Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
MAbs ; 7(1): 120-8, 2015.
Article in English | MEDLINE | ID: mdl-25524207

ABSTRACT

While antibody engineering improves the properties of therapeutic antibodies, optimization of regions that do not contact antigens has been mainly focused on modifying the effector functions and pharmacokinetics of antibodies. We recently reported an asymmetric anti-FIXa/FX bispecific IgG4 antibody, ACE910, which mimics the cofactor function of FVIII by placing the two factors into spatial proximity for the treatment of hemophilia A. During the optimization process, we found that the activity was significantly affected by IgG subclass and by modifications to the inter-chain disulfide bonds, upper hinge region, elbow hinge region, and Fc glycan, even though these regions were unlikely to come into direct contact with the antigens. Of these non-antigen-contacting regions, the tertiary structure determined by the inter-chain disulfide bonds was found to strongly affect the FVIII-mimetic activity. Interestingly, IgG4-like disulfide bonds between Cys131 in the heavy chain and Cys114 in the light chain, and disulfide bonds between the two heavy chains at the hinge region were indispensable for the high FVIII-mimetic activity. Moreover, proline mutations in the upper hinge region and removal of the Fc glycan enhanced the FVIII-mimetic activity, suggesting that flexibility of the upper hinge region and the Fc portion structure are important for the FVIII-mimetic activity. This study suggests that these non-antigen-contacting regions can be engineered to improve the biological activity of IgG antibodies with functions similar to ACE910, such as placing two antigens into spatial proximity, retargeting effector cells to target cells, or co-ligating two identical or different antigens on the same cell.


Subject(s)
Antibodies, Bispecific/chemistry , Biomimetic Materials/chemistry , Factor IXa/chemistry , Factor VIII , Factor X/chemistry , Antibodies, Bispecific/genetics , Humans
2.
Blood ; 124(20): 3165-71, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25274508

ABSTRACT

ACE910 is a humanized anti-factor IXa/X bispecific antibody mimicking the function of factor VIII (FVIII). We previously demonstrated in nonhuman primates that a single IV dose of ACE910 exerted hemostatic activity against hemophilic bleeds artificially induced in muscles and subcutis, and that a subcutaneous (SC) dose of ACE910 showed a 3-week half-life and nearly 100% bioavailability, offering support for effective prophylaxis for hemophilia A by user-friendly SC dosing. However, there was no direct evidence that such SC dosing of ACE910 would prevent spontaneous bleeds occurring in daily life. In this study, we newly established a long-term primate model of acquired hemophilia A by multiple IV injections of an anti-primate FVIII neutralizing antibody engineered in mouse-monkey chimeric form to reduce its antigenicity. The monkeys in the control group exhibited various spontaneous bleeding symptoms as well as continuous prolongation of activated partial thromboplastin time; notably, all exhibited joint bleeds, which are a hallmark of hemophilia. Weekly SC doses of ACE910 (initial 3.97 mg/kg followed by 1 mg/kg) significantly prevented these bleeding symptoms; notably, no joint bleeding symptoms were observed. ACE910 is expected to prevent spontaneous bleeds and joint damage in hemophilia A patients even with weekly SC dosing, although appropriate clinical investigation is required.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor IXa/immunology , Factor X/immunology , Hemophilia A/complications , Hemorrhage/complications , Hemorrhage/prevention & control , Animals , Antibodies, Bispecific/immunology , Antibodies, Monoclonal, Humanized/immunology , Cell Line , Disease Models, Animal , Hemorrhage/pathology , Humans , Joints/drug effects , Joints/pathology , Macaca fascicularis , Male , Mice
3.
PLoS One ; 8(2): e57479, 2013.
Article in English | MEDLINE | ID: mdl-23468998

ABSTRACT

In hemophilia A, routine prophylaxis with exogenous factor VIII (FVIII) requires frequent intravenous injections and can lead to the development of anti-FVIII alloantibodies (FVIII inhibitors). To overcome these drawbacks, we screened asymmetric bispecific IgG antibodies to factor IXa (FIXa) and factor X (FX), mimicking the FVIII cofactor function. Since the therapeutic potential of the lead bispecific antibody was marginal, FVIII-mimetic activity was improved by modifying its binding properties to FIXa and FX, and the pharmacokinetics was improved by engineering the charge properties of the variable region. Difficulties in manufacturing the bispecific antibody were overcome by identifying a common light chain for the anti-FIXa and anti-FX heavy chains through framework/complementarity determining region shuffling, and by pI engineering of the two heavy chains to facilitate ion exchange chromatographic purification of the bispecific antibody from the mixture of byproducts. Engineering to overcome low solubility and deamidation was also performed. The multidimensionally optimized bispecific antibody hBS910 exhibited potent FVIII-mimetic activity in human FVIII-deficient plasma, and had a half-life of 3 weeks and high subcutaneous bioavailability in cynomolgus monkeys. Importantly, the activity of hBS910 was not affected by FVIII inhibitors, while anti-hBS910 antibodies did not inhibit FVIII activity, allowing the use of hBS910 without considering the development or presence of FVIII inhibitors. Furthermore, hBS910 could be purified on a large manufacturing scale and formulated into a subcutaneously injectable liquid formulation for clinical use. These features of hBS910 enable routine prophylaxis by subcutaneous delivery at a long dosing interval without considering the development or presence of FVIII inhibitors. We expect that hBS910 (investigational drug name: ACE910) will provide significant benefit for severe hemophilia A patients.


Subject(s)
Antibodies, Bispecific/immunology , Factor VIII/physiology , Immunoglobulin G/immunology , Epitopes/immunology , Factor VIII/immunology , Factor VIII/pharmacokinetics , Humans , Isoelectric Point , Solubility , T-Lymphocytes/immunology
4.
Nat Med ; 18(10): 1570-4, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23023498

ABSTRACT

Hemophilia A is a bleeding disorder resulting from coagulation factor VIII (FVIII) deficiency. Exogenously provided FVIII effectively reduces bleeding complications in patients with severe hemophilia A. In approximately 30% of such patients, however, the 'foreignness' of the FVIII molecule causes them to develop inhibitory antibodies against FVIII (inhibitors), precluding FVIII treatment in this set of patients. Moreover, the poor pharmacokinetics of FVIII, attributed to low subcutaneous bioavailability and a short half-life of 0.5 d, necessitates frequent intravenous injections. To overcome these drawbacks, we generated a humanized bispecific antibody to factor IXa (FIXa) and factor X (FX), termed hBS23, that places these two factors into spatially appropriate positions and mimics the cofactor function of FVIII. hBS23 exerted coagulation activity in FVIII-deficient plasma, even in the presence of inhibitors, and showed in vivo hemostatic activity in a nonhuman primate model of acquired hemophilia A. Notably, hBS23 had high subcutaneous bioavailability and a 2-week half-life and would not be expected to elicit the development of FVIII-specific inhibitory antibodies, as its molecular structure, and hence antigenicity, differs from that of FVIII. A long-acting, subcutaneously injectable agent that is unaffected by the presence of inhibitors could markedly reduce the burden of care for the treatment of hemophilia A.


Subject(s)
Antibodies, Bispecific , Factor IXa/immunology , Factor VIII/physiology , Factor X/immunology , Hemophilia A/therapy , Hemostasis , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Hemophilia A/immunology , Macaca fascicularis
5.
Chem Pharm Bull (Tokyo) ; 58(1): 38-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20045964

ABSTRACT

Selective factor VIIa-tissue factor complex (FVIIa/TF) inhibition is regarded as a promising target for developing new anticoagulant drugs. In previous reports, we described a S3 subsite found in the X-ray crystal structure of compound 2 that bound to FVIIa/soluble tissue factor (sTF). Based on the X-ray crystal structure information and with the aim of improving the inhibition activity for FVIIa/TF and selectivity against other serine proteases, we synthesized derivatives by introducing substituents at position 5 of the indole ring of compound 2. Among them, compound 16 showed high selectivity against other serine proteases. Contrary to our expectations, compound 16 did not occupy the S3-subsite; X-ray structure analysis revealed that compound 16 improved selectivity by forming hydrogen bonds with Gln217, Thr99 and Asn100.


Subject(s)
Factor VIIa/antagonists & inhibitors , Factor VIIa/metabolism , Peptides/chemistry , Peptides/pharmacology , Biomimetics , Crystallography, X-Ray , Factor VIIa/chemistry , Models, Molecular , Protein Binding , Thromboplastin/antagonists & inhibitors , Thromboplastin/chemistry , Thromboplastin/metabolism
6.
Bioorg Med Chem Lett ; 18(16): 4533-7, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18674905

ABSTRACT

Selective factor VIIa-tissue factor complex (FVIIa/TF) inhibition is regarded as a promising target for developing new anticoagulant drugs. Compound 1 was discovered from focused screening of serine protease-directed compounds from our internal collection. Using parallel synthesis supported by structure-based drug design, we identified peptidemimetic FVIIa/TF inhibitors (compounds 4-11) containing L-Gln or L-Met as the P2 moiety. However, these compounds lacked the selectivity of other serine proteases in the coagulation cascade, especially thrombin. Further optimization of these compounds was carried out with a focus on the P4 moiety. Among the optimized compounds, 12b-f showed improved selectivity.


Subject(s)
Chemistry, Pharmaceutical/methods , Factor VIIa/antagonists & inhibitors , Serine Endopeptidases/pharmacology , Serine Proteinase Inhibitors/chemical synthesis , Thromboembolism/drug therapy , Blood Coagulation/drug effects , Crystallography, X-Ray/methods , Drug Design , Humans , Kinetics , Models, Chemical , Molecular Conformation , Peptides/chemistry , Serine Endopeptidases/chemistry , Serine Proteinase Inhibitors/chemistry , Thromboembolism/enzymology
7.
Biochem Biophys Res Commun ; 327(2): 589-96, 2005 Feb 11.
Article in English | MEDLINE | ID: mdl-15629154

ABSTRACT

Selective factor VIIa-tissue factor complex (FVIIa/TF) inhibition is seen as a promising target for developing new anticoagulant drugs. Structure-based designs of the P3 moiety in the peptide mimetic factor VIIa inhibitor successfully lead to novel inhibitors with selectivity for FVIIa/TF and extrinsic coagulation the same as or even higher than those of previously reported peptide mimetic factor VIIa inhibitors. X-ray crystal structure analysis reveals that one of the novel inhibitors shows improved selectivity by forming interactions between the inhibitor and FVIIa as expected. Another of the novel inhibitors achieves improved selectivity through an unexpected hydrogen bond with Gln217, with a unique bent conformation in FVIIa/TF accompanied by conformational changes of the inhibitor and the protein.


Subject(s)
Biomimetic Materials/chemistry , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Factor VIIa/antagonists & inhibitors , Peptides/chemical synthesis , Peptides/pharmacology , Amino Acid Sequence , Binding Sites , Biomimetic Materials/chemical synthesis , Biomimetic Materials/pharmacology , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Factor VIIa/chemistry , Factor VIIa/metabolism , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary , Sequence Alignment , Structure-Activity Relationship
8.
Biochem Biophys Res Commun ; 326(4): 859-65, 2005 Jan 28.
Article in English | MEDLINE | ID: mdl-15607748

ABSTRACT

Selective factor VIIa-tissue factor complex (FVIIa/TF) inhibition is seen as a promising target for developing new anticoagulant drugs. A novel peptide mimetic factor VIIa inhibitor, ethylsulfonamide-d-biphenylalanine-Gln-p-aminobenzamidine, shows 100-fold selectivity against thrombin in spite of its large P3 moiety, unlike previously reported FVIIa/TF selective inhibitors. X-ray crystal structure analysis reveals that the large P3 moiety, d-biphenylalanine, and the small P4 moiety, ethylsulfonamide, make novel interactions with the 170-loop and Lys192 of FVIIa/TF, respectively, accompanying ligand-induced conformational changes of the 170-loop, Gln217, and Lys192. Structural comparisons of FVIIa with thrombin and amino acid sequence comparisons among coagulation serine proteases suggest that these interactions play an important role in achieving selective inhibition for FVIIa/TF.


Subject(s)
Biomimetics/methods , Blood Coagulation Factor Inhibitors/chemistry , Factor VIIa/antagonists & inhibitors , Models, Molecular , Peptides/chemistry , Thrombin/chemistry , Amino Acid Sequence , Binding Sites , Computer Simulation , Crystallography, X-Ray , Enzyme Activation , Humans , Models, Chemical , Molecular Sequence Data , Protein Binding , Protein Conformation , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...