Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 51(8): 2074-2085, 2021 08.
Article in English | MEDLINE | ID: mdl-33945643

ABSTRACT

The aberrant release of inflammatory mediators often referred to as a cytokine storm or cytokine release syndrome (CRS), is a common and sometimes fatal complication in acute infectious diseases including Ebola, dengue, COVID-19, and influenza. Fatal CRS occurrences have also plagued the development of highly promising cancer therapies based on T-cell engagers and chimeric antigen receptor (CAR) T cells. CRS is intimately linked with dysregulated and excessive cytokine release, including IFN-γ, TNF-α, IL 1, IL-6, and IL-10, resulting in a systemic inflammatory response leading to multiple organ failure. Here, we show that mice intravenously administered the agonistic hamster anti-mouse CD3ε monoclonal antibody 145-2C11 develop clinical and laboratory manifestations seen in patients afflicted with CRS, including body weight loss, hepatosplenomegaly, thrombocytopenia, increased vascular permeability, lung inflammation, and hypercytokinemia. Blood cytokine levels and gene expression analysis from lung, liver, and spleen demonstrated a hierarchy of inflammatory cytokine production and infiltrating immune cells with differentiating organ-dependent kinetics. IL-2, IFN-γ, TNF-α, and IL-6 up-regulation preceded clinical signs of CRS. The co-treatment of mice with a neutralizing anti-cytokine antibody cocktail transiently improved early clinical and laboratory features of CRS. We discuss the predictive use of this model in the context of new anti-cytokine strategies to treat human CRS.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies/immunology , CD3 Complex/antagonists & inhibitors , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/metabolism , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Animals , Antibodies/adverse effects , Antibodies, Monoclonal/therapeutic use , Cytokine Release Syndrome/diagnosis , Cytokine Release Syndrome/drug therapy , Cytokines/blood , Disease Models, Animal , Drug Therapy, Combination , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Lymphocyte Activation/immunology , Mice , Phenotype , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
2.
Mol Cancer Ther ; 17(8): 1739-1751, 2018 08.
Article in English | MEDLINE | ID: mdl-29743205

ABSTRACT

CD47, an ubiquitously expressed innate immune checkpoint receptor that serves as a universal "don't eat me" signal of phagocytosis, is often upregulated by hematologic and solid cancers to evade immune surveillance. Development of CD47-targeted modalities is hindered by the ubiquitous expression of the target, often leading to rapid drug elimination and hemotoxicity including anemia. To overcome such liabilities, we have developed a fully human bispecific antibody, NI-1701, designed to coengage CD47 and CD19 selectively on B cells. NI-1701 demonstrates favorable elimination kinetics with no deleterious effects seen on hematologic parameters following single or multiple administrations to nonhuman primates. Potent in vitro and in vivo activity is induced by NI-1701 to kill cancer cells across a plethora of B-cell malignancies and control tumor growth in xenograft mouse models. The mechanism affording maximal tumor growth inhibition by NI-1701 is dependent on the coengagement of CD47/CD19 on B cells inducing potent antibody-dependent cellular phagocytosis of the targeted cells. NI-1701-induced control of tumor growth in immunodeficient NOD/SCID mice was more effective than that achieved with the anti-CD20 targeted antibody, rituximab. Interestingly, a synergistic effect was seen when tumor-implanted mice were coadministered NI-1701 and rituximab leading to significantly improved tumor growth inhibition and regression in some animals. We describe herein, a novel bispecific antibody approach aimed at sensitizing B cells to become more readily phagocytosed and eliminated thus offering an alternative or adjunct therapeutic option to patients with B-cell malignancies refractory/resistant to anti-CD20-targeted therapy. Mol Cancer Ther; 17(8); 1739-51. ©2018 AACR.


Subject(s)
Antibodies, Bispecific/genetics , Leukemia/genetics , Leukemia/therapy , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/therapy , Animals , Antigens, CD19 , CD47 Antigen , Humans , Leukemia/pathology , Lymphoma, B-Cell/pathology , Mice , Xenograft Model Antitumor Assays
3.
J Allergy Clin Immunol ; 141(4): 1439-1449, 2018 04.
Article in English | MEDLINE | ID: mdl-28807602

ABSTRACT

BACKGROUND: The pathogenesis of macrophage activation syndrome (MAS) is not clearly understood: a large body of evidence supports the involvement of mechanisms similar to those implicated in the setting of primary hemophagocytic lymphohistiocytosis. OBJECTIVE: We sought to investigate the pathogenic role of IFN-γ and the therapeutic efficacy of IFN-γ neutralization in an animal model of MAS. METHODS: We used an MAS model established in mice transgenic for human IL-6 (IL-6TG mice) challenged with LPS (MAS mice). Levels of IFN-γ and IFN-γ-inducible chemokines were evaluated by using real-time PCR in the liver and spleen and by means of ELISA in plasma. IFN-γ neutralization was achieved by using the anti-IFN-γ antibody XMG1.2 in vivo. RESULTS: Mice with MAS showed a significant upregulation of the IFN-γ pathway, as demonstrated by increased mRNA levels of Ifng and higher levels of phospho-signal transducer and activator of transcription 1 in the liver and spleen and increased expression of the IFN-γ-inducible chemokines Cxcl9 and Cxcl10 in the liver and spleen, as well as in plasma. A marked increase in Il12a and Il12b expression was also found in livers and spleens of mice with MAS. In addition, mice with MAS had a significant increase in numbers of liver CD68+ macrophages. Mice with MAS treated with an anti-IFN-γ antibody showed a significant improvement in survival and body weight recovery associated with a significant amelioration of ferritin, fibrinogen, and alanine aminotransferase levels. In mice with MAS, treatment with the anti-IFN-γ antibody significantly decreased circulating levels of CXCL9, CXCL10, and downstream proinflammatory cytokines. The decrease in CXCL9 and CXCL10 levels paralleled the decrease in serum levels of proinflammatory cytokines and ferritin. CONCLUSION: These results provide evidence for a pathogenic role of IFN-γ in the setting of MAS.


Subject(s)
Antibodies, Neutralizing/immunology , Interferon-gamma/immunology , Macrophage Activation Syndrome/immunology , Macrophage Activation/immunology , Macrophages/immunology , Alanine Transaminase/immunology , Animals , Chemokine CXCL10/immunology , Chemokine CXCL9/immunology , Cytokines/immunology , Disease Models, Animal , Ferritins/immunology , Fibrinogen/immunology , Inflammation/immunology , Lymphohistiocytosis, Hemophagocytic/immunology , Mice
4.
Antibodies (Basel) ; 7(1)2018 Jan 03.
Article in English | MEDLINE | ID: mdl-31544856

ABSTRACT

CD47 serves as an anti-phagocytic receptor that is upregulated by cancer to promote immune escape. As such, CD47 is the focus of intense immuno-oncology drug development efforts. However, as CD47 is expressed ubiquitously, clinical development of conventional drugs, e.g., monoclonal antibodies, is confronted with patient safety issues and poor pharmacology due to the widespread CD47 "antigen sink". A potential solution is tumor-directed blockade of CD47, which can be achieved with bispecific antibodies (biAbs). Using mouse CD47-blocking biAbs in a syngeneic tumor model allowed us to evaluate the efficacy of tumor-directed blockade of CD47 in the presence of the CD47 antigen sink and a functional adaptive immune system. We show here that CD47-targeting biAbs inhibited tumor growth in vivo, promoting durable antitumor responses and stimulating CD8+ T cell activation in vitro. In vivo efficacy of the biAbs could be further enhanced when combined with chemotherapy or PD-1/PD-L1 immune checkpoint blockade. We also show that selectivity and pharmacological properties of the biAb are dependent on the affinity of the anti-CD47 arm. Taken together, our study validates the approach to use CD47-blocking biAbs either as a monotherapy or part of a multi-drug approach to enhance antitumor immunity.

5.
Transl Res ; 180: 37-52.e2, 2017 02.
Article in English | MEDLINE | ID: mdl-27559680

ABSTRACT

Life-threatening cytokine release syndromes include primary (p) and secondary (s) forms of hemophagocytic lymphohistiocytosis (HLH). Below detection in healthy individuals, interferon γ (IFNγ) levels are elevated to measurable concentrations in these afflictions suggesting a central role for this cytokine in the development and maintenance of HLH. Mimicking an infection-driven model of sHLH in mice, we observed that the tissue-derived levels of IFNγ are actually 500- to 2000-fold higher than those measured in the blood. To identify a blood biomarker, we postulated that the IFNγ gene products, CXCL9 and CXCL10 would correlate with disease parameters in the mouse model. To translate this into a disease relevant biomarker, we investigated whether CXCL9 and CXCL10 levels correlated with disease activity in pediatric sHLH patients. Our data demonstrate that disease control in mice correlates with neutralization of IFNγ activity in tissues and that the 2 chemokines serve as serum biomarkers to reflect disease status. Importantly, CXCL9 and CXCL10 levels in pediatric sHLH were shown to correlate with key disease parameters and severity in these patients. Thus, the translatability of the IFNγ-biomarker correlates from mouse to human, advocating the use of serum CXCL9 or CXCL10 as a means to monitor total IFNγ activity in patients with sHLH.


Subject(s)
Interferon-gamma/blood , Lymphohistiocytosis, Hemophagocytic/blood , Animals , Biomarkers/blood , Chemokine CXCL10/blood , Chemokine CXCL9/blood , Child , Disease Models, Animal , Female , Humans , Lymphohistiocytosis, Hemophagocytic/pathology , Mice, Inbred C57BL , Neutralization Tests , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Syndrome
6.
J Immunol Res ; 2016: 5069678, 2016.
Article in English | MEDLINE | ID: mdl-27243038

ABSTRACT

The aim of this study was, at the assay development stage and thus with an appropriate degree of rigor, to select the most appropriate technology platform and sample pretreatment procedure for a clinical ADA assay. Thus, ELISA, MSD, Gyrolab, and AlphaLISA immunoassay platforms were evaluated in association with target depletion and acid dissociation sample pretreatment steps. An acid dissociation step successfully improved the drug tolerance for all 4 technology platforms and the required drug tolerance was achieved with the Gyrolab and MSD platforms. The target tolerance was shown to be better for the ELISA format, where an acid dissociation treatment step alone was sufficient to achieve the desired target tolerance. However, inclusion of a target depletion step in conjunction with the acid treatment raised the target tolerance to the desired level for all of the technologies. A higher sensitivity was observed for the MSD and Gyrolab assays and the ELISA, MSD, and Gyrolab all displayed acceptable interdonor variability. This study highlights the usefulness of evaluating the performance of different assay platforms at an early stage in the assay development process to aid in the selection of the best fit-for-purpose technology platform and sample pretreatment steps.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoassay/methods , Antibodies, Monoclonal/therapeutic use , Drug Tolerance , Enzyme-Linked Immunosorbent Assay , Enzymes, Immobilized/chemistry , Humans , Immunoassay/standards , Immunologic Tests , Molecular Targeted Therapy , Sensitivity and Specificity
7.
Ann Rheum Dis ; 75(6): 1177-86, 2016 06.
Article in English | MEDLINE | ID: mdl-26698852

ABSTRACT

BACKGROUND: Interferon γ (IFNγ) is considered a seminal cytokine in the pathogenesis of giant cell arteritis (GCA), but its functional role has not been investigated. We explored changes in infiltrating cells and biomarkers elicited by blocking IFNγ with a neutralising monoclonal antibody, A6, in temporal arteries from patients with GCA. METHODS: Temporal arteries from 34 patients with GCA (positive histology) and 21 controls were cultured on 3D matrix (Matrigel) and exposed to A6 or recombinant IFNγ. Changes in gene/protein expression were measured by qRT-PCR/western blot or immunoassay. Changes in infiltrating cells were assessed by immunohistochemistry/immunofluorescence. Chemotaxis/adhesion assays were performed with temporal artery-derived vascular smooth muscle cells (VSMCs) and peripheral blood mononuclear cells (PBMCs). RESULTS: Blocking endogenous IFNγ with A6 abrogated STAT-1 phosphorylation in cultured GCA arteries. Furthermore, selective reduction in CXCL9, CXCL10 and CXCL11 chemokine expression was observed along with reduction in infiltrating CD68 macrophages. Adding IFNγ elicited consistent opposite effects. IFNγ induced CXCL9, CXCL10, CXCL11, CCL2 and intracellular adhesion molecule-1 expression by cultured VSMC, resulting in increased PBMC chemotaxis/adhesion. Spontaneous expression of chemokines was higher in VSMC isolated from GCA-involved arteries than in those obtained from controls. Incubation of IFNγ-treated control arteries with PBMC resulted in adhesion/infiltration by CD68 macrophages, which did not occur in untreated arteries. CONCLUSIONS: Our ex vivo system suggests that IFNγ may play an important role in the recruitment of macrophages in GCA by inducing production of specific chemokines and adhesion molecules. Vascular wall components (ie, VSMC) are mediators of these functions and may facilitate progression of inflammatory infiltrates through the vessel wall.


Subject(s)
Chemokines, CXC/metabolism , Giant Cell Arteritis/immunology , Interferon-gamma/antagonists & inhibitors , Macrophages/immunology , Aged , Aged, 80 and over , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Chemokines, CXC/genetics , Chemotaxis/immunology , Down-Regulation/immunology , Female , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/pharmacology , Male , Muscle, Smooth, Vascular/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Temporal Arteries/immunology , Tissue Culture Techniques
8.
J Immunol ; 192(4): 1641-50, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442438

ABSTRACT

B cells play a major role in the pathogenesis of many autoimmune disorders, including rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and type I diabetes mellitus, as indicated by the efficacy of B cell-targeted therapies in these diseases. Therapeutic effects of the most commonly used B cell-targeted therapy, anti-CD20 mAb, are contingent upon long-term depletion of peripheral B cells. In this article, we describe an alternative approach involving the targeting of CD79, the transducer subunit of the B cell AgR. Unlike anti-CD20 mAbs, the protective effects of CD79-targeted mAbs do not require cell depletion; rather, they act by inducing an anergic-like state. Thus, we describe a novel B cell-targeted approach predicated on the induction of B cell anergy.


Subject(s)
Autoimmune Diseases/prevention & control , B-Lymphocytes/immunology , CD79 Antigens/immunology , Clonal Anergy/immunology , Animals , Antibodies, Monoclonal/immunology , Autoimmunity/immunology , Female , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout
9.
J Biol Chem ; 286(50): 42959-70, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-21990364

ABSTRACT

IL-6 trans-signaling via the soluble IL-6 receptor (sIL-6R) plays a critical role in chronic inflammation and cancer. Soluble gp130 (sgp130) specifically inhibits IL-6 trans-signaling but was described to not interfere with classic signaling via the membrane-bound IL-6R. Physiological and most pathophysiological conditions are characterized by a molar excess of serum sIL-6R over IL-6 characterized by free IL-6 and IL-6 found in IL-6·sIL-6R complexes allowing both classic and trans-signaling. Surprisingly, under these conditions, sgp130 was able to trap all free IL-6 molecules in IL-6·sIL-6R·sgp130 complexes, resulting in inhibition of classic signaling. Because a significant fraction of IL-6 molecules did not form complexes with sIL-6R, our results demonstrate that compared with the anti-IL-6R antibody tocilizumab or the anti-trans-signaling monoclonal antibody 25F10, much lower concentrations of the dimeric sgp130Fc were sufficient to block trans-signaling. In vivo, sgp130Fc blocked IL-6 signaling in the colon but not in liver and lung, indicating that the colon is a prominent target of IL-6 trans-signaling. Our results point to a so far unanticipated role of sgp130 in the blockade of classic signaling and indicate that in vivo only low therapeutic concentrations of sgp130Fc guarantee blockade of IL-6 trans-signaling without affecting IL-6 classic signaling.


Subject(s)
Cytokine Receptor gp130/metabolism , Interleukin-6/metabolism , Receptors, Interleukin-6/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Cytokine Receptor gp130/antagonists & inhibitors , Flow Cytometry , Humans , Immunohistochemistry , Interleukin-6/pharmacology , Male , Mice , Mice, Inbred C57BL , Receptors, Interleukin-6/antagonists & inhibitors , Signal Transduction/drug effects
10.
J Immunol ; 185(9): 5512-21, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20870936

ABSTRACT

IL-6-mediated T cell-driven immune responses are associated with signaling occurring through the membrane-bound cognate receptor α-chain (mIL-6Rα). Once formed, IL-6-mIL-6Rα complexes induce the homodimerization and subsequent phosphorylation of the ubiquitously expressed signal-transducing protein, gp130. This signaling event is defined as classical IL-6 signaling. However, many inflammatory processes assigned to IL-6 may be mediated via binding a naturally occurring soluble IL-6Rα, which forms an agonistic complex (IL-6/soluble IL-6Rα) capable of evoking responses on a wide range of cell types that lack mIL-6Rα (IL-6 trans-signaling). To dissect the differential contribution of the two IL-6 signaling pathways in cell-mediated inflammatory processes, we pharmaceutically targeted each using two murine models of human arthritis. Whereas intra-articular neutralization of trans-signaling attenuated local inflammatory responses, the classical pathway was found to be obligate and sufficient to induce pathogenic T cells and humoral responses, leading to systemic disease. Our data illustrate that mechanisms occurring in the secondary lymphoid organs underlying arthropathies are mediated via the classical pathway of IL-6 signaling, whereas trans-signaling contributes only at the local site, that is, in the affected tissues.


Subject(s)
Arthritis, Experimental/immunology , Autoimmunity/immunology , Interleukin-6/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Arthritis, Experimental/metabolism , Cell Separation , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Interleukin-6/metabolism , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Receptors, Interleukin-6/immunology , Receptors, Interleukin-6/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Surface Plasmon Resonance , Transfection
11.
J Immunol ; 185(4): 2544-54, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20644170

ABSTRACT

Chemokines are key regulators of leukocyte trafficking and play a crucial role under homeostatic and inflammatory conditions. Because chemokines are involved in multiple pathologies, they represent an attractive class of therapeutic targets. However, because of the redundancy of this system, neutralizing a single chemokine may be insufficient to achieve therapeutic benefit. Our strategy was to use a Fc-fusion recombinant protein form of the poxvirus-derived viral CC chemokine inhibitor protein (vCCI-Fc) that has the ability to specifically bind to multiple CC chemokines and neutralize their activity. In this study, we demonstrate first that, in vivo, vCCI-Fc prevents CC chemokine-dependent migration of macrophages into inflamed tissue of carageenan-challenged mice. We next studied this effect of inhibiting CC chemokine activity in a model more relevant to human disease, collagen-induced arthritis. Mice receiving vCCI-Fc revealed a striking retention of splenocytes, including activated and IFN-gamma-secreting CD4(+) and CD8(+) T cells, that was associated with a concomitant decrease of cells in the draining lymph nodes. These phenomena resulted in a significant decrease in the incidence of disease and a reduction in clinical score, joint inflammation, and cartilage destruction as compared with mice receiving isotype control. Taken together, these results define a role for CC chemokines in the control of disease, as interfering with their function leads to a previously unappreciated role of controlling inflammatory cell trafficking in and out of secondary lymphoid organs.


Subject(s)
Arthritis, Experimental/immunology , Chemokines, CC/immunology , Inflammation/immunology , Recombinant Fusion Proteins/immunology , Animals , Arthritis, Experimental/prevention & control , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/immunology , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/metabolism , Female , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Inflammation/prevention & control , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Count , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Protein Binding , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Eur J Immunol ; 34(10): 2656-63, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15368281

ABSTRACT

Although non-obese diabetic (NOD) mice spontaneously develop T cell autoimmunity, it is not clear whether this phenomenon results from a defect in tolerance to self-Ag. Furthermore, as autoimmunity has been postulated to result from T cell responses directed toward self-peptides that bind with low affinity to NOD I-A(g7) MHC class II molecules, it is important to determine whether the expression of such peptides induces tolerance. We have constructed NOD transgenic (Tg) mice expressing the Leishmania antigen receptor for C kinase (LACK) Ag in either the thymus or pancreatic beta cells. We identified LACK peptides that were the targets of T cells in LACK-immunized NOD mice while binding to I-A(g7) with low affinity. While CD4(+) T cells from NOD mice secreted IFN-gamma, IL-4, IL-5 and IL-10 in response to LACK, those from LACK-expressing Tg mice secreted reduced levels of cytokines. Experiments using peptide/MHC multimers showed that LACK-expressing Tg mice exhibited self-reactive CD4(+) T cells with impaired proliferation capabilities. Hence, even self-peptides that bind to I-A(g7) with low affinity can induce tolerance in NOD mice. This result is important in light of the commonly held hypothesis that T cells reacting to peptides that bind to MHC with low affinity escape tolerance induction and cause autoimmunity.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class II/metabolism , Immune Tolerance , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antigens, Protozoan/immunology , Cytokines/immunology , Cytokines/metabolism , Female , Flow Cytometry , Mice , Mice, Inbred NOD , Mice, Transgenic , Molecular Sequence Data , Peptides/immunology , Protozoan Proteins/immunology
13.
Trends Immunol ; 24(4): 162-4, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12697440

ABSTRACT

Natural killer (NK)-cell receptors (NKRs) on CD8+ T cells can modulate antigen-specific T-cell activity but their function and rules that govern their expression remain unclear. Recent work has provided new insight into CD94-NKG2A expression on T cells. Whereas upregulation on the cell surface is induced following T-cell receptor (TCR) engagement, commitment to CD94-NKG2A expression is a clonal attribute. NKRs can therefore shape cytotoxic T-lymphocyte (CTL) responses and should be considered in designing vaccine therapies.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/metabolism , Lectins, C-Type/metabolism , Lymphocyte Subsets/metabolism , Receptors, Immunologic/metabolism , Animals , Antigens/immunology , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation , Humans , Lectins, C-Type/immunology , Lymphocyte Subsets/immunology , NK Cell Lectin-Like Receptor Subfamily C , NK Cell Lectin-Like Receptor Subfamily D , Receptors, Immunologic/immunology , Receptors, Natural Killer Cell , Thymus Gland/cytology , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...