Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Virol ; : e0029424, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829139

ABSTRACT

Arenaviral vaccine vectors encoding simian immunodeficiency virus (SIV) immunogens are capable of inducing efficacious humoral and cellular immune responses in nonhuman primates. Several studies have evaluated the use of immune modulators to further enhance vaccine-induced T-cell responses. The hematopoietic growth factor Flt3L drives the expansion of various bone marrow progenitor populations, and administration of Flt3L was shown to promote expansion of dendritic cell populations in spleen and blood, which are targets of arenaviral vectors. Therefore, we evaluated the potential of Flt3 signaling to enhance the immunogenicity of arenaviral vaccines encoding SIV immunogens (SIVSME543 Gag, Env, and Pol) in rhesus macaques, with a rhesus-specific engineered Flt3L-Fc fusion protein. In healthy animals, administration of Flt3L-Fc led to a 10- to 100-fold increase in type 1 dendritic cells 7 days after dosing, with no antidrug antibody (ADA) generation after repeated dosing. We observed that administration of Flt3L-Fc fusion protein 7 days before arenaviral vaccine increased the frequency and activation of innate immune cells and enhanced T-cell activation with no treatment-related adverse events. Flt3L-Fc administration induced early innate immune activation, leading to a significant enhancement in magnitude, breadth, and polyfunctionality of vaccine-induced T-cell responses. The Flt3L-Fc enhancement in vaccine immunogenicity was comparable to a combination with αCTLA-4 and supports the use of safe and effective variants of Flt3L to augment therapeutic vaccine-induced T-cell responses.IMPORTANCEInduction of a robust human immunodeficiency virus (HIV)-specific CD4+ and CD8+ T-cell response through therapeutic vaccination is considered essential for HIV cure. Arenaviral vaccine vectors encoding simian immunodeficiency virus (SIV) immunogens have demonstrated strong immunogenicity and efficacy in nonhuman primates. Here, we demonstrate that the immunogenicity of arenaviral vectors encoding SIV immunogens can be enhanced by administration of Flt3L-Fc fusion protein 7 days before vaccination. Flt3L-Fc-mediated increase in dendritic cells led to robust improvements in vaccine-induced T- and B-cell responses compared with vaccine alone, and Flt3L-Fc dosing was not associated with any treatment-related adverse events. Importantly, immune modulation by either Flt3L-Fc or αCTLA-4 led to comparable enhancement in vaccine response. These results indicate that the addition of Flt3L-Fc fusion protein before vaccine administration can significantly enhance vaccine immunogenicity. Thus, safe and effective Flt3L variants could be utilized as part of a combination therapy for HIV cure.

2.
Anal Bioanal Chem ; 412(7): 1693-1700, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31993727

ABSTRACT

Major histocompatibility complex class I chain-related A and B (MICA/B) are cell-surface proteins that act as ligands to natural killer cell receptors, NKG2D, expressed on immune cells. Prevention of proteolytic shedding of MICA/B to retain their integrity on the cell surface has become a therapeutic strategy in immuno-oncology. Given the unique mechanism of MICA/B shedding, structural characterization of MICA/B and therapeutic agent interaction is important in the drug discovery process. In this study, we describe the practical utility of hydrogen/deuterium exchange mass spectrometry (HDX-MS) in epitope mapping studies of a cohort of four monoclonal antibodies targeting MICA in a rapid manner. HDX-MS followed by electron-transfer dissociation allows high-resolution refinement of binding epitopes. This integrated strategy offers, for the first time, molecular-level understanding of MICA's conformational dynamics in solution as well as the unique mechanism of actions of these antibodies in targeting MICA. Graphical abstract.


Subject(s)
Antibodies, Monoclonal/immunology , Deuterium Exchange Measurement/methods , Epitope Mapping/methods , Histocompatibility Antigens Class I/immunology , Mass Spectrometry/methods , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Electron Transport , Humans
3.
Blood ; 123(26): 4120-31, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24711662

ABSTRACT

The C-X-C chemokine receptor type 4 (CXCR4) plays a crucial role in modulating cell trafficking in hematopoietic stem cells and clonal B cells. We screened 418 patients with B-cell lymphoproliferative disorders and described the presence of the C1013G/CXCR4 warts, hypogammaglobulinemia, infections, and myelokathexis-associated mutation in 28.2% (37/131) of patients with lymphoplasmacytic lymphoma (Waldenström macroglobulinemia [WM]), being either absent or present in only 7% of other B-cell lymphomas. In vivo functional characterization demonstrates its activating role in WM cells, as demonstrated by significant tumor proliferation and dissemination to extramedullary organs, leading to disease progression and decreased survival. The use of a monoclonal antibody anti-CXCR4 led to significant tumor reduction in a C1013G/CXCR4 WM model, whereas drug resistance was observed in mutated WM cells exposed to Bruton's tyrosine kinase, mammalian target of rapamycin, and phosphatidylinositol 3-kinase inhibitors, but not proteasome inhibitors. These findings demonstrate that C1013G/CXCR4 is an activating mutation in WM and support its role as a critical regulator of WM molecular pathogenesis and as an important therapeutic target.


Subject(s)
Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Mutation, Missense , Receptors, CXCR4/metabolism , Waldenstrom Macroglobulinemia/metabolism , Animals , Cell Proliferation/drug effects , Disease-Free Survival , Female , Heterografts , Humans , Male , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Receptors, CXCR4/genetics , Survival Rate , Waldenstrom Macroglobulinemia/drug therapy , Waldenstrom Macroglobulinemia/genetics , Waldenstrom Macroglobulinemia/mortality , Waldenstrom Macroglobulinemia/pathology
4.
Clin Cancer Res ; 30(14): 2954-2963, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38295150

ABSTRACT

PURPOSE: GS-3583, an FMS-like tyrosine kinase 3 (FLT3) agonist Fc fusion protein, expanded conventional dendritic cells (cDC) in the periphery of healthy volunteers, suggesting potential for GS-3583 to increase cDCs in the tumor microenvironment and promote T cell-mediated antitumor activity in cancer patients. This phase Ib open-label study assessed GS-3583 in adults with advanced solid tumors. PATIENTS AND METHODS: Multiple escalating doses of GS-3583 (standard 3+3 design) were administered intravenously on days 1 and 15 of cycle 1 and day 1 of each subsequent 28-day cycle for up to 52 weeks. Dose-limiting toxicity (DLT) was evaluated during the first 28 days of GS-3583 at each dose level. RESULTS: Thirteen participants enrolled in four dose-escalation cohorts, after which the study was terminated following safety review. Median (range) age was 71 (44-79), and 7 (54%) participants were male. There were no DLTs. Seven participants had grade ≥3 AEs; 2 participants had grade 5 AEs, including a second primary malignancy (acute myeloid leukemia) considered treatment-related. Dose-dependent increase in GS-3583 serum exposure was observed in the dose range of 2-20 mg with GS-3583 accumulation at higher dose levels. Expansions of cDCs occurred at all four doses with a dose-dependent trend in the durability of the cDC expansion. CONCLUSIONS: GS-3583 was relatively well tolerated and induced dose-dependent expansion of cDCs in the periphery of patients with advanced solid tumors. However, development of a second primary malignancy provides a cautionary tale for the FLT3 agonist mechanism. See related commentary by Raeder and Drazer, p. 2857.


Subject(s)
Neoplasms , Recombinant Fusion Proteins , fms-Like Tyrosine Kinase 3 , Humans , Male , Neoplasms/drug therapy , Neoplasms/pathology , Female , Middle Aged , Aged , Adult , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/therapeutic use , Maximum Tolerated Dose , Immunoglobulin Fc Fragments/adverse effects , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/administration & dosage , Treatment Outcome
5.
Oncoimmunology ; 11(1): 2141007, 2022.
Article in English | MEDLINE | ID: mdl-36352891

ABSTRACT

The presence of T regulatory (Treg) cells in the tumor microenvironment is associated with poor prognosis and resistance to therapies aimed at reactivating anti-tumor immune responses. Therefore, depletion of tumor-infiltrating Tregs is a potential approach to overcome resistance to immunotherapy. However, identifying Treg-specific targets to drive such selective depletion is challenging. CCR8 has recently emerged as one of these potential targets. Here, we describe GS-1811, a novel therapeutic monoclonal antibody that specifically binds to human CCR8 and is designed to selectively deplete tumor-infiltrating Tregs. We validate previous findings showing restricted expression of CCR8 on tumor Tregs, and precisely quantify CCR8 receptor densities on tumor and normal tissue T cell subsets, demonstrating a window for selective depletion of Tregs in the tumor. Importantly, we show that GS-1811 depleting activity is limited to cells expressing CCR8 at levels comparable to tumor-infiltrating Tregs. Targeting CCR8 in mouse tumor models results in robust anti-tumor efficacy, which is dependent on Treg depleting activity, and synergizes with PD-1 inhibition to promote anti-tumor responses in PD-1 resistant models. Our data support clinical development of GS-1811 to target CCR8 in cancer and drive tumor Treg depletion in order to promote anti-tumor immunity.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Mice , Animals , Humans , T-Lymphocytes, Regulatory/metabolism , Programmed Cell Death 1 Receptor , Immunotherapy/methods , Neoplasms/therapy , Tumor Microenvironment , Immunoglobulin Fc Fragments/metabolism , Receptors, CCR8/metabolism
6.
Cancer Immunol Immunother ; 59(2): 257-65, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19657637

