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










Publication year range
1.
PLoS One ; 14(7): e0219999, 2019.
Article in English | MEDLINE | ID: mdl-31323052

ABSTRACT

While pigs and rabbits are used as models for human immune diseases, FcγR binding is poorly characterized in both test species. To evaluate antibody binding to FcγRIIIA, a receptor involved in antibody-dependent cellular cytotoxicity, chimerized antibodies were generated by grafting the variable regions of a human IgG1 onto scaffolds from both species. The affinities of the parent and chimeric antibodies to the FcγRIIIA proteins from all three species were determined. While the human IgG1 and rabbit IgG had similar affinities for each FcγRIIIA with notable differences across species, pig IgG1 only bound pig FcγRIIIA with appreciable affinity. Also, the functional pig and rabbit proteins described here can be used in future experiments, such as pharmacology and mechanism of action studies.


Subject(s)
Antibody Affinity/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Receptors, IgG/metabolism , Recombinant Fusion Proteins , Animals , Enzyme-Linked Immunosorbent Assay , Gene Expression , Humans , Immunoglobulin G/isolation & purification , Protein Binding , Rabbits , Receptors, IgG/genetics , Receptors, IgG/isolation & purification , Surface Plasmon Resonance , Swine
2.
Mol Cancer Ther ; 17(11): 2412-2426, 2018 11.
Article in English | MEDLINE | ID: mdl-30126944

ABSTRACT

The primary function of tissue factor (TF) resides in the vasculature as a cofactor of blood clotting; however, multiple solid tumors aberrantly express this transmembrane receptor on the cell surface. Here, we developed anti-TF antibody-drug conjugates (ADC) that did not interfere with the coagulation cascade and benchmarked them against previously developed anti-TF ADCs. After screening an affinity-matured antibody panel of diverse paratopes and affinities, we identified one primary paratope family that did not inhibit conversion of Factor X (FX) to activated Factor X (FXa) and did not affect conversion of prothrombin to thrombin. The rest of the antibody panel and previously developed anti-TF antibodies were found to perturb coagulation to varying degrees. To compare the anticancer activity of coagulation-inert and -inhibitory antibodies as ADCs, a selection of antibodies was conjugated to the prototypic cytotoxic agent monomethyl auristatin E (MMAE) through a protease-cleavable linker. The coagulation-inert and -inhibitory anti-TF ADCs both killed cancer cells effectively. Importantly, the coagulation-inert ADCs were as efficacious as tisotumab vedotin, a clinical stage ADC that affected blood clotting, including in patient-derived xenografts from three solid tumor indications with a need for new therapeutic treatments-squamous cell carcinoma of the head and neck (SCCHN), ovarian, and gastric adenocarcinoma. Furthermore, a subset of the anti-TF antibodies could also be considered for the treatment of other diseases associated with upregulation of membranous TF expression, such as macular degeneration. Mol Cancer Ther; 17(11); 2412-26. ©2018 AACR.


Subject(s)
Blood Coagulation , Immunoconjugates/pharmacology , Thromboplastin/metabolism , Animals , Antibodies/metabolism , Antibody Affinity , Antibody Specificity , Blood Coagulation/drug effects , Endocytosis , Humans , Macaca fascicularis , Mice, Nude , Oligopeptides/toxicity , Protein Binding , Xenograft Model Antitumor Assays
3.
Mol Pharm ; 12(11): 3986-98, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26393951

ABSTRACT

Conventional antibody-drug conjugates (ADCs) are heterogeneous mixtures of chemically distinct molecules that vary in both drugs/antibody (DAR) and conjugation sites. Suboptimal properties of heterogeneous ADCs have led to new site-specific conjugation methods for improving ADC homogeneity. Most site-specific methods require extensive antibody engineering to identify optimal conjugation sites and introduce unique functional groups for conjugation with appropriately modified linkers. Alternative nonrecombinant methods have emerged in which bifunctional linkers are utilized to cross-link antibody interchain cysteines and afford ADCs containing four drugs/antibody. Although these methods have been shown to improve ADC homogeneity and stability in vitro, their effect on the pharmacological properties of ADCs in vivo is unknown. In order to determine the relative impact of interchain cysteine cross-linking on the therapeutic window and other properties of ADCs in vivo, we synthesized a derivative of the known ADC payload, MC-MMAF, that contains a bifunctional dibromomaleimide (DBM) linker instead of a conventional maleimide (MC) linker. The DBM-MMAF derivative was conjugated to trastuzumab and a novel anti-CD98 antibody to afford ADCs containing predominantly four drugs/antibody. The pharmacological properties of the resulting cross-linked ADCs were compared with analogous heterogeneous ADCs derived from conventional linkers. The results demonstrate that DBM linkers can be applied directly to native antibodies, without antibody engineering, to yield highly homogeneous ADCs via cysteine cross-linking. The resulting ADCs demonstrate improved pharmacokinetics, superior efficacy, and reduced toxicity in vivo compared to analogous conventional heterogeneous ADCs.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cysteine/chemistry , Immunoconjugates/pharmacology , Lung Neoplasms/drug therapy , Trastuzumab/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cross-Linking Reagents , Female , Flow Cytometry , Fluorescent Antibody Technique , Fusion Regulatory Protein-1/immunology , Humans , Immunoconjugates/chemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Rats , Rats, Sprague-Dawley , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Am J Transl Res ; 7(6): 1181-8, 2015.
Article in English | MEDLINE | ID: mdl-26279761

ABSTRACT

Soft tissue sarcoma (STS) is a heterogenous tumor arising from the embryonic mesoderm represented by approximately 50 histological subtypes. Effective therapeutic intervention is lacking for recurrent, late stage and metastatic disease. CD39, a cell-surface ectonucleotidase, has previously been shown to be upregulated in hematological malignancies and various epithelial tumors, but not in STS. Here, we show by mass spectrometry and immunohistochemistry that CD39 is highly expressed in primary patient sarcoma samples. Moreover, CD39 nucleotidase activity is enhanced in fibrosarcoma compared with normal control cells. We demonstrate that an inhibitory monoclonal anti-CD39 antibody, abrogates CD39 enzymatic activity significantly and prolongs survival in a lethal metastatic patient-derived sarcoma model. Taken together, the data suggest CD39 is a novel therapeutic target for the treatment of STS.

5.
PLoS One ; 10(4): e0124708, 2015.
Article in English | MEDLINE | ID: mdl-25894652

ABSTRACT

Antibodies against cell surface antigens may be internalized through their specific interactions with these proteins and in some cases may induce or perturb antigen internalization. The anti-cancer efficacy of antibody-drug conjugates is thought to rely on their uptake by cancer cells expressing the surface antigen. Numerous techniques, including microscopy and flow cytometry, have been used to identify antibodies with desired cellular uptake rates. To enable quantitative measurements of internalization of labeled antibodies, an assay based on internalized and quenched fluorescence was developed. For this approach, we generated novel anti-Alexa Fluor monoclonal antibodies (mAbs) that effectively and specifically quench cell surface-bound Alexa Fluor 488 or Alexa Fluor 594 fluorescence. Utilizing Alexa Fluor-labeled mAbs against the EphA2 receptor tyrosine kinase, we showed that the anti-Alexa Fluor reagents could be used to monitor internalization quantitatively over time. The anti-Alexa Fluor mAbs were also validated in a proof of concept dual-label internalization assay with simultaneous exposure of cells to two different mAbs. Importantly, the unique anti-Alexa Fluor mAbs described here may also enable other single- and dual-label experiments, including label detection and signal enhancement in macromolecules, trafficking of proteins and microorganisms, and cell migration and morphology.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Flow Cytometry , Fluorescent Dyes/chemistry , Maleimides/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antigens, Surface/immunology , Cell Line, Tumor , Humans , Mice , Molecular Sequence Data , Protein Transport , Receptor, EphA2/immunology , Spectrometry, Fluorescence
6.
J Immunol Methods ; 405: 1-14, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24380699

ABSTRACT

The diversity of a panel of antibodies that target a specific antigen can be established in various assay formats. In conventional epitope binning assays purified antibodies are tested in a pairwise manner: two antibodies that compete with each other for binding to an antigen are grouped into the same cluster or bin, while they are assigned to two different clusters when they do not compete. Here we present a high through put assay that enables grouping of crude hybridoma supernatants without a need for antibody purification. In addition, the assay does not require recombinant protein, because it is conducted on cells that express the antigen of interest. Hence, one can use the antibody-clustering assay for cell surface proteins that are not amenable to purification. Heavy chain variable region (VH) sequencing shows that VH composition within clusters is conserved. Finally, the assay is in good agreement with a conventional epitope binning assay with purified antigen.


Subject(s)
Antibodies, Monoclonal/immunology , Binding Sites, Antibody/immunology , Cluster Analysis , Epitopes/immunology , Animals , Antibodies, Monoclonal/classification , Antigens/immunology , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/classification , Humans , Hybridomas/immunology , Hybridomas/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin Variable Region/classification , Immunoglobulin Variable Region/immunology , Mice , Mice, 129 Strain
7.
Cancer Res ; 69(15): 6007-10, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19638582

ABSTRACT

Ligand-dependent and ligand-independent activation of the Hedgehog (Hh) signaling pathway is involved in tumorigenesis. The importance of mutations in Hh pathway components leading to constitutive signaling has been well established in basal cell carcinoma and medulloblastoma. However, the role of ligand-driven Hh pathway activation in cancer remains to be established. Three recent articles support a model in which, in the absence of mutations in the Hh pathway, Hh ligands expressed by a subset of epithelial cancers, including colon, pancreatic, and ovarian cancer, promote tumor growth indirectly by activating Hh signaling in the surrounding stroma, which, in turn, provides a more favorable environment for tumor growth. These data have important implications for the use of Hh pathway inhibitors currently in development and for selection of tumors likely to respond to such inhibitors.


Subject(s)
Hedgehog Proteins/metabolism , Neoplasms/metabolism , Animals , Humans , Signal Transduction
8.
Curr Biol ; 19(15): 1320-6, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19592253

ABSTRACT

The Hedgehog (Hh) signaling pathway regulates development in animals ranging from flies to humans. Although its framework is conserved, differences in pathway components have been reported. A kinesin-like protein, Costal2 (Cos2), plays a central role in the Hh pathway in flies. Knockdown of a zebrafish homolog of Cos2, Kif7, results in ectopic Hh signaling, suggesting that Kif7 acts primarily as a negative regulator of Hh signal transduction. However, in vitro analysis of the function of mammalian Kif7 and the closely related Kif27 has led to the conclusion that neither protein has a role in Hh signaling. Using Kif7 knockout mice, we demonstrate that mouse Kif7, like its zebrafish and Drosophila homologs, plays a role in transducing the Hh signal. We show that Kif7 accumulates at the distal tip of the primary cilia in a Hh-dependent manner. We also demonstrate a requirement for Kif7 in the efficient localization of Gli3 to cilia in response to Hh and for the processing of Gli3 to its repressor form. These results suggest a role for Kif7 in coordinating Hh signal transduction at the tip of cilia and preventing Gli3 cleavage into a repressor form in the presence of Hh.


Subject(s)
Cilia/metabolism , Embryonic Development/physiology , Hedgehog Proteins/metabolism , Kinesins/genetics , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cells, Cultured , DNA Primers/genetics , Genotype , Green Fluorescent Proteins , Immunoprecipitation , Kinesins/physiology , Mice , Mice, Knockout , Zinc Finger Protein Gli3
9.
Methods Enzymol ; 409: 251-84, 2006.
Article in English | MEDLINE | ID: mdl-16793406

ABSTRACT

Dramatic progress in understanding the mediators and mechanisms of chromosome break metabolism has been made in recent years. As a result, the links between disease and defects in chromosome dynamics have become clearer. In this chapter, we discuss techniques employed in our laboratory to study chromosome break metabolism, which include assessments at the molecular and cellular level. In our laboratory, we use the Mre11 complex as a tool to study this process, but the techniques discussed are of general relevance.


Subject(s)
Chromosomes , DNA Damage , DNA-Binding Proteins/genetics , Animals , Cell Cycle , Cell Line, Transformed , DNA Repair Enzymes , Fluorescent Antibody Technique, Indirect , Humans , MRE11 Homologue Protein , Mice , Spleen/cytology , Thymus Gland/cytology
10.
Genes Dev ; 19(24): 3043-54, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16357220

ABSTRACT

Genetic and cytologic data from Saccharomyces cerevisiae and mammals implicate the Mre11 complex, consisting of Mre11, Rad50, and Nbs1, as a sensor of DNA damage, and indicate that the complex influences the activity of ataxia-telangiectasia mutated (ATM) in the DNA damage response. Rad50(S/S) mice exhibit precipitous apoptotic attrition of hematopoietic cells. We generated ATM- and Chk2-deficient Rad50(S/S) mice and found that Rad50(S/S) cellular attrition was strongly ATM and Chk2 dependent. The hypomorphic Mre11(ATLD1) and Nbs1(Delta)(B) alleles conferred similar rescue of Rad50(S/S)-dependent hematopoietic failure. These data indicate that the Mre11 complex activates an ATM-Chk2-dependent apoptotic pathway. We find that apoptosis and cell cycle checkpoint activation are parallel outcomes of the Mre11 complex-ATM pathway. Conversely, the Rad50(S) mutation mitigated several phenotypic features of ATM deficiency. We propose that the Rad50(S) allele is hypermorphic for DNA damage signaling, and that the resulting constitutive low-level activation of the DNA damage response accounts for the partial suppression of ATM deficiency in Rad50(S/S) Atm(-/-) mice.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , ATP-Binding Cassette Transporters/genetics , Acid Anhydride Hydrolases , Alleles , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins , Cells, Cultured , Checkpoint Kinase 2 , DNA Repair Enzymes , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation , Hematopoiesis , Hematopoietic Stem Cells/cytology , MRE11 Homologue Protein , Male , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/deficiency
11.
DNA Repair (Amst) ; 3(8-9): 845-54, 2004.
Article in English | MEDLINE | ID: mdl-15279769

ABSTRACT

The conserved Mre11 complex (Mre11, Rad50, and Nbs1) plays a role in each aspect of chromosome break metabolism. The complex acts as a break sensor and functions in the activation and propagation of signaling pathways that govern cell cycle checkpoint functions in response to DNA damage. In addition, the Mre11 complex influences recombinational DNA repair through promoting recombination between sister chromatids. The Mre11 complex is required for mammalian cell viability but hypomorphic mutants of Mre11 and Nbs1 have been identified in human genetic instability disorders. These hypomorphic mutations, as well as those identified in yeast, have provided a benchmark for establishing mouse models of Mre11 complex deficiency. In addition to consideration of Mre11 complex functions in human cells and yeast, this review will discuss the characterization of mouse models and insight gleaned from those models regarding the metabolism of chromosome breaks. The current picture of break metabolism supports a central role for the Mre11 complex at the interface of chromosome stability and the regulation of cell growth. Further genetic analysis of the Mre11 complex will be an invaluable tool for dissecting its function on an organismal level and determining its role in the prevention of malignancy.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Chromosomes/ultrastructure , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Acid Anhydride Hydrolases , Alleles , Amino Acid Sequence , Animals , Cell Cycle , Cell Survival , DNA/genetics , DNA Damage , DNA Repair , DNA Repair Enzymes/physiology , DNA, Single-Stranded/genetics , Genome , Humans , MRE11 Homologue Protein , Meiosis , Mice , Models, Genetic , Molecular Sequence Data , Mutation , Neoplasms/etiology , Phenotype , Recombination, Genetic , Saccharomyces cerevisiae/metabolism
12.
Cell Cycle ; 3(5): 541-2, 2004 May.
Article in English | MEDLINE | ID: mdl-15034299

ABSTRACT

Hypomorphic mutants affecting the Mre11 complex components Mre11 (Mre11(ATLD1/ATLD1)) and Nbs1 (Nbs1(DeltaB/DeltaB)) have been established in the mouse. These mutations recapitulate those inherited in human chromosome fragility syndromes, the ataxia-telangiectasia like disorder and Nijmegen breakage syndrome. At the cellular level, the human and murine mutants exhibit defects in the intra S and G2/M checkpoints and marked chromosome instability. Whereas these outcomes are associated with predisposition to malignancy in humans, similar predisposition was not observed in either Mre11(ATLD1/ATLD1) or Nbs1(DeltaB/DeltaB) mice. These data demonstrate that chromosome breakage per se is insufficient to significantly enhance the initiation of tumorigenesis. However, these mutations greatly enhanced the risk of malignancy in p53+/- mice. We propose that proper metabolism of chromosome breaks arising during DNA replication is uniquely important for suppressing loss of heterozygosity and thus the penetrance of recessive oncogenic lesions.


Subject(s)
Cell Cycle/physiology , DNA Damage , Neoplasms/metabolism , Animals , Ataxia Telangiectasia/genetics , DNA Repair Enzymes , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Susceptibility , Genotype , Humans , MRE11 Homologue Protein , Mice , Mutation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Mol Cell ; 12(6): 1511-23, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14690604

ABSTRACT

In this study, mice expressing one of the two Mre11 alleles inherited in the human ataxia-telangiectasia like disorder (A-TLD) were derived. The mutation had a profound maternal effect on embryonic viability, revealing an acute requirement for Mre11 complex function in early embryogenesis. Mre11(ATLD1/ATLD1) mice exhibited several indices of impaired ATM function. The mice also exhibited pronounced chromosomal instability. Despite this phenotypic spectrum, the animals were not prone to malignancy. These data indicate that defective cell cycle checkpoints and chromosomal instability are insufficient to significantly enhance the initiation of tumorigenesis. In contrast, the latency of malignancy in p53(+/-) mice was dramatically reduced. We propose that in Mre11(ATLD1/ATLD1) mice, genome instability and cell cycle checkpoint defects reduce viability in early embryos and in proliferating cells, while promoting malignancy in the context of an initiating lesion.


Subject(s)
Ataxia Telangiectasia/genetics , Cell Cycle/physiology , Chromosomal Instability , Chromosomes, Mammalian/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Lymphoma/metabolism , Animals , Cells, Cultured , DNA Damage , DNA Repair Enzymes , Embryo, Mammalian/physiology , Female , Fertility , Fibroblasts/cytology , Fibroblasts/physiology , Genes, cdc , Humans , Karyotyping , MRE11 Homologue Protein , Mice , Pregnancy , Survival Rate , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...