Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Virol ; 97(5): e0030923, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37070982

ABSTRACT

Coxsackievirus A21 (CVA21) is a naturally occurring RNA virus that, in preclinical studies and clinical trials, has demonstrated promising potential in treating a range of malignancies. Other oncolytic viruses, such as adenovirus, vesicular stomatitis virus, herpesvirus, and vaccinia virus, all can be engineered to carry one or more transgenes for various purposes, including immune modulation, virus attenuation, and induction of apoptosis of tumor cells. However, it remained unknown whether CVA21 can express therapeutic or immunomodulatory payloads due to its small size and high mutation rate. Using reverse genetics techniques, we demonstrated that a transgene encoding a truncated green fluorescent protein (GFP) of up to 141 amino acids (aa) can be successfully carried in the 5' end of the coding region. Furthermore, a chimeric virus carrying an eel fluorescent protein, UnaG (139 aa), was also made and shown to be stable, and it maintained efficient tumor cell-killing activity. Similar to other oncolytic viruses, the likelihood of delivering CVA21 by the intravenous route is low due to issues like blood absorption, neutralizing antibodies, and liver clearance. To address this problem, we designed the CVA21 cDNA under the control of a weak RNA polymerase II promoter, and subsequently, a stable cell pool in 293T cells was made by integrating the resulting CVA21 cDNA into the cell genome. We showed that the cells are viable and able to persistently generate rCVA21 de novo. The carrier cell approach described here may pave the way to designing new cell therapy strategies by arming with oncolytic viruses. IMPORTANCE As a naturally occurring virus, coxsackievirus A21 is a promising oncolytic virotherapy modality. In this study, we first used reverse genetics to determine whether A21 can stably carry transgenes and found that it could express up to 141 amino acids of foreign GFP. The chimeric virus carrying another fluorescent eel protein UnaG (139 amino acids) gene also appeared to be stable over at least 7 passages. Our results provided guidance on how to select and engineer therapeutic payloads for future A21 anticancer research. Second, the challenges of delivering oncolytic viruses by the intravenous route hamper the broader use of oncolytic viruses in the clinic. Here, we used A21 to show that cells could be engineered to stably carry and persistently release the virus by harboring the viral cDNA in the genome. The approach we presented here may pave a new way for oncolytic virus administration using cells as carriers.


Subject(s)
Enterovirus A, Human , Oncolytic Viruses , Amino Acids/genetics , Cell Line, Tumor , DNA, Complementary , Enterovirus A, Human/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Transgenes
2.
Cancer Immunol Immunother ; 64(1): 61-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25287778

ABSTRACT

Elotuzumab is a humanized monoclonal antibody specific for signaling lymphocytic activation molecule-F7 (SLAMF7, also known as CS1, CD319, or CRACC) that enhances natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC) of SLAMF7-expressing myeloma cells. This study explored the mechanisms underlying enhanced myeloma cell killing with elotuzumab as a single agent and in combination with lenalidomide, to support ongoing phase III trials in patients with relapsed/refractory or newly-diagnosed multiple myeloma (MM). An in vitro peripheral blood lymphocyte (PBL)/myeloma cell co-culture model was developed to evaluate the combination of elotuzumab and lenalidomide. Expression of activation markers and adhesion receptors was evaluated by flow cytometry, cytokine expression by Luminex and ELISPOT assays, and cytotoxicity by myeloma cell counts. Elotuzumab activated NK cells and promoted myeloma cell death in PBL/myeloma cell co-cultures. The combination of elotuzumab plus lenalidomide demonstrated superior anti-myeloma activity on established MM xenografts in vivo and in PBL/myeloma cell co-cultures in vitro than either agent alone. The combination enhanced myeloma cell killing by modulating NK cell function that coincided with the upregulation of adhesion and activation markers, including interleukin (IL)-2Rα expression, IL-2 production by CD3(+)CD56(+) lymphocytes, and tumor necrosis factor (TNF)-α production. In co-culture assays, TNF-α directly increased NK cell activation and myeloma cell death with elotuzumab or elotuzumab plus lenalidomide, and neutralizing TNF-α decreased NK cell activation and myeloma cell death with elotuzumab. These results demonstrate that elotuzumab activates NK cells and induces myeloma cell death via NK cell-mediated ADCC, which is further enhanced when combined with lenalidomide.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Interleukin-2/metabolism , Killer Cells, Natural/immunology , Multiple Myeloma/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Flow Cytometry , Humans , Immunoenzyme Techniques , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lymphocyte Activation , Mice , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cancer Immunol Immunother ; 62(12): 1841-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24162108

