Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Bioorg Chem ; 148: 107463, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776649

ABSTRACT

Thrombosis leads to elevated mortality rates and substantial medical expenses worldwide. Human factor IXa (HFIXa) protease is pivotal in tissue factor (TF)-mediated thrombin generation, and represents a promising target for anticoagulant therapy. We herein isolated novel DNA aptamers that specifically bind to HFIXa through systematic evolution of ligands by exponential enrichment (SELEX) method. We identified two distinct aptamers, seq 5 and seq 11, which demonstrated high binding affinity to HFIXa (Kd = 74.07 ± 2.53 nM, and 4.93 ± 0.15 nM, respectively). Computer software was used for conformational simulation and kinetic analysis of DNA aptamers and HFIXa binding. These aptamers dose-dependently prolonged activated partial thromboplastin time (aPTT) in plasma. We further rationally optimized the aptamers by truncation and site-directed mutation, and generated the truncated forms (Seq 5-1t, Seq 11-1t) and truncated-mutated forms (Seq 5-2tm, Seq 11-2tm). They also showed good anticoagulant effects. The rationally and structurally designed antidotes (seq 5-2b and seq 11-2b) were competitively bound to the DNA aptamers and effectively reversed the anticoagulant effect. This strategy provides DNA aptamer drug-antidote pair with effective anticoagulation and rapid reversal, developing advanced therapies by safe, regulatable aptamer drug-antidote pair.


Subject(s)
Antidotes , Aptamers, Nucleotide , Factor IXa , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Humans , Factor IXa/antagonists & inhibitors , Factor IXa/metabolism , Antidotes/pharmacology , Antidotes/chemistry , Antidotes/chemical synthesis , Dose-Response Relationship, Drug , Anticoagulants/pharmacology , Anticoagulants/chemistry , Structure-Activity Relationship , Molecular Structure , SELEX Aptamer Technique
2.
Int J Biol Macromol ; 269(Pt 2): 131952, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692541

ABSTRACT

Thromboembolic diseases pose a serious risk to human health worldwide. Fucosylated chondroitin sulfate (FCS) is reported to have good anticoagulant activity with a low bleeding risk. Molecular weight plays a significant role in the anticoagulant activity of FCS, and FCS smaller than octasaccharide in size has no anticoagulant activity. Therefore, identifying the best candidate for developing novel anticoagulant FCS drugs is crucial. Herein, native FCS was isolated from sea cucumber Cucumaria frondosa (FCScf) and depolymerized into a series of lower molecular weights (FCScfs). A comprehensive assessment of the in vitro anticoagulant activity and in vivo bleeding risk of FCScfs with different molecule weights demonstrated that 10 kDa FCScf (FCScf-10 K) had a greater intrinsic anticoagulant activity than low molecular weight heparin (LMWH) without any bleeding risk. Using molecular modeling combined with experimental validation, we revealed that FCScf-10 K can specifically inhibit the formation of the Xase complex by binding the negatively charged sulfate group of FCScf-10 K to the positively charged side chain of arginine residues on the specific surface of factor IXa. Thus, these data demonstrate that the intermediate molecular weight FCScf-10 K is a promising candidate for the development of novel anticoagulant drugs.


Subject(s)
Anticoagulants , Chondroitin Sulfates , Factor IXa , Molecular Weight , Animals , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Chondroitin Sulfates/isolation & purification , Anticoagulants/pharmacology , Anticoagulants/chemistry , Anticoagulants/isolation & purification , Factor IXa/metabolism , Factor IXa/antagonists & inhibitors , Factor IXa/chemistry , Cucumaria/chemistry , Sea Cucumbers/chemistry , Blood Coagulation/drug effects , Humans , Models, Molecular
3.
Molecules ; 26(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34500804

