Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
ERJ Open Res ; 9(6)2023 Nov.
Article in English | MEDLINE | ID: mdl-38152083

ABSTRACT

Background: Exertional breathlessness is a major symptom in cardiorespiratory disease and is often assessed using the modified Medical Research Council (mMRC) questionnaire. The mMRC might underestimate exertional breathlessness in people with impaired exercise capacity who have reduced their physical activity to avoid the symptom. We aimed to evaluate the ability of mMRC to detect abnormally high exertional breathlessness or abnormally low exercise capacity during incremental cycle exercise testing (IET). Methods: A secondary analysis of data from a randomised controlled trial of outpatients aged 18 years or older referred for IET was carried out. Participants completed the mMRC before IET. Abnormally high exertional breathlessness was defined as a breathlessness (Borg 0-10) intensity response more than the upper limit of normal. Abnormally low exercise capacity was defined using published reference equations. The sensitivity, specificity, accuracy and discriminative ability of each mMRC rating to detect each outcomewas calculated. Results: 92 participants were included; the mean age was 59 years, 61% were male, and 64% and 15% had mMRC 1 and ≥2, respectively. An mMRC ≥2 had the highest accuracy (71%) to detect abnormally high exertional breathlessness, with a specificity of 93% but a sensitivity of only 28%, failing to identify 72% of people with abnormally high exertional breathlessness. The accuracy, specificity and sensitivity for abnormally low exercise capacity was 64%, 88% and 19%, respectively. Conclusion: Among people referred for clinical exercise testing, the mMRC dyspnoea scale misclassified exertional breathlessness and exercise capacity assessed using cycle IET, with substantial underdetection. A mMRC dyspnoea rating of 0-1 does not preclude the presence of abnormally high exertional breathlessness or abnormally low exercise capacity.

2.
Acta Neurochir (Wien) ; 164(3): 615-624, 2022 03.
Article in English | MEDLINE | ID: mdl-34936014

ABSTRACT

BACKGROUND: A major challenge in management of traumatic brain injury (TBI) is to assess the heterogeneity of TBI pathology and outcome prediction. A reliable outcome prediction would have both great value for the healthcare provider, but also for the patients and their relatives. A well-known prediction model is the International Mission for Prognosis and Analysis of Clinical Trials (IMPACT) prognostic calculator. The aim of this study was to externally validate all three modules of the IMPACT calculator on TBI patients admitted to Uppsala University hospital (UUH). METHOD: TBI patients admitted to UUH are continuously enrolled into the Uppsala neurointensive care unit (NICU) TBI Uppsala Clinical Research (UCR) quality register. The register contains both clinical and demographic data, radiological evaluations, and outcome assessments based on the extended Glasgow outcome scale extended (GOSE) performed at 6 months to 1 year. In this study, we included 635 patients with severe TBI admitted during 2008-2020. We used IMPACT core parameters: age, motor score, and pupillary reaction. RESULTS: The patients had a median age of 56 (range 18-93), 142 female and 478 male. Using the IMPACT Core model to predict outcome resulted in an AUC of 0.85 for mortality and 0.79 for unfavorable outcome. The CT module did not increase AUC for mortality and slightly decreased AUC for unfavorable outcome to 0.78. However, the lab module increased AUC for mortality to 0.89 but slightly decreased for unfavorable outcome to 0.76. Comparing the predicted risk to actual outcomes, we found that all three models correctly predicted low risk of mortality in the surviving group of GOSE 2-8. However, it produced a greater variance of predicted risk in the GOSE 1 group, denoting general underprediction of risk. Regarding unfavorable outcome, all models once again underestimated the risk in the GOSE 3-4 groups, but correctly predicts low risk in GOSE 5-8. CONCLUSIONS: The results of our study are in line with previous findings from centers with modern TBI care using the IMPACT model, in that the model provides adequate prediction for mortality and unfavorable outcome. However, it should be noted that the prediction is limited to 6 months outcome and not longer time interval.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/therapy , Cohort Studies , Female , Glasgow Outcome Scale , Humans , Male , Prognosis , Sweden/epidemiology
3.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808272

ABSTRACT

Traumatic brain injury is one of the leading causes of mortality and morbidity in the world with no current pharmacological treatment. The role of BDNF in neural repair and regeneration is well established and has also been the focus of TBI research. Here, we review experimental animal models assessing BDNF expression following injury as well as clinical studies in humans including the role of BDNF polymorphism in TBI. There is a large heterogeneity in experimental setups and hence the results with different regional and temporal changes in BDNF expression. Several studies have also assessed different interventions to affect the BDNF expression following injury. Clinical studies highlight the importance of BDNF polymorphism in the outcome and indicate a protective role of BDNF polymorphism following injury. Considering the possibility of affecting the BDNF pathway with available substances, we discuss future studies using transgenic mice as well as iPSC in order to understand the underlying mechanism of BDNF polymorphism in TBI and develop a possible pharmacological treatment.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Brain-Derived Neurotrophic Factor/pharmacology , Animals , Brain Injuries/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Humans , Models, Animal , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Recovery of Function/drug effects
4.
J Biol Chem ; 295(15): 5136-5151, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32132173

ABSTRACT

Increased plasma concentrations of lipoprotein(a) (Lp(a)) are associated with an increased risk for cardiovascular disease. Lp(a) is composed of apolipoprotein(a) (apo(a)) covalently bound to apolipoprotein B of low-density lipoprotein (LDL). Many of apo(a)'s potential pathological properties, such as inhibition of plasmin generation, have been attributed to its main structural domains, the kringles, and have been proposed to be mediated by their lysine-binding sites. However, available small-molecule inhibitors, such as lysine analogs, bind unselectively to kringle domains and are therefore unsuitable for functional characterization of specific kringle domains. Here, we discovered small molecules that specifically bind to the apo(a) kringle domains KIV-7, KIV-10, and KV. Chemical synthesis yielded compound AZ-05, which bound to KIV-10 with a Kd of 0.8 µm and exhibited more than 100-fold selectivity for KIV-10, compared with the other kringle domains tested, including plasminogen kringle 1. To better understand and further improve ligand selectivity, we determined the crystal structures of KIV-7, KIV-10, and KV in complex with small-molecule ligands at 1.6-2.1 Å resolutions. Furthermore, we used these small molecules as chemical probes to characterize the roles of the different apo(a) kringle domains in in vitro assays. These assays revealed the assembly of Lp(a) from apo(a) and LDL, as well as potential pathophysiological mechanisms of Lp(a), including (i) binding to fibrin, (ii) stimulation of smooth-muscle cell proliferation, and (iii) stimulation of LDL uptake into differentiated monocytes. Our results indicate that a small-molecule inhibitor targeting the lysine-binding site of KIV-10 can combat the pathophysiological effects of Lp(a).


Subject(s)
Apolipoproteins A/antagonists & inhibitors , Apolipoproteins A/metabolism , Fibrin/metabolism , Kringles/drug effects , Small Molecule Libraries/pharmacology , Amino Acid Sequence , High-Throughput Screening Assays , Humans , Ligands , Models, Molecular , Protein Binding , Protein Domains , Sequence Homology
5.
J Pharmacol Exp Ther ; 368(2): 255-261, 2019 02.
Article in English | MEDLINE | ID: mdl-30482795

ABSTRACT

Fewer new medicines have become available to patients during the last decades. Clinical efficacy failures in late-phase development have been identified as a common cause of this decline. Improved ways to ensure early selection of the right drug targets when it comes to efficacy is therefore a highly desirable goal. The aim of this work was to develop a strategy to facilitate selection of novel targets already in the discovery phase that later on in clinical development would demonstrate efficacy. A cross-functional team at AstraZeneca with extensive experience in drug discovery and development participated in several workshops to identify the critical elements that contribute to building human target validation [(HTV); the relevance of the target from a human perspective]. The elements were consolidated into a 10-point HTV classification system that was ranked from lowest to highest in terms of perceived impact on future clinical efficacy. Using 50 years of legacy research and development data, the ability of the 10-point HTV classification to predict future clinical efficacy was evaluated. Drug targets were classified as having low, medium, or high HTV at the time of candidate drug selection. Comparing this HTV classification with later clinical development efficacy data showed that HTV classification was highly predictive of future clinical efficacy success. This new strategy for HTV assessment provides a novel approach to early prediction of clinical efficacy and a better understanding of portfolio risk.


Subject(s)
Drug Delivery Systems/classification , Drug Delivery Systems/trends , Drug Development/classification , Drug Development/trends , Drug Delivery Systems/methods , Drug Development/methods , Forecasting , Humans , Reproducibility of Results , Treatment Outcome
6.
Neurosci Lett ; 681: 37-43, 2018 08 10.
Article in English | MEDLINE | ID: mdl-29758302

ABSTRACT

Plasminogen binding inhibitors (PBIs) reduce the risk of bleeding in hemorrhagic conditions. However, generic PBIs are also associated with an increased risk of seizures, an adverse effect linked to unwanted activities towards inhibitory neuronal receptors. Development of novel PBIs serve to remove compounds with such properties, but progress is limited by a lack of higher throughput methods with human translatability. Herein we apply human induced pluripotent stem cell (hiPSC) derived neurons in combination with dynamic mass redistribution (DMR) technology to demonstrate robust and reproducible modulation of both GABAA and glycine receptors. These cells respond to GABA (EC50 0.33 ±â€¯0.18 µM), glycine (EC50 11.0 ±â€¯3.7 µM) and additional ligands in line with previous reports from patch clamp technologies. Additionally, we identify and characterize a competitive antagonistic behavior of the prototype inhibitor and drug tranexamic acid (TXA). Finally, we demonstrate proof of concept for effective counter-screening of lead series compounds towards unwanted GABAA receptor activities. No activity was observed for a previously identified PBI candidate drug, AZD6564, whereas a discontinued analog, AZ13267257, could be characterized as a potent GABAA receptor agonist.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Plasminogen Inactivators/pharmacology , Receptors, GABA-A/metabolism , Receptors, Glycine/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Glycine/pharmacology , Humans , Induced Pluripotent Stem Cells/drug effects , Neurons/drug effects , Protein Binding/physiology , Receptors, Glycine/agonists , Tranexamic Acid/pharmacology , gamma-Aminobutyric Acid/pharmacology
7.
J Chem Inf Model ; 57(7): 1703-1714, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28653850

ABSTRACT

Inhibition of plasmin has been found to effectively reduce fibrinolysis and to avoid hemorrhage. This can be achieved by addressing its kringle 1 domain with the known drug and lysine analogue tranexamic acid. Guided by shape similarities toward a previously discovered lead compound, 5-(4-piperidyl)isoxazol-3-ol, a set of 16 structurally similar compounds was assembled and investigated. Successfully, in vitro measurements revealed one compound, 5-(4-piperidyl)isothiazol-3-ol, superior in potency compared to the initial lead. Furthermore, a strikingly high correlation (R2 = 0.93) between anti-fibrinolytic activity and kringle 1 binding affinity provided strong support for the hypothesized inhibition mechanism, as well as revealing opportunities to fine-tune biological effects through minor structural modifications. Several different ligand-based (Freeform, shape, and electrostatic-based similarities) and structure-based methods (e.g., Posit, MM/GBSA, FEP+) were used to retrospectively predict the binding affinities. A combined method, molecular alignment using Posit and scoring with Tcombo, lead to the highest coefficient of determination (R2 = 0.6).


Subject(s)
Antifibrinolytic Agents/chemistry , Antifibrinolytic Agents/pharmacology , Drug Discovery , Fibrinolysin/antagonists & inhibitors , Isoxazoles/chemistry , Isoxazoles/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Antifibrinolytic Agents/metabolism , Fibrinolysin/chemistry , Fibrinolysin/metabolism , Isoxazoles/metabolism , Molecular Docking Simulation , Piperidines/metabolism , Protein Domains , Quantitative Structure-Activity Relationship , Thermodynamics
8.
Adv Mater ; 29(8)2017 Feb.
Article in English | MEDLINE | ID: mdl-28004860

ABSTRACT

A new device architecture for fast organic transistor memory is developed, based on a vertical organic transistor configuration incorporating high-performance ambipolar conjugated polymers and unipolar small molecules as the transport layers, to achieve reliable and fast programming and erasing of the threshold voltage shift in less than 200 ns.

9.
J Med Chem ; 59(14): 6658-70, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27347787

ABSTRACT

The only oral direct thrombin inhibitors that have reached the market, ximelagatran and dabigatran etexilat, are double prodrugs with low bioavailability in humans. We have evaluated an alternative strategy: the preparation of a nonpeptidic, polar direct thrombin inhibitor as a single, macrocyclic esterase-cleavable (acyloxy)alkoxy prodrug. Two homologous prodrugs were synthesized and displayed high solubilities and Caco-2 cell permeabilities, suggesting high absorption from the intestine. In addition, they were rapidly and completely converted to the active zwitterionic thrombin inhibitor in human hepatocytes. Unexpectedly, the most promising prodrug displayed only moderately higher oral bioavailability in rat than the polar direct thrombin inhibitor, most likely due to rapid metabolism in the intestine or the intestinal wall. To the best of our knowledge, this is the first in vivo ADME study of macrocyclic (acyloxy)alkoxy prodrugs, and it remains to be established if the modest increase in bioavailability is a general feature of this category of prodrugs or not.


Subject(s)
Macrocyclic Compounds/pharmacology , Prodrugs/pharmacology , Serine Proteinase Inhibitors/pharmacology , Thrombin/antagonists & inhibitors , Animals , Biological Availability , Caco-2 Cells , Dose-Response Relationship, Drug , Hepatocytes/chemistry , Hepatocytes/metabolism , Humans , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/metabolism , Molecular Structure , Prodrugs/chemistry , Prodrugs/metabolism , Rats , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Solubility , Structure-Activity Relationship , Thrombin/metabolism
10.
PLoS One ; 10(1): e0113705, 2015.
Article in English | MEDLINE | ID: mdl-25629509

ABSTRACT

Activated factor XI (FXIa) inhibitors are anticipated to combine anticoagulant and profibrinolytic effects with a low bleeding risk. This motivated a structure aided fragment based lead generation campaign to create novel FXIa inhibitor leads. A virtual screen, based on docking experiments, was performed to generate a FXIa targeted fragment library for an NMR screen that resulted in the identification of fragments binding in the FXIa S1 binding pocket. The neutral 6-chloro-3,4-dihydro-1H-quinolin-2-one and the weakly basic quinolin-2-amine structures are novel FXIa P1 fragments. The expansion of these fragments towards the FXIa prime side binding sites was aided by solving the X-ray structures of reported FXIa inhibitors that we found to bind in the S1-S1'-S2' FXIa binding pockets. Combining the X-ray structure information from the identified S1 binding 6-chloro-3,4-dihydro-1H-quinolin-2-one fragment and the S1-S1'-S2' binding reference compounds enabled structure guided linking and expansion work to achieve one of the most potent and selective FXIa inhibitors reported to date, compound 13, with a FXIa IC50 of 1.0 nM. The hydrophilicity and large polar surface area of the potent S1-S1'-S2' binding FXIa inhibitors compromised permeability. Initial work to expand the 6-chloro-3,4-dihydro-1H-quinolin-2-one fragment towards the prime side to yield molecules with less hydrophilicity shows promise to afford potent, selective and orally bioavailable compounds.


Subject(s)
Drug Design , Drug Evaluation, Preclinical , Factor XIa/chemistry , Quantitative Structure-Activity Relationship , Serine Proteinase Inhibitors/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Drug Evaluation, Preclinical/methods , Factor XIa/antagonists & inhibitors , Humans , Ligands , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Library , Protein Binding , Serine Proteinase Inhibitors/pharmacology
11.
ACS Med Chem Lett ; 5(5): 538-43, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24900876

ABSTRACT

A class of novel oral fibrinolysis inhibitors has been discovered, which are lysine mimetics containing an isoxazolone as a carboxylic acid isostere. As evidenced by X-ray crystallography the inhibitors bind to the lysine binding site in plasmin thus preventing plasmin from binding to fibrin, hence blocking the protein-protein interaction. Optimization of the series, focusing on potency in human buffer and plasma clotlysis assays, permeability, and GABAa selectivity, led to the discovery of AZD6564 (19) displaying an in vitro human plasma clot lysis IC50 of 0.44 µM, no detectable activity against GABAa, and with DMPK properties leading to a predicted dose of 340 mg twice a day oral dosing in humans.

12.
Sci Rep ; 4: 5318, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24937703

ABSTRACT

Lipoprotein (a) [Lp(a)] is a low density lipoprotein (LDL) with one apolipoprotein (a) molecule bound to the apolipoprotein B-100 of LDL. Lp(a) is an independent risk factor for cardiovascular disease (CVD). However, the relationship of Lp(a) to diabetes and metabolic syndrome, both known for increased CVD risk, is controversial. In a population based study on type two diabetes mellitus (T2DM) development in women, Lp(a) plasma levels showed the well known skewed distribution without any relation to diabetes or impaired glucose tolerance. A modified clot lysis assay on a subset of 274 subjects showed significantly increased clot lysis times in T2DM subjects, despite inhibition of PAI-1 and TAFI. Lp(a) plasma levels significantly increased the maximal peak height of the clot lysis curve, indicating a change in clot structure. In this study Lp(a) is not related to the development of T2DM but may affect clot structure ex vivo without a prolongation of the clot lysis time.


Subject(s)
Diabetes Mellitus, Type 2/blood , Lipoprotein(a)/blood , Aged , Analysis of Variance , Blood Coagulation , Blood Coagulation Tests , Carboxypeptidase B2/blood , Female , Fibrinolysis , Humans , Middle Aged , Plasminogen Activator Inhibitor 1/metabolism
13.
Bioorg Med Chem Lett ; 24(3): 821-7, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24418773

ABSTRACT

A design strategy was used to identify inhibitors of activated protein C with selectivity over thrombin featured by a basic and/or aromatic functionality for binding to the S2 pocket. Our strongest inhibitor showed an IC50-material value and selectivity for APC vs thrombin similar to a compound previously reported in the literature. However, in contrast to the reference compound, our compound showed a retained coagulant effect of thrombin with increasing substrate concentration in a modified Calibrated Automated Thrombogram (CAT) method. This was likely related to our compound being inactive against FVIIa, while the reference compound showed an IC50 of 8.9 µM. Thus, the higher selectivity of our compound against all relevant coagulation factors likely explained its higher therapeutic potential in comparison to the reference compound. The data indicate that at least a 100-fold selectivity over other serine proteases in the coagulation cascade will be required for an effective APC inhibitor.


Subject(s)
Drug Design , Protein C Inhibitor/chemical synthesis , Protein C Inhibitor/pharmacology , Thrombin/antagonists & inhibitors , Binding Sites , Coagulants/chemical synthesis , Coagulants/chemistry , Coagulants/pharmacology , Hemophilia A/drug therapy , Inhibitory Concentration 50 , Protein Binding/drug effects , Protein C Inhibitor/chemistry , Structure-Activity Relationship , Substrate Specificity
14.
Platelets ; 25(4): 257-63, 2014.
Article in English | MEDLINE | ID: mdl-23855479

ABSTRACT

Abstract Platelets play an important role in abdominal sepsis and P2Y12 receptor antagonists have been reported to exert anti-inflammatory effects. Herein, we assessed the impact of platelet inhibition with the P2Y12 receptor antagonist ticagrelor on pulmonary neutrophil recruitment and tissue damage in a model of abdominal sepsis. Wild-type C57BL/6 mice were subjected to cecal ligation and puncture (CLP). Animals were treated with ticagrelor (100 mg/kg) or vehicle prior to CLP induction. Edema formation and bronchoalveolar neutrophils as well as lung damage were quantified. Flow cytometry was used to determine expression of platelet-neutrophil aggregates, neutrophil activation and CD40L expression on platelets. CLP-induced pulmonary infiltration of neutrophils at 24 hours was reduced by 50% in ticagrelor-treated animals. Moreover, ticagrelor abolished CLP-provoked lung edema and decreased lung damage score by 41%. Notably, ticagrelor completely inhibited formation of platelet-neutrophil aggregates and markedly reduced thrombocytopenia in CLP animals. In addition, ticagrelor reduced platelet shedding of CD40L in septic mice. Our data indicate that ticagrelor can reduce CLP-induced pulmonary neutrophil recruitment and lung damage suggesting a potential role for platelet antagonists, such as ticagrelor, in the management of patients with abdominal sepsis.


Subject(s)
Adenosine/analogs & derivatives , Lung Injury/pathology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Sepsis/metabolism , Adenosine/pharmacology , Animals , Blood Platelets/metabolism , Chemokines, CXC/metabolism , Disease Models, Animal , Lung Injury/etiology , Lung Injury/metabolism , Macrophage-1 Antigen/metabolism , Male , Mice , Neutrophils/pathology , Platelet Aggregation/drug effects , Pulmonary Edema/etiology , Pulmonary Edema/pathology , Sepsis/complications , Sepsis/etiology , Ticagrelor
15.
Thromb Res ; 132(2): 248-55, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23830061

ABSTRACT

INTRODUCTION: Oral treatment is lacking for haemophilia, the rare bleeding disorders, and some severe forms of von Willebrand's disease. We have serendipitously identified a small molecule procoagulant compound (AZ10047130). This publication describes some characteristics of AZ10047130 and a systematic search for novel hits using a, human plasma-based, high-throughput screening (HTS) assay. MATERIAL AND METHODS: Coagulation, thrombin generation, chromogenic assays and surface plasmon resonance (SPR) experiments were used to characterise AZ10047130. A 1536-well formatted human plasma coagulation assay for HTS was developed. RESULTS: In the plasma clot assay (re-calcified plasma with low tissue factor) AZ10047130 shortened time to coagulation with an EC50 value of 3.9 µM (assay concentration). AZ10047130 was similarly effective in immunodepleted human and haemophilia A plasmas. SPR and chromogenic substrate experiments indicated that AZ10047130 binds to the heparin binding site of several coagulation factors. The HTS screened in excess of one million compounds. It generated some hits belonging to the same pharmacophore as AZ10047130 but also some entirely novel hits. CONCLUSION: These novel small molecule procoagulant compounds may serve as templates for discovery of oral procoagulant drugs.


Subject(s)
Benzofurans/pharmacology , Blood Chemical Analysis/methods , Blood Coagulation Factors/pharmacology , Blood Coagulation/drug effects , High-Throughput Screening Assays/methods , Sulfonamides/pharmacology , Benzofurans/chemistry , Blood Coagulation Factors/chemistry , Hemophilia A/blood , Hemophilia A/drug therapy , Hemostasis/drug effects , Humans , Sulfonamides/chemistry , Thrombin/biosynthesis
16.
BMC Nephrol ; 14: 164, 2013 Jul 29.
Article in English | MEDLINE | ID: mdl-23895142

ABSTRACT

Uric acid is the end product of purine metabolism in humans. High levels are causative in gout and urolithiasis. Hyperuricaemia has also been implicated in the pathophysiology of hypertension, chronic kidney disease (CKD), congestive heart failure (CHF), the metabolic syndrome, type 2 diabetes mellitus (T2DM), and atherosclerosis, with or without cardiovascular events. This article briefly reviews uric acid metabolism and summarizes the current literature on hyperuricaemia in cardiovascular disease and related co-morbidities, and emerging treatment options.


Subject(s)
Cardiovascular Diseases/mortality , Cardiovascular Diseases/physiopathology , Hyperuricemia/mortality , Hyperuricemia/physiopathology , Animals , Cardiovascular Diseases/epidemiology , Humans , Hyperuricemia/epidemiology , Morbidity
17.
J Med Chem ; 56(8): 3273-80, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23521080

ABSTRACT

Protein-protein interfaces provide an important class of drug targets currently receiving increased attention. The typical design strategy to inhibit protein-protein interactions usually involves large molecules such as peptides and macrocycles. One exception is tranexamic acid (TXA), which, as a lysine mimetic, inhibits binding of plasminogen to fibrin. However, the daily dose of TXA is high due to its modest potency and pharmacokinetic properties. In this study, we report a computational approach, where the focus was on finding electrostatic potential similarities to TXA. Coupling this computational technique with a high-quality low-throughput screen identified 5-(4-piperidyl)-3-isoxazolol (4-PIOL) as a potent plasminogen binding inhibitor with the potential for the treatment of various bleeding disorders. Remarkably, 4-PIOL was found to be more than four times as potent as the drug TXA.


Subject(s)
Antifibrinolytic Agents/pharmacology , Fibrinolysis/drug effects , Isoxazoles/pharmacology , Piperidines/pharmacology , Antifibrinolytic Agents/chemistry , Computational Biology , Drug Evaluation, Preclinical , Humans , Isoxazoles/chemistry , Models, Molecular , Piperidines/chemistry , Plasminogen/metabolism , Protein Binding , Static Electricity , Tranexamic Acid/pharmacology
18.
J Biol Chem ; 288(2): 873-85, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23155046

ABSTRACT

A novel class of small molecule inhibitors for plasminogen activator inhibitor type 1 (PAI-1), represented by AZ3976, was identified in a high throughput screening campaign. AZ3976 displayed an IC(50) value of 26 µm in an enzymatic chromogenic assay. In a plasma clot lysis assay, the compound was active with an IC(50) of 16 µm. Surprisingly, AZ3976 did not bind to active PAI-1 but bound to latent PAI-1 with a K(D) of 0.29 µm at 35 °C and a binding stoichiometry of 0.94, as measured by isothermal calorimetry. Reversible binding was confirmed by surface plasmon resonance direct binding experiments. The x-ray structure of AZ3976 in complex with latent PAI-1 was determined at 2.4 Å resolution. The inhibitor was bound in the flexible joint region with the entrance to the cavity located between α-helix D and ß-strand 2A. A set of surface plasmon resonance experiments revealed that AZ3976 inhibited PAI-1 by enhancing the latency transition of active PAI-1. Because AZ3976 only had measurable affinity for latent PAI-1, we propose that its mechanism of inhibition is based on binding to a small fraction in equilibrium with active PAI-1, a latent-like prelatent form, from which latent PAI-1 is then generated more rapidly. This mode of action, with induced accelerated latency transition of active PAI-1 may, together with supporting x-ray data, provide improved opportunities for small molecule drug design in the hunt for therapeutically useful PAI-1 inhibitors.


Subject(s)
Azetidines/pharmacology , Plasminogen Activator Inhibitor 1/chemistry , Pyrimidinones/pharmacology , Animals , Azetidines/chemistry , CHO Cells , Calorimetry , Cricetinae , Cricetulus , Humans , Models, Molecular , Protein Conformation , Pyrimidinones/chemistry , Rats , Surface Plasmon Resonance , Thermodynamics
19.
Circ Res ; 111(7): 920-9, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22982873

ABSTRACT

Anticoagulants are the cornerstone of therapy for conditions associated with arterial and venous thrombosis. Direct thrombin inhibitors (DTIs) are anticoagulants that bind to thrombin and block its enzymatic activity. The bivalent parenteral DTIs hirudin and bivalirudin were based on the observation that the salivary extracts of medicinal leeches prevented blood from clotting. Key events that facilitated the subsequent development of small molecule active site inhibitors, such as argatroban, were the observation that fibrinopeptide A had antithrombotic properties and determination of the crystal structure of thrombin. Hirudin and argatroban have found their niche for the treatment of patients with heparin-induced thrombocytopenia, whereas bivalirudin is approved as an alternative to heparin for patients undergoing percutaneous coronary intervention. The development of orally active direct thrombin inhibitors was challenging because of the need to convert water-soluble, poorly absorbable, active site inhibitors into fat-soluble prodrugs that were then transformed back to the active drug after intestinal absorption. Dabigatran etexilate was the first new oral anticoagulant to be approved for long-term anticoagulant treatment in 6 decades. This Review highlights the development of DTIs as a translational success story; an example in which the combination of scientific ingenuity, structure-based design, and rigorous clinical trials has created a new class of anticoagulants that has improved patient care.


Subject(s)
Anticoagulants/therapeutic use , Antithrombins/therapeutic use , Thrombosis/prevention & control , Translational Research, Biomedical/trends , Arginine/analogs & derivatives , Hirudin Therapy , Hirudins , Humans , Peptide Fragments/therapeutic use , Pipecolic Acids/therapeutic use , Recombinant Proteins/therapeutic use , Sulfonamides
SELECTION OF CITATIONS
SEARCH DETAIL
...