Comparison Of Plasma Kallikrein Inhibition By The Endogenous C1-Inhibitor Versus DX-2930, a Monoclonal Antibody Inhibitor

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1066-1066 ◽  
Author(s):  
Daniel J Sexton ◽  
Jon A Kenniston ◽  
Ryan R Faucette ◽  
Andrew E Nixon ◽  
Chris TenHoor ◽  
...  

Abstract Dysregulated plasma kallikrein proteolytic activity leads to edematous attacks in hereditary angioedema (HAE) and has been associated with inflammation and thrombosis. Plasma kallikrein (pKal) is a serine protease that circulates as prekallikrein, a zymogen, which, together with factor XII (FXII) and high molecular weight kininogen (HMWK), constitutes the contact system. Activation of the contact system following assembly of FXII, HMWK, and prekallikrein on a negatively charged surface promotes inflammation via the generation of bradykinin and triggers intrinsic pathway coagulation via formation of activated coagulation factor XIa. Normal hemostasis appears not to be mediated by the contact system as individuals deficient in contact system proteins are not at risk for bleeding. However, the contact system has been shown to be pathologically activated by agents that include misfolded proteins, platelet polyphosphate, and implanted devices. Therefore, pharmacologic modulation of the contact system may attenuate thrombosis and inflammation without disrupting normal hemostasis. C1 inhibitor (C1-INH) is a serpin and a key endogenous, protein-based, inhibitor of pKal activity. HAE is caused by autosomal dominant mutations in the C1-INH gene resulting in functional protein levels that are approximately 30% or less than normal (16-33 mg/dL or 1.6-3.3 μM). Prekallikrein is present in plasma at a concentration of approximately 500 nM and it has been estimated that only 30-110 nM is converted to active pKal during an HAE attack. This study investigates the requirement for super-stoichiometric amounts of endogenous C1-INH to adequately regulate pKal activity. In vitro enzyme inhibition kinetics experiments with purified proteins show that the need for high concentrations of C1-INH is likely due to its relatively slow association rate constant (1.7 x 104 M-1s-1). In contrast, DX-2930, a human monoclonal antibody inhibitor of pKal being developed for prophylactic treatment of HAE, potently inhibited pKal (Ki = 125 pM) with a faster association rate constant (3.4 x 106 M-1s-1). Contact activation was observed in human plasma activated by the addition of ellagic acid and monitored using a pKal-selective synthetic peptide substrate. Consistent with the data obtained using purified proteins, the apparent IC50 observed upon adding exogenous C1-INH to normal human plasma was approximately 100-fold higher than that of DX-2930. Using a Western blot assay to monitor endogenous HMWK cleavage in activated plasma we similarly observed that stoichiometric additions of DX-2930 were sufficient to prevent HMWK proteolysis by active pKal; whereas significantly higher concentrations of C1-INH (e.g. 1 µM) were required to block HMWK proteolysis. Active pKal can bind endothelial cells via interactions between the non-catalytic domain of pKal with HMWK, which binds receptors (urokinase receptor, cytokeratin 1, and the globular C1q receptor) present on endothelial cells. Cell bound pKal is likely to be a physiologically relevant form of the enzyme and may provide an explanation for attack localization in HAE. In this study, active pKal was assembled in vitro on cultured human umbilical vein endothelial cells (HUVEC) and binding of a range of concentrations of either biotinylated C1-INH or biotinylated DX-2930 was observed using streptavidin-horseradish peroxidase as detection. The data obtained demonstrates that C1-INH bound cell-associated pKal with > 200-fold less potency than DX-2930. Regarding protease inhibition specificity, while DX-2930 did not inhibit any of 20 tested serine proteases at a concentration of 1 µM, C1-INH is known to inhibit multiple serine proteases. This study demonstrates that effective regulation of pKal activity requires high concentrations of C1-INH (≥ 1 µM), which are necessary to drive the kinetics of this second order, irreversible interaction. These high inhibitory concentrations of C1-INH match the normal range and provide a potential kinetic mechanism for why HAE attacks can occur at C1-INH levels that exceed expected levels of activated pKal. Furthermore, the broad specificity of C1-INH towards other proteases that could be activated during disease could sufficiently deplete C1-INH levels and thereby lead to dysregulated pKal activity. Disclosures: Sexton: Dyax Corp: Employment. Kenniston:Dyax Corp: Employment. Faucette:Dyax Corp: Employment. Nixon:Dyax Corp: Employment. TenHoor:Dyax Corp: Employment. Chyung:Dyax Corp: Employment. Adelman:Dyax Corp: Employment.

2010 ◽  
pp. 711-719 ◽  
Author(s):  
H Maxová ◽  
L Bačáková ◽  
V Lisá ◽  
J Novotná ◽  
H Tomášová ◽  
...  

Matrix metalloproteinases (MMPs) is a family of proteolytic enzymes involved in remodeling of extracellular matrix. Although proteolytic enzymes are produced by many cell types, mast cells seem to be more important than other types in remodeling of pulmonary arteries during hypoxia. Therefore, we tested in vitro production of MMPs and serine proteases in four cell types (mast cells, fibroblasts, vascular smooth muscle cells and endothelial cells) cultivated for 48 h under normoxic or hypoxic (3 % O2) conditions. MMP-13 was visualized by immunohistochemistry, MMP-2 and MMP-9 were detected by zymography in cell lysates. Enzymatic activities (MMPs, tryptase and chymase) were estimated in the cultivation media. Hypoxia had a minimal effect on total MMP activity in the cultivation media of all types of cells, but immunofluorescence revealed higher intensity of MMP-13 in the cells exposed to hypoxia except of fibroblasts. Tryptase activity was three times higher and chymase activity twice higher in mast cells cultivated in hypoxia than in those cultured in normoxia. Among all cell types studied here, mast cells are the most abundant source of proteolytic enzymes under normoxic and hypoxic conditions. Moreover, in these cells hypoxia increases the production of both specific serine proteases tryptase and chymase, which can act as MMPs activators.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1067-1067
Author(s):  
Jon A Kenniston ◽  
Daniel J Sexton ◽  
Diana Martik ◽  
Ryan R Faucette ◽  
Malini Viswanathan ◽  
...  

Abstract The plasma-kallikrein kinin (contact) system contributes to the physiological and pathophysiological reactions of vascular biology. Activation of this pathway causes the release of the potent nonapeptide vasodilator bradykinin following proteolytic cleavage of high-molecular weight kininogen (HMWK) by the serine protease plasma kallikrein (pKal). Normal vascular homeostasis requires regulation of pKal activity by interactions with the C1-inhibitor (C1-INH). This is most apparent in individuals with hereditary angioedema (HAE), a disease characterized by a genetic deficiency in C1-INH that results in persistent pKal activity and consequent bradykinin release. These events can ultimately manifest as unpredictable and potentially fatal attacks of subcutaneous and mucosal edema. Inhibition of pKal proteolytic activity has proven to be a viable therapeutic option for HAE, however there remains an unmet medical need for a long-lasting prophylactic treatment for this disease. Given the potential for target specificity and long serum half-life with antibody therapeutics, we used phage display to select a fully human antibody inhibitor (DX-2930) specific for pKal. In vitro enzyme inhibition and affinity assays demonstrate that DX-2930 is a potent antibody inhibitor of pKal (Ki = 125 pM) that binds the active form of pKal, but not the proenzyme form (prekallikrein) or any other serine protease tested. DX-2930 binding consequently prohibits pKal from cleaving bradykinin out of HMWK and thereby prevents the activation of the bradykinin receptor B2. A 2.1Å resolution X-ray crystallographic structure of pKal complexed to a DX-2930 Fab construct supports these findings, demonstrating that the pKal proteolytic active site is intimately bound - and thereby occluded - by the Fab. This structural analysis provides both a rationale for the potency and specificity of DX-2930, and demonstrates the utility of using antibodies to specifically target an antigen among a family of related proteins (e.g. serine proteases). To further address the functional activity of DX-2930, we demonstrate that subcutaneous dosing of DX-2930 effectively reduces carrageenan-induced paw edema in vivo in rats when injected 24 hours prior to challenge. Combined with our finding that DX-2930 has a prolonged serum residence time in cynomolgus monkeys (t1/2 = 301 hours, SC), the data presented here demonstrates the potential of DX-2930 for the prophylactic inhibition of pKal-mediated diseases, such as HAE. Disclosures: Kenniston: Dyax Corp: Employment. Sexton:Dyax Corp: Employment. Martik:Dyax Corp: Employment, former employee of Dyax Corp Other. Faucette:Dyax Corp: Employment. Viswanathan:Dyax Corp: Employment. Kastrapeli:Dyax: Employment. Kopacz:Dyax Corp: Employment. Conley:Dyax Corp: Employment. Lindberg:Dyax Corp: Employment. Cosic:Dyax Corp: Employment. Comeau:Dyax Corp: Employment. Mason:Dyax Corp: Employment. DiLeo:Dyax Corp: Employment. Chen:Dyax Corp: Employment. Ladner:Dyax Corp: Employment. Edwards:Emerald Biostructures: Employment. TenHoor:Dyax Corp: Employment. Nixon:Dyax Corp: Employment. Adelman:Dyax Corp: Employment.


Author(s):  
Gaurav Girdhar ◽  
Sulan Xu ◽  
Jolyon Jesty ◽  
Danny Bluestein

Cigarette smoking is a risk factor for development of cardiovascular (CV) disease [1], with increased platelet activation due to cigarette smoke involving a major part of this risk.[2] We have shown that this smoke-induced platelet activation is largely due to the non-nicotine smoke components and their effects can be effectively modulated in the presence of nicotine.[3] However, the effects of nicotine and non-nicotine cigarette smoke components need to be confirmed more physiologically in the presence of endothelial cells (ECs). Prior in-vitro studies have shown that high concentrations of cigarette smoke extract (CSE) increase adhesion molecule expression on ECs.[4] These studies however preclude the involvement of physiological shear stresses and are performed on ECs alone. To overcome these limitations and investigate ECs-platelets together in one system under shear stress, we use a hemodynamic shear device (HSD) that combines features of the cone and plate, and the annular Couette viscometer (to facilitate platelet sampling). We test the following hypotheses — (1) smoke-activated platelets and the nicotine-free extract would confer a synergistic E-selectin expression on ECs, and (2) in contrast to conventional cigarette extracts, nicotine-free smoke extracts would increase platelet activation more significantly, and that this effect may be independent of the presence of ECs.


2004 ◽  
Vol 92 (12) ◽  
pp. 1277-1283 ◽  
Author(s):  
Sriram Ravindran ◽  
Thomas Grys ◽  
Rodney A.Welch ◽  
Marc Schapira ◽  
Philip Patston

SummaryActivation of plasma prekallikein and generation of bradykinin are responsible for the angioedema attacks observed with C1inhibitor deficiency. Heterozygous individuals with <50% levels of active C1-inhibitor are susceptible to angioedema attacks indicating a critical need for C1-inhibitor to be present at maximum levels to prevent unwanted prekallikrein activation. Studies with purified proteins do not adequately explain this observation. Therefore to investigate why reduction of C1inhibitor to levels seen in angioedema patients results in excessive kallikrein generation we examined the effect of endothelial cells on the inhibition of kallikrein by C1-inhibitor. Surprisingly, it was found that a C1-inhibitor concentration of greater than 1 µM was needed to inhibit 3 nM kallikrein. We propose that this apparent protection from inhibition was mediated by kallikrein binding to the cells via the heavy chain in a high molecular weight kininogen and zinc independent manner. Protection of kallikrein from inhibition was not observed when C1-inhibitor truncated in the amino-terminal domain by the StcE metalloproteinase was used, which suggests a novel function for this unique domain. The requirement for high concentrations of C1-inhibitor to fully inhibit kallikrein is consistent with the fact that reduced levels of C1-inhibitor result in the kallikrein activation seen in angioedema.


Blood ◽  
2003 ◽  
Vol 102 (9) ◽  
pp. 3224-3231 ◽  
Author(s):  
Hiroshi Kataoka ◽  
Justin R. Hamilton ◽  
David D. McKemy ◽  
Eric Camerer ◽  
Yao-Wu Zheng ◽  
...  

AbstractDefining the relative importance of protease-activated receptors (PARs) for thrombin signaling in mouse endothelial cells is critical for a basic understanding of thrombin signaling in these cells and for the rational use of knockout mice to probe the roles of thrombin's actions on endothelial cells in vivo. We examined thrombin- and PAR agonist–induced increases in cytoplasmic calcium, phosphoinositide hydrolysis, extracellular signal-regulated kinase (ERK) phosphorylation, and gene expression in endothelial cells from wild-type and PAR-deficient mice. PAR1 and PAR4 agonists triggered responses in wild-type but not in Par1–/– and Par4–/– endothelial cells, respectively. Calcium imaging confirmed that a substantial fraction of individual endothelial cells responded to both agonists. Compared with wild-type cells, Par1–/– endothelial cells showed markedly decreased responses to low concentrations of thrombin, and cells that lacked both PAR1 and PAR4 showed no responses to even high concentrations of thrombin. Similar results were obtained when endothelial-dependent vasorelaxation of freshly isolated mouse aorta was used as an index of signaling in native endothelial cells. Thus PAR1 is the major thrombin receptor in mouse endothelial cells, but PAR4 also contributes. These receptors serve at least partially redundant roles in endothelial cells in vitro and in vivo and together are necessary for the thrombin responses measured.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 921-921
Author(s):  
Enriqueta Coll-Sangrona ◽  
Ali Amirkhosravi ◽  
Alshad S. Lalani ◽  
Liza Robles ◽  
Hina Desai ◽  
...  

Abstract Calcitriol, the hormonally-active metabolite of Vitamin D3, plays critical roles in calcium homeostasis, cell growth and differentiation, and immunoregulation. The anti-tumor activities of high-dose calcitriol have been demonstrated in a variety of preclinical models of solid tumors, leukemias and lymphomas. Recently, a new dose-intense formulation of calcitriol, termed DN-101 (Asentar™), was developed specifically for cancer therapy which allows for supraphysiological concentrations of calcitriol to be safely delivered in vivo to patients with cancer. In a recent Phase 2 clinical trial, DN-101 significantly increased overall survival and also reduced the incidence of thromboembolic events in men with androgen-independent prostate cancer receiving docetaxel-based chemotherapy. Based on previous observations we hypothesized that calcitriol’s anti-thrombotic effects in vivo may be due to the downregulation of Tissue Factor (TF) antigen and activity and/or upregulation of Thrombomodulin (TM). To test this hypothesis, we incubated A549 lung carcinoma, A375-C15 metastatic melanoma, THP-1 monocytic leukemia, and Eahy926 endothelial cells with increasing concentrations of calcitriol for 24 hrs. For TF induction, tumor cells were stimulated with TNFα for 5 hrs and activity was measured by a clotting assay and a thrombin generation assay (TGA). TM activity was measured by a chromogenic assay. TF and TM surface antigen were assessed by flow cytometry. Calcitriol prevented the induction of TF in TNFα-stimulated THP-1 cells in a dose-dependent manner (from 33% at 1 nM to 94% at 100 nM) as evidenced by a prolongation of plasma clotting time, a decrease in endogenous thrombin potential (ETP), and a reduction of surface TF antigen. In addition, the activity and surface expression of TM on THP-1 cells was increased significantly (40% and 3-fold respectively, P < 0.01) following 100 nM calcitriol treatment. Similarly, in TNFα-stimulated melanoma cells, calcitriol prevented the induction of TF activity (from 26% at 1 nM to 60% at 1 μM) and expression in a dose-dependent manner. High-dose calcitriol treatment also increased melanoma cell TM activity between 8% and 62%. In contrast, constitutively expressed TF activity and antigen were less affected by calcitriol in A549 lung carcinoma cells (12 to 28% reduction at concentrations between 1–100 nM) whilst TM activity and antigen were unaffected. In comparison to the tumor cells, calcitriol had no significant effect on TM or TF activity or antigen in TNFα-stimulated EAhy926 endothelial cells. In conclusion, we have demonstrated that high concentrations of calcitriol inhibit the induction of surface TF expression and upregulates TM in multiple tumor cell lines in vitro. The degree of the inhibition is proportional to the extent of TF induction by TNF-α. These in vitro results provide further support for the anticoagulant properties associated with high concentrations of calcitriol and may provide a rationale for understanding the lower incidence of thromboembolic complications observed in patients with metastatic prostate cancer treated with DN-101.


1987 ◽  
Vol 58 (02) ◽  
pp. 778-785 ◽  
Author(s):  
J H Nuijens ◽  
C C M Huijbregts ◽  
M Cohen ◽  
G O Navis ◽  
A de Vries ◽  
...  

SummaryRadioimmunoassays (RIAs) for the detection of C1-inhihitor (C1-Inh) complexed to either kallikrein or activated Hageman factor (factor XIIa) are described. Kallikrein-C1-Inh or factor XIIa-C1-Inh complexes were bound to Scpharosc to which monospecific antibodies against (pre)kallikrein or factor XII, respectively, were coupled. Bound complexes were subsequently detected by an incubation with affinity purified 125I-labeled antibodies against Ci-Inh. These RIAs were used to detect activation of the contact system of coagulation in vitro and in vivo. Addition of dextran sulfate (DXS) (20 μg/ml) to fresh plasma resulted at 37° C in the rapid generation of amidolytic kallikrein activity, which was maximal after 1 to 2 min of incubation and subsequently decreased within a few minutes. The generation of kallikrein activity coincided with the appearance of both kallikrein-C1-Inh and factor XIIa-C1-Inh complexes. However, in contrast to kallikrein activity, both types of complexes remained detectable in the incubation mixtures during the incubation period. Experiments with purified kallikrein, C1-Inh and partly purified β-factor XIIa, and activation experiments in plasmas deficient in either factor XII or prekallikrein, demonstrated the specificity of both RIAs. The minimal amount of DXS that resulted in the generation of measurable amounts of both types of complexes in plasma was 2-3 μg per ml. Similar experiments with kaolin showed that with limiting amounts of activator (1-2 mg/ ml), only kallikrein-C1-Inh complexes were detected in plasma. When larger amounts of kaolin were added to plasma, factor XIIa-C1-Inh complexes were additionally detected in plasma. In plasma samples obtained from healthy donors under conditions that prevented activation of the contact system in vitro, very low levels of both factor XIIa-C1-Inh and kallikrein-C1-Inh complexes were measured, representing approximately 0.3% activation of both factor XII and prekallikrein. In serial plasma samples from a patient with adult respiratory distress syndrome, increased levels of both types of complexes were detected. The radioimmunoassays described in this paper provide useful tools to detect activation of the contact system in vitroas well as in vivo.


1981 ◽  
Author(s):  
L Jørgensen ◽  
A G Grøthe ◽  
T Larsen ◽  
R L Kinlough-Rathbone ◽  
J F Mustard

Platelets stimulated in vivo may cause endothelial injury. The question is whether