Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Protein Eng Des Sel ; 29(5): 159-67, 2016 May.
Article in English | MEDLINE | ID: mdl-26968452

ABSTRACT

The CTLA4-Ig therapeutics abatacept and belatacept inhibit CD28-mediated T cell activation by binding CD80 (B7-1) and CD86 (B7-2) co-stimulatory ligands. Both compounds preferentially bind CD80, yet CD86 has been implicated as the dominant co-stimulatory ligand. Using directed evolution methods, novel CTLA4-Ig variants were created with selective CD86 binding affinity, a property that confers increased immunosuppressive potency and potentially improved efficacy and safety profiles. Relative to abatacept (wild-type CTLA4-Ig), ASP2408 and ASP2409 have 83-fold and 220-fold enhanced binding affinity to CD86 while retaining 1.5-fold and 5.6-fold enhanced binding affinity to CD80, respectively. Improvements in CD86 binding affinity correlates with increased immunosuppressive potencyin vitroandin vivo Our results highlight the power of directed evolution methods to obtain non-intuitive protein engineering solutions and represent the first examples of CD86-selective CTLA4-Ig compounds that have entered clinical trials.


Subject(s)
Abatacept/genetics , Abatacept/pharmacology , B7-2 Antigen/metabolism , Directed Molecular Evolution , Immunoconjugates/metabolism , Immunoconjugates/pharmacology , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Abatacept/chemistry , Abatacept/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Female , Humans , Immunoconjugates/chemistry , Immunosuppressive Agents/chemistry , Ligands , Mice , Models, Molecular , Protein Conformation , Substrate Specificity
2.
Anal Biochem ; 376(2): 163-72, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18342613

ABSTRACT

This study illustrates the application of asymmetrical flow field-flow fractionation (AF4) and light scattering analysis during the development of a gene delivery vehicle based on virus-like particles (VLPs) derived from the human polyoma JC virus. The analytical system was created by connecting an AF4 apparatus to the following detectors: diode array, fluorescence, multiangle light scattering, dynamic light scattering, and refractometer. From a single analysis, the molar mass, root mean square and hydrodynamic radii, composition, and purity of the sample could be determined. The VLPs were purified from baculovirus-infected Sf158 insect cells overexpressing the recombinant VP1 protein using weak anion exchange chromatography. The VLPs were dissociated to VP1 pentamers, and the contaminating DNA and proteins were removed using strong anion exchange chromatography. The gene delivery vehicle was created by reassembling the VP1 pentamers in the presence of the desired DNA. The newly formed VLPs encapsulated the DNA and were shown to be capable of delivering the gene of interest to target cells where it was translated into protein. This paper describes the scalable process that was derived to produce the VLPs and demonstrates how the AF4-based analytical characterization was indispensable during the development process.


Subject(s)
DNA/genetics , Fractionation, Field Flow/methods , Gene Transfer Techniques , Polyomavirus/genetics , Scattering, Radiation , Animals , Cell Line , Humans , Light , Recombinant Proteins/metabolism , Spodoptera
3.
Am J Physiol Heart Circ Physiol ; 293(1): H69-76, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17434974

ABSTRACT

The present study tested the hypothesis that murine (m)IFN-beta or mIFN-alpha(2) can eliminate cardiac viral load and protect cardiomyocytes from injury in animals infected with coxsackievirus B3 (CVB3). CVB3-inoculated male Balb/c mice exhibited signs of illness, including lethargy, progressive weight loss, and death (10% on day 3 and 100% on day 8). Cardiac viral load was high [4,277 +/- 1,009 plaque-forming units and 25 +/- 5 copies CVB3/hypoxanthine guanine phosphoribosyl transferase 1 mRNA] on day 4. The cardiac tissue exhibited severe inflammatory infiltration and myocyte damage with an average myocarditis integrated pathology score of 2.1 +/- 0.2 on day 7. Most of the mice infected with CVB3 also developed epicarditis, and 55% had intraventricular thrombi present. Treatment with mIFN-beta [2.5 to 10 million international units (MIU)/kg] dose-dependently improved the general health status in CVB3-inoculated mice, as evidenced by reduction in weight loss, prevention of death, elimination of cardiac viral load, protection of myocytes from injury, decrease in inflammatory cell infiltration, and attenuation of intraventricular thrombus formation. Treatment with 10 MIU/kg mIFN-alpha(2) resulted in a similar level of efficacy as that induced by 5 MIU/kg mIFN-beta, with the exception that mIFN-alpha(2) did not reduce cardiac CVB3 mRNA. However, mIFN-alpha(2) , but not any dose group of mIFN-beta, significantly attenuated CVB3-induced epicarditis. These data demonstrate antiviral effects for both mIFN-beta and mIFN-alpha(2), which lead to protection of the mice from CVB3-induced myocarditis. However, the potential mechanisms leading to a differential host response for the two isoforms of mIFN remain to be elucidated.


Subject(s)
Enterovirus/drug effects , Interferon-alpha/administration & dosage , Interferon-beta/administration & dosage , Myocarditis/drug therapy , Myocarditis/virology , Pericardium/drug effects , Pericardium/virology , Animals , Antiviral Agents/administration & dosage , Cardiotonic Agents/administration & dosage , Cells, Cultured , Male , Mice , Mice, Inbred BALB C , Muscle Cells/drug effects , Muscle Cells/virology , Treatment Outcome
4.
J Biomol Screen ; 11(8): 985-95, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17092910

ABSTRACT

A therapeutic antibody candidate (AT-19) isolated using multivalent phage display binds native tomoregulin (TR) as a mul-timer not as a monomer. This report raises the importance of screening and selecting phage antibodies on native antigen and reemphasizes the possibility that potentially valuable antibodies are discarded when a monomeric phage display system is used for screening. A detailed live cell panning selection and screening method to isolate multivalently active antibodies is described. AT-19 is a fully human antibody recognizing the cell surface protein TR, a proposed prostate cancer target for therapeutic antibody internalization. AT-19 was isolated from a multivalent single-chain variable fragment (scFv) antibody library rescued with hyperphage. The required multivalency for isolation of AT-19 is supported by fluorescence activated cell sorting data demonstrating binding of the multivalent AT-19 phage particles at high phage concentrations and failure of monovalent particles to bind. Pure monomeric scFv AT-19 does not bind native receptor on cells, whereas dimeric scFv or immunoglobulin G binds with nanomolar affinity. The isolation of AT-19 antibody with obligate bivalent binding activity to native TR is attributed to the use of a multivalent display of scFv on phage and the method for selecting and screening by alternate use of 2 recombinant cell lines.


Subject(s)
Antigens, Surface/immunology , Bacteriophages/immunology , Immunoglobulin Variable Region/immunology , Membrane Proteins/pharmacology , Neoplasm Proteins/pharmacology , Peptide Library , Animals , Antibodies , Bacteriophages/genetics , CHO Cells , Cell Line , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay/methods , Humans , Immunoglobulin G/metabolism , Sequence Analysis, Protein , Transfection , Viral Load
5.
Thromb Haemost ; 96(3): 317-24, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16953273

ABSTRACT

Tissue factor (TF) exposure is a potent pro-thrombotic trigger that initiates activation of the coagulation cascade, while thrombomodulin (TM) is a potent anticoagulant protein that limits the extent of activation. Both TF neutralizing antibodies and soluble TM (sTM) are effective anticoagulants. We have developed a novel anticoagulant fusion protein, Ab(TF)-TM, by fusing a TFneutralizing single-chain antibody, Ab(TF), to an active fragment of TM. Ab(TF)-TM is a novel anticoagulant targeting to sites of TF exposure with a dual mechanism of action. The Ab(TF) portion of the molecule inhibitsTF/factorVIIa mediated activation of FIX and FX, and the TM portion of the molecule acts as a cofactor for activation of protein C. In-vitro coagulation assays show that Ab(TF)-TM more potently inhibits TF-initiated coagulation (prothrombin time) than can its individual components, Ab(TF) (20-fold) and sTM (80-fold) alone, or in combination (10-fold). In contrast, the potency of Ab(TF)-TM in the activated partial thromboplastin and thrombin clotting time assays was similar to sTM alone. In a rat model of disseminated intravascular coagulation (DIC), intravenous injection of a human TF-containing thromboplastin reagent (0.5 ml/kg) resulted in an immediate death in approximately 60% of the animals and a clinical score of approximately 2.5. Pre-injection of Ab(TF)-TM or Ab(TF) and sTM, given alone or in combination, showed dose-dependent efficacy. At a dose of 0.7 nmol/kg, Ab(TF)-TM completely prevented death and reduced clinical scores by 79%, while neitherAb(TF) nor sTM, given alone or in combination, showed significant therapeutic effects. Calculated effective doses that reduced mortality by 50% relative to that in the control group (ED(50), nmol/kg) were 0.21 for Ab(TF)-TM, 3.2 for an equimolar mixture of Ab(TF) and sTM, 4.3 for sTM and 20 for Ab(TF). Thus, Ab(TF)-TM presented 10- to 100-fold enhancement of the anticoagulant potency, relative to the ED(50) in Ab(TF) and sTM given either alone or in combination, in a rat DIC model.


Subject(s)
Anticoagulants/pharmacology , Thrombomodulin/blood , Thromboplastin/metabolism , Animals , Anticoagulants/chemistry , Blood Coagulation , Dimerization , Dose-Response Relationship, Drug , Humans , Male , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Thromboembolism , Thrombomodulin/chemistry , Thromboplastin/immunology , Time Factors
6.
Gene ; 380(2): 111-9, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16879932

ABSTRACT

The detection and purification of proteins are often time-consuming and frequently involve complicated protocols. The addition of a peptide tag to recombinant proteins can make this process more efficient. Many of the commonly used tags, such as Flagtrade mark, Myc, HA and V5 are recognized by specific monoclonal antibodies and therefore, allow immunoaffinity-based purification. Enhancing the current scope of flexibility in using diverse peptide tags, we report here the development of a novel, short polypeptide tag (Tab2) for detection and purification of recombinant proteins. The Tab2 epitope corresponds to the NH2-terminal seven amino acid residues of human TGFalpha. A monoclonal anti-Tab2 antibody was raised and characterized. To investigate the potential of this peptide sequence as a novel tag for recombinant proteins, we expressed several different recombinant proteins containing this tag in E. coli, baculovirus, and mammalian cells. The data presented demonstrates the Tab2 tag-anti-Tab2 antibody combination is a reliable tool enabling specific Western blot detection, FACS analysis, and immunoprecipitation as well as non-denaturing protein affinity purification.


Subject(s)
Chromatography, Affinity/methods , Epitopes/chemistry , Peptide Fragments/immunology , Recombinant Fusion Proteins/isolation & purification , Transforming Growth Factor alpha/genetics , Amino Acid Motifs/genetics , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Cells, Cultured , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Genetic Vectors/chemical synthesis , Humans , Immunoprecipitation/methods , Insecta , Peptide Fragments/genetics , Phosphotransferases/genetics , Phosphotransferases/isolation & purification , Phosphotransferases/metabolism , Protein Binding/immunology , Protein Structure, Tertiary/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/isolation & purification , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/immunology , Transforming Growth Factor alpha/chemistry , Transforming Growth Factor alpha/metabolism
7.
Neoplasia ; 8(1): 18-30, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16533422

ABSTRACT

The EphA2 receptor tyrosine kinase has been shown to be over-expressed in cancer and a monoclonal antibody (mAb) that activates and down-modulates EphA2 was reported to inhibit the growth of human breast and lung tumor xenografts in nude mice. Reduction of EphA2 levels by treatment with anti-EphA2 siRNA also inhibited tumor growth, suggesting that the anti-tumor effects of these agents are mediated by decreasing the levels of EphA2. As these studies employed human tumor xenograft models in nude mice with reagents whose cross reactivity with murine EphA2 is unknown, we generated a mAb (Ab20) that preferentially binds, activates, and induces the degradation of murine EphA2. Treatment of established murine CT26 colorectal tumors with Ab20 reduced EphA2 protein levels to approximately 12% of control tumor levels, yet had no effect on tumor growth. CT26 tumor cell colonization of the lung was also not affected by Ab20 administration despite having barely detectable levels of EphA2. We also generated and tested a potent agonistic mAb against human EphA2 (1G9-H7). No inhibition of humanMDA-231 breast tumor xenograft growth was observed despite evidence for >85% reduction of EphA2 protein levels in the tumors. These results suggest that molecular characteristics of the tumors in addition to EphA2 over-expression may be important for predicting responsiveness to EphA2-directed therapies.


Subject(s)
Breast Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Immunotherapy/methods , Mammary Neoplasms, Animal/metabolism , Receptor, EphA2/chemistry , Animals , Antibodies, Monoclonal/chemistry , Breast Neoplasms/therapy , Cell Line, Tumor , Colorectal Neoplasms/therapy , Female , Humans , Mammary Neoplasms, Animal/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Receptor, EphA2/immunology
8.
Cancer Res ; 65(18): 8397-405, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16166318

ABSTRACT

Gene expression analysis showed that a human mindin homologue, mindin/RG-1, is expressed selectively in prostate tissues and that its expression level is elevated in some prostate tumors. Mindin/RG-1 protein expression is maintained in >80% of prostate cancers metastatic to bone or lymph nodes as well as in locally recurrent tumors in androgen-unresponsive patients. In contrast, mindin/RG-1 expression in other normal tissues is significantly lower than that seen in the prostate. A fully human antibody, 19G9, was generated against mindin/RG-1 protein and was shown to accumulate at high abundance in LNCaP tumor xenografts. Conjugates of this antibody with the chelator CHX-A''-DTPA were generated and radiolabeled with either 111In, 90Y, or 86Y. Small animal positron emission tomography imaging with the 86Y-radiolabeled conjugate showed very specific accumulation of the antibody in LNCaP tumor xenografts with clear tumor delineation apparent at 4 hours. The therapeutic efficacy of [90Y]-CHX-A''-DTPA-19G9 was evaluated in mice bearing LNCaP xenografts. A dose-finding study identified a nontoxic therapeutic dose to be approximately 75 microCi. Significant antitumor effects were seen with a single administration of radiolabeled antibody to animals bearing 200 to 400 mm3 tumors. Inhibition of tumor growth was observed in all treated animals over a 49-day period. At 49 days posttreatment, slow tumor growth recurred but this could be prevented for an additional 40-day period by a second administration of a 75 microCi dose at day 49. We conclude that [90Y]-CHX-A''-DTPA-19G9 is a novel antibody conjugate that has considerable promise for therapy of metastatic prostate cancer in androgen-unresponsive patients.


Subject(s)
Antibodies, Monoclonal/immunology , Extracellular Matrix Proteins/immunology , Immunotoxins/immunology , Prostatic Neoplasms/radiotherapy , Radioimmunotherapy/methods , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , CHO Cells , Cricetinae , Dose-Response Relationship, Immunologic , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/pharmacology , Isothiocyanates/immunology , Isothiocyanates/pharmacokinetics , Isothiocyanates/pharmacology , Male , Molecular Sequence Data , Pentetic Acid/analogs & derivatives , Pentetic Acid/immunology , Pentetic Acid/pharmacokinetics , Pentetic Acid/pharmacology , Positron-Emission Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tissue Distribution , Xenograft Model Antitumor Assays , Yttrium Radioisotopes/administration & dosage , Yttrium Radioisotopes/pharmacokinetics , Yttrium Radioisotopes/pharmacology
9.
Biochemistry ; 44(26): 9339-47, 2005 Jul 05.
Article in English | MEDLINE | ID: mdl-15982000

ABSTRACT

This paper presents the crystal structure of porcine pancreatic carboxypeptidase B (pp-CpB) in complex with a variety of thiol-based inhibitors that were developed as antagonists of activated thrombin-activatable fibrinolysis inhibitor (TAFIa). Recent studies have indicated that a selective inhibitor of TAFIa could enhance the efficacy of existing thrombolytic agents for the treatment of acute myocardial infarction, one of the most prevalent forms of heart attacks. Unfortunately, activated TAFIa rapidly degrades in solution and cannot be used for crystallographic studies. In contrast, porcine pancreatic CpB is stable at room temperature and is available from commercial sources. Both pancreatic CpB and TAFIa are zinc-based exopeptidases, and the proteins share a 47% sequence identity. The homology improves considerably in the active site where nearly all of the residues are conserved. The inhibitors used in this study were designed to mimic a C-terminal arginine residue, one of the natural substrates of TAFIa. The X-ray structures show that the thiol group chelates the active site zinc, the carboxylic acid forms a salt bridge to Arg145, and the guanidine group forms two hydrogen bonds to Asp255. A meta-substituted phenyl was introduced into our inhibitors to reduce conformational freedom. This modification vastly improved the selectivity of compounds against other exopeptidases that cleave basic residues. Comparisons between structures indicate that selectivity derives from the interaction between the guanidine group in the inhibitors and an acidic active site residue. The location of this acidic residue is not conserved in the various carboxypeptidases.


Subject(s)
Carboxypeptidase B/antagonists & inhibitors , Protease Inhibitors/chemistry , Sulfhydryl Compounds/chemistry , Animals , Carboxypeptidase B/metabolism , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Pancreas/enzymology , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Protein Conformation , Sulfhydryl Compounds/metabolism , Sulfhydryl Compounds/pharmacology , Swine
10.
Protein Expr Purif ; 42(2): 295-304, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15925519

ABSTRACT

Prolyl-4-hydroxylase domain-containing enzymes (PHDs) mediate the oxygen-dependent regulation of the heterodimeric transcription factor hypoxia-inducible factor-1 (HIF-1). Under normoxic conditions, one of the subunits of HIF-1, HIF-1alpha, is hydroxylated on specific proline residues to target HIF-1alpha for degradation by the ubiquitin-proteasome pathway. Under hypoxic conditions, the hydroxylation by the PHDs is attenuated by lack of the oxygen substrate, allowing HIF-1 to accumulate, translocate to the nucleus, and mediate HIF-mediated gene transcription. In several mammalian species including humans, three PHDs have been identified. We report here the cloning of a full-length rat cDNA that is highly homologous to the human and murine PHD-1 enzymes and encodes a protein that is 416 amino acids long. Both cDNA and protein are widely expressed in rat tissues and cell types. We demonstrate that purified and crude baculovirus-expressed rat PHD-1 exhibits HIF-1alpha specific prolyl hydroxylase activity with similar substrate affinities and is comparable to human PHD-1 protein.


Subject(s)
Cloning, Molecular , Procollagen-Proline Dioxygenase/chemistry , Procollagen-Proline Dioxygenase/genetics , Amino Acid Sequence , Animals , Humans , Kinetics , Mice , Molecular Sequence Data , Organ Specificity/genetics , Procollagen-Proline Dioxygenase/biosynthesis , RNA, Messenger/metabolism , Rats , Spodoptera
11.
Thromb Haemost ; 92(5): 956-65, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15543321

ABSTRACT

During thrombosis, P-selectin is expressed on the surface of activated endothelial cells and platelets. We hypothesized that targeting a plasminogen activator (PA) to P-selectin would enhance local thrombolysis and reduce bleeding risk. Previously, a urokinase (uPA)/anti-P-selectin antibody (HuSZ51) fusion protein was shown to increase fibrinolysis in a hamster pulmonary embolism model. To explore the therapeutic potential of this targeting strategy, we fused the fibrin-selective Desmodus rotundus salivary PA alpha1 (dsPA alpha 1) to HuSZ51 and compared the fibrinolytic activity of P-selectin-targeted dsPA alpha 1 (HuSZ51-dsPA alpha 1) to unmodified dsPA alpha 1 in vitro and in vivo. HuSZ51-dsPA alpha 1 and dsPA alpha 1 were expressed in CHO cells and purified to homogeneity by affinity chromatography. HuSZ51-dsPA alpha 1 bound to thrombin-activated human and dog platelets with comparable affinities to that of parental antibody SZ51. The fusion protein retained the catalytic activities of dsPA alpha 1 in chromogenic and clot lysis assays, indicating that dsPA alpha 1 is fully functional when fused to HuSZ51. Compared to dsPA alpha 1, HuSZ51-dsPA alpha 1 had similar thrombolytic efficacy in a rat pulmonary embolism model and anti-thrombotic potency in a dog model of femoral artery thrombosis. However, HuSZ51-dsPA alpha 1 was less effective in lysis of preexisting arterial thrombi in the dog model. The reduced arterial thrombolysis was not due to the pharmacokinetic properties of HuSZ51-dsPA alpha 1 because antigen level and amidolytic activity were higher in plasma from HuSZ51-dsPA alpha 1-treated groups than corresponding dsPA alpha 1-treated groups. These data indicate that the thrombolytic efficacy of HuSZ51-dsPA alpha 1 varied dependent on the physical composition of thrombi. The lack of stimulation by fibrin in arterial thrombi may contribute to the attenuated thrombolytic efficacy of HuSZ51-dsPA alpha 1 in the dog model.


Subject(s)
Blood Platelets/drug effects , Fibrin/drug effects , Immunoconjugates/therapeutic use , P-Selectin/immunology , Plasminogen Activators/administration & dosage , Thrombolytic Therapy/methods , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Blood Platelets/chemistry , Blood Platelets/physiology , Disease Models, Animal , Dogs , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacokinetics , Fibrinolytic Agents/pharmacology , Genetic Engineering , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Plasminogen Activators/genetics , Plasminogen Activators/therapeutic use , Platelet Aggregation , Pulmonary Embolism/drug therapy , Rats , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Thrombin , Thrombosis/drug therapy , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...