ABSTRACT

Elotuzumab is a monoclonal antibody in development for multiple myeloma (MM) that targets CS1, a cell surface glycoprotein expressed on MM cells. In preclinical models, elotuzumab exerts anti-MM efficacy via natural killer (NK)-cell-mediated antibody-dependent cellular cytotoxicity (ADCC). CS1 is also expressed at lower levels on NK cells where it acts as an activating receptor. We hypothesized that elotuzumab may have additional mechanisms of action via ligation of CS1 on NK cells that complement ADCC activity. Herein, we show that elotuzumab appears to induce activation of NK cells by binding to NK cell CS1 which promotes cytotoxicity against CS1(+) MM cells but not against autologous CS1(+) NK cells. Elotuzumab may also promote CS1-CS1 interactions between NK cells and CS1(+) target cells to enhance cytotoxicity in a manner independent of ADCC. NK cell activation appears dependent on differential expression of the signaling intermediary EAT-2 which is present in NK cells but absent in primary, human MM cells. Taken together, these data suggest elotuzumab may enhance NK cell function directly and confer anti-MM efficacy by means beyond ADCC alone.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Killer Cells, Natural/immunology , Multiple Myeloma/immunology , Receptors, Immunologic/metabolism , Blotting, Western , Cell Proliferation , Flow Cytometry , Humans , Immunoprecipitation , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lymphocyte Activation , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signaling Lymphocytic Activation Molecule Family , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
4.
Mol Cancer Ther ; 19(6): 1298-1307, 2020 06.
Article in English | MEDLINE | ID: mdl-32229606

ABSTRACT

The programmed cell death 1 (PD-1) pathway represents a major immune checkpoint, which may be engaged by cells in the tumor microenvironment to overcome active T-cell immune surveillance. Pembrolizumab (Keytruda®, MK-3475) is a potent and highly selective humanized mAb of the IgG4/kappa isotype designed to directly block the interaction between PD-1 and its ligands, PD-L1 and PD-L2. This blockade enhances the functional activity of T cells to facilitate tumor regression and ultimately immune rejection. Pembrolizumab binds to human and cynomolgus monkey PD-1 with picomolar affinity and blocks the binding of human and cynomolgus monkey PD-1 to PD-L1 and PD-L2 with comparable potency. Pembrolizumab binds both the C'D and FG loops of PD-1. Pembrolizumab overcomes human and cynomolgus monkey PD-L1-mediated immune suppression in T-cell cultures by enhancing IL2 production following staphylococcal enterotoxin B stimulation of healthy donor and cancer patient cells, and IFNγ production in human primary tumor histoculture. Ex vivo and in vitro studies with human and primate T cells show that pembrolizumab enhances antigen-specific T-cell IFNγ and IL2 production. Pembrolizumab does not mediate FcR or complement-driven effector function against PD-1-expressing cells. Pembrolizumab displays dose-dependent clearance and half-life in cynomolgus monkey pharmacokinetic and toxicokinetic studies typical for human IgG4 antibodies. In nonhuman primate toxicology studies, no findings of toxicologic significance were observed. The preclinical data for pembrolizumab are consistent with the clinical anticancer activity and safety that has been demonstrated in human clinical trials.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Leukocytes, Mononuclear/drug effects , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , Animals , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Female , Humans , Immune Checkpoint Inhibitors/pharmacokinetics , Immune Checkpoint Inhibitors/pharmacology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Macaca fascicularis , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tissue Distribution , Toxicity Tests
5.
J Immunol Res ; 2017: 5737159, 2017.
Article in English | MEDLINE | ID: mdl-29075649

ABSTRACT

Enavatuzumab is a humanized IgG1 anti-TWEAK receptor monoclonal antibody that was evaluated in a phase I clinical study for the treatment of solid malignancies. The current study was to determine whether and how myeloid effector cells were involved in postulated mechanisms for its potent antitumor activity in xenograft models. The initial evidence for a role of effector cells was obtained in a subset of tumor xenograft mouse models whose response to enavatuzumab relied on the binding of Fc of the antibody to Fcγ receptor. The involvement of effector cells was further confirmed by immunohistochemistry, which revealed strong infiltration of CD45+ effector cells into tumor xenografts in responding models, but minimal infiltration in nonresponders. Consistent with the xenograft studies, human effector cells preferentially migrated toward in vivo-responsive tumor cells treated by enavatuzumab in vitro, with the majority of migratory cells being monocytes. Conditioned media from enavatuzumab-treated tumor cells contained elevated levels of chemokines, which might be responsible for enavatuzumab-triggered effector cell migration. These preclinical studies demonstrate that enavatuzumab can exert its potent antitumor activity by actively recruiting and activating myeloid effectors to kill tumor cells. Enavatuzumab-induced chemokines warrant further evaluation in clinical studies as potential biomarkers for such activity.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunotherapy/methods , Lymphocytes/immunology , Monocytes/immunology , Myeloid Cells/immunology , Neoplasms, Experimental/drug therapy , Animals , Antibody-Dependent Cell Cytotoxicity , Cell Movement , Cytokine TWEAK/immunology , Cytokines/metabolism , HCT116 Cells , Humans , Immunity, Innate , Mice , Mice, SCID , Receptors, Fc/metabolism , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Leuk Lymphoma ; 45(2): 301-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15101715

ABSTRACT

Excess apoptosis leading to ineffective hematopoiesis is a common feature of myelodysplastic syndrome (MDS). CD178 (Fas ligand/APO-1 ligand) and CD137 ligand (CD137L), 2 molecules involved in the regulation of apoptosis, have previously been found in sera of patients with malignancies and have been hypothesized to participate in the pathogenesis of various diseases. We analyzed sera of patients with MDS and found that while time to progression of MDS correlated with the IPSS score there was no correlation of CD137L or CD178 serum levels with this score or with karyotype, bone marrow blast count or cytopenia. However, when cut-off-values for significant differentiation between cases with higher/lower levels of these molecules were determined we found that high levels of soluble CD137L (sCD137L) and low serum levels of soluble CD178 (sCD178) correlate with statistical significance to rapid progression of disease as estimated by log-rank-test. Conversely, low levels of sCD137L and high levels of sCD178 correlate significantly with prolongation of time to progression of disease. Our results indicate that serum levels of sCD137L and sCD178 represent valuable novel indicators for prognosis and disease progression and may be a useful parameter for treatment decisions in patients with MDS.


Subject(s)
Membrane Glycoproteins/blood , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/pathology , Prognosis , Receptors, Nerve Growth Factor/blood , Receptors, Tumor Necrosis Factor/blood , Adult , Aged , Aged, 80 and over , Antigens, CD , Disease Progression , Fas Ligand Protein , Female , Humans , Ligands , Male , Middle Aged , Time Factors , Tumor Necrosis Factor Receptor Superfamily, Member 9
7.
Front Immunol ; 4: 505, 2014.
Article in English | MEDLINE | ID: mdl-24409185

ABSTRACT

TweakR is a TNF receptor family member, whose natural ligand is the multifunctional cytokine TWEAK. The growth inhibitory activity observed following TweakR stimulation in certain cancer cell lines and the overexpression of TweakR in many solid tumor types led to the development of enavatuzumab (PDL192), a humanized IgG1 monoclonal antibody to TweakR. The purpose of this study was to determine the mechanism of action of enavatuzumab's tumor growth inhibition and to provide insight into the biology behind TweakR as a cancer therapeutic target. A panel of 105 cancer lines was treated with enavatuzumab in vitro; and 29 cell lines of varying solid tumor backgrounds had >25% growth inhibition in response to the antibody. Treatment of sensitive cell lines with enavatuzumab resulted in the in vitro and in vivo (xenograft) activation of both classical (p50, p65) and non-classical (p52, RelB) NFκB pathways. Using NFκB DNA binding functional ELISAs and microarray analysis, we observed increased activation of NFκB subunits and NFκB-regulated genes in sensitive cells over that observed in resistant cell lines. Inhibiting NFκB subunits (p50, p65, RelB, p52) and upstream kinases (IKK1, IKK2) with siRNA and chemical inhibitors consistently blocked enavatuzumab's activity. Furthermore, enavatuzumab treatment resulted in NFκB-dependent reduction in cell division as seen by the activation of the cell cycle inhibitor p21 both in vitro and in vivo. The finding that NFκB drives the growth inhibitory activity of enavatuzumab suggests that targeting TweakR with enavatuzumab may represent a novel cancer treatment strategy.

8.
Mol Immunol ; 56(4): 819-28, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24036152

ABSTRACT

The role of the B7 family molecules in the regulation of the immune response is well documented. A large body of experimental evidence indicates that costimulatory molecules such as B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3 and B7-H4 are critical for initiation, maintenance and down-regulation of the immune response. However the immunological function of butyrophilin (BTN)-like molecules, which are a part of the expanded B7 family, is not known. Here, we demonstrate that the extracellular portion of human BTNL8 can augment Ag-induced activation of T lymphocytes. BTNL8 has two alternatively spliced forms: B7-like and BTN-like. Both isoforms of BTNL8 were expressed concurrently in various human tissues. A putative BTNL8 receptor was detected only on resting T lymphocytes. Administration of BTNL8Ig fusion protein into mice promoted production of Ag-specific IgG during the primary, but not the secondary immune responses. BTNL8 may therefore play an essential role in priming of naïve T lymphocytes.


Subject(s)
Antigens/immunology , Lymphocyte Activation/immunology , Membrane Proteins/immunology , T-Lymphocytes/immunology , Alternative Splicing , Amino Acid Sequence , Animals , B7 Antigens/genetics , B7 Antigens/immunology , Butyrophilins , CHO Cells , Cricetinae , Cricetulus , Female , HEK293 Cells , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Jurkat Cells , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , T-Lymphocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology
9.
J Cancer Res Clin Oncol ; 139(2): 315-25, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23073510

ABSTRACT

BACKGROUND: The receptor for the cytokine TWEAK (TweakR) is a cell surface member of the tumor necrosis factor receptor superfamily with diverse biological roles. TNFRSF family members are appealing therapeutic targets in oncology due to their aberrant expression and function in tumor cells. The goal of the current study was to examine the potential of TweakR as a therapeutic target in breast cancer. METHODS: Expression of TweakR in primary breast cancer tissues and metastases was characterized using immunohistochemistry. To determine the functional relevance of TweakR, breast cancer cell lines were treated in vitro and in vivo with enavatuzumab, a humanized mAb against TweakR. RESULTS: Overexpression of TweakR was observed in infiltrating tumors compared to normal adjacent breast tissues, and strong staining of TweakR was observed in all subtypes of invasive ductal breast cancer. In addition, a positive correlation of TweakR and HER2 expression and co-localization were observed, irrespective of ER status. TweakR expression was also observed in bone metastasis samples from primary breast cancer but rarely in benign tumors. Enavatuzumab inhibited the in vitro growth of TweakR-expressing breast cancer cell lines, and this activity was augmented by cross-linking the mAb. In addition, enavatuzumab significantly inhibited the in vivo growth of multiple breast cancer xenograft models including a model of metastasis. CONCLUSIONS: TweakR is highly expressed in all subtypes of invasive ductal breast cancer, and enavatuzumab administration exhibited a dose-dependent inhibition of primary tumor growth and lung metastasis and enhanced the antitumor activity of several chemotherapy agents currently used to treat breast cancer. These data provide the rationale to evaluate enavatuzumab as a potential therapy for the treatment of breast cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Ductal, Breast/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Female , Gene Expression , Humans , Mice , Neoplasm Invasiveness/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Tumor Necrosis Factor/genetics , TWEAK Receptor , Trastuzumab , Xenograft Model Antitumor Assays
10.
Arthritis Res Ther ; 15(6): R207, 2013.
Article in English | MEDLINE | ID: mdl-24299175

ABSTRACT

INTRODUCTION: Targeting the CD20 antigen has been a successful therapeutic intervention in the treatment of rheumatoid arthritis (RA). However, in some patients with an inadequate response to anti-CD20 therapy, a persistence of CD20- plasmablasts is noted. The strong expression of CD319 on CD20- plasmablast and plasma cell populations in RA synovium led to the investigation of the potential of CD319 as a therapeutic target. METHODS: PDL241, a novel humanized IgG1 monoclonal antibody (mAb) to CD319, was generated and examined for its ability to inhibit immunoglobulin production from plasmablasts and plasma cells generated from peripheral blood mononuclear cells (PBMC) in the presence and absence of RA synovial fibroblasts (RA-SF). The in vivo activity of PDL241 was determined in a human PBMC transfer into NOD scid IL-2 gamma chain knockout (NSG) mouse model. Finally, the ability of PDL241 to ameliorate experimental arthritis was evaluated in a collagen-induced arthritis (CIA) model in rhesus monkeys. RESULTS: PDL241 bound to plasmablasts and plasma cells but not naïve B cells. Consistent with the binding profile, PDL241 inhibited the production of IgM from in vitro PBMC cultures by the depletion of CD319+ plasmablasts and plasma cells but not B cells. The activity of PDL241 was dependent on an intact Fc portion of the IgG1 and mediated predominantly by natural killer cells. Inhibition of IgM production was also observed in the human PBMC transfer to NSG mouse model. Treatment of rhesus monkeys in a CIA model with PDL241 led to a significant inhibition of anti-collagen IgG and IgM antibodies. A beneficial effect on joint related parameters, including bone remodeling, histopathology, and joint swelling was also observed. CONCLUSIONS: The activity of PDL241 in both in vitro and in vivo models highlights the potential of CD319 as a therapeutic target in RA.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibody Formation/drug effects , Arthritis, Rheumatoid/immunology , Plasma Cells/immunology , Receptors, Immunologic/immunology , Animals , Flow Cytometry , Heterografts , Humans , Immunoglobulins/biosynthesis , Immunohistochemistry , Macaca mulatta , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Signaling Lymphocytic Activation Molecule Family , Synovial Membrane/immunology , Synovial Membrane/metabolism
11.
J Neuroimmunol ; 212(1-2): 65-73, 2009 Jul 25.
Article in English | MEDLINE | ID: mdl-19477024

ABSTRACT

Humanization and modification of the Fc region of anti-human CD3 mAbs have greatly expanded their potential use in chronic T cell mediated diseases. However, low levels of cytokine release and immunogenicity may still impact a chronic dosing strategy. We investigated the use of an Fc-modified murine chimeric anti-mouse CD3 (N297A) in the chronic MOG(35-55)-induced EAE mouse model of MS. Two daily doses of 10 microg at the onset of clinical symptoms led to both a reduction in T cell numbers in the blood and a significant, prolonged reduction in the symptoms. Histological examination of the spinal cords at the peak of efficacy confirmed a reduction of infiltrating T cells in the CNS. Analysis of the cerebral spinal fluid from EAE mice showed biologically active levels of N297A. Analysis of the cytokine/chemokine levels in cerebrospinal fluid showed a decrease in GM-CSF, IL-6 and IP-10. The combination of N297A dosing with cyclosporine A (CSA) pretreatment showed a significant decrease of TNFalpha, IL-6 and IP-10 without effect on clinical efficacy. However, pretreatment of CSA significantly reduced the immunogenic response observed following a second course of N297A treatment. Therefore, the side effects of an Fc-modified anti-CD3 mAb may be modulated without affecting efficacy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD3 Complex/immunology , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/therapy , Immunoglobulin Fc Fragments/therapeutic use , Animals , Blood-Brain Barrier , Chemokines/biosynthesis , Cyclosporine/pharmacology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spinal Cord/immunology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology
12.
Int Immunol ; 18(3): 473-84, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16481347

ABSTRACT

Members of the T cell, Ig domain and mucin domain (Tim) family of proteins have recently been implicated in the control of T cell-mediated immune responses. Tim-1 (HUGO designation HAVCR1) polymorphisms have been linked to the regulation of atopy in mice and humans, suggestive of a role in immune regulation. Tim-1 is expressed upon activation of T cells. In concert with the increased expression of Tim-1, a binding partner for the extracellular domain of Tim-1 (eTim-1) was induced on activated T cells, and mRNA expression data was consistent with the binding partner being Tim-4. We found that co-immobilized recombinant eTim-1 was able to inhibit T cell activation mediated by CD3 + CD28 mAb. eTim-1 mediated its inhibitory effects on proliferation by arresting cell cycle at G(0)/G(1) phase through regulation of cell cycle proteins. In vivo, administration of eTim-1 proteins led to a decrease in both ear (contact hypersensitivity to oxazolone) and joint (methylated BSA antigen-induced arthritis) swelling. The inhibitory activity of eTim-1 in the T(h)1-dependent models was evidence that eTim-1 is able to modulate T cell responses. Manipulation of the Tim-1 interaction with its binding partner on T cells may therefore provide a novel target for therapeutic intervention in T cell-mediated diseases.


Subject(s)
Membrane Glycoproteins/immunology , Receptors, Virus/immunology , T-Lymphocytes/immunology , Animals , Arthritis/immunology , Arthritis/prevention & control , CHO Cells , Cell Cycle , Cricetinae , Cricetulus , Dermatitis, Contact/prevention & control , G1 Phase , Hepatitis A Virus Cellular Receptor 1 , Humans , Lymphocyte Activation , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Receptors, Interleukin-2/biosynthesis , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombinant Fusion Proteins/immunology , Resting Phase, Cell Cycle , T-Lymphocytes/cytology
14.
Br J Haematol ; 117(1): 76-85, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11918536

ABSTRACT

The survival of leukaemic blasts contributes to the pathological mechanism of acute promyelocytic leukaemia (APL). While treatment of APL using retinoic acid (RA) is a model of differentiation therapy, little is known about possible effects of this treatment on the Fas/FasL system. Investigation of APL cells from patients undergoing differentiation therapy with RA and of promyelocytic HL-60 and monoblastic U-937 cells cultured with RA revealed a reduction of surface expression of both Fas and its ligand. Accordingly, the sensitivity of the cells to anti-Fas-induced apoptosis decreased proportionally and the reduced expression of FasL resulted in a decreased ability of the leukaemic cells to induce apoptosis in T cells. Our findings demonstrate that there are significant changes in Fas and FasL expression during RA treatment of APL, which probably have consequences for the interaction between host immune and leukaemia cells, and thus may be involved in the beneficial effects of differentiation therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/immunology , Membrane Glycoproteins/analysis , Tretinoin/therapeutic use , fas Receptor/analysis , Apoptosis/drug effects , Blotting, Western/methods , Cell Differentiation/drug effects , Coculture Techniques , Fas Ligand Protein , Flow Cytometry , HL-60 Cells , Humans , Jurkat Cells , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL