Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
1.
J Thromb Haemost ; 22(5): 1433-1446, 2024 May.
Article in English | MEDLINE | ID: mdl-38331196

ABSTRACT

BACKGROUND: Cardiovascular implantable devices, such as vascular stents, are critical for the treatment of cardiovascular diseases. However, their success is dependent on robust and often long-term antithrombotic therapies. Yet, the current standard-of-care therapies often pose significant bleeding risks to patients. Coagulation factor (F)XI and FXII have emerged as potentially safe and efficacious targets to safely reduce pathologic thrombin generation in medical devices. OBJECTIVES: To study the efficacy of monoclonal antibody-targeting FXII and FXI of the contact pathway in preventing vascular device-related thrombosis. METHODS: The effects of inhibition of FXII and FXI using function-blocking monoclonal antibodies were examined in a nonhuman primate model of nitinol stent-related thrombosis under arterial and venous flow conditions. RESULTS: We found that function-blocking antibodies of FXII and FXI reduced markers of stent-induced thrombosis in vitro and ex vivo. However, FXI inhibition resulted in more effective mitigation of thrombosis markers under varied flow conditions. CONCLUSION: This work provides further support for the translation of contact pathway of coagulation inhibitors for their adjunctive clinical use with cardiovascular devices.


Subject(s)
Alloys , Antibodies, Monoclonal , Factor XII , Factor XI , Stents , Thrombosis , Animals , Thrombosis/prevention & control , Thrombosis/blood , Factor XII/metabolism , Factor XII/antagonists & inhibitors , Factor XII/immunology , Factor XI/antagonists & inhibitors , Factor XI/immunology , Factor XI/metabolism , Antibodies, Monoclonal/pharmacology , Humans , Blood Coagulation/drug effects , Disease Models, Animal , Male , Regional Blood Flow , Fibrinolytic Agents/pharmacology
2.
Molecules ; 26(14)2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34299400

ABSTRACT

The goal of this study was to assess the pharmacological effects of black tea (Camellia sinensis var. assamica) water extract on human kinin-forming enzymes in vitro. Tea is a highly consumed beverage in the world. Factor XII (FXII, Hageman factor)-independent- and -dependent activation of prekallikrein to kallikrein leads to the liberation of bradykinin (BK) from high-molecular-weight kininogen (HK). The excessive BK production causes vascular endothelial and nonvascular smooth muscle cell permeability, leading to angioedema. The prevalence of angiotensin-converting enzyme inhibitor (ACEI)-induced angioedema appears to be through BK. Both histamine and BK are potent inflammatory mediators. However, the treatments for histamine-mediated angioedema are unsuitable for BK-mediated angioedema. We hypothesized that long-term consumption of tea would reduce bradykinin-dependent processes within the systemic and pulmonary vasculature, independent of the anti-inflammatory actions of polyphenols. A purified fraction of the black tea water extract inhibited both kallikrein and activated FXII. The black tea water extracts inhibited factor XII-induced cell migration and inhibited the production of kallikrein on the endothelial cell line. We compared the inhibitory effects of the black tea water extract and twenty-three well-known anti-inflammatory medicinal herbs, in inhibiting both kallikrein and FXII. Surprisingly, arjunglucoside II specifically inhibited the activated factor XII (FXIIa), but not the kallikrein and the activated factor XI. Taken together, the black tea water extract exerts its anti-inflammatory effects, in part, by inhibiting kallikrein and activated FXII, which are part of the plasma kallikrein-kinin system (KKS), and by decreasing BK production. The inhibition of kallikrein and activated FXII represents a unique polyphenol-independent anti-inflammatory mechanism of action for the black tea.


Subject(s)
Bradykinin/metabolism , Camellia/chemistry , Endothelium, Vascular/drug effects , Factor XII/antagonists & inhibitors , Kallikrein-Kinin System/drug effects , Plant Extracts/pharmacology , Pulmonary Artery/drug effects , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/metabolism , Humans , Pulmonary Artery/metabolism
3.
Expert Opin Ther Pat ; 31(12): 1155-1176, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34142629

ABSTRACT

Introduction: Blood coagulation factor XII (FXII) is an emerging and potentially safe drug target, which dysregulation is associated with thrombosis, hereditary angioedema, and (neuro)inflammation. At the same time, FXII-deficiency is practically asymptomatic. Industrial and academic institutions have developed a number of potential therapeutic agents targeting either FXII zymogen or its active form FXIIa for the treatment of thrombotic and inflammatory conditions associated with the activity of this enzyme.Areas covered: A short overview of the FXII(a) structure and function, underlining its suitability as a drug target, is given. The article reviews patents reported over the last three decades on FXII(a)-targeting therapeutic agents. These agents include small molecules, proteins, peptides, oligonucleotides, siRNAs, and monoclonal antibodies.Expert opinion: The performed analysis of patents revealed that many FXII(a) inhibitors are in the early preclinical stage, while several already showed efficacy in vivo animal models of thrombosis, sepsis, hereditary angioedema, and multiple sclerosis. Two anti-FXIIa agents namely tick protein Ir-CPI and monoclonal antibody CSL312 are currently in human clinical trials. The results of these trials and further studies of FXII(a) pathophysiological functions will encourage the development of new FXII(a) inhibitors.


Subject(s)
Factor XII/antagonists & inhibitors , Factor XIIa/antagonists & inhibitors , Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/pathology , Animals , Drug Development , Factor XII/metabolism , Factor XIIa/metabolism , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Patents as Topic , Thrombosis/drug therapy , Thrombosis/pathology
4.
Biomaterials ; 272: 120778, 2021 05.
Article in English | MEDLINE | ID: mdl-33812214

ABSTRACT

Blood contact with high surface area medical devices, such as dialysis and extracorporeal life support (ECLS), induces rapid surface coagulation. Systemic anticoagulation, such as heparin, is thus necessary to slow clot formation, but some patients suffer from bleeding complications. Both problems might be reduced by 1) replacing heparin anticoagulation with artificial surface inhibition of the protein adsorption that initiates coagulation and 2) selective inhibition of the intrinsic branch of the coagulation cascade. This approach was evaluated by comparing clot formation and bleeding times during short-term ECLS using zwitterionic polycarboxybetaine (PCB) surface coatings combined with either a potent, selective, bicyclic peptide inhibitor of activated Factor XII (FXII900) or standard heparin anticoagulation. Rabbits underwent venovenous ECLS with small sham oxygenators for 60 min using three means of anticoagulation (n = 4 ea): (1) PCB coating + FXII900 infusion, (2) PCB coating + heparin infusion with an activated clotting time of 220-300s, and (3) heparin infusion alone. Sham oxygenator blood clot weights in the PCB + FXII900 and PCB + heparin groups were 4% and 25% of that in the heparin group (p < 10-6 and p < 10-5), respectively. At the same time, the bleeding time remained normal in the PCB + FXII900 group (2.4 ± 0.2 min) but increased to 4.8 ± 0.5 and 5.1 ± 0.7 min in the PCB + heparin and heparin alone groups (p < 10-4 and 0.01). Sham oxygenator blood flow resistance was significantly lower in the PCB + FXII900 and PCB + heparin groups than in the heparin only group (p < 10-6 and 10-5). These results were confirmed by gross and scanning electron microscopy (SEM) images and fibrinopeptide A (FPA) concentrations. Thus, the combined use of PCB coating and FXII900 markedly reduced sham oxygenator coagulation and tissue bleeding times versus the clinical standard of heparin anticoagulation and is a promising anticoagulation method for clinical ECLS.


Subject(s)
Anticoagulants/pharmacology , Extracorporeal Membrane Oxygenation , Factor XII/antagonists & inhibitors , Animals , Betaine , Blood Coagulation , Heparin/pharmacology , Humans , Polymethacrylic Acids , Rabbits , Renal Dialysis
5.
Eur J Med Chem ; 208: 112753, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32883641

ABSTRACT

Coagulation factor XII (FXII), a S1A serine protease, was discovered more than fifty years ago. However, its in vivo functions and its three-dimensional structure started to be disclosed in the last decade. FXII was found at the crosstalk of several physiological pathways including the intrinsic coagulation pathway, the kallikrein-kinin system, and the immune response. The FXII inhibition emerges as a therapeutic strategy for the safe prevention of artificial surface-induced thrombosis and in patients suffering from hereditary angioedema. The anti-FXII antibody garadacimab discovered by phage-display library technology is actually under phase II clinical evaluation for the prophylactic treatment of hereditary angioedema. The implication of FXII in neuro-inflammatory and neurodegenerative disorders is also an emerging research field. The FXII or FXIIa inhibitors currently under development include peptides, proteins, antibodies, RNA-based technologies, and, to a lesser extent, small-molecular weight inhibitors. Most of them are proteins, mainly isolated from hematophagous arthropods and plants. The discovery and development of these FXII inhibitors and their potential indications are discussed in the review.


Subject(s)
Anticoagulants/pharmacology , Factor XII/antagonists & inhibitors , Factor XIIa/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Animals , Anticoagulants/chemistry , Drug Discovery , Factor XII/chemistry , Factor XIIa/chemistry , Humans , Serine Proteinase Inhibitors/chemistry
6.
Nat Commun ; 11(1): 3890, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753636

ABSTRACT

Inhibiting thrombosis without generating bleeding risks is a major challenge in medicine. A promising solution may be the inhibition of coagulation factor XII (FXII), because its knock-out or inhibition in animals reduced thrombosis without causing abnormal bleeding. Herein, we have engineered a macrocyclic peptide inhibitor of activated FXII (FXIIa) with sub-nanomolar activity (Ki = 370 ± 40 pM) and a high stability (t1/2 > 5 days in plasma), allowing for the preclinical evaluation of a first synthetic FXIIa inhibitor. This 1899 Da molecule, termed FXII900, efficiently blocks FXIIa in mice, rabbits, and pigs. We found that it reduces ferric-chloride-induced experimental thrombosis in mice and suppresses blood coagulation in an extracorporeal membrane oxygenation (ECMO) setting in rabbits, all without increasing the bleeding risk. This shows that FXIIa activity is controllable in vivo with a synthetic inhibitor, and that the inhibitor FXII900 is a promising candidate for safe thromboprotection in acute medical conditions.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Factor XIIa/antagonists & inhibitors , Peptides, Cyclic/drug effects , Thrombosis/prevention & control , Animals , Chlorides/adverse effects , Cloning, Molecular , Disease Models, Animal , Drug Discovery , Extracorporeal Membrane Oxygenation/methods , Factor XII/antagonists & inhibitors , Female , Ferric Compounds/adverse effects , Humans , Lung , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rabbits , Recombinant Proteins/pharmacology , Swine
7.
Clin Sci (Lond) ; 134(9): 1049-1061, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32309850

ABSTRACT

Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. Chronic inflammation and excessive matrix remodelling are considered important in AAA pathogenesis. Kinins are bioactive peptides important in regulating inflammation. Stimulation of the kinin B2 receptor has been previously reported to promote AAA development and rupture in a mouse model. The endogenous B2 receptor agonist, bradykinin, is generated from the kallikrein-kinin system following activation of plasma kallikrein by Factor XII (FXII). In the current study whole-body FXII deletion, or neutralisation of activated FXII (FXIIa), inhibited expansion of the suprarenal aorta (SRA) of apolipoprotein E-deficient mice in response to angiotensin II (AngII) infusion. FXII deficiency or FXIIa neutralisation led to decreased aortic tumor necrosis factor-α-converting enzyme (TACE/a disintegrin and metalloproteinase-17 (aka tumor necrosis factor-α-converting enzyme) (ADAM-17)) activity, plasma kallikrein concentration, and epithelial growth factor receptor (EGFR) phosphorylation compared with controls. FXII deficiency or neutralisation also reduced Akt1 and Erk1/2 phosphorylation and decreased expression and levels of active matrix metalloproteinase (Mmp)-2 and Mmp-9. The findings suggest that FXII, kallikrein, ADAM-17, and EGFR are important molecular mediators by which AngII induces aneurysm in apolipoprotein E-deficient mice. This could be a novel pathway to target in the design of drugs to limit AAA progression.


Subject(s)
Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Apolipoproteins E/deficiency , Factor XII/antagonists & inhibitors , ADAM17 Protein/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Aortic Aneurysm, Abdominal/metabolism , Disease Models, Animal , Factor XII/metabolism , Mice
8.
Nucleic Acid Ther ; 29(4): 218-223, 2019 08.
Article in English | MEDLINE | ID: mdl-31063435

ABSTRACT

Recently, our group reported that a small interfering RNA (siRNA) targeting coagulation factor XII (siF12) leads to an unexpected prothrombotic response in a mouse model where venous thrombosis follows inhibition of endogenous anticoagulants. In this study, we aimed to clarify this unexpected response by evaluating the effects of this siF12 (here, siF12-A) on plasma coagulation through thrombin generation (TG). Besides a routine negative control siRNA (siNEG), we included extra siRNA controls: one siRNA similar to siF12-A except for positions 9-11 of the siRNA that are replaced with its complementary base pairs (siF12-AC9/11), and a second siRNA against F12 (siF12-B). Three days after injection, a significant increase in TG peak height was observed solely for animals injected with siF12-A and siF12-AC9/11, which is considered prothrombotic. As this change in coagulation was unrelated to FXII we conclude that it was off-target. For siRNA studies we now recommend to include mismatch siRNA controls, such as the C9/11 mismatch control used in this study, and to consider plasma coagulation in off-target analysis.


Subject(s)
Anticoagulants/pharmacology , Factor XII/genetics , RNA, Small Interfering/genetics , Venous Thrombosis/drug therapy , Animals , Anticoagulants/chemistry , Blood Coagulation/drug effects , DNA Mismatch Repair/genetics , Disease Models, Animal , Factor XII/antagonists & inhibitors , Humans , Mice , RNA, Small Interfering/pharmacology , Thrombin/genetics , Venous Thrombosis/genetics , Venous Thrombosis/prevention & control
9.
Circ Res ; 124(3): 426-436, 2019 02.
Article in English | MEDLINE | ID: mdl-30702990

ABSTRACT

Recent advances in our understanding of the contribution of thrombin generation to arterial thrombosis and the role of platelets in venous thrombosis have prompted new treatment paradigms. Nonetheless, bleeding remains the major side effect of such treatments spurring the quest for new antithrombotic regimens with better benefit-risk profiles and for safer anticoagulants for existing and new indications. The aims of this article are to review the results of recent trials aimed at enhancing the benefit-risk profile of antithrombotic therapy and explain how these findings are changing our approach to the management of arterial and venous thrombosis. Focusing on these 2 aspects of thrombosis management, this article discusses 4 advances: (1) the observation that in some indications, lowering the dose of some direct oral anticoagulants reduces the risk of bleeding without compromising efficacy, (2) the recognition that aspirin is not only effective for secondary prevention of atherothrombosis but also for prevention of venous thromboembolism, (3) the finding that dual pathway inhibition with the combination of low-dose rivaroxaban to attenuate thrombin generation plus aspirin to reduce thromboxane A2-mediated platelet activation is superior to aspirin or rivaroxaban alone for prevention of atherothrombosis in patients with coronary or peripheral artery disease, and (4) the development of inhibitors of factor XI or XII as potentially safer anticoagulants.


Subject(s)
Aspirin/therapeutic use , Factor Xa Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/therapeutic use , Rivaroxaban/administration & dosage , Thrombosis/drug therapy , Administration, Oral , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Antithrombins/administration & dosage , Antithrombins/adverse effects , Atrial Fibrillation/complications , Clinical Trials as Topic , Coronary Disease/complications , Drug Therapy, Combination , Factor XI/antagonists & inhibitors , Factor XII/antagonists & inhibitors , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Humans , Peripheral Arterial Disease/complications , Plaque, Atherosclerotic/complications , Platelet Aggregation , Primary Prevention , Risk Assessment , Secondary Prevention , Thrombin/metabolism , Thrombosis/etiology , Thrombosis/prevention & control , Venous Thrombosis/prevention & control
10.
Rev Cardiovasc Med ; 20(4): 245-253, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31912715

ABSTRACT

Atherosclerosis is a leading cause of cardiovascular and neurological ischemic events. Plaque rupture leads to the exposure of highly thrombogenic material with blood and results in the activation of the coagulation cascade, thrombus formation, and embolic events. Although antiplatelets and anticoagulants are used to prevent thromboembolic episodes, bleeding episodes remain the major adverse effect. Decreased ischemic events have been reported while comparing oral rivaroxaban and apixaban with aspirin to improve the therapeutic outcome in several clinical trials, including Anti-Xa Therapy to Lower Cardiovascular Events in Addition to Standard Therapy in Subjects with Acute Coronary Syndrome-Thrombolysis in Myocardial Infarction 51, Apixaban for Prevention of Acute Ischemic and Safety Events, and GEMINI-ACS-1 phase II clinical trials. However, there were bleeding episodes. Thus, there is an unmet need for better therapeutic strategies. Therefore, the current focus is to target Factors IX, XI, and XII to develop safer and efficient strategies. In this article, we critically reviewed and discussed the limitations of current therapies and the potential of targeting Factors IX, XI, and XII for anticoagulant therapy in atherothrombosis.


Subject(s)
Anticoagulants/therapeutic use , Atherosclerosis/drug therapy , Blood Coagulation/drug effects , Factor IX/antagonists & inhibitors , Factor XII/antagonists & inhibitors , Factor XI/antagonists & inhibitors , Thrombosis/drug therapy , Animals , Anticoagulants/adverse effects , Atherosclerosis/blood , Atherosclerosis/diagnosis , Factor IX/metabolism , Factor XI/metabolism , Factor XII/metabolism , Hemorrhage/chemically induced , Humans , Molecular Targeted Therapy , Plaque, Atherosclerotic , Risk Factors , Thrombosis/blood , Thrombosis/diagnosis , Treatment Outcome
11.
mBio ; 9(6)2018 12 18.
Article in English | MEDLINE | ID: mdl-30563903

ABSTRACT

Antibiotic-resistant Acinetobacter baumannii is increasingly recognized as a cause of difficult-to-treat nosocomial infections, including pneumonia, wound infections, and bacteremia. Previous studies have demonstrated that the metalloprotease CpaA contributes to virulence and prolongs clotting time when added to human plasma as measured by the activated partial thromboplastin time (aPTT) assay. Here, we show that CpaA interferes with the intrinsic coagulation pathway, also called the contact activation system, in human as well as murine plasma, but has no discernible effect on the extrinsic pathway. By utilizing a modified aPTT assay, we demonstrate that coagulation factor XII (fXII) is a target of CpaA. In addition, we map the cleavage by CpaA to two positions, 279-280 and 308-309, within the highly glycosylated proline-rich region of human fXII, and show that cleavage at the 308-309 site is responsible for inactivation of fXII. At both sites, cleavage occurs between proline and an O-linked glycosylated threonine, and deglycosylation of fXII prevents cleavage by CpaA. Consistent with this, mutant fXII (fXII-Thr309Lys) from patients with hereditary angioedema type III (HAEIII) is protected from CpaA inactivation. This raises the possibility that individuals with HAEIII who harbor this mutation may be partially protected from A. baumannii infection if CpaA contributes to human disease. By inactivating fXII, CpaA may attenuate important antimicrobial defense mechanisms such as intravascular thrombus formation, thus allowing A. baumannii to disseminate.IMPORTANCE Ventilator-associated pneumonia and catheter-related bacteremia are the most common and severe infections caused by Acinetobacter baumannii Besides the capsule, lipopolysaccharides, and the outer membrane porin OmpA, little is known about the contribution of secreted proteins to A. baumannii survival in vivo Here we focus on CpaA, a potentially recently acquired virulence factor that inhibits blood coagulation in vitro We identify coagulation factor XII as a target of CpaA, map the cleavage sites, and show that glycosylation is a prerequisite for CpaA-mediated inactivation of factor XII. We propose adding CpaA to a small, but growing list of bacterial proteases that are specific for highly glycosylated components of the host defense system.


Subject(s)
Acinetobacter baumannii/enzymology , Bacterial Proteins/metabolism , Factor XII/antagonists & inhibitors , Metalloproteases/metabolism , Acinetobacter baumannii/pathogenicity , Angioedemas, Hereditary/blood , Animals , Blood Coagulation , Factor XII/metabolism , Female , Humans , Mice , Mice, Inbred CBA , Middle Aged , Mutation , Partial Thromboplastin Time , Prothrombin Time , Virulence Factors
12.
Blood Adv ; 2(5): 549-558, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29519898

ABSTRACT

Activated factor XIIa (FXIIa) is a serine protease that has received a great deal of interest in recent years as a potential target for the development of new antithrombotics. Despite the strong interest in obtaining structural information, only the structure of the FXIIa catalytic domain in its zymogen conformation is available. In this work, reproducible experimental conditions found for the crystallization of human plasma ß-FXIIa and crystal growth optimization have led to determination of the first structure of the active form of the enzyme. Two crystal structures of human plasma ß-FXIIa complexed with small molecule inhibitors are presented herein. The first is the noncovalent inhibitor benzamidine. The second is an aminoisoquinoline containing a boronic acid-reactive group that targets the catalytic serine. Both benzamidine and the aminoisoquinoline bind in a canonical fashion typical of synthetic serine protease inhibitors, and the protease domain adopts a typical chymotrypsin-like serine protease active conformation. This novel structural data explains the basis of the FXII activation, provides insights into the enzymatic properties of ß-FXIIa, and is a great aid toward the further design of protease inhibitors for human FXIIa.


Subject(s)
Factor XII/chemistry , Benzamidines/chemistry , Boronic Acids/chemistry , Crystallization/methods , Crystallography, X-Ray , Databases, Protein , Factor XII/antagonists & inhibitors , Humans , Molecular Structure , Protein Binding , Software
13.
Semin Thromb Hemost ; 44(1): 60-69, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28898903

ABSTRACT

Exposure of blood to a variety of artificial surface induces contact activation, a process that contributes to the host innate response to foreign substances. On the foreign surface, the contact factors, factor XII (FXII), and plasma prekallikrein undergo reciprocal conversion to their fully active protease forms (FXIIa and α-kallikrein, respectively) by a process supported by the cofactor high-molecular-weight kininogen. Contact activation can trigger blood coagulation by conversion of factor XI (FXI) to the protease FXIa. There is interest in developing therapeutic inhibitors to FXIa and FXIIa because these activated factors can contribute to thrombosis in certain situations. Drugs targeting these proteases may be particularly effective in thrombosis triggered by exposure of blood to the surfaces of implantable medical devices. Here, we review clinical data supporting roles for FXII and FXI in thrombosis induced by medical devices, and preclinical data suggesting that therapeutic targeting of these proteins may limit surface-induced thrombosis.


Subject(s)
Factor XII/antagonists & inhibitors , Factor XI/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Disease Models, Animal , Factor XI/metabolism , Factor XII/metabolism , Humans , Mice
14.
Immunol Allergy Clin North Am ; 37(3): 585-595, 2017 08.
Article in English | MEDLINE | ID: mdl-28687111

ABSTRACT

Remarkable progress has been made in the treatment of bradykinin-mediated angioedema with the advent of multiple new therapies. Patients now have effective medications available for prophylaxis and treatment of acute attacks. However, hereditary angioedema is a burdensome disease that can lead to debilitating and dangerous angioedema episodes associated with significant costs for individuals and society. The burden of treatment must be addressed regarding medication administration difficulties, treatment complications, and adverse side effects. New therapies are being investigated and may offer solutions to these challenges. This article reviews the emerging therapeutic options for the treatment of HAE.


Subject(s)
Angioedemas, Hereditary/therapy , Angioedemas, Hereditary/diagnosis , Angioedemas, Hereditary/etiology , Angioedemas, Hereditary/metabolism , Animals , Bradykinin/metabolism , Clinical Trials as Topic , Combined Modality Therapy , Complement C1 Inhibitor Protein/therapeutic use , Drug Discovery , Drug Evaluation, Preclinical , Factor XII/antagonists & inhibitors , Factor XII/metabolism , Histamine/metabolism , Humans , Signal Transduction , Treatment Outcome
15.
Nat Commun ; 8: 16092, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28714475

ABSTRACT

The rapid renal clearance of peptides in vivo limits this attractive platform for the treatment of a broad range of diseases that require prolonged drug half-lives. An intriguing approach for extending peptide circulation times works through a 'piggy-back' strategy in which peptides bind via a ligand to the long-lived serum protein albumin. In accordance with this strategy, we developed an easily synthesized albumin-binding ligand based on a peptide-fatty acid chimera that has a high affinity for human albumin (Kd=39 nM). This ligand prolongs the elimination half-life of cyclic peptides in rats 25-fold to over seven hours. Conjugation to a peptide factor XII inhibitor developed for anti-thrombotic therapy extends the half-life from 13 minutes to over five hours, inhibiting coagulation for eight hours in rabbits. This high-affinity albumin ligand could potentially extend the half-life of peptides in human to several days, substantially broadening the application range of peptides as therapeutics.


Subject(s)
Drug Carriers/chemistry , Fatty Acids/chemistry , Peptides/administration & dosage , Serum Albumin, Human/chemistry , Acylation , Animals , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Factor XII/antagonists & inhibitors , Fatty Acids/metabolism , Fatty Acids/pharmacokinetics , Half-Life , Humans , Ligands , Metabolic Clearance Rate , Peptides/metabolism , Peptides/pharmacokinetics , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacokinetics , Plasma Kallikrein/antagonists & inhibitors , Protein Binding , Rabbits , Rats , Serum Albumin, Human/metabolism , Serum Albumin, Human/pharmacokinetics , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
16.
Thromb Haemost ; 117(7): 1283-1288, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28594426

ABSTRACT

Thrombosis is a leading cause of death and disability worldwide, and anticoagulants are the mainstay of its prevention and treatment. Starting with unfractionated heparin (UFH) and vitamin K antagonists (VKAs) such as warfarin, the choices of anticoagulants have exploded in the past 20 years. With over 90 % subcutaneous bioavailability, no need for coagulation monitoring and dose adjustment, and a lower risk of heparin-induced thrombocytopenia, low-molecular-weight heparin and fondaparinux have replaced UFH for prevention and initial treatment of venous thromboembolism and for secondary prevention in cancer patients. In patients undergoing percutaneous interventions, bivalirudin is often used instead of UFH. Oral anticoagulation therapy has advanced with the introduction of the non-vitamin K antagonist oral anticoagulants (NOACs), which include dabigatran, rivaroxaban, apixaban and edoxaban. With efficacy at least equal to that of VKAs but with greater safety and convenience, the NOACs are now replacing VKAs for many indications. This paper a) highlights these advances, b) outlines how specific reversal agents for the NOACs will enhance their safety, c) reviews some of the ongoing trials with the NOACs, and d) describes the inhibitors of factor XII and XI that are under investigation as anticoagulants.


Subject(s)
Anticoagulants/therapeutic use , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Antidotes/therapeutic use , Coronary Artery Disease/drug therapy , Drug Discovery , Factor XI/antagonists & inhibitors , Factor XII/antagonists & inhibitors , Heart Failure/drug therapy , Heparin/adverse effects , Humans , Peripheral Arterial Disease/drug therapy , Stroke/drug therapy , Thrombosis/blood , Thrombosis/drug therapy , Thrombosis/prevention & control , Venous Thromboembolism/drug therapy , Vitamin K/antagonists & inhibitors , Warfarin/adverse effects
17.
J Med Chem ; 60(3): 1151-1158, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28045547

ABSTRACT

Factor XII (FXII) is a plasma protease that has emerged in recent years as a potential target to treat or prevent pathological thrombosis, to inhibit contact activation in extracorporeal circulation, and to treat the swelling disorder hereditary angioedema. While several protein based inhibitors with high affinity for activated FXII (FXIIa) were developed, the generation of small molecule inhibitors has been challenging. In this work, we have generated a potent and selective FXIIa inhibitor by optimizing a peptide macrocycle that was recently evolved by phage display (Ki = 0.84 ± 0.03 nM). A fluorine atom introduced in the para-position of phenylalanine enhanced the binding affinity as much as 10-fold. Furthermore, we improved the proteolytic stability by substituting the N-terminal arginine by norarginine. The resulting inhibitor combines high inhibitory affinity and selectivity with a good stability in plasma (Ki = 1.63 ± 0.18 nM, >27 000-fold selectivity, t1/2(plasma) =16 ± 4 h). The inhibitor efficiently blocked activation of the intrinsic coagulation pathway in human blood ex vivo.


Subject(s)
Factor XII/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Peptides/chemistry , Humans , Macrocyclic Compounds/chemistry , Proteolysis
18.
Ann Biomed Eng ; 45(5): 1328-1340, 2017 05.
Article in English | MEDLINE | ID: mdl-27933406

ABSTRACT

In the contact activation pathway of the coagulation, zymogen factor XII (FXII) is converted to FXIIa, which triggers activation of FXI leading to the activation of FIX and subsequent thrombin generation and fibrin formation. Feedback activation of FXI by thrombin has been shown to promote thrombin generation in a FXII-independent manner and FXIIa can bypass FXI to directly activate FX and prothrombin in the presence of highly negatively charged molecules, such as long-chain polyphosphates (LC polyP). We sought to determine whether activation of FXII or FXI differentially regulate the physical biology of fibrin formation. Fibrin formation was initiated with tissue factor, ellagic acid (EA), or LC polyP in the presence of inhibitors of FXI and FXII. Our data demonstrated that inhibition of FXI decreased the rate of fibrin formation and fiber network density, and increased the fibrin network strength and rate of fibrinolysis when gelation was initiated via the contact activation pathway with EA. FXII inhibition decreased the fibrin formation and fibrin density, and increased the fibrinolysis rate only when fibrin formation was initiated via the contact activation pathway with LC polyP. Overall, we demonstrate that inhibition of FXI and FXII distinctly alter the biophysical properties of fibrin.


Subject(s)
Blood Coagulation Factor Inhibitors/chemistry , Factor XII , Factor XI , Fibrin/chemistry , Fibrinolysis , Factor XI/antagonists & inhibitors , Factor XI/chemistry , Factor XII/antagonists & inhibitors , Factor XII/chemistry , Humans , Polyphosphates/chemistry
19.
Arterioscler Thromb Vasc Biol ; 37(1): 13-20, 2017 01.
Article in English | MEDLINE | ID: mdl-27834692

ABSTRACT

Coagulation factor XII (FXII, Hageman factor) is a plasma protease that in its active form (FXIIa) initiates the procoagulant and proinflammatory contact system. This name arises from FXII's unique mechanism of activation that is induced by binding (contact) to negatively charged surfaces. Various substances have the capacity to trigger FXII contact-activation in vivo including mast cell-derived heparin, misfolded protein aggregates, collagen, nucleic acids, and polyphosphate. FXII deficiency is not associated with bleeding, and for decades, the factor was considered to be dispensable for coagulation in vivo. However, despite the fact that humans and animals with deficiency in FXII have a normal hemostatic capacity, animal models revealed a critical role of FXIIa-driven coagulation in thromboembolic diseases. In addition to its role in thrombosis, FXIIa contributes to inflammation through the activation of the inflammatory bradykinin-producing kallikrein-kinin system. Pharmacological inhibition of FXII/FXIIa interferes with thrombosis and inflammation in animal models. Thus, targeting the FXIIa-driven contact system seems to be a promising and safe therapeutic anticoagulation treatment strategy, with additional anti-inflammatory effects. Here, we discuss novel functions of FXIIa in cardiovascular thrombotic and inflammatory disorders.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Blood Coagulation/drug effects , Factor XII/antagonists & inhibitors , Fibrinolytic Agents/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Inflammation/drug therapy , Kallikrein-Kinin System/drug effects , Thromboembolism/drug therapy , Animals , Disease Models, Animal , Factor XII/genetics , Factor XII/metabolism , Factor XIIa/antagonists & inhibitors , Factor XIIa/metabolism , Genetic Predisposition to Disease , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/blood , Mice, Knockout , Molecular Targeted Therapy , Thromboembolism/blood , Thromboembolism/genetics , Thromboembolism/immunology
20.
Blood ; 129(2): 147-154, 2017 01 12.
Article in English | MEDLINE | ID: mdl-27780803

ABSTRACT

Despite the introduction of direct oral anticoagulants (DOACs), the search for more effective and safer antithrombotic strategies continues. Better understanding of the pathogenesis of thrombosis has fostered 2 new approaches to achieving this goal. First, evidence that thrombin may be as important as platelets to thrombosis at sites of arterial injury and that platelets contribute to venous thrombosis has prompted trials comparing anticoagulants with aspirin for secondary prevention in arterial thrombosis and aspirin with anticoagulants for primary and secondary prevention of venous thrombosis. These studies will help identify novel treatment strategies. Second, emerging data that naturally occurring polyphosphates activate the contact system and that this system is critical for thrombus stabilization and growth have identified factor XII (FXII) and FXI as targets for new anticoagulants that may be even safer than the DOACs. Studies are needed to determine whether FXI or FXII is the better target and to compare the efficacy and safety of these new strategies with current standards of care for the prevention or treatment of thrombosis. Focusing on these advances, this article outlines how treatment strategies for thrombosis are evolving and describes the rationale and approaches to targeting FXII and FXI. These emerging anticoagulant strategies should address unmet needs and reduce the systemic underuse of anticoagulation because of the fear of bleeding.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/physiology , Thrombosis/metabolism , Animals , Blood Coagulation/drug effects , Factor XI/antagonists & inhibitors , Factor XII/antagonists & inhibitors , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...