivolumab (NIVO), or pembrolizumab (PEMBRO) at Virginia Mason Medical Center between 2011 and 2019 were evaluated. irAEs were graded according to the Common Terminology Criteria for Adverse Events (Version 5) and verified independently. Statistical analyses were performed to assess associations between irAEs, pre-treatment factors, and OS. Of the 288 patients evaluated, 59% developed irAEs of any grade, and 19% developed irAEs of grade 3 or 4. A time-dependent survival analysis demonstrated a clear association between the occurrence of irAEs and OS (P < 0.001). A 6-week landmark analysis adjusted for body mass index confirmed an association between irAEs and OS in non-Small Cell Lung Cancer (NSCLC) (P < 0.03). An association between melanoma and skin irAEs (P < 0.01) and between NSCLC and respiratory irAEs (P = 0.03) was observed, independent of CPI administered. Patients with preexisting autoimmune disease experienced a higher incidence of severe irAEs (P = 0.01), but not a higher overall incidence of irAEs (P = 0.6). A significant association between irAEs and OS was observed in this diverse patient population. No correlation was observed between preexisting comorbid conditions and the type of irAE observed. However, a correlation between skin-related irAEs and melanoma and between respiratory irAEs and NSCLC was observed, suggesting that many irAEs are driven by a specific response to the primary tumor. In patients with NSCLC, the respiratory irAEs were associated with a survival benefit.

18.
J Clin Invest ; 130(1): 480-490, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31815738

RESUMEN

Although most patients with type 1 diabetes (T1D) retain some functional insulin-producing islet ß cells at the time of diagnosis, the rate of further ß cell loss varies across individuals. It is not clear what drives this differential progression rate. CD8+ T cells have been implicated in the autoimmune destruction of ß cells. Here, we addressed whether the phenotype and function of autoreactive CD8+ T cells influence disease progression. We identified islet-specific CD8+ T cells using high-content, single-cell mass cytometry in combination with peptide-loaded MHC tetramer staining. We applied a new analytical method, DISCOV-R, to characterize these rare subsets. Autoreactive T cells were phenotypically heterogeneous, and their phenotype differed by rate of disease progression. Activated islet-specific CD8+ memory T cells were prevalent in subjects with T1D who experienced rapid loss of C-peptide; in contrast, slow disease progression was associated with an exhaustion-like profile, with expression of multiple inhibitory receptors, limited cytokine production, and reduced proliferative capacity. This relationship between properties of autoreactive CD8+ T cells and the rate of T1D disease progression after onset make these phenotypes attractive putative biomarkers of disease trajectory and treatment response and reveal potential targets for therapeutic intervention.


Asunto(s)
Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Memoria Inmunológica , Islotes Pancreáticos/inmunología , Activación de Linfocitos , Adolescente , Adulto , Linfocitos T CD8-positivos/patología , Niño , Preescolar , Diabetes Mellitus Tipo 1/patología , Femenino , Humanos , Lactante , Islotes Pancreáticos/patología , Masculino , Persona de Mediana Edad
19.
Sci Transl Med ; 11(521)2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801887

RESUMEN

CCR5 is thought to play a central role in orchestrating migration of cells in response to inflammation. CCR5 antagonists can reduce inflammatory disease processes, which has led to an increased interest in using CCR5 antagonists in a wide range of inflammation-driven diseases. Paradoxically, these antagonists appear to function without negatively affecting host immunity at barrier sites. We reasoned that the resolution to this paradox may lie in the CCR5+ T cell populations that permanently reside in tissues. We used a single-cell analysis approach to examine the human CCR5+ T cell compartment in the blood, healthy, and inflamed mucosal tissues to resolve these seemingly contradictory observations. We found that 65% of the CD4 tissue-resident memory T (TRM) cell compartment expressed CCR5. These CCR5+ TRM cells were enriched in and near the epithelial layer and not only limited to TH1-type cells but also contained a large TH17-producing and a stable regulatory T cell population. The CCR5+ TRM compartment was stably maintained even in inflamed tissues including the preservation of TH17 and regulatory T cell populations. Further, using tissues from the CHARM-03 clinical trial, we found that CCR5+ TRM are preserved in human mucosal tissue during treatment with the CCR5 antagonist Maraviroc. Our data suggest that the human CCR5+ TRM compartment is functionally and spatially equipped to maintain barrier immunity even in the absence of CCR5-mediated, de novo T cell recruitment from the periphery.


Asunto(s)
Compartimento Celular , Inflamación/inmunología , Receptores CCR5/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Compartimento Celular/efectos de los fármacos , Citocinas/biosíntesis , Femenino , Humanos , Lectinas Tipo C/metabolismo , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Masculino , Maraviroc/farmacología , Persona de Mediana Edad , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/inmunología , Mucosa Bucal/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Células Th17/efectos de los fármacos , Células Th17/inmunología , Transcriptoma/genética , Adulto Joven
20.
PLoS One ; 13(1): e0192082, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29377961

RESUMEN

Globally, diarrheal diseases are a leading cause of death in children under five and disproportionately affect children in developing countries. Children who contract diarrheal diseases are rarely screened to identify the etiologic agent due to time and cost considerations associated with pathogen-specific screening and hence pathogen-directed therapy is uncommon. The development of biomarkers to rapidly identify underlying pathogens could improve treatment options and clinical outcomes in childhood diarrheal diseases. Here, we perform RNA sequencing on blood samples collected from children evaluated in an emergency room setting with diarrheal disease where the pathogen(s) present are known. We determine host response gene signatures specific to Salmonella, Shigella and rotavirus, but not E. coli, infections that distinguish them from each other and from healthy controls. Specifically, we observed differential expression of genes related to chemokine receptors or inflammasome signaling in Shigella cases, such as CCR3, CXCR8, and NLRC4, and interferon response genes, such as IFI44 and OASL, in rotavirus cases. Our findings add insight into the host peripheral immune response to these pathogens, and suggest strategies and limitations for the use host response transcript signatures for diagnosing the etiologic agent of childhood diarrheal diseases.


Asunto(s)
Diarrea/inmunología , Perfilación de la Expresión Génica , ARN Mensajero/sangre , Niño , Diarrea/sangre , Diarrea/genética , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/microbiología , Humanos , Rotavirus/aislamiento & purificación , Shigella/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...