Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Brain Behav Immun ; 27(1): 185-92, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23103445

ABSTRACT

Neuropathic pain is a debilitating condition of the somatosensory system caused by pathology of the nervous system. Current drugs treat symptoms but largely fail to target the underlying mechanisms responsible for the pathological changes seen in the central or peripheral nervous system. We investigated the therapeutic effects of PDA-001, a culture expanded placenta-derived adherent cell, in the rat neuritis model. Pain is induced in the model by applying carrageenan to the sciatic nerve trunk, causing perineural inflammation of the sciatic nerve. PDA-001, at doses ranging from 0.4×10(6) to 4×10(6) cells/animal, or vehicle control was intravenously administrated to assess the biological activity of the cells. A dose-dependent effect of PDA-001 on pain relief was demonstrated. PDA-001 at doses of 1×10(6) and 4×10(6), but not 0.4×10(6), reduced mechanical hyperalgesia within 24h following treatment and through day 8 after induction of neuritis. The mechanism underlying PDA-001-mediated reduction of neuroinflammatory pain was also explored. Ex vivo tissue analyses demonstrated that PDA-001 suppressed homing, maturation and differentiation of dendritic cells, thus inhibiting T-cell priming and activation in draining lymph nodes. PDA-001 also reduced interferon gamma and IL-17 in draining lymph nodes and in the ispilateral sciatic nerve, and increased the levels of IL-10 in draining lymph nodes and plasma, pointing to T-cell modulation as a possible mechanism mediating the observed anti-hyperalgesic effects. Furthermore, in the ipsilateral sciatic nerve, significantly less leukocyte infiltration was observed in PDA-001-treated animals. The results suggest that PDA-001may provide a novel therapeutic approach in the management of inflammatory neuropathic pain and similar conditions.


Subject(s)
Cell Transplantation/methods , Hyperalgesia , Neuralgia , Neuritis , Placenta , Sciatic Neuropathy , Animals , Carrageenan/adverse effects , Cell Differentiation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Humans , Hyperalgesia/chemically induced , Hyperalgesia/immunology , Hyperalgesia/therapy , Male , Neuralgia/chemically induced , Neuralgia/immunology , Neuralgia/therapy , Neuritis/chemically induced , Neuritis/immunology , Neuritis/therapy , Placenta/cytology , Placenta/immunology , Placenta/transplantation , Pregnancy , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/chemically induced , Sciatic Neuropathy/immunology , Sciatic Neuropathy/therapy , T-Lymphocytes/cytology , T-Lymphocytes/immunology
2.
Clin Cancer Res ; 11(8): 3126-35, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15837769

ABSTRACT

PURPOSE: Substantial evidence indicates that supraoligomerization of the death receptors for Fas ligand and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is necessary for efficient activation of the apoptotic pathway. Bivalent IgG antibodies can induce the efficient apoptosis by mimicking the natural ligands but only after these antibodies are further oligomerized by cross-linking. In this study, we generated a novel agonist antibody to TRAIL receptor 2 (TRAIL-R2) capable of inducing apoptosis without cross-linking and elucidated its mode of action and efficacy. EXPERIMENTAL DESIGN: A fully human antibody to TRAIL-R2, KMTR2, was generated from KM Mouse immunized with TRAIL-R2 ectodomain. Apoptosis-inducing activities of unfractionated or purified monomeric IgG of KMTR2 was evaluated in the presence or absence of cross-linkers, secondary antibodies or Fc receptor-expressing effector cells, against human colorectal adenocarcinoma Colo205. Oligomerization of TRAIL-R2 was analyzed by size exclusion chromatography and confocal microscopy, and in vivo efficacy was examined in Colo205 xenograft model. RESULTS: KMTR2 specifically recognized TRAIL-R2 and induced apoptosis with or without cross-linking. Size exclusion chromatography showed that the apoptosis activity coeluted with monomeric IgG and was effective independent of secondary antibody or Fc receptor-expressing effector cells. The antibody formed supracomplexes with soluble recombinant and membrane-anchored TRAIL-R2 and enhanced clustering of TRAIL-R2 on cell surface without cross-linking. KMTR2 was dramatically efficacious in reducing established human tumor. CONCLUSION: Our findings indicate that novel agonist antibody KMTR2 can direct antibody-dependent oligomerization of TRAIL-R2 and initiates efficient apoptotic signaling and tumor regression independent of host effector function. Thus, the direct agonist would be a lead candidate for cancer therapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Receptors, Tumor Necrosis Factor/immunology , Xenograft Model Antitumor Assays/methods , Animals , Antibody Specificity , Cell Line, Tumor , Cell Survival/drug effects , Dimerization , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/chemistry , Remission Induction
3.
J Mol Biol ; 334(1): 103-18, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-14596803

ABSTRACT

It is well established that the humoral immune response can generate antibodies to many different antigens. The antibody diversity required to achieve this is believed to be substantial. However, the extent to which the immune repertoire can generate structural diversity against a single target antigen has never been addressed. Here, we have used phage display to demonstrate the extraordinary capacity of the human antibody repertoire. Over 1000 antibodies, all different in amino acid sequence, were generated to a single protein, B-lymphocyte stimulator (BLyS protein). This is a highly diverse panel of antibodies as exemplified by the extensive heavy and light chain germline usage: 42/49 functional heavy chain germlines and 19/33 V(lambda) and 13/35 V(kappa) light chain germlines were all represented in the panel of antibodies. Moreover, a high level of sequence diversity was observed in the V(H) CDR3 domains of these antibodies, with 568 different amino acid sequences identified. Thus we have demonstrated that specific recognition of a single antigen can be achieved from many different VDJ combinations, illustrating the remarkable problem-solving ability of the human immune repertoire. When studied in a biochemical assay, around 500 (40%) of these antibodies inhibited the binding of BLyS to its receptors on B-cell lines. The most potent antibodies inhibited BLyS binding with sub-nanomolar IC(50) values and with sub-nanomolar affinities. Such antibodies provide excellent choices as candidates for the treatment of BLyS-associated autoimmune diseases.


Subject(s)
Antibodies , Membrane Proteins/immunology , Tumor Necrosis Factor-alpha/immunology , Amino Acid Sequence , Antibodies/chemistry , Antibodies/classification , Antibodies/genetics , Antibodies/immunology , B-Cell Activating Factor , Complementarity Determining Regions , Humans , Immunoglobulin Heavy Chains , Immunoglobulin Light Chains , Molecular Sequence Data , Peptide Library , Phylogeny , Point Mutation
4.
Clin Transl Immunology ; 3(5): e14, 2014 May.
Article in English | MEDLINE | ID: mdl-25505962

ABSTRACT

Human placenta-derived adherent cells (PDAC cells) are a culture expanded, undifferentiated mesenchymal-like population derived from full-term placental tissue, with immunomodulatory and anti-inflammatory properties. PDA-001 (cenplacel-L), an intravenous formulation of PDAC cells, is in clinical development for the treatment of autoimmune and inflammatory diseases. To elucidate the mechanisms underlying the immunoregulatory properties of PDAC cells, we investigated their effects on immune cell populations, including T cells and dendritic cells (DC) in vitro and in vivo. PDAC cells suppressed T-cell proliferation in an OT-II T-cell adoptive transfer model, reduced the severity of myelin oligodendrocyte glycoprotein peptide-induced experimental autoimmune encephalomyelitis and ameliorated inflammation in a delayed type hypersensitivity response model. In vitro, PDAC cells suppressed T-cell proliferation and inhibited Th1 and Th17 differentiation. Analysis of tissues derived from PDAC cell-treated animals revealed diminished CD86 expression on splenic DC, suggesting that they can also modulate DC populations. Furthermore, PDAC cells modulate the differentiation and maturation of mouse bone marrow-derived DC. Similarly, human DC differentiated from CD14(+) monocytes in the presence of PDAC cells acquired a tolerogenic phenotype. These tolerogenic DC failed to induce allogeneic T-cell proliferation and differentiation toward Th1, but skewed T-cell differentiation toward Th2. Inhibition of cyclo-oxygenase-2 activity resulted in a significant, but not complete, abrogation of PDAC cells' effects on DC phenotype and function, implying a role for prostaglandin E2 in PDAC-mediated immunomodulation. This study identifies modulation of DC differentiation toward immune tolerance as a key mechanism underlying the immunomodulatory activities of PDAC cells.

5.
MAbs ; 1(6): 552-62, 2009.
Article in English | MEDLINE | ID: mdl-20068388

ABSTRACT

Apoptosis through the TRAIL receptor pathway can be induced via agonistic IgG to either TRAIL-R1 or TRAIL-R2. Here we describe the use of phage display to isolate a substantive panel of fully human anti-TRAIL receptor single chain Fv fragments (scFvs); 234 and 269 different scFvs specific for TRAIL-R1 and TRAIL-R2 respectively. In addition, 134 different scFvs that were cross-reactive for both receptors were isolated. To facilitate screening of all 637 scFvs for potential agonistic activity in vitro, a novel high-throughput surrogate apoptosis assay was developed. Ten TRAIL-R1 specific scFv and 6 TRAIL-R2 specific scFv were shown to inhibit growth of tumor cells in vitro in the absence of any cross-linking agents. These scFv were all highly specific for either TRAIL-R1 or TRAIL-R2, potently inhibited tumor cell proliferation, and were antagonists of TRAIL binding. Moreover, further characterization of TRAIL-R1 agonistic scFv demonstrated significant anti-tumor activity when expressed and purified as a monomeric Fab fragment. Thus, scFv and Fab fragments, in addition to whole IgG, can be agonistic and induce tumor cell death through specific binding to either TRAIL-R1 or TRAIL-R2. These potent agonistic scFv were all isolated directly from the starting phage antibody library and demonstrated significant tumor cell killing properties without any requirement for affinity maturation. Some of these selected scFv have been converted to IgG format and are being studied extensively in clinical trials to investigate their potential utility as human monoclonal antibody therapeutics for the treatment of human cancer.


Subject(s)
Immunoglobulin Fab Fragments/pharmacology , Immunotherapy , Neoplasms/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Single-Chain Antibodies/pharmacology , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Apoptosis/drug effects , Apoptosis/immunology , Binding, Competitive , Caspase 3/metabolism , Cell Proliferation/drug effects , Genetic Engineering , HeLa Cells , High-Throughput Screening Assays , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/metabolism , Neoplasms/immunology , Neoplasms/pathology , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics , Single-Chain Antibodies/isolation & purification
6.
Urology ; 69(2): 395-401, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17320696

ABSTRACT

OBJECTIVES: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in a variety of tumor cells through two of its receptors: TRAIL-R1 and TRAIL-R2. In this study, we investigated the susceptibility of human prostate cancer and bladder cancer cells to HGS-ETR2, a human monoclonal agonistic antibody specific for TRAIL-R2. METHODS: The cell surface expression of TRAIL-R1 and TRAIL-R2 on prostate cancer and bladder cancer cells was determined using flow cytometry. Cytotoxicity was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and caspase activities were measured by a quantitative colorimetric assay. RESULTS: HGS-ETR2 effectively induced apoptotic cell death in DU145, PC3, and LNCaP human prostate cancer cells and J82 and T24 human bladder cancer cells. The increased effectiveness of HGS-ETR2 for inducing cell death might have been affected by differences in the cell surface expression of the two TRAIL receptors, in that TRAIL-R2, but not TRAIL-R1, was frequently expressed in the prostate cancer and bladder cancer cells. HGS-ETR2 significantly activated the caspase cascade, including caspase-3, -6, -8, and -9, which were the downstream molecules of the death receptors in prostate cancer cells. Caspase-3, -6, and -9 were also significantly activated with HGS-ETR2-induced apoptosis in the bladder cancer cells. CONCLUSIONS: These findings suggest the potential utility of TRAIL-R2 antibody as a novel therapeutic agent against prostate cancer and bladder cancer.


Subject(s)
Apoptosis/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/pharmacology , Receptors, Tumor Necrosis Factor, Type II/pharmacology , Apoptosis/physiology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Cell Line, Tumor/drug effects , Flow Cytometry , Humans , Male , Probability , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Sensitivity and Specificity , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology
7.
Br J Haematol ; 130(4): 501-10, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16098063

ABSTRACT

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) is a death protein that preferentially kills tumour cells while sparing normal cells. TRAIL has four exclusive receptors, two of which (TRAIL-R1, TRAIL-R2) are death receptors. Both TRAIL/Apo2L and agonistic antibodies to the TRAIL death receptors are currently being explored for cancer therapy. Although the activity of TRAIL/Apo2L in a variety of haematological malignancies has been examined, the activity of anti-TRAIL receptor agonistic antibodies in primary and cultured lymphoma cells has not. Using two fully human selective agonistic monoclonal antibodies to the TRAIL death receptors TRAIL-R1 (HGS-ETR1) and TRAIL-R2 (HGS-ETR2) this study demonstrated that both monoclonal antibodies activated caspase-8 and induced cell death in five of nine human lymphoma cell lines, and induced >10% cell death in 67% and 70%, respectively, of 27 primary lymphoma cells, and >20% cell death in at least one-thirds of the samples. HGS-ETR1 and HGS-ETR2 demonstrated comparable activity in the fresh tumour samples, which was independent of TRAIL receptor surface expression, Bax, cFLIP, or procaspase-8 expression, or exposure to prior therapy. Furthermore, both antibodies enhanced the killing effect of doxorubicin and bortezomib. Our data demonstrate that HGS-ETR1 and HGS-ETR2 monoclonal antibodies can induce cell death in a variety of cultured and primary lymphoma cells, and may have therapeutic value in lymphoma.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/therapeutic use , Lymphoma/drug therapy , Lymphoma/immunology , Receptors, Tumor Necrosis Factor/agonists , Tumor Cells, Cultured/pathology , Analysis of Variance , Apoptosis/drug effects , Boronic Acids/therapeutic use , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/therapeutic use , Drug Synergism , Fluorescent Antibody Technique , Humans , Pyrazines/therapeutic use , Receptors, TNF-Related Apoptosis-Inducing Ligand , Statistics, Nonparametric , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/immunology
8.
J Immunol ; 169(8): 4314-21, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12370363

ABSTRACT

BLyS and APRIL are two members of the TNF superfamily that are secreted by activated myeloid cells and have costimulatory activity on B cells. BLyS and APRIL share two receptors, TACI and BCMA, whereas a third receptor, BAFF-R, specifically binds BLyS. Both BLyS and APRIL have been described as homotrimeric molecules, a feature common to members of the TNF superfamily. In this study, we show that APRIL and BLyS can form active heterotrimeric molecules when coexpressed and that circulating heterotrimers are present in serum samples from patients with systemic immune-based rheumatic diseases. These findings raise the possibility that active BLyS/APRIL heterotrimers may play a role in rheumatic and other autoimmune diseases and that other members of the TNF ligand superfamily may also form active soluble heterotrimers.


Subject(s)
B-Lymphocytes/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/physiology , Rheumatic Diseases/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/physiology , Animals , Arthritis, Psoriatic/blood , Arthritis, Psoriatic/immunology , Arthritis, Reactive/blood , Arthritis, Reactive/immunology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , B-Cell Activation Factor Receptor , B-Lymphocytes/metabolism , Cell Line , Cells, Cultured , Female , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/genetics , Membrane Proteins/blood , Membrane Proteins/isolation & purification , Mice , Mice, Inbred BALB C , Polymyositis/blood , Polymyositis/immunology , Receptors, Tumor Necrosis Factor/blood , Receptors, Tumor Necrosis Factor/isolation & purification , Rheumatic Diseases/blood , Spondylitis, Ankylosing/blood , Spondylitis, Ankylosing/immunology , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor Ligand Superfamily Member 13 , Tumor Necrosis Factor-alpha/isolation & purification
9.
Arthritis Rheum ; 48(11): 3253-65, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14613291

ABSTRACT

OBJECTIVE: To identify and characterize a fully human antibody directed against B lymphocyte stimulator (BLyS), a tumor necrosis factor-related cytokine that plays a critical role in the regulation of B cell maturation and development. Elevated levels of BLyS have been implicated in the pathogenesis of autoimmune diseases. METHODS: A human phage display library was screened for antibodies against human BLyS. A human monoclonal antibody, LymphoStat-B, specific for human BLyS was obtained from the library screening and subsequent affinity optimization mutagenesis. The antibody was tested for inhibition of human BLyS in vitro and in an in vivo murine model. Additionally, the consequences of BLyS inhibition were tested in vivo by administration of LymphoStat-B to cynomolgus monkeys. RESULTS: LymphoStat-B bound with high affinity to human BLyS and inhibited the binding of BLyS to its 3 receptors, TACI, BCMA, and BLyS receptor 3/BAFF-R. LymphoStat-B potently inhibited BLyS-induced proliferation of B cells in vitro, and administration of LymphoStat-B to mice prevented human BLyS-induced increases in splenic B cell numbers and IgA titers. In cynomolgus monkeys, administration of LymphoStat-B resulted in decreased B cell representation in both spleen and mesenteric lymph nodes. CONCLUSION: A fully human monoclonal antibody has been isolated that binds to BLyS with high affinity and neutralizes human BLyS bioactivity in vitro and in vivo. Administration of this antibody to cynomolgus monkeys resulted in B cell depletion in spleen and lymph node. This antibody may prove therapeutically useful in the treatment of autoimmune diseases in humans.


Subject(s)
Antibodies, Monoclonal/biosynthesis , B-Lymphocytes/metabolism , Membrane Proteins , Receptors, Tumor Necrosis Factor/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , B-Cell Activation Factor Receptor , B-Cell Maturation Antigen , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Division/drug effects , Dose-Response Relationship, Drug , Female , Humans , Injections, Intravenous , Leukocytes, Mononuclear/drug effects , Lymph Nodes/cytology , Lymph Nodes/drug effects , Macaca fascicularis , Male , Mice , Mutagenesis, Site-Directed , Neutralization Tests , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/immunology , Spleen/cytology , Spleen/drug effects , Transmembrane Activator and CAML Interactor Protein
SELECTION OF CITATIONS
SEARCH DETAIL