ABSTRACT

A human anti-CD19 antibody was expressed in fucosyltransferase-deficient CHO cells to generate nonfucosylated MDX-1342. Binding of MDX-1342 to human CD19-expressing cells was similar to its fucosylated parental antibody. However, MDX-1342 exhibited increased affinity for FcγRIIIa-Phe158 and FcγRIIIa-Val158 receptors as well as enhanced effector cell function, as demonstrated by increased potency and efficacy in antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis assays. MDX-1342 showed dose-dependent improvement in survival using a murine B-cell lymphoma model in which Ramos cells were administered systemically. In addition, low nanomolar binding to cynomolgus monkey CD19 and increased affinity for cynomolgus monkey FcγRIIIa was observed. In vivo administration of MDX-1342 in cynomolgus monkeys revealed potent B-cell depletion, suggesting its potential utility as a B-lymphocyte depletive therapy for malignancies and autoimmune indications.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD19/immunology , B-Lymphocytes/immunology , Lymphocyte Depletion , Lymphoma, B-Cell/therapy , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibody Affinity , CHO Cells , Cricetinae , Cricetulus , Humans , Macaca fascicularis , Mice , Mice, SCID , Mice, Transgenic , Phagocytosis , Receptors, IgG/immunology , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 15(10): 3376-83, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19401346

ABSTRACT

PURPOSE: This study was undertaken to evaluate the effects of MDX-1401, a nonfucosylated fully human monoclonal antibody that binds to human CD30, and to determine whether it exhibits greater in vitro and in vivo activity than its parental antibody. EXPERIMENTAL DESIGN: Assays measuring antibody binding to CD30-expressing cells and FcgammaRIIIa (CD16) transfectants as well as antibody-dependent cellular cytotoxicity (ADCC) were conducted. Antitumor activity was determined using a Karpas-299 systemic model. RESULTS: The binding of MDX-1401 to CD30 antigen was identical to fucose-containing parental anti-CD30 antibody (MDX-060). In contrast, MDX-1401 showed increased binding affinity to FcgammaRIIIa-transfected cells resulting in increased effector function. MDX-1401 greatly improved ADCC activity as evidenced by a decrease in half-maximal effective concentration (EC(50)) and an increase in maximum cell lysis when compared with MDX-060. Increased ADCC activity was observed among a panel of cell lines, including one with very low CD30 antigen expression in which parental antibody failed to induce any detectable ADCC. MDX-1401 activity with all FcgammaRIIIa polymorphic variants, including less active Phe/Phe158 and Phe/Val158 effector cells, was shown. Furthermore, MDX-1401 was efficacious in inhibiting tumor growth in CD30(+) lymphoma xenografts. CONCLUSIONS: The low doses of antibody required for ADCC activity irrespective of donor genotype, the ability to mediate ADCC in target cells expressing low levels of CD30, and increased in vivo efficacy support the development of MDX-1401 for treatment of malignant lymphoma.


Subject(s)
Antibodies, Monoclonal/pharmacology , Lymphoma/drug therapy , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibody Affinity/drug effects , Antibody Affinity/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Binding Sites, Antibody/immunology , CHO Cells , Carbohydrates/chemistry , Carbohydrates/immunology , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Fucose/chemistry , Fucose/immunology , Humans , Ki-1 Antigen/immunology , Lymphoma/immunology , Lymphoma/pathology , Male , Mice , Mice, SCID , Receptors, IgG/chemistry , Receptors, IgG/immunology
8.
Oncotarget ; 7(3): 2809-22, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26646452

ABSTRACT

The CXCR4 receptor (Chemokine C-X-C motif receptor 4) is highly expressed in different hematological malignancies including chronic lymphocytic leukemia (CLL). The CXCR4 ligand (CXCL12) stimulates CXCR4 promoting cell survival and proliferation, and may contribute to the tropism of leukemia cells towards lymphoid tissues. Therefore, strategies targeting CXCR4 may constitute an effective therapeutic approach for CLL. To address that question, we studied the effect of Ulocuplumab (BMS-936564), a fully human IgG4 anti-CXCR4 antibody, using a stroma--CLL cells co-culture model. We found that Ulocuplumab (BMS-936564) inhibited CXCL12 mediated CXCR4 activation-migration of CLL cells at nanomolar concentrations. This effect was comparable to AMD3100 (Plerixafor--Mozobil), a small molecule CXCR4 inhibitor. However, Ulocuplumab (BMS-936564) but not AMD3100 induced apoptosis in CLL at nanomolar concentrations in the presence or absence of stromal cell support. This pro-apoptotic effect was independent of CLL high-risk prognostic markers, was associated with production of reactive oxygen species and did not require caspase activation. Overall, these findings are evidence that Ulocuplumab (BMS-936564) has biological activity in CLL, highlight the relevance of the CXCR4-CXCL12 pathway as a therapeutic target in CLL, and provide biological rationale for ongoing clinical trials in CLL and other hematological malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chemokine CXCL12/biosynthesis , Imino Furanoses/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Pyrimidinones/pharmacology , Reactive Oxygen Species/metabolism , Receptors, CXCR4/antagonists & inhibitors , Actins/metabolism , Benzylamines , Cell Movement/drug effects , Cell Proliferation , Cell Survival , Chemokine CXCL12/metabolism , Cyclams , Enzyme Activation/drug effects , Heterocyclic Compounds/pharmacology , Humans , Jurkat Cells , Leukocytes, Mononuclear , Receptors, CXCR4/biosynthesis , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
9.
Cell Rep ; 12(4): 622-35, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26190113

ABSTRACT

Extra-medullary disease (EMD) in multiple myeloma (MM) is associated with poor prognosis and resistance to chemotherapy. However, molecular alterations that lead to EMD have not been well defined. We developed bone marrow (BM)- and EMD-prone MM syngeneic cell lines; identified that epithelial-to-mesenchymal transition (EMT) transcriptional patterns were significantly enriched in both clones compared to parental cells, together with higher levels of CXCR4 protein; and demonstrated that CXCR4 enhanced the acquisition of an EMT-like phenotype in MM cells with a phenotypic conversion for invasion, leading to higher bone metastasis and EMD dissemination in vivo. In contrast, CXCR4 silencing led to inhibited tumor growth and reduced survival. Ulocuplumab, a monoclonal anti-CXCR4 antibody, inhibited MM cell dissemination, supported by suppression of the CXCR4-driven EMT-like phenotype. These results suggest that targeting CXCR4 may act as a regulator of EMD through EMT-like transcriptional modulation, thus representing a potential therapeutic strategy to prevent MM disease progression.


Subject(s)
Epithelial-Mesenchymal Transition , Receptors, CXCR4/metabolism , Sarcoma, Myeloid/metabolism , Transcriptional Activation , Animals , Antibodies, Monoclonal/therapeutic use , Bone Neoplasms/secondary , Cells, Cultured , Humans , Mice , Mice, SCID , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Sarcoma, Myeloid/pathology , Sarcoma, Myeloid/therapy
10.
Cancer Immunol Res ; 2(9): 846-56, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24872026

ABSTRACT

The programmed death-1 (PD-1) receptor serves as an immunologic checkpoint, limiting bystander tissue damage and preventing the development of autoimmunity during inflammatory responses. PD-1 is expressed by activated T cells and downmodulates T-cell effector functions upon binding to its ligands, PD-L1 and PD-L2, on antigen-presenting cells. In patients with cancer, the expression of PD-1 on tumor-infiltrating lymphocytes and its interaction with the ligands on tumor and immune cells in the tumor microenvironment undermine antitumor immunity and support its rationale for PD-1 blockade in cancer immunotherapy. This report details the development and characterization of nivolumab, a fully human IgG4 (S228P) anti-PD-1 receptor-blocking monoclonal antibody. Nivolumab binds to PD-1 with high affinity and specificity, and effectively inhibits the interaction between PD-1 and its ligands. In vitro assays demonstrated the ability of nivolumab to potently enhance T-cell responses and cytokine production in the mixed lymphocyte reaction and superantigen or cytomegalovirus stimulation assays. No in vitro antibody-dependent cell-mediated or complement-dependent cytotoxicity was observed with the use of nivolumab and activated T cells as targets. Nivolumab treatment did not induce adverse immune-related events when given to cynomolgus macaques at high concentrations, independent of circulating anti-nivolumab antibodies where observed. These data provide a comprehensive preclinical characterization of nivolumab, for which antitumor activity and safety have been demonstrated in human clinical trials in various solid tumors.