ABSTRACT

Blood coagulation is an essential physiological process for hemostasis; however, abnormal coagulation can lead to various potentially fatal disorders, generally known as thromboembolic disorders, which are a major cause of mortality in the modern world. Recently, the FDA has approved several anticoagulant drugs for Factor Xa (FXa) which work via the common pathway of the coagulation cascade. A main side effect of these drugs is the potential risk for bleeding in patients. Coagulation Factor IXa (FIXa) has recently emerged as the strategic target to ease these risks as it selectively regulates the intrinsic pathway. These aforementioned coagulation factors are highly similar in structure, functional architecture, and inhibitor binding mode. Therefore, it remains a challenge to design a selective inhibitor which may affect only FIXa. With the availability of a number of X-ray co-crystal structures of these two coagulation factors as protein-ligand complexes, structural alignment, molecular docking, and pharmacophore modeling were employed to derive the relevant criteria for selective inhibition of FIXa over FXa. In this study, six ligands (three potent, two selective, and one inactive) were selected for FIXa inhibition and six potent ligands (four FDA approved drugs) were considered for FXa. The pharmacophore hypotheses provide the distribution patterns for the principal interactions that take place in the binding site. None of the pharmacophoric patterns of the FXa inhibitors matched with any of the patterns of FIXa inhibitors. Based on pharmacophore analysis, a selectivity of a ligand for FIXa over FXa may be defined quantitatively as a docking score of lower than -8.0 kcal/mol in the FIXa-grids and higher than -7.5 kcal/mol in the FXa-grids.


Subject(s)
Anticoagulants/pharmacology , Factor IXa/antagonists & inhibitors , Factor Xa Inhibitors/pharmacology , Factor Xa/metabolism , Anticoagulants/chemistry , Crystallography, X-Ray , Factor IXa/genetics , Factor IXa/metabolism , Factor Xa/genetics , Factor Xa Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure
4.
Blood ; 138(14): 1258-1268, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34077951

ABSTRACT

Hemophilia A is a bleeding disorder resulting from deficient factor VIII (FVIII), which normally functions as a cofactor to activated factor IX (FIXa) that facilitates activation of factor X (FX). To mimic this property in a bispecific antibody format, a screening was conducted to identify functional pairs of anti-FIXa and anti-FX antibodies, followed by optimization of functional and biophysical properties. The resulting bispecific antibody (Mim8) assembled efficiently with FIXa and FX on membranes, and supported activation with an apparent equilibrium dissociation constant of 16 nM. Binding affinity with FIXa and FX in solution was much lower, with equilibrium dissociation constant values for FIXa and FX of 2.3 and 1.5 µM, respectively. In addition, the activity of Mim8 was dependent on stimulatory activity contributed by the anti-FIXa arm, which enhanced the proteolytic activity of FIXa by 4 orders of magnitude. In hemophilia A plasma and whole blood, Mim8 normalized thrombin generation and clot formation, with potencies 13 and 18 times higher than a sequence-identical analogue of emicizumab. A similar potency difference was observed in a tail vein transection model in hemophilia A mice, whereas reduction of bleeding in a severe tail-clip model was observed only for Mim8. Furthermore, the pharmacokinetic parameters of Mim8 were investigated and a half-life of 14 days shown in cynomolgus monkeys. In conclusion, Mim8 is an activated FVIII mimetic with a potent and efficacious hemostatic effect based on preclinical data.


Subject(s)
Antibodies, Bispecific/therapeutic use , Hemophilia A/drug therapy , Hemorrhage/drug therapy , Animals , Factor IXa/antagonists & inhibitors , Factor VIIIa/therapeutic use , Factor X/antagonists & inhibitors , Female , Humans , Male , Mice, Inbred C57BL
5.
Bioorg Med Chem Lett ; 30(15): 127279, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32527459

ABSTRACT

The synthesis and structure activity relationship development of a pyrimidine series of heterocyclic Factor IXa inhibitors is described. Increased selectivity over Factor Xa inhibition was achieved through SAR expansion of the P1 element. Select compounds were evaluated in vivo to assess their plasma levels in rat.


Subject(s)
Drug Discovery , Factor IXa/antagonists & inhibitors , Factor Xa Inhibitors/pharmacology , Pyrimidines/pharmacology , Dose-Response Relationship, Drug , Factor IXa/metabolism , Factor Xa Inhibitors/chemical synthesis , Factor Xa Inhibitors/chemistry , Humans , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
6.
Carbohydr Polym ; 222: 115025, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31320079

ABSTRACT

Fucosylated chondroitin sulfate (FCS) oligosaccharides of specific molecular weight have shown potent anticoagulant activities with selectivity towards intrinsic factor Xase complex. However, the preparation of FCS oligosaccharides by traditional methods requires multiple purification steps consuming large amounts of time and significant resources. The current study focuses on developing a method for the rapid preparation of FCS oligomers from sea cucumber Pearsonothuria graeffei having 6-18 saccharide residues. The key steps controlling molecular weight (Mw) and purity of these FCS oligomers were evaluated. Structural analysis showed the resulting FCS oligomers were primarily l-Fuc3,4diS-α1,3-d-GlcA-ß1,3-(d-GalNAc4,6diS-ß1,4-[l-Fuc3,4diS-α1,3-]d-GlcA-ß1,3-)nd-anTal-ol4,6diS (n = 1˜5) accompanied by partial de-fucosylation and/or de-sulfation. In vitro and in vivo experiments demonstrate that these FCS oligomers selectively inhibit intrinsic factor Xase complex and exhibit remarkable antithrombotic activity without hemorrhagic and hypotension side effects. This method is suitable for large-scale preparation of FCS oligosaccharides as clinical anticoagulants.


Subject(s)
Anticoagulants/therapeutic use , Chondroitin Sulfates/therapeutic use , Factor IXa/antagonists & inhibitors , Factor VIIIa/antagonists & inhibitors , Fibrinolytic Agents/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacology , Carbohydrate Sequence , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Cysteine Endopeptidases , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Male , Mice , Rabbits , Rats, Sprague-Dawley , Sea Cucumbers/chemistry , Venous Thrombosis/drug therapy
7.
Niger J Clin Pract ; 22(3): 416-421, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30837433

ABSTRACT

INTRODUCTION: Inhibitor formation is a major complication of hemophilia treatment because it interferes with the clinical response to factor replacement and causes significant morbidity. This cross-sectional study was conducted to assess the presence and frequency of inhibitors among registered person with hemophilia and to identify risk factors associated with inhibitor development. PATIENTS AND METHODS: A total of 143 hemophilics, 118 with hemophilia A (HA) and 25 with hemophilia B (HB), were enrolled for the study. Participant's clinical data were obtained through patient's medical records. Factor VIII and IX levels and the presence of inhibitors were assessed using a fully automated coagulometer. From the results of a Bethesda assay, patients were divided into those with high titers (≥5 BU) and those with low titers (<5 BU). RESULTS: The patient's age ranged from 1 to 67 years with median of 13.8 years. Inhibitors were detected in 18.6% and none of HA and HB patients, respectively. Of the 22 patients with HA and inhibitors, 18 (82%) had high titer inhibitors. The frequency of inhibitors was significantly higher among patients with severe hemophilia, a history of early exposure (≤3 months) to factor VIII concentrate, and family histories of autoimmune disease and immune system challenges (P < 0.05). The independent risk factors associated with inhibitor development were severe hemophilia (95% CIs = 1.02-55.6, OR = 7.5) and immune system challenges (95% CIs = 1.14-5.99, OR = 2.6). CONCLUSION: Inhibitors were common among HA patients, and both severe HA and immune system challenges (surgery and trauma) are independent risk factors for inhibitor development.


Subject(s)
Factor IXa/antagonists & inhibitors , Factor VIII/antagonists & inhibitors , Hemophilia A/blood , Hemophilia B/blood , Adolescent , Adult , Aged , Child , Child, Preschool , Cross-Sectional Studies , Factor IXa/metabolism , Factor IXa/therapeutic use , Factor VIII/metabolism , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemophilia B/drug therapy , Hemostatics/adverse effects , Hemostatics/therapeutic use , Humans , Infant , Iraq , Middle Aged , Risk Factors , Young Adult
8.
Eur Heart J Acute Cardiovasc Care ; 8(6): 520-526, 2019 Sep.
Article in English | MEDLINE | ID: mdl-28403626

ABSTRACT

BACKGROUND: Residual platelet reactivity is a predictor of poor prognosis in patients with acute coronary syndromes (ACSs) undergoing percutaneous coronary intervention. Thrombin is a major platelet activator and upon initiation of the coagulation cascade, it is subsequently produced downstream of factor IXa, which itself is known to be increased in ACS. Pegnivacogin is a novel RNA-aptamer based factor IXa inhibitor featuring a reversal agent, anivamersen. We hypothesized that pegnivacogin could reduce platelet reactivity. METHODS: Whole blood samples from healthy volunteers were incubated in vitro in the presence and absence of pegnivacogin and platelet reactivity was analysed. In addition, platelet aggregometry was performed in blood samples from ACS patients in the RADAR trial featuring the intravenous administration of pegnivacogin as well as reversal by anivamersen. RESULTS: In vitro, pegnivacogin significantly reduced adenosine diphosphate-induced CD62P-expression (100% vs. 89.79±4.04%, p=0.027, n=9) and PAC-1 binding (100% vs. 83.02±4.08%, p=0.010, n=11). Platelet aggregation was reduced (97.71±5.30% vs. 66.53±9.92%, p=0.013, n=10) as evaluated by light transmission aggregometry. In the presence of the RNA-aptamer reversal agent anivamersen, neither CD62P-expression nor platelet aggregation was attenuated. In patients with ACS treated with aspirin and clopidogrel, residual platelet aggregation was significantly reduced 20 min after intravenous bolus of 1 mg/kg pegnivacogin (100% versus 43.21±8.23%, p=0.020). CONCLUSION: Inhibition of factor IXa by pegnivacogin decreases platelet activation and aggregation in vitro. This effect was negated by anivamersen. In ACS patients, platelet aggregation was significantly reduced after intravenous pegnivacogin. An aptamer-based anticoagulant inhibiting factor IXa therefore might be a promising antithrombotic strategy in ACS patients.


Subject(s)
Acute Coronary Syndrome/therapy , Aptamers, Nucleotide/therapeutic use , Factor IXa/antagonists & inhibitors , Percutaneous Coronary Intervention/methods , Acute Coronary Syndrome/mortality , Administration, Intravenous , Anticoagulants/therapeutic use , Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/pharmacology , Case-Control Studies , Humans , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacology , Platelet Aggregation/drug effects , Thrombin/pharmacology
9.
Carbohydr Res ; 467: 45-51, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30114596

ABSTRACT

Fucosylated chondroitin sulfate (FuCS) is a structurally complex glycosaminoglycan found in sea cucumbers with a wide spectrum of biological activities, among which anticoagulant activity is particularly attractive for the development of alternative anticoagulant drugs with decreased adverse effects and risks of bleeding. Previous studies show that FuCS glycomimetics bearing several trisaccharide epitopes displayed promising anticoagulant activity and did not change the mode of action of FuCS. To simplify synthetic difficulty of high valent glycoclusters and obtain candidate compounds with relatively low molecular weights, here we report the synthesis of two FuCS glycoclusters with low valence and more compact structures. Anticoagulation studies showed that these simplified "short-armed" glycoclusters demonstrated comparable potency with "long-armed" high valent glycoclusters, offering a concise approach for the development of novel anticoagulant agents.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Chondroitin Sulfates/pharmacology , Enzyme Inhibitors/pharmacology , Glycosides/pharmacology , Anticoagulants/chemical synthesis , Anticoagulants/chemistry , Carbohydrate Conformation , Chondroitin Sulfates/chemical synthesis , Chondroitin Sulfates/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Factor IXa/antagonists & inhibitors , Factor IXa/metabolism , Factor VIIIa/antagonists & inhibitors , Factor VIIIa/metabolism , Glycosides/chemical synthesis , Glycosides/chemistry , Humans
10.
Curr Opin Hematol ; 25(5): 382-388, 2018 09.
Article in English | MEDLINE | ID: mdl-30015643

ABSTRACT

PURPOSE OF REVIEW: Since the selection of the first thrombin-binding aptamer in 1992, the use of nucleic acid aptamers to target specific coagulation factors has emerged as a valuable approach for generating novel anticoagulant and procoagulant therapeutics. Herein, we highlight the most recent discoveries involving application of aptamers for those purposes. RECENT FINDINGS: Learning from the successes and pitfalls of the FIXa-targeting aptamer pegnivacogin in preclinical and clinical studies, the latest efforts to develop antidote-controllable anticoagulation strategies for cardiopulmonary bypass that avoid unfractionated heparin involve potentiation of the exosite-binding factor X (FX)a aptamer 11F7t by combination with either a small molecule FXa catalytic site inhibitor or a thrombin aptamer. Recent work has also focused on identifying aptamer inhibitors of contact pathway factors such as FXIa and kallikrein, which may prove to be well tolerated and effective antithrombotic agents in certain clinical settings. Finally, new approaches to develop procoagulant aptamers to control bleeding associated with hemophilia and other coagulopathies involve targeting activated protein C and tissue plasminogen activator. SUMMARY: Overall, these recent findings exemplify the versatility of aptamers to modulate a variety of procoagulant and anticoagulant factors, along with their capacity to be used complementarily with other aptamers or drugs for wide-ranging applications.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Factor IXa , Factor Xa Inhibitors/therapeutic use , Hemostasis , Animals , Antidotes/pharmacokinetics , Antidotes/therapeutic use , Aptamers, Nucleotide/adverse effects , Aptamers, Nucleotide/pharmacokinetics , Cardiopulmonary Bypass , Catalytic Domain , Factor IXa/antagonists & inhibitors , Factor IXa/metabolism , Factor Xa Inhibitors/adverse effects , Factor Xa Inhibitors/pharmacokinetics , Hemophilia A/blood , Hemophilia A/drug therapy , Humans , Kallikreins/metabolism
11.
J Recept Signal Transduct Res ; 38(3): 213-224, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29724133

ABSTRACT

Human Coagulation Factor IXa (FIXa), specifically inhibited at the initiation stage of the blood coagulation cascade, is an excellent target for developing selective and safe anticoagulants. To explore this inhibitory mechanism, 86 FIXa inhibitors were selected to generate pharmacophore models and subsequently SAR models. Both best pharmacophore model and ROC curve were built through the Receptor-Ligand Pharmacophore Generation module. CoMFA model based on molecular docking and PLS factor analysis methods were developed. Model propagations values are q2 = 0.709, r2 = 0.949, and r2pred = 0.905. The satisfactory q2 value of 0.609, r2 value of 0.962, and r2pred value of 0.819 for CoMSIA indicated that the CoMFA and CoMSIA models are both available to predict the inhibitory activity on FIXa. On the basis of pharmacophore modeling, molecular docking, and 3D-QSAR modeling screening, six molecules are screened as potential FIXa inhibitors.


Subject(s)
Drug Design , Factor IXa/chemistry , Fibrinolytic Agents/chemistry , Thrombosis/drug therapy , Blood Coagulation/drug effects , Factor IXa/antagonists & inhibitors , Fibrinolytic Agents/therapeutic use , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Models, Molecular , Molecular Docking Simulation , Protein Binding/drug effects , Quantitative Structure-Activity Relationship , Thrombosis/genetics , Thrombosis/pathology
12.
J Thromb Haemost ; 16(6): 1078-1088, 2018 06.
Article in English | MEDLINE | ID: mdl-29645406

ABSTRACT

Essentials The activated partial prothrombin time (aPTT) cannot predict the activity of emicizumab (Emi). Adjusted clot waveform analyses using a prothrombin time (PT)/aPTT initiator were developed. Activity of Emi in the co-presence of factor VIII or bypassing agents was quantified. This assay is useful for assessing coagulation potential in Emi-treated hemophilia A. SUMMARY: Background Emicizumab is an anti-activated factor IX/FX bispecific antibody that mimics activated FVIII cofactor function. Emicizumab does not require activation by thrombin, and its effect on shortening the activated partial thromboplastin time (APTT) is much greater than that of FVIII. Therefore, the APTT has limited utility in hemophilia A (HA) patients treated with emicizumab. Aim To evaluate the global coagulation potential of emicizumab. Methods Clot waveform analysis (CWA) with prothrombin time (PT)/APTT mixed reagents was used to define hemostatic monitoring protocols in HA patients. A modified parameter, adjusted-|min1| (Ad|min1|), was developed. Maximum and minimum percentage transmittance were defined as 100% and 0% in the precoagulation and postcoagulation phases, respectively. Ad|min1| was calculated as an index of the maximum velocity of the coagulation process. Results Ad|min1| obtained with mixed-trigger reagent (PT/APTT/buffer, 1 : 15 : 135) in the presence of emicizumab optimally corresponded to the conversion rate estimated in animals; 0.2-0.4 IU dL-1 equivalent FVIII per 1 µg mL-1 emicizumab). Ex vivo addition of emicizumab to HA plasma with or without inhibitors resulted in concentration-dependent increases in Ad|min1|, with some individual variations. The addition of various concentrations of FVIII to HA plasma mixed with emicizumab resulted in dose-dependent increases in Ad|min1|. Similarly, mixtures of activated prothrombin complex concentrate and emicizumab added to HA plasma resulted in dose-dependent increases in Ad|min1|. In contrast, enhanced coagulation potential appeared to be better defined by the clot time than by Ad|min1| in experiments using recombinant activated FVII. Conclusion The PT/APTT reagent-triggered adjusted CWA could provide a useful means of assessing global coagulation potential in emicizumab-treated HA patients, with enhanced activity neither masking nor being masked by FVIII or bypassing agents.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Blood Coagulation/drug effects , Coagulants/pharmacology , Factor IXa/antagonists & inhibitors , Factor X/antagonists & inhibitors , Hemophilia A/diagnosis , Partial Thromboplastin Time , Prothrombin Time , Case-Control Studies , Hemophilia A/blood , Hemophilia A/drug therapy , Humans , Predictive Value of Tests , Reproducibility of Results
13.
Thromb Haemost ; 117(7): 1348-1357, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28451690

ABSTRACT

Emicizumab, a humanised bispecific antibody recognising factors (F) IX/IXa and X/Xa, can accelerate FIXa-catalysed FX activation by bridging FIXa and FX in a manner similar to FVIIIa. However, details of the emicizumab-antigen interactions have not been reported so far. In this study, we first showed by surface plasmon resonance analysis that emicizumab bound FIX, FIXa, FX, and FXa with moderate affinities (KD = 1.58, 1.52, 1.85, and 0.978 µM, respectively). We next showed by immunoblotting analysis that emicizumab recognised the antigens' epidermal growth factor (EGF)-like domains. We then performed KD-based simulation of equilibrium states in plasma for quantitatively predicting the ways that emicizumab would interact with the antigens. The simulation predicted that only a small part of plasma FIX, FX, and emicizumab would form antigen-bridging FIX-emicizumab-FX ternary complex, of which concentration would form a bell-shaped relationship with emicizumab concentration. The bell-shaped concentration dependency was reproduced by plasma thrombin generation assays, suggesting that the plasma concentration of the ternary complex would correlate with emicizumab's cofactor activity. The simulation also predicted that at 10.0-100 µg/ml of emicizumab-levels shown in a previous study to be clinically effective-the majority of plasma FIX, FX, and emicizumab would exist as monomers. In conclusion, emicizumab binds FIX/FIXa and FX/FXa with micromolar affinities at their EGF-like domains. The KD-based simulation predicted that the antigen-bridging ternary complex formed in circulating plasma would correlate with emicizumab's cofactor activity, and the majority of FIX and FX would be free and available for other coagulation reactions.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Factor VIIIa/immunology , Antibodies, Bispecific/blood , Antibodies, Monoclonal, Humanized/blood , Antibody Specificity , Antigen-Antibody Reactions , Binding Sites , Biomimetic Materials/pharmacology , Computer Simulation , Factor IX/antagonists & inhibitors , Factor IX/immunology , Factor IXa/antagonists & inhibitors , Factor IXa/immunology , Factor X/antagonists & inhibitors , Factor X/immunology , Factor Xa/immunology , Factor Xa Inhibitors/blood , Factor Xa Inhibitors/immunology , Factor Xa Inhibitors/pharmacology , Humans , Models, Immunological
15.
Anal Bioanal Chem ; 408(28): 8241-8251, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27655335

ABSTRACT

Heparins are widely used anticoagulant drugs. The current monitoring practice for heparin in plasma, such as the chromogenic anti-factor Xa assay, relies on heparin-triggered activation of antithrombin, an inhibitor of coagulation proteases. Such assays are not applicable to the detection of non-anticoagulant heparins, an emerging class of drug candidates for therapeutic applications unrelated to anticlotting activity. This study describes the application of a commercially available fluorescent probe assay (Heparin Red) for the direct and sensitive detection of the "chemical" heparin in plasma, independent of any anticoagulant activity. The quantification range is about 0-5 µg/mL for both unfractionated heparin (corresponding to 0-1 IU/mL) and the low molecular weight heparin enoxaparin. The Heparin Red assay is of particular value for the quantification of non-anticoagulant heparins, as exemplified by the low molecular weight heparin derivative tafoxiparin and a N-desulfated-N-reacetylated heparin. Heparin octa- and decasaccharides are also detected. Graphical abstract Heparin quantification in plasma by mixing the sample with the Heparin Red reagent and fluorescence readout.


Subject(s)
Factor IXa/antagonists & inhibitors , Fluorescent Dyes/chemistry , Fluorometry/methods , Heparin/analogs & derivatives , Heparin/blood , Healthy Volunteers , Humans , In Vitro Techniques
16.
PLoS One ; 11(5): e0154127, 2016.
Article in English | MEDLINE | ID: mdl-27196067

ABSTRACT

BACKGROUND: Thrombin generation (TG) is a pivotal process in achieving hemostasis. Coagulation profiles during pregnancy and early neonatal period are different from that of normal (non-pregnant) adults. In this ex vivo study, the differences in TG in maternal and cord plasma relative to normal adult plasma were studied. METHODS: Twenty consented pregnant women and ten consented healthy adults were included in the study. Maternal and cord blood samples were collected at the time of delivery. Platelet-poor plasma was isolated for the measurement of TG. In some samples, anti-FIXa aptamer, RB006, or a TFPI inhibitor, BAX499 were added to elucidate the contribution of intrinsic and extrinsic pathway to TG. Additionally, procoagulant and inhibitor levels were measured in maternal and cord plasma, and these values were used to mathematically simulate TG. RESULTS: Peak TG was increased in maternal plasma (393.6±57.9 nM) compared to adult and cord samples (323.2±38.9 nM and 209.9±29.5 nM, respectively). Inhibitory effects of RB006 on TG were less robust in maternal or cord plasma (52% vs. 12% respectively) than in adult plasma (81%). Likewise the effectiveness of BAX499 as represented by the increase in peak TG was much greater in adult (21%) than in maternal (10%) or cord plasma (12%). Further, BAX499 was more effective in reversing RB006 in adult plasma than in maternal or cord plasma. Ex vivo data were reproducible with the results of the mathematical simulation of TG. CONCLUSION: Normal parturient plasma shows a large intrinsic pathway reserve for TG compared to adult and cord plasma, while TG in cord plasma is sustained by extrinsic pathway, and low levels of TFPI and AT.


Subject(s)
Fetal Blood/metabolism , Thrombin/biosynthesis , Adult , Aptamers, Nucleotide/chemistry , Blood Coagulation , Blood Coagulation Tests , Calibration , Case-Control Studies , Coagulants/chemistry , Computer Simulation , Factor IXa/antagonists & inhibitors , Female , Hemostasis , Humans , Models, Statistical , Models, Theoretical , Plasma/metabolism , Pregnancy , Young Adult
19.
J Med Chem ; 59(5): 1818-29, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26871940

ABSTRACT

A potent and selective Factor IXa (FIXa) inhibitor was subjected to a series of liver microsomal incubations, which generated a number of metabolites. Using automated ligand identification system-affinity selection (ALIS-AS) methodology, metabolites in the incubation mixture were prioritized by their binding affinities to the FIXa protein. Microgram quantities of the metabolites of interest were then isolated through microisolation analytical capabilities, and structurally characterized using MicroCryoProbe heteronuclear 2D NMR techniques. The isolated metabolites recovered from the NMR experiments were then submitted directly to an in vitro FIXa enzymatic assay. The order of the metabolites' binding affinity to the Factor IXa protein from the ALIS assay was completely consistent with the enzymatic assay results. This work showcases an innovative and efficient approach to uncover structure-activity relationships (SARs) and guide drug design via microisolation-structural characterization and ALIS capabilities.


Subject(s)
Automation , Drug Design , Factor IXa/antagonists & inhibitors , Fibrinolytic Agents/pharmacology , Nuclear Magnetic Resonance, Biomolecular , Animals , Dose-Response Relationship, Drug , Factor IXa/metabolism , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/metabolism , Humans , Ligands , Molecular Structure , Rats , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...