Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cancer Cell ; 42(1): 35-51.e8, 2024 01 08.
Article in English | MEDLINE | ID: mdl-38134936

ABSTRACT

Chimeric antigen receptor T cells (CAR-Ts) have remarkable efficacy in liquid tumors, but limited responses in solid tumors. We conducted a Phase I trial (NCT02107963) of GD2 CAR-Ts (GD2-CAR.OX40.28.z.iC9), demonstrating feasibility and safety of administration in children and young adults with osteosarcoma and neuroblastoma. Since CAR-T efficacy requires adequate CAR-T expansion, patients were grouped into good or poor expanders across dose levels. Patient samples were evaluated by multi-dimensional proteomic, transcriptomic, and epigenetic analyses. T cell assessments identified naive T cells in pre-treatment apheresis associated with good expansion, and exhausted T cells in CAR-T products with poor expansion. Myeloid cell assessment identified CXCR3+ monocytes in pre-treatment apheresis associated with good expansion. Longitudinal analysis of post-treatment samples identified increased CXCR3- classical monocytes in all groups as CAR-T numbers waned. Together, our data uncover mediators of CAR-T biology and correlates of expansion that could be utilized to advance immunotherapies for solid tumor patients.


Subject(s)
Neuroblastoma , Receptors, Chimeric Antigen , Child , Young Adult , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Antigen, T-Cell/genetics , Proteomics , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , T-Lymphocytes , Neuroblastoma/pathology , Cell- and Tissue-Based Therapy
2.
Clin Cancer Res ; 28(10): 2069-2081, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35046061

ABSTRACT

PURPOSE: Improving our understanding of the immunologic response to cancer cells within the sentinel lymph nodes (SLN) of primary tumors is expected to identify new approaches to stimulate clinically meaningful cancer immunity. EXPERIMENTAL DESIGN: We used mass cytometry by time-of-flight (CyTOF), flow cytometry, and T-cell receptor immunosequencing to conduct simultaneous single-cell analyses of immune cells in the SLNs of patients with melanoma. RESULTS: We found increased effector-memory αß T cells, TCR clonality, and γδ T cells selectively in the melanoma-bearing SLNs relative to non-melanoma-bearing SLNs, consistent with possible activation of an antitumor immune response. However, we also observed a markedly immunotolerant environment in the melanoma-bearing SLNs indicated by reduced and impaired NK cells and increased levels of CD8+CD57+PD-1+ cells, which are known to display low melanoma killing capabilities. Other changes observed in melanoma-bearing SLNs when compared with non-melanoma-bearing SLNs include (i) reduced CD8+CD69+ T cell/T regulatory cell ratio, (ii) high PD-1 expression on CD4+ and CD8+ T cells, and (iii) high CTLA-4 expression on γδ T cells. CONCLUSIONS: Our data suggest that these immunologic changes compromise antimelanoma immunity and contribute to a high relapse rate. We propose the development of clinical trials to test the neo-adjuvant administration of anti-PD-1 antibodies prior to SLN resection in patients with stage III melanoma. See related commentary by Lund, p. 1996.


Subject(s)
Melanoma , Sentinel Lymph Node , Skin Neoplasms , Humans , Immune Tolerance , Melanoma/pathology , Programmed Cell Death 1 Receptor/therapeutic use , Sentinel Lymph Node/pathology , Sentinel Lymph Node Biopsy , Skin Neoplasms/pathology , Tumor Microenvironment
3.
Gastroenterology ; 161(6): 2014-2029.e14, 2021 12.
Article in English | MEDLINE | ID: mdl-34450180

ABSTRACT

BACKGROUND AND AIMS: Acute pancreatitis (AP) is an inflammatory disease with mild to severe course that is associated with local and systemic complications and significant mortality. Uncovering inflammatory pathways that lead to progression and recovery will inform ways to monitor and/or develop effective therapies. METHODS: We performed single-cell mass Cytometry by Time Of Flight (CyTOF) analysis to identify pancreatic and systemic inflammatory signals during mild AP (referred to as AP), severe AP (SAP), and recovery using 2 independent experimental models and blood from patients with AP and recurrent AP. Flow cytometric validation of monocytes subsets identified using CyTOF analysis was performed independently. RESULTS: Ly6C+ inflammatory monocytes were the most altered cells in the pancreas during experimental AP, recovery, and SAP. Deep profiling uncovered heterogeneity among pancreatic and blood monocytes and identified 7 novel subsets during AP and recovery, and 6 monocyte subsets during SAP. Notably, a dynamic shift in pancreatic CD206+ macrophage population was observed during AP and recovery. Deeper profiling of the CD206+ macrophage identified 7 novel subsets during AP, recovery, and SAP. Differential expression analysis of these novel monocyte and CD206+ macrophage subsets revealed significantly altered surface (CD44, CD54, CD115, CD140a, CD196, podoplanin) and functional markers (interferon-γ, interleukin 4, interleukin 22, latency associated peptide-transforming growth factor-ß, tumor necrosis factor-α, T-bet, RoRγt) that were associated with recovery and SAP. Moreover, a targeted functional analysis further revealed distinct expression of pro- and anti-inflammatory cytokines by pancreatic CD206+ macrophage subsets as the disease either progressed or resolved. Similarly, we identified heterogeneity among circulating classical inflammatory monocytes (CD14+CD16-) and novel subsets in patients with AP and recurrent AP. CONCLUSIONS: We identified several novel monocyte/macrophage subsets with unique phenotype and functional characteristics that are associated with AP, recovery, and SAP. Our findings highlight differential innate immune responses during AP progression and recovery that can be leveraged for future disease monitoring and targeting.


Subject(s)
Immunity, Innate , Macrophages/immunology , Monocytes/immunology , Pancreas/immunology , Pancreatitis/immunology , Animals , Biomarkers/blood , Cell Separation , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunophenotyping , Macrophages/metabolism , Mice, Inbred BALB C , Monocytes/metabolism , Pancreas/metabolism , Pancreatitis/blood , Pancreatitis/diagnosis , Phenotype , Recovery of Function , Severity of Illness Index , Time Factors
4.
Methods Mol Biol ; 2285: 49-63, 2021.
Article in English | MEDLINE | ID: mdl-33928542

ABSTRACT

CD4+ T cells or helper T cells play various roles in the immune response to pathogens, tumors, as well as in asthma, allergy, and autoimmunity. Consequently, there is great interest in the comprehensive investigation of different T helper cell subsets. Here, we use mass cytometry (CyTOF), which is similar to flow cytometry but uses metal ion-tagged antibodies, which are detected using time-of-flight mass spectrometry. CyTOF allows the simultaneous detection of over 40 different antibodies, allowing us to collect high-dimensional single-cell proteomic data on T helper subsets. We use an extensive staining panel with a large number of lineage markers, cytokines, and other functional markers to identify and characterize CD4+ T cell subsets. In this method, human peripheral blood mononuclear cells are stimulated ex vivo with PMA and ionomycin, which activates T cells. The activated CD4+ T cells can then be identified as Th1, Th2, or Th17 cells based on their production of IFNγ, IL-4, and IL-17, respectively. Tregs are identified as CD4+CD25+CD127lo. Once Th1, Th2, Th17, and Tregs have been identified, they can be characterized in more detail using the large number of phenotypic and functional markers included in the CyTOF staining panel. Finally, automated and unbiased high-dimensional data analysis tools can be employed to comprehensively characterize T helper cells and discover novel features.


Subject(s)
Cell Separation , Flow Cytometry , Mass Spectrometry , T-Lymphocytes, Helper-Inducer/immunology , Animals , Biomarkers/metabolism , Humans , Immunophenotyping , Phenotype , Research Design , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Workflow
5.
Immunohorizons ; 4(12): 774-788, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33310880

ABSTRACT

The antiviral response to influenza virus is complex and multifaceted, involving many immune cell subsets. There is an urgent need to understand the role of CD4+ T cells, which orchestrate an effective antiviral response, to improve vaccine design strategies. In this study, we analyzed PBMCs from human participants immunized with influenza vaccine, using high-dimensional single-cell proteomic immune profiling by mass cytometry. Data were analyzed using a novel clustering algorithm, denoised ragged pruning, to define possible influenza virus-specific clusters of CD4+ T cells. Denoised ragged pruning identified six clusters of cells. Among these, one cluster (Cluster 3) was found to increase in abundance following stimulation with influenza virus peptide ex vivo. A separate cluster (Cluster 4) was found to expand in abundance between days 0 and 7 postvaccination, indicating that it is vaccine responsive. We examined the expression profiles of all six clusters to characterize their lineage, functionality, and possible role in the response to influenza vaccine. Clusters 3 and 4 consisted of effector memory cells, with high CD154 expression. Cluster 3 expressed cytokines like IL-2, IFN-γ, and TNF-α, whereas Cluster 4 expressed IL-17. Interestingly, some participants had low abundance of Clusters 3 and 4, whereas others had higher abundance of one of these clusters compared with the other. Taken together, we present an approach for identifying novel influenza virus-reactive CD4+ T cell subsets, a method that could help advance understanding of the immune response to influenza, predict responsiveness to vaccines, and aid in better vaccine design.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Influenza Vaccines/immunology , Influenza, Human/prevention & control , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD/analysis , Child , Cluster Analysis , Cytokines/metabolism , Female , Flow Cytometry , Humans , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Male , Middle Aged , Young Adult
6.
Clin Cancer Res ; 26(13): 3193-3201, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32205463

ABSTRACT

PURPOSE: Preclinical data suggest that radiotherapy (RT) is beneficial in combination with immune checkpoint blockade. Clinical trials have explored RT with single-agent immune checkpoint blockade, but no trials have reported RT with the combination of nivolumab and ipilimumab. PATIENTS AND METHODS: We conducted a phase 1 study of patients with stage IV melanoma receiving nivolumab and ipilimumab with two different dose-fractionation schemes of RT. Patients had at least one melanoma metastasis that would benefit from palliative RT and one metastasis that would not be irradiated. Nivolumab 1 mg/kg + ipilimumab 3 mg/kg and extracranial RT with a dose of 30 Gy in 10 fractions was administered in Cohort A, and then 27 Gy in 3 fractions was administered in Cohort B. The primary outcome was safety. RESULTS: Twenty patients were treated (10 in each cohort). The rates of treatment-related grade 3-4 adverse events in Cohort A and B were 40% and 30%, respectively. There were no grade ≥3 adverse events attributed to RT. Patients responded to treatment outside of the irradiated volume (Cohort A 5/10; Cohort B 1/9). No evaluable patients had progression of irradiated metastases. Immunologic changes were seen in the peripheral blood with increases in T-cell receptor diversity in some responding patients. CONCLUSIONS: RT with nivolumab and ipilimumab was safe compared with historical data of nivolumab and ipilimumab alone. Immunologic effects were observed in the peripheral blood. Randomized studies are ongoing to assess whether RT increases the efficacy of nivolumab and ipilimumab.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/pathology , Melanoma/therapy , Neoadjuvant Therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Chemoradiotherapy , Female , Humans , Ipilimumab/administration & dosage , Male , Melanoma/etiology , Melanoma/mortality , Middle Aged , Neoadjuvant Therapy/methods , Neoplasm Metastasis , Neoplasm Staging , Nivolumab/administration & dosage , Prognosis , Treatment Outcome
7.
J Clin Oncol ; 38(1): 20-28, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31532724

ABSTRACT

PURPOSE: To assess the efficacy of pembrolizumab in patients with advanced relapsed or refractory mycosis fungoides (MF) or Sézary syndrome (SS). PATIENTS AND METHODS: CITN-10 is a single-arm, multicenter phase II trial of 24 patients with advanced MF or SS. Patients were treated with pembrolizumab 2 mg/kg every 3 weeks for up to 24 months. The primary end point was overall response rate by consensus global response criteria. RESULTS: Patients had advanced-stage disease (23 of 24 with stage IIB to IV MF/SS) and were heavily pretreated with a median of four prior systemic therapies. The overall response rate was 38% with two complete responses and seven partial responses. Of the nine responding patients, six had 90% or more improvement in skin disease by modified Severity Weighted Assessment Tool, and eight had ongoing responses at last follow-up. The median duration of response was not reached, with a median response follow-up time of 58 weeks. Immune-related adverse events led to treatment discontinuation in four patients. A transient worsening of erythroderma and pruritus occurred in 53% of patients with SS. This cutaneous flare reaction did not result in treatment discontinuation for any patient. The flare reaction correlated with high PD-1 expression on Sézary cells but did not associate with subsequent clinical responses or lack of response. Treatment responses did not correlate with expression of PD-L1, total mutation burden, or an interferon-γ gene expression signature. CONCLUSION: Pembrolizumab demonstrated significant antitumor activity with durable responses and a favorable safety profile in patients with advanced MF/SS.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Mycosis Fungoides/drug therapy , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , Biomarkers, Tumor/metabolism , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Mycosis Fungoides/immunology , Mycosis Fungoides/metabolism , Mycosis Fungoides/pathology , Neoplasm Staging , Recurrence , Sezary Syndrome/immunology , Sezary Syndrome/metabolism , Sezary Syndrome/pathology , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
8.
Viral Immunol ; 32(2): 102-109, 2019 03.
Article in English | MEDLINE | ID: mdl-30698511

ABSTRACT

An immune cell's phenotype expresses through its high-dimensional marker signature. Cluster analyses of data from high-throughput mass and flow cytometry marker panels permit discovery of previously undescribed immune cell phenotypes. Impactful reporting of new phenotypes demands low-dimensional visualization tools that preserve with integrity phenotypes' original high-dimensional structure. For this purpose, we introduce penalized supervised star plots. As designed and as we demonstrate, penalized supervised star plots are two-dimensional projections that tend to preserve separation of clusters as well as information on the relative contributions of various markers in differentiating phenotypes. The new method is robust to markers that do not differentiate phenotypes at all, as shown in a challenge data set. Results include comparison with other popular procedures. Penalized supervised star plots incorporate cross-validation to permit portability of estimated optimal projections to new samples. Supervised star plots are further illustrated with a featured influenza-specific T cell data set as well as a peripheral blood mononuclear cell phenotyping data set.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Computational Biology/methods , Immunophenotyping/methods , Influenza, Human/immunology , Leukocytes, Mononuclear/immunology , Algorithms , Biomarkers , Flow Cytometry , Humans
9.
J Immunother Cancer ; 6(1): 18, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29510697

ABSTRACT

BACKGROUND: While immune checkpoint blockade has greatly improved clinical outcomes in diseases such as melanoma, there remains a need for predictive biomarkers to determine who will likely benefit most from which therapy. To date, most biomarkers of response have been identified in the tumors themselves. Biomarkers that could be assessed from peripheral blood would be even more desirable, because of ease of access and reproducibility of sampling. METHODS: We used mass cytometry (CyTOF) to comprehensively profile peripheral blood of melanoma patients, in order to find predictive biomarkers of response to anti-CTLA-4 or anti-PD-1 therapy. Using a panel of ~ 40 surface and intracellular markers, we performed in-depth phenotypic and functional immune profiling to identify potential predictive biomarker candidates. RESULTS: Immune profiling of baseline peripheral blood samples using CyTOF revealed that anti-CTLA-4 and anti-PD-1 therapies have distinct sets of candidate biomarkers. The distribution of CD4+ and CD8+ memory/non-memory cells and other memory subsets was different between responders and non-responders to anti-CTLA-4 therapy. In anti-PD-1 (but not anti-CTLA-4) treated patients, we discovered differences in CD69 and MIP-1ß expressing NK cells between responders and non-responders. Finally, multivariate analysis was used to develop a model for the prediction of response. CONCLUSIONS: Our results indicate that anti-CTLA-4 and anti-PD-1 have distinct predictive biomarker candidates. CD4+ and CD8+ memory T cell subsets play an important role in response to anti-CTLA-4, and are potential biomarker candidates. For anti-PD-1 therapy, NK cell subsets (but not memory T cell subsets) correlated with clinical response to therapy. These functionally active NK cell subsets likely play a critical role in the anti-tumor response triggered by anti-PD-1.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , Killer Cells, Natural/immunology , Melanoma/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/immunology , Adult , Aged , Biomarkers/blood , Female , Humans , Immunotherapy , Male , Melanoma/blood , Melanoma/drug therapy , Middle Aged , Skin Neoplasms/blood , Skin Neoplasms/drug therapy , Treatment Outcome
10.
Curr Protoc Cytom ; 82: 9.54.1-9.54.12, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28967988

ABSTRACT

The central role of the immune system is becoming appreciated in a wide variety of diseases. Cancer immunotherapy is one area that has yielded much recent success, although not all patients benefit equally. At the same time, recent studies have highlighted the heterogeneity of the human immune system. Despite this heterogeneity, we do not routinely measure immune competence in clinical practice, and there are no consensus assays of healthy immune function. Using mass cytometry (CyTOF), we can simultaneously detect ∼40 markers to identify various cell subsets and determine their function by the expression of cytokines, cytotoxicity, and activation markers. This can help assess 'immunocompetence' and facilitate better implementation of immunotherapies, both in specific disease settings and perhaps eventually as a prognostic tool in healthy subjects. Here we introduce the concepts behind this assay and provide a protocol that we have successfully implemented to identify possible predictive biomarkers of immunotherapy outcome. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Antigens, Differentiation , Cytokines , Flow Cytometry/methods , Immunotherapy , Animals , Antigens, Differentiation/blood , Antigens, Differentiation/immunology , Cytokines/blood , Cytokines/immunology , Humans
11.
Int J Radiat Oncol Biol Phys ; 96(3): 578-88, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27681753

ABSTRACT

PURPOSE: Local radiation therapy (RT) combined with systemic anti-cytotoxic T-lymphocyte-associated protein-4 immunotherapy may enhance induction of systemic antimelanoma immune responses. The primary objective of the present trial was to assess the safety and efficacy of combining ipilimumab with RT in patients with stage IV melanoma. The secondary objectives included laboratory assessment of induction of antimelanoma immune responses. METHODS AND MATERIALS: In our prospective clinical trial, 22 patients with stage IV melanoma were treated with palliative RT and ipilimumab for 4 cycles. RT to 1 to 2 disease sites was initiated within 5 days after starting ipilimumab. Patients had ≥1 nonirradiated metastasis measuring ≥1.5 cm available for response assessment. Tumor imaging studies were obtained at baseline, 2 to 4 weeks after cycle 4 of ipilimumab, and every 3 months until progression. Laboratory immune response parameters were measured before and during treatment. RESULTS: Combination therapy was well-tolerated without unexpected toxicities. Eleven patients (50.0%) experienced clinical benefit from therapy, including complete and partial responses and stable disease at median follow-up of 55 weeks. Three patients (27.3%) achieved an ongoing systemic complete response at a median follow-up of 55 weeks (range 32-65), and 3 (27.3%) had an initial partial response for a median of 40 weeks. Analysis of immune response data suggested a relationship between elevated CD8-activated T-cells and response. CONCLUSION: This is the second prospective clinical trial of treatment of metastatic melanoma using the combination of RT and systemic immunotherapy and the first using this sequence of therapy. The results from the present trial demonstrate that a subset of patients may benefit from combination therapy, arguing for continued clinical investigation of the use of RT combined with immunotherapy, including programmed cell death 1 inhibitors, which might have the potential to be even more effective in combination with RT.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Melanoma/radiotherapy , Melanoma/secondary , Radioimmunotherapy/methods , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Ipilimumab , Male , Melanoma/diagnosis , Middle Aged , Prospective Studies , Treatment Outcome , Young Adult
12.
Pathog Dis ; 74(6)2016 08.
Article in English | MEDLINE | ID: mdl-27354294

ABSTRACT

Sphingosine 1-phosphate (S1P) is a sphingosine containing lipid intermediate obtained from ceramide. S1P is known to be an important signaling molecule and plays multiple roles in the context of immunity. This lysophospholipid binds and activates G-protein-coupled receptors (GPCRs) known as S1P receptors 1-5 (S1P1-5). Once activated, these GPCRs mediate signaling that can lead to alterations in cell proliferation, survival or migration, and can also have other effects such as promoting angiogenesis. In this review, we will present evidence demonstrating a role for S1P in lymphocyte migration, inflammation and infection, as well as in cancer. The therapeutic potential of targeting S1P receptors, kinases and lyase will also be discussed.


Subject(s)
Chemotaxis, Leukocyte/immunology , Infections/etiology , Infections/metabolism , Inflammation/etiology , Inflammation/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Cytokines/metabolism , Disease Models, Animal , Fingolimod Hydrochloride/pharmacology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Host-Pathogen Interactions , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunosuppressive Agents/pharmacology , Signal Transduction/drug effects , Sphingosine/metabolism , Transcription Factors/metabolism
13.
Front Microbiol ; 6: 262, 2015.
Article in English | MEDLINE | ID: mdl-25904903

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) is a leading cause of childhood renal disease Hemolytic Uremic Syndrome (HUS). The involvement of renal cytokines and chemokines is suspected to play a critical role in disease progression. In current article, we tested the hypothesis that NKT cells are involved in Stx2-induced pathology in vivo. To address this hypothesis we compared Stx2 toxicity in WT and CD1 knockout (KO) mice. In CD1KO mice, which lack natural killer T (NKT) cells, Stx2-induced pathologies such as weight loss, renal failure, and death were delayed. In WT mice, Stx2-specific selective increase in urinary albumin occurs in later time points, and this was also delayed in NKT cell deficient mice. NKT cell-associated cytokines such as IL-2, IL-4, IFN-γ, and IL-17 were detected in kidney lysates of Stx2-injected WT mice with the peak around 36 h after Stx2 injection. In CD1KO, there was a delay in the kinetics, and increases in these cytokines were observed 60 h post Stx2 injection. These data suggest that NKT cells accelerate Stx2-induced pathology in mouse kidneys. To determine the mechanism by which NKT cells promote Stx2-associated disease, in vitro studies were performed using murine renal cells. We found that murine glomerular endothelial cells and podocytes express functional CD1d molecules and can present exogenous antigen to NKT cells. Moreover, we observed the direct interaction between Stx2 and the receptor Gb3 on the surface of mouse renal cells by 3D STORM-TIRF which provides single molecule imaging. Collectively, these data suggest that Stx2 binds to Gb3 on renal cells and leads to aberrant CD1d-mediated NKT cell activation. Therefore, strategies targeting NKT cells could have a significant impact on Stx2-associated renal pathology in STEC disease.

14.
J Immunol ; 193(5): 2096-105, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25070854

ABSTRACT

NKT cells are a unique subset of T cells that recognize glycolipid Ags presented in the context of CD1d molecules. NKT cells mount strong antitumor responses and are a major focus in developing effective cancer immunotherapy. It is known that CD1d molecules are constantly internalized from the cell surface, recycled through the endocytic compartments, and re-expressed on the cell surface. However, little is known about the regulation of CD1d-mediated Ag processing and presentation in B cell lymphoma. Prosurvival factors of the Bcl-2 family, such as Bcl-xL, are often upregulated in B cell lymphomas and are intimately linked to sphingolipid metabolism, as well as the endocytic compartments. We hypothesized that Bcl-xL can regulate CD1d-mediated Ag presentation to NKT cells. We found that overexpression or induction of Bcl-xL led to increased Ag presentation to NKT cells. Conversely, the inhibition or knockdown of Bcl-xL led to decreased NKT cell activation. Furthermore, knockdown of Bcl-xL resulted in the loss of CD1d trafficking to lysosome-associated membrane protein 1(+) compartments. Rab7, a late endosomal protein, was upregulated and CD1d molecules accumulated in the Rab7(+) late endosomal compartment. These results demonstrate that Bcl-xL regulates CD1d-mediated Ag processing and presentation to NKT cells by altering the late endosomal compartment and changing the intracellular localization of CD1d.


Subject(s)
Antigen Presentation/physiology , Antigens, CD1d/immunology , Endocytosis/immunology , Lymphocyte Activation/physiology , Natural Killer T-Cells/immunology , bcl-X Protein/immunology , Animals , Antigens, CD1d/genetics , Cell Line , Female , Humans , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/immunology , Mice , Natural Killer T-Cells/pathology , Protein Transport/physiology , Up-Regulation/physiology , bcl-X Protein/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/immunology , rab7 GTP-Binding Proteins
15.
Med Sci (Basel) ; 2(2): 82-97, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24955247

ABSTRACT

Natural killer T (NKT) cells are a unique subset of CD1d-restricted T lymphocytes that express characteristics of both T cells and natural killer cells. NKT cells mediate tumor immune-surveillance; however, NKT cells are numerically reduced and functionally impaired in lymphoma patients. Many hematologic malignancies express CD1d molecules and co-stimulatory proteins needed to induce anti-tumor immunity by NKT cells, yet most tumors are poorly immunogenic. In this study, we sought to investigate NKT cell responses to B cell lymphoma. In the presence of exogenous antigen, both mouse and human NKT cell lines produce cytokines following stimulation by B cell lymphoma lines. NKT cell populations were examined ex vivo in mouse models of spontaneous B cell lymphoma, and it was found that during early stages, NKT cell responses were enhanced in lymphoma-bearing animals compared to disease-free animals. In contrast, in lymphoma-bearing animals with splenomegaly and lymphadenopathy, NKT cells were functionally impaired. In a mouse model of blastoid variant mantle cell lymphoma, treatment of tumor-bearing mice with a potent NKT cell agonist, α-galactosylceramide (α-GalCer), resulted in a significant decrease in disease pathology. Ex vivo studies demonstrated that NKT cells from α-GalCer treated mice produced IFN-γ following α-GalCer restimulation, unlike NKT cells from vehicle-control treated mice. These data demonstrate an important role for NKT cells in the immune response to an aggressive hematologic malignancy like mantle cell lymphoma.

16.
J Interferon Cytokine Res ; 32(11): 505-16, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23050947

ABSTRACT

Natural killer T (NKT) cells constitute an important subset of T cells that can both directly and indirectly mediate antitumor immunity. However, we and others have reported that cancer patients have a reduction in both NKT cell number and function. NKT cells can be stimulated and expanded with α-GalCer and cytokines and these expanded NKT cells retain their phenotype, remain responsive to antigenic stimulation, and display cytotoxic function against tumor cell lines. These data strongly favor the use of ex vivo expanded NKT cells in adoptive immunotherapy. NKT cell based-immunotherapy has been limited by the use of autologous antigen-presenting cells, which can vary substantially in their quantity and quality. A standardized system that relies on artificial antigen-presenting cells (aAPCs) could produce the stimulating effects of dendritic cell (DC) without the pitfalls of allo- or xenogeneic cells. In this review, we discuss the progress that has been made using CD1d-based aAPC and how this acellular antigen presenting system can be used in the future to enhance our understanding of NKT cell biology and to develop NKT cell-specific adoptive immunotherapeutic strategies.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , Immunotherapy, Adoptive , Natural Killer T-Cells/immunology , Neoplasms/immunology , Neoplasms/therapy , Antigens, CD1d/metabolism , Cell Line, Tumor/immunology , Cytokines/metabolism , Galactosylceramides/metabolism , Humans , Lymphocyte Activation , Lymphocyte Count
17.
J Vaccines Vaccin ; 3: 144, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-24089657

ABSTRACT

Natural killer T (NKT) cells play an important immunoregulatory role and are thought to bridge the innate and adaptive immune responses. Following activation through cognate interactions with lipid antigen presented in the context of CD1d molecules, NKT cells rapidly produce a plethora of cytokines and can also mediate cytotoxicity. Due to their potent effector functions, extensive research has been performed to increase our understanding on how to effectively modulate these cells. In fact, NKT cell agonists have been used as vaccine adjuvants to enhance antigen specific T and B cell responses to infections and malignancy. In this review, we will focus on recent advances in NKT cell-based vaccination strategies. Given the role that NKT cells play in autoimmune disease, infectious diseases, cancer, transplant immunology and dermatology, it is important to understand how to effectively guide their effector functions in order to develop novel immunotherapeutic strategies.

SELECTION OF CITATIONS
SEARCH DETAIL
...