Subject(s)
Antibodies, Monoclonal/immunology , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Female , Humans , Immunotherapy , Macaca fascicularis , Male , Mice , Mice, Transgenic , Neoplasms/therapy , Nivolumab , Toxicity Tests , Tumor Microenvironment
11.
Clin Cancer Res ; 19(2): 357-66, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23213054

ABSTRACT

PURPOSE: CXCR4 has been identified as a prognostic marker for acute myeloid leukemia (AML) and other malignancies. We describe the development and characterization of a fully human antibody to CXCR4 and its application for therapy of AML, non-Hodgkin lymphoma (NHL), chronic lymphoid leukemia (CLL), and multiple myeloma. EXPERIMENTAL DESIGN: Human transgenic mice were immunized with CXCR4-expressing cells, and antibodies reactive with CXCR4 were analyzed for apoptosis induction and ability to interfere with CXCL12-induced migration and calcium flux. In vivo efficacy was determined in multiple AML, NHL, and multiple myeloma xenograft tumors in severe combined immunodeficient mice. RESULTS: BMS-936564/MDX-1338 is a fully human IgG(4) monoclonal antibody that specifically recognizes human CXCR4. In vitro studies show that MDX-1338 binds to CXCR4-expressing cells with low nanomolar affinity, blocks CXCL12 binding to CXCR4-expressing cells, and inhibits CXCL12-induced migration and calcium flux with low nanomolar EC(50) values. When given as monotherapy, MDX-1338 exhibits antitumor activity in established tumors including AML, NHL, and multiple myeloma xenograft models. In addition, we show that MDX-1338 induced apoptosis on a panel of cell lines and propose that antibody-induced apoptosis is one of the mechanisms of tumor growth inhibition. CONCLUSIONS: BMS-936564/MDX-1338 is a potent CXCR4 antagonist which is efficacious as monotherapy in tumor-bearing mice and is currently in phase I for the treatment of relapsed/refractory AML, NHL, CLL, and multiple myeloma.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Hematologic Neoplasms/immunology , Receptors, CXCR4/immunology , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Calcium/metabolism , Cell Line, Tumor , Chemokine CXCL12/immunology , Chemokine CXCL12/metabolism , Disease Models, Animal , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Humans , Ligands , Mice , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
J Immunol ; 171(2): 860-6, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12847255

ABSTRACT

Engagement of the T cell with Ag on an APC results in a series of immediate signaling events emanating from the stimulation of the TCR. These events include the induced phosphorylation of a number of cellular proteins with a subsequent increase in intracellular calcium and the restructuring of the microtubule and actin cytoskeleton within the T cell. This restructuring of the cytoskeleton culminates in the polarization of the T cell's secretory apparatus toward the engaging APC, allowing the T cell to direct secretion of cytokines toward the appropriate APC. This polarization can be monitored by analyzing the position of the microtubule-organizing center (MTOC), as it moves toward the interface of the T cell and APC. The requirements for MTOC polarization were examined at a single-cell level by studying the interaction of a Jurkat cell line expressing a fluorescently labeled MTOC with Staphylococcal enterotoxin superantigen-bound Raji B cell line, which served as the APC. We found that repolarization of the MTOC substantially followed fluxes in calcium. We also used immobilized anti-TCR mAb and Jurkat signaling mutants, defective in TCR-induced calcium increases, to determine whether signaling components that are necessary for a calcium response also play a role in MTOC polarization. We found that zeta-associated protein-70 as well as its substrate adaptor proteins linker for activation of T cells and Src homology 2 domain-containing leukocyte protein-76 are required for MTOC polarization. Moreover, our studies revealed that a calcium-dependent event not requiring calcineurin or calcium/calmodulin-dependent kinase is required for TCR-induced polarization of the MTOC.


Subject(s)
Adaptor Proteins, Signal Transducing , CD3 Complex , Carrier Proteins/physiology , Lymphocyte Activation , Membrane Proteins , Microtubule-Organizing Center/metabolism , Phosphoproteins/physiology , Protein-Tyrosine Kinases/physiology , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Antibodies, Monoclonal/pharmacology , Antigen-Presenting Cells/metabolism , Benzylamines/pharmacology , Calcineurin/physiology , Calcineurin Inhibitors , Calcium/metabolism , Calcium/physiology , Calcium Signaling/immunology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Polarity/immunology , Humans , Jurkat Cells , Microtubule-Organizing Center/enzymology , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/immunology , Sulfonamides/pharmacology , T-Lymphocytes/enzymology , Tacrolimus/pharmacology , Tumor Cells, Cultured , ZAP-70 Protein-Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL