Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
MAbs ; 8(1): 150-62, 2016.
Article in English | MEDLINE | ID: mdl-26431358

ABSTRACT

Semaphorin 4D (SEMA4D or CD100) is a member of the semaphorin family of proteins and an important mediator of the movement and differentiation of multiple cell types, including those of the immune, vascular, and nervous systems. Blocking the binding of SEMA4D to its receptors can result in physiologic changes that may have implications in cancer, autoimmune, and neurological disease. To study the effects of blocking SEMA4D, we generated, in SEMA4D-deficient mice, a panel of SEMA4D-specific hybridomas that react with murine, primate, and human SEMA4D. Utilizing the complementarity-determining regions from one of these hybridomas (mAb 67-2), we generated VX15/2503, a humanized IgG4 monoclonal antibody that is currently in clinical development for the potential treatment of various malignancies and neurodegenerative disorders, including multiple sclerosis and Huntington's disease. This work describes the generation and characterization of VX15/2503, including in vitro functional testing, epitope mapping, and an in vivo demonstration of efficacy in an animal model of rheumatoid arthritis.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neutralizing/immunology , Antibody Specificity , Semaphorins/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Neutralizing/pharmacology , Antigens, CD/immunology , Humans , Mice , Mice, Knockout , Semaphorins/immunology
2.
BMC Immunol ; 16: 6, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25879435

ABSTRACT

BACKGROUND: Homeostatic B Cell-Attracting chemokine 1 (BCA-1) otherwise known as CXCL13 is constitutively expressed in secondary lymphoid organs by follicular dendritic cells (FDC) and macrophages. It is the only known ligand for the CXCR5 receptor, which is expressed on mature B cells, follicular helper T cells (Tfh), Th17 cells and regulatory T (Treg) cells. Aberrant expression of CXCL13 within ectopic germinal centers has been linked to the development of autoimmune disorders (e.g. Rheumatoid Arthritis, Multiple Sclerosis, Systemic Lupus Erythematosis). We, therefore, hypothesized that antibody-mediated disruption of the CXCL13 signaling pathway would interfere with the formation of ectopic lymphoid follicles in the target organs and inhibit autoimmune disease progression. This work describes pre-clinical development of human anti-CXCL13 antibody MAb 5261 and includes therapeutic efficacy data of its mouse counterpart in murine models of autoimmunity. RESULTS: We developed a human IgG1 monoclonal antibody, MAb 5261 that specifically binds to human, rodent and primate CXCL13 with an affinity of approximately 5 nM and is capable of neutralizing the activity of CXCL13 from these various species in in vitro functional assays. For in vivo studies we have engineered a chimeric antibody to contain the same human heavy and light chain variable genes along with mouse constant regions. Treatment with this antibody led to a reduction in the number of germinal centers in mice immunized with 4-Hydroxy-3-nitrophenylacetyl hapten conjugated to Keyhole Limpet Hemocyanin (NP-KLH) and, in adoptive transfer studies, interfered with the trafficking of B cells to the B cell areas of mouse spleen. Furthermore, this mouse anti-CXCL13 antibody demonstrated efficacy in a mouse model of Rheumatoid arthritis (Collagen-Induced Arthritis (CIA)) and Th17-mediated murine model of Multiple Sclerosis (passively-induced Experimental Autoimmune Encephalomyelitis (EAE)). CONCLUSIONS: We developed a novel therapeutic antibody targeting CXCL13-mediated signaling pathway for the treatment of autoimmune disorders.


Subject(s)
Antibodies, Blocking/administration & dosage , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , B-Lymphocytes/drug effects , Chemokine CXCL13/metabolism , Dendritic Cells, Follicular/drug effects , Encephalomyelitis, Autoimmune, Experimental/therapy , Immunoglobulin G/administration & dosage , Immunotherapy/methods , Macrophages/drug effects , Multiple Sclerosis/therapy , Recombinant Fusion Proteins/administration & dosage , Th17 Cells/drug effects , Animals , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , B-Lymphocytes/immunology , Cell Movement/drug effects , Cells, Cultured , Chemokine CXCL13/immunology , Dendritic Cells, Follicular/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Genetic Engineering , Germinal Center/drug effects , Hemocyanins/chemistry , Hemocyanins/immunology , Humans , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Multiple Sclerosis/immunology , Nitrophenols/chemistry , Nitrophenols/immunology , Phenylacetates/chemistry , Phenylacetates/immunology , Receptors, CXCR5/metabolism , Signal Transduction/drug effects , Th17 Cells/immunology
3.
Neurobiol Dis ; 76: 46-56, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25662335

ABSTRACT

Huntington disease (HD) is an inherited, fatal neurodegenerative disease with no disease-modifying therapy currently available. In addition to characteristic motor deficits and atrophy of the caudate nucleus, signature hallmarks of HD include behavioral abnormalities, immune activation, and cortical and white matter loss. The identification and validation of novel therapeutic targets that contribute to these degenerative cellular processes may lead to new interventions that slow or even halt the course of this insidious disease. Semaphorin 4D (SEMA4D) is a transmembrane signaling molecule that modulates a variety of processes central to neuroinflammation and neurodegeneration including glial cell activation, neuronal growth cone collapse and apoptosis of neural precursors, as well as inhibition of oligodendrocyte migration, differentiation and process formation. Therefore, inhibition of SEMA4D signaling could reduce CNS inflammation, increase neuronal outgrowth and enhance oligodendrocyte maturation, which may be of therapeutic benefit in the treatment of several neurodegenerative diseases, including HD. To that end, we evaluated the preclinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody, which prevents the interaction between SEMA4D and its receptors, in the YAC128 transgenic HD mouse model. Anti-SEMA4D treatment ameliorated neuropathological signatures, including striatal atrophy, cortical atrophy, and corpus callosum atrophy and prevented testicular degeneration in YAC128 mice. In parallel, a subset of behavioral symptoms was improved in anti-SEMA4D treated YAC128 mice, including reduced anxiety-like behavior and rescue of cognitive deficits. There was, however, no discernible effect on motor deficits. The preservation of brain gray and white matter and improvement in behavioral measures in YAC128 mice treated with anti-SEMA4D suggest that this approach could represent a viable therapeutic strategy for the treatment of HD. Importantly, this work provides in vivo demonstration that inhibition of pathways initiated by SEMA4D constitutes a novel approach to moderation of neurodegeneration.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Huntington Disease/therapy , Semaphorins/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Brain/metabolism , Brain/pathology , Cognition Disorders/etiology , Cognition Disorders/therapy , Disease Models, Animal , Huntington Disease/complications , Immunotherapy , Mice , Mice, Transgenic , Motor Activity/drug effects , Signal Transduction/drug effects
4.
Cancer Immunol Res ; 3(6): 689-701, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25614511

ABSTRACT

Semaphorin 4D (SEMA4D, CD100) and its receptor plexin-B1 (PLXNB1) are broadly expressed in murine and human tumors, and their expression has been shown to correlate with invasive disease in several human tumors. SEMA4D normally functions to regulate the motility and differentiation of multiple cell types, including those of the immune, vascular, and nervous systems. In the setting of cancer, SEMA4D-PLXNB1 interactions have been reported to affect vascular stabilization and transactivation of ERBB2, but effects on immune-cell trafficking in the tumor microenvironment (TME) have not been investigated. We describe a novel immunomodulatory function of SEMA4D, whereby strong expression of SEMA4D at the invasive margins of actively growing tumors influences the infiltration and distribution of leukocytes in the TME. Antibody neutralization of SEMA4D disrupts this gradient of expression, enhances recruitment of activated monocytes and lymphocytes into the tumor, and shifts the balance of cells and cytokines toward a proinflammatory and antitumor milieu within the TME. This orchestrated change in the tumor architecture was associated with durable tumor rejection in murine Colon26 and ERBB2(+) mammary carcinoma models. The immunomodulatory activity of anti-SEMA4D antibody can be enhanced by combination with other immunotherapies, including immune checkpoint inhibition and chemotherapy. Strikingly, the combination of anti-SEMA4D antibody with antibody to CTLA-4 acts synergistically to promote complete tumor rejection and survival. Inhibition of SEMA4D represents a novel mechanism and therapeutic strategy to promote functional immune infiltration into the TME and inhibit tumor progression.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Neoplasms/immunology , Semaphorins/antagonists & inhibitors , Semaphorins/immunology , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Cyclophosphamide/pharmacology , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Female , Humans , Immunologic Memory , Immunomodulation/drug effects , Immunotherapy , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
5.
Neurobiol Dis ; 73: 254-68, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25461192

ABSTRACT

Multiple sclerosis (MS) is a chronic neuroinflammatory disease characterized by immune cell infiltration of CNS, blood-brain barrier (BBB) breakdown, localized myelin destruction, and progressive neuronal degeneration. There exists a significant need to identify novel therapeutic targets and strategies that effectively and safely disrupt and even reverse disease pathophysiology. Signaling cascades initiated by semaphorin 4D (SEMA4D) induce glial activation, neuronal process collapse, inhibit migration and differentiation of oligodendrocyte precursor cells (OPCs), and disrupt endothelial tight junctions forming the BBB. To target SEMA4D, we generated a monoclonal antibody that recognizes mouse, rat, monkey and human SEMA4D with high affinity and blocks interaction between SEMA4D and its cognate receptors. In vitro, anti-SEMA4D reverses the inhibitory effects of recombinant SEMA4D on OPC survival and differentiation. In vivo, anti-SEMA4D significantly attenuates experimental autoimmune encephalomyelitis in multiple rodent models by preserving BBB integrity and axonal myelination and can be shown to promote migration of OPC to the site of lesions and improve myelin status following chemically-induced demyelination. Our study underscores SEMA4D as a key factor in CNS disease and supports the further development of antibody-based inhibition of SEMA4D as a novel therapeutic strategy for MS and other neurologic diseases with evidence of demyelination and/or compromise to the neurovascular unit.


Subject(s)
Blood-Brain Barrier/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Oligodendroglia/metabolism , Semaphorins/metabolism , Animals , Antibodies, Monoclonal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Semaphorins/antagonists & inhibitors , Semaphorins/immunology
6.
EMBO J ; 29(2): 410-23, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20019666

ABSTRACT

We have shown earlier that DNA polymerase beta (Pol beta) localizes to the synaptonemal complex (SC) during Prophase I of meiosis in mice. Pol beta localizes to synapsed axes during zygonema and pachynema, and it associates with the ends of bivalents during late pachynema and diplonema. To test whether these localization patterns reflect a function for Pol beta in recombination and/or synapsis, we used conditional gene targeting to delete the PolB gene from germ cells. We find that Pol beta-deficient spermatocytes are defective in meiotic chromosome synapsis and undergo apoptosis during Prophase I. We also find that SPO11-dependent gammaH2AX persists on meiotic chromatin, indicating that Pol beta is critical for the repair of SPO11-induced double-strand breaks (DSBs). Pol beta-deficient spermatocytes yielded reduced steady-state levels of the SPO11-oligonucleotide complexes that are formed when SPO11 is removed from the ends of DSBs, and cytological experiments revealed that chromosome-associated foci of replication protein A (RPA), RAD51 and DMC1 are less abundant in Pol beta-deficient spermatocyte nuclei. Localization of Pol beta to meiotic chromosomes requires the formation of SPO11-dependent DSBs. Taken together, these findings strongly indicate that Pol beta is required at a very early step in the processing of meiotic DSBs, at or before the removal of SPO11 from DSB ends and the generation of the 3' single-stranded tails necessary for subsequent strand exchange. The chromosome synapsis defects and Prophase I apoptosis of Pol beta-deficient spermatocytes are likely a direct consequence of these recombination defects.


Subject(s)
Chromosome Pairing , DNA Polymerase beta/metabolism , Meiosis , Mice/metabolism , Spermatocytes/enzymology , Animals , Chromosomes/metabolism , DNA Breaks, Double-Stranded , DNA Polymerase beta/genetics , DNA Repair , Endodeoxyribonucleases , Esterases/metabolism , Female , Gene Deletion , Male , Seminiferous Tubules/cytology , Seminiferous Tubules/ultrastructure
7.
Mol Cancer Ther ; 5(11): 2919-30, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17121940

ABSTRACT

Identification of shared tumor-specific targets is useful in developing broadly applicable therapies. In a study designed to identify genes up-regulated in breast cancer, a cDNA clone corresponding to a novel gene C35 (C17orf37) was selected by representational difference analysis of tumor and normal human mammary cell lines. Abundant expression of C35 transcript in tumors was confirmed by Northern blot and real-time PCR. The C35 gene is located on chromosome 17q12, 505 nucleotides from the 3' end of the ERBB2 oncogene, the antigenic target for trastuzumab (Herceptin) therapy. The chromosomal arrangement of the genes encoding C35 and ERBB2 is tail to tail. An open reading frame encodes a 12-kDa protein of unknown function. Immunohistochemical analysis detected robust and frequent expression of C35 protein, including 32% of grade 1 and 66% of grades 2 and 3 infiltrating ductal carcinomas of the breast (in contrast to 20% overexpressing HER-2/neu), 38% of infiltrating lobular carcinoma (typically HER-2/neu negative), as well as tumors arising in other tissues. C35 was not detected in 38 different normal human tissues, except Leydig cells in the testes and trace levels in a small percentage of normal breast tissue samples. The distinct and favorable expression profile of C35 spanning early through late stages of disease, including high frequency of overexpression in various breast carcinoma, abundant expression in distant metastases, and either absence or low level expression in normal human tissues, warrants further investigation of the relevance of C35 as a biomarker and/or a target for development of broadly applicable cancer-specific therapies.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Neoplasm Proteins/genetics , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , DNA, Complementary/metabolism , Female , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Neoplasm Proteins/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Cells, Cultured
8.
PLoS Biol ; 2(11): e327, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15502868

ABSTRACT

The hallmark of Parkinson's disease (PD) is the selective loss of dopamine neurons in the ventral midbrain. Although the cause of neurodegeneration in PD is unknown, a Mendelian inheritance pattern is observed in rare cases, indicating a genetic factor. Furthermore, pathological analyses of PD substantia nigra have correlated cellular oxidative stress and altered proteasomal function with PD. Homozygous mutations in DJ-1 were recently described in two families with autosomal recessive Parkinsonism, one of which is a large deletion that is likely to lead to loss of function. Here we show that embryonic stem cells deficient in DJ-1 display increased sensitivity to oxidative stress and proteasomal inhibition. The accumulation of reactive oxygen species in toxin-treated DJ-1-deficient cells initially appears normal, but these cells are unable to cope with the consequent damage that ultimately leads to apoptotic death. Furthermore, we find that dopamine neurons derived from in vitro-differentiated DJ-1-deficient embryonic stem cells display decreased survival and increased sensitivity to oxidative stress. These data are consistent with a protective role for DJ-1, and demonstrate the utility of genetically modified embryonic stem cell-derived neurons as cellular models of neuronal disorders.


Subject(s)
Dopamine/metabolism , Embryo, Mammalian/cytology , Neurons/metabolism , Oncogene Proteins/genetics , Oxidative Stress , Parkinson Disease/metabolism , Stem Cells/cytology , Animals , Apoptosis , Cell Differentiation , Cell Survival , DNA, Complementary/metabolism , Disease Models, Animal , Gene Deletion , Genetic Vectors , Heterozygote , Homozygote , Humans , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Fluorescence , Models, Genetic , Molecular Sequence Data , Mutation , Neurodegenerative Diseases/metabolism , Peroxiredoxins , Proteasome Endopeptidase Complex/metabolism , Protein Deglycase DJ-1 , RNA Interference , Reactive Oxygen Species , Reverse Transcriptase Polymerase Chain Reaction , Substantia Nigra/pathology
9.
PLoS Biol ; 2(11): e362, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15502874

ABSTRACT

Parkinson's disease (PD) pathology is characterized by the degeneration of midbrain dopamine neurons (DNs) ultimately leading to a progressive movement disorder in patients. The etiology of DN loss in sporadic PD is unknown, although it is hypothesized that aberrant protein aggregation and cellular oxidative stress may promote DN degeneration. Homozygous mutations in DJ-1 were recently described in two families with autosomal recessive inherited PD (Bonifati et al. 2003). In a companion article (Martinat et al. 2004), we show that mutations in DJ-1 alter the cellular response to oxidative stress and proteasomal inhibition. Here we show that DJ-1 functions as a redox-sensitive molecular chaperone that is activated in an oxidative cytoplasmic environment. We further demonstrate that DJ-1 chaperone activity in vivo extends to alpha-synuclein, a protein implicated in PD pathogenesis.


Subject(s)
Molecular Chaperones/chemistry , Oncogene Proteins/physiology , Oxidation-Reduction , alpha-Synuclein/chemistry , Antioxidants/chemistry , Cell Differentiation , Cell Line, Tumor , Cysteine/chemistry , Cytoplasm/metabolism , Dimerization , Dopamine/metabolism , Genetic Vectors , HeLa Cells , Homozygote , Humans , Intracellular Signaling Peptides and Proteins , Molecular Chaperones/metabolism , Mutation , Neurons/metabolism , Oncogene Proteins/metabolism , Oxidative Stress , Oxygen/chemistry , Parkinson Disease/metabolism , Protein Deglycase DJ-1 , Spectrophotometry , Synucleins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...