Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 26(23): 6196-6203, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32887725

ABSTRACT

PURPOSE: The safety and preliminary efficacy of MEDI1873, an agonistic IgG1 fusion protein targeting glucocorticoid-induced TNF receptor-related protein (GITR), were evaluated in an open-label, first-in-human, phase I, dose escalation study in previously treated patients with advanced solid tumors. PATIENTS AND METHODS: Two single-patient cohorts at 1.5 and 3 mg i.v. were followed by 3+3 dose escalation in six cohorts at 7.5, 25, 75, 250, 500, and 750 mg, all every 2 weeks, for up to 52 weeks. Primary endpoints were safety and tolerability, dose-limiting toxicities (DLT), and MTD. Secondary endpoints included antitumor activity, pharmacokinetics, immunogenicity, and pharmacodynamics. RESULTS: Forty patients received MEDI1873. Three experienced DLTs: grade 3 worsening tumor pain (250 mg); grade 3 nausea, vomiting, and headache (500 mg); and grade 3 non-ST segment elevation myocardial infarction (750 mg). An MTD was not reached and treatment was well tolerated up to 500 mg. Most common treatment-related adverse events were headache (25%), infusion-related reaction (17.5%), and decreased appetite (17.5%). MEDI1873 exposure was dose proportional. Antidrug-antibody incidence was low. MEDI1873 increased peripheral CD4+ effector memory T-cell proliferation as well as cytokines associated with effector T-cell activation at dose levels ≥75 mg. The best response was stable disease (SD) in 17 patients (42.5%), including 1 unconfirmed partial response. Eight patients (20.0%) had SD ≥24 weeks. CONCLUSIONS: MEDI1873 showed acceptable safety up to 500 mg i.v. every 2 weeks with pharmacodynamics activity, and prolonged SD in some patients. However, further development is not planned because of lack of demonstrated tumor response.


Subject(s)
Antineoplastic Agents/therapeutic use , Glucocorticoid-Induced TNFR-Related Protein/agonists , Immunoglobulin G/chemistry , Neoplasms/drug therapy , Adult , Aged , Case-Control Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasms/pathology , Prognosis , Retrospective Studies
2.
Sci Rep ; 9(1): 1605, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733557

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is a serine protease inhibitor (serpin) that regulates fibrinolysis, cell adhesion and cell motility via its interactions with plasminogen activators and vitronectin. PAI-1 has been shown to play a role in a number of diverse pathologies including cardiovascular diseases, obesity and cancer and is therefore an attractive therapeutic target. However the multiple patho-physiological roles of PAI-1, and understanding the relative contributions of these in any one disease setting, make the development of therapeutically relevant molecules challenging. Here we describe the identification and characterisation of fully human antibody MEDI-579, which binds with high affinity and specificity to the active form of human PAI-1. MEDI-579 specifically inhibits serine protease interactions with PAI-1 while conserving vitronectin binding. Crystallographic analysis reveals that this specificity is achieved through direct binding of MEDI-579 Fab to the reactive centre loop (RCL) of PAI-1 and at the same exosite used by both tissue and urokinase plasminogen activators (tPA and uPA). We propose that MEDI-579 acts by directly competing with proteases for RCL binding and as such is able to modulate the interaction of PAI-1 with tPA and uPA in a way not previously described for a human PAI-1 inhibitor.


Subject(s)
Antibodies, Neutralizing/immunology , Plasminogen Activator Inhibitor 1/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/chemistry , Antibody Specificity , Humans , Mice , Models, Molecular , Plasminogen Activator Inhibitor 1/chemistry , Protein Conformation , Rats
3.
Clin Cancer Res ; 23(13): 3416-3427, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28069723

ABSTRACT

Purpose: To generate and characterize a murine GITR ligand fusion protein (mGITRL-FP) designed to maximize valency and the potential to agonize the GITR receptor for cancer immunotherapy.Experimental Design: The EC50 value of the mGITRL-FP was compared with an anti-GITR antibody in an in vitro agonistic cell-based reporter assay. We assessed the impact of dose, schedule, and Fc isotype on antitumor activity and T-cell modulation in the CT26 tumor model. The activity of the mGITRL-FP was compared with an agonistic murine OX40L-FP targeting OX40, in CT26 and B16F10-Luc2 tumor models. Combination of the mGITRL-FP with antibodies targeting PD-L1, PD-1, or CTLA-4 was analyzed in mice bearing CT26 tumors.Results: The mGITRL-FP had an almost 50-fold higher EC50 value compared with an anti-murine GITR antibody. Treatment of CT26 tumor-bearing mice with mGITRL-FP-mediated significant antitumor activity that was dependent on isotype, dose, and duration of exposure. The antitumor activity could be correlated with the increased proliferation of peripheral CD8+ and CD4+ T cells and a significant decrease in the frequency of intratumoral Tregs. The combination of mGITRL-FP with mOX40L-FP or checkpoint inhibitor antagonists enhanced antitumor immunity above that of monotherapy treatment.Conclusions: These results suggest that therapeutically targeting GITR represents a unique approach to cancer immunotherapy and suggests that a multimeric fusion protein may provide increased agonistic potential versus an antibody. In addition, these data provide, for the first time, early proof of concept for the potential combination of GITR targeting agents with OX40 agonists and PD-L1 antagonists. Clin Cancer Res; 23(13); 3416-27. ©2017 AACR.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/immunology , Melanoma, Experimental/immunology , Oncogene Proteins, Fusion/administration & dosage , Tumor Necrosis Factors/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Disease Models, Animal , Glucocorticoid-Induced TNFR-Related Protein/administration & dosage , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Membrane Glycoproteins/agonists , Membrane Glycoproteins/immunology , Mice , OX40 Ligand , Oncogene Proteins, Fusion/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Tumor Necrosis Factors/agonists , Tumor Necrosis Factors/genetics
4.
Am J Respir Crit Care Med ; 185(1): 34-43, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21997333

ABSTRACT

RATIONALE: Inflammation and oxidative stress are linked to the deleterious effects of cigarette smoke in producing chronic obstructive pulmonary disease (COPD). Myeloperoxidase (MPO), a neutrophil and macrophage product, is important in bacterial killing, but also drives inflammatory reactions and tissue oxidation. OBJECTIVES: To determine the role of MPO in COPD. METHODS: We treated guinea pigs with a 2-thioxanthine MPO inhibitor, AZ1, in a 6-month cigarette smoke exposure model, with one group receiving compound from Smoking Day 1 and another group treated after 3 months of smoke exposure. RESULTS: At 6 months both treatments abolished smoke-induced increases in lavage inflammatory cells, largely ameliorated physiological changes, and prevented or stopped progression of morphologic emphysema and small airway remodeling. Cigarette smoke caused a marked increase in immunohistochemical staining for the myeloperoxidase-generated protein oxidation marker dityrosine, and this effect was considerably decreased with both treatment arms. Serum 8-isoprostane, another marker of oxidative stress, showed similar trends. Both treatments also prevented muscularization of the small intrapulmonary arteries, but only partially ameliorated smoke-induced pulmonary hypertension. Acutely, AZ1 prevented smoke-induced increases in expression of cytokine mediators and nuclear factor-κB binding. CONCLUSIONS: We conclude that an MPO inhibitor is able to stop progression of emphysema and small airway remodeling and to partially protect against pulmonary hypertension, even when treatment starts relatively late in the course of long-term smoke exposure, suggesting that inhibition of MPO may be a novel and useful therapeutic treatment for COPD. Protection appears to relate to inhibition of oxidative damage and down-regulation of the smoke-induced inflammatory response.


Subject(s)
Enzyme Inhibitors/pharmacology , Peroxidase/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Purines/therapeutic use , Smoking/adverse effects , Thiones/therapeutic use , Airway Remodeling/drug effects , Animals , Dinoprost/analogs & derivatives , Dinoprost/blood , Disease Models, Animal , Disease Progression , Female , Guinea Pigs , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/prevention & control , Inflammation/etiology , Inflammation/metabolism , Inflammation/prevention & control , Lung/drug effects , Lung/metabolism , Oxidative Stress/drug effects , Peroxidase/metabolism , Pulmonary Disease, Chronic Obstructive/etiology , Thioxanthenes/antagonists & inhibitors , Thioxanthenes/metabolism , Tyrosine/analogs & derivatives , Tyrosine/drug effects
5.
J Mol Biol ; 411(4): 791-807, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21723291

ABSTRACT

The differentiation of therapeutic monoclonal antibodies in an increasingly competitive landscape requires optimization of clinical efficacy combined with increased patient convenience. We describe here the generation of MEDI5117, a human anti-interleukin (IL)-6 antibody generated by variable domain engineering, to achieve subpicomolar affinity for IL-6, combined with Fc (fragment crystallizable) engineering to enhance pharmacokinetic half-life. MEDI5117 was shown to be highly potent in disease-relevant cellular assays. The pharmacokinetics of MEDI5117 were evaluated and compared to those of its progenitor, CAT6001, in a single-dose study in cynomolgus monkeys. The antibodies were administered, either subcutaneously or intravenously, as a single dose of 5 mg/kg. The half-life of MEDI5117 was extended by approximately 3-fold, and clearance was reduced by approximately 4-fold when compared to CAT6001. MEDI5117 therefore represents a potential 'next-generation' antibody; future studies are planned to determine the potential for affinity-driven efficacy and/or less frequent administration.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Interleukin-6/antagonists & inhibitors , Interleukin-6/immunology , Macaca fascicularis/immunology , Animals , Antibodies, Anti-Idiotypic/administration & dosage , Antibodies, Monoclonal/administration & dosage , Cells, Cultured , Half-Life , Humans , Interleukin-6/genetics , Kidney/cytology , Kidney/metabolism , Models, Chemical , Mutagenesis , Protein Engineering , Receptors, Interleukin/immunology , Surface Plasmon Resonance , T-Lymphocytes/metabolism , Tissue Distribution , Vascular Endothelial Growth Factor A/metabolism
6.
J Mol Biol ; 406(1): 160-75, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21167836

ABSTRACT

Interleukin (IL) 15 is an inflammatory cytokine that plays an essential role in the activation, proliferation, and maintenance of specific natural killer cell and T-cell populations, and has been implicated as a mediator of inflammatory diseases. An anti-IL-15 antibody that blocked IL-15-dependent cellular responses was isolated by phage display and optimised via mutagenesis of the third complementarity-determining regions (CDRs) of variable heavy (VH) and variable light chains. Entire repertoires of improved variants were recombined with each other to explore the maximum potential sequence space. DISC0280, the most potent antibody isolated using this comprehensive strategy, exhibits a 228-fold increase in affinity and a striking 40,000-fold increase in cellular potency compared to its parent. Such a wholesale recombination strategy therefore represents a useful method for exploiting synergistic potency gains as part of future antibody engineering efforts. The crystal structure of DISC0280 Fab (fragment antigen binding), in complex with human IL-15, was determined in order to map the structural epitope and paratope. The most remarkable feature revealed lies within the paratope and is a novel six-amino-acid α-helix that sits within the VH CDR3 loop at the center of the antigen binding site. This is the first report to describe an α-helix as a principal component of a naturally derived VH CDR3 following affinity maturation.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/isolation & purification , Complementarity Determining Regions/chemistry , Interleukin-15/antagonists & inhibitors , Interleukin-15/immunology , Protein Engineering , Amino Acid Sequence , Antibodies, Neutralizing/genetics , Binding Sites, Antibody/genetics , Complementarity Determining Regions/genetics , Epitopes/chemistry , Epitopes/genetics , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Molecular Sequence Data , Mutation
7.
Protein Expr Purif ; 68(1): 42-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19435602

ABSTRACT

The physiological activities of Interleukin-15 (IL-15) suggest that it could be useful as an immunomodulator to activate the innate immune system, however, the expression and purification yields of recombinant mature IL-15 have typically been low. In this report, a method was optimised to generate milligram quantities of this cytokine. Human IL-15 with an N-terminal (His)(6)-tag was expressed in Escherichia coli as an insoluble protein. The IL-15 material was purified from other cellular proteins by dissolution in 6M guanidine HCl, followed by Ni-NTA chromatography in a buffer containing 8M urea. Use of a multi-component screen identified the optimal conditions for folding (His)(6)-tagged human IL-15 and the method was scaled up to produce milligram quantities of folded material in its native conformation, with two intra-molecular disulphides as determined by electrospray mass spectrometry. Mature IL-15 was generated by cleavage with recombinant enterokinase, which was subsequently removed by Ni-NTA chromatography. Identical methods were used to produce mature cynomolgus monkey (Macaca fascicularis) IL-15 in similar quantities. Human and cynomolgus IL-15 were both active in two IL-15 dependent assays; mouse CTLL2 cell proliferation and human and cynomolgus CD69 upregulation on CD3(-) CD8+ lymphocytes in whole blood. Despite being 96% identical at the amino acid level the human IL-15 was 10-fold more potent than the cynomolgus IL-15 in both assays. The methods described here are useful for producing both mature IL-15 proteins in sufficient quantity for in vivo and in vitro studies, including X-ray crystallography.


Subject(s)
Escherichia coli/metabolism , Interleukin-15/isolation & purification , Interleukin-15/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Line , Cell Proliferation , Chromatography, Gel , Dialysis , Escherichia coli/genetics , Humans , Interleukin-15/chemistry , Interleukin-15/genetics , Lectins, C-Type , Macaca fascicularis/genetics , Molecular Weight , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Up-Regulation
8.
J Biomol Screen ; 14(3): 263-72, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19211775

ABSTRACT

Inducible nitric oxide synthase (iNOS) is active as a homodimer. A cell-based assay suitable for high-throughput screening (HTS) was generated to identify inhibitors of iNOS dimerization using the InteraX enzyme complementation technology of Applied Biosystems. The cells contain 2 chimeric proteins of complementing deletion mutants of beta-galactosidase, each fused to the oxygenase domain of human iNOS. The assay was characterized using known iNOS dimerization inhibitors, which gave a decrease in beta-galactosidase activity. Surprisingly, the assay was also able to identify compounds that have the same profile as known inhibitors of fully formed dimeric iNOS by causing an increase in beta-galactosidase activity. The iNOS InteraX assay was used to screen approximately 800,000 compounds in a 384-well format. After hit confirmation, 3359 compounds were taken forward for full IC50 determination in InteraX and cytotoxicity assays. Of these compounds 40.5% were confirmed as greater than 10-fold more active in InteraX compared to a cytotoxicity assay and were classified as potential iNOS dimerization inhibitors as they did not inhibit beta-galactosidase alone. In the same primary screen, 901 compounds gave a significant increase in beta-galactosidase activity. Many of these were known inhibitors of iNOS. After IC50 determination in InteraX and cytotoxicity assays, 182 novel compounds remained as potential arginine-competitive inhibitors of dimeric iNOS.


Subject(s)
Biological Assay/methods , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Animals , Binding Sites , Carcinoma/metabolism , Carcinoma/pathology , Cell Line , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dimerization , Enzyme Inhibitors/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Indicators and Reagents/metabolism , Inhibitory Concentration 50 , Kidney/cytology , Mice , Models, Biological , Molecular Structure , Oxazines/metabolism , Protein Binding , Proteins/metabolism , Reproducibility of Results , Retroviridae/genetics , Transduction, Genetic , Xanthenes/metabolism , beta-Galactosidase/analysis , beta-Galactosidase/metabolism
9.
Nat Chem Biol ; 4(11): 700-7, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18849972

ABSTRACT

Nitric oxide synthase (NOS) enzymes synthesize nitric oxide, a signal for vasodilatation and neurotransmission at low concentrations and a defensive cytotoxin at higher concentrations. The high active site conservation among all three NOS isozymes hinders the design of selective NOS inhibitors to treat inflammation, arthritis, stroke, septic shock and cancer. Our crystal structures and mutagenesis results identified an isozyme-specific induced-fit binding mode linking a cascade of conformational changes to a new specificity pocket. Plasticity of an isozyme-specific triad of distant second- and third-shell residues modulates conformational changes of invariant first-shell residues to determine inhibitor selectivity. To design potent and selective NOS inhibitors, we developed the anchored plasticity approach: anchor an inhibitor core in a conserved binding pocket, then extend rigid bulky substituents toward remote specificity pockets, which become accessible upon conformational changes of flexible residues. This approach exemplifies general principles for the design of selective enzyme inhibitors that overcome strong active site conservation.


Subject(s)
Drug Design , Enzyme Inhibitors , Inflammation/drug therapy , Inflammation/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Amino Acid Sequence , Aminopyridines/chemistry , Aminopyridines/pharmacology , Animals , Cattle , Crystallography, X-Ray , Disease Models, Animal , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Isoenzymes/antagonists & inhibitors , Male , Mice , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutation , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Rats
10.
Mol Pharmacol ; 74(5): 1193-202, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18676678

ABSTRACT

The chemokine receptors CXCR1 and CXCR2 are G-protein-coupled receptors (GPCRs) implicated in mediating cellular functions associated with the inflammatory response. Potent CXCR2 receptor antagonists have been discovered, some of which have recently entered clinical development. The aim of this study was to identify key amino acid residue differences between CXCR1 and CXCR2 that influence the relative antagonism by two compounds that have markedly different chemical structures. By investigating the effects of domain switching and point mutations, we found that the second extracellular loop, which contained significant amino acid sequence diversity, was not important for compound antagonism. We were surprised to find that switching the intracellular C-terminal 60 amino acid domains of CXCR1 and CXCR2 caused an apparent reversal of antagonism at these two receptors. Further investigation showed that a single amino acid residue, lysine 320 in CXCR2 and asparagine 311 in CXCR1, plays a predominant role in describing the relative antagonism of the two compounds. Homology modeling studies based on the structure of bovine rhodopsin indicated a potential intracellular antagonist binding pocket involving lysine 320. We conclude that residue 320 in CXCR2 forms part of a potential allosteric binding pocket on the intracellular side of the receptor, a site that is distal to the orthosteric site commonly assumed to be the location of antagonist binding to GPCRs. The existence of a common intracellular allosteric binding site at GPCRs related to CXCR2 may be of value in the design of novel antagonists for therapeutic intervention.


Subject(s)
Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Allosteric Site , Amino Acid Sequence , Cell Line , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Radioligand Assay , Receptors, Interleukin-8A/chemistry , Receptors, Interleukin-8A/drug effects , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8B/chemistry , Receptors, Interleukin-8B/drug effects , Receptors, Interleukin-8B/genetics , Sequence Homology, Amino Acid
11.
Mutat Res ; 572(1-2): 1-13, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15790486

ABSTRACT

A well-characterised gain-of-function point mutation within exon 17 of the c-kit proto-oncogene known as Asp816Val is present in patients with mastocytosis. Activation of mast cells through this receptor primes them for IgE-dependent activation, and patients with mastocytosis are at increased risk of anaphylaxis. We hypothesised that the Asp816Val mutation is associated with a history of anaphylaxis in the general population. A mismatch amplification real-time PCR assay was developed and validated to test for the Asp816Val mutation. Subjects were recruited to four subject groups: normal non-atopics, atopics without anaphylaxis, food-induced anaphylactics and non-food anaphylactics. Blood samples collected from forty subjects were tested for the presence of Asp816Val. Thirteen subjects were found to carry the mutation; normals (2/9), atopics (2/10), food anaphylactics (5/11) and non-food anaphylactics (4/10). Statistical analysis of the data determined that there was no significant difference between the numbers of subjects found to carry the Asp816Val mutation in each of the groups although a trend towards an increased occurrence in anaphylactics was observed. In summary, the hypothesis that the presence of the Asp816Val mutation is linked to the occurrence of anaphylaxis was not supported, but interestingly, we have shown for the first time Asp816Val may occur more frequently than previously reported within the general population.


Subject(s)
Anaphylaxis/genetics , Point Mutation , Polymerase Chain Reaction/methods , Proto-Oncogene Proteins c-kit/genetics , Aspartic Acid/genetics , Base Sequence , Blotting, Western , Cell Line , Cloning, Molecular , DNA Primers , Humans , Proto-Oncogene Mas , Valine/genetics
12.
Protein Expr Purif ; 25(2): 256-62, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12135558

ABSTRACT

Rat mast cell protease 7 (rMCP7) is a neutral serine protease and a component of mast cells, where it is stored in secretory granules. Mast cells express numerous proteases so in order to characterize rMCP7, it was cloned and expressed as a recombinant protein in Pichia pastoris. During expression, rMCP7 protein was cleaved from the alpha-mating factor signal at the engineered KEX2 cleavage site to produce active rMCP7. The protein produced was stable at pH 5.5 and active in the absence of heparin. The rMCP7 was glycosylated and treatment with N-glycosidase F resulted in a protein of the predicted molecular mass of 30 kDa. The rMCP7 was purified via an ammonium sulfate precipitation, using casein as a carrier protein, followed by cation exchange chromatography. The purified protein was assayed using a range of substrates and where possible, k(m) and k(cat) values were determined. The substrate profile displayed by the recombinant rMCP7 was consistent with that of tryptase isolated from rat skin. The expression and purification of recombinant rMCP7 offer an efficient, low-cost method of producing large amounts of protein. It also offers the opportunity of easy manipulation and mutagenesis of rMCP7 for further biochemical, structural, and physiological studies.


Subject(s)
Mast Cells/enzymology , Pichia/genetics , Serine Endopeptidases/isolation & purification , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Chromatography, Gel , Chymases , Electrophoresis, Polyacrylamide Gel , Glycosylation , Kinetics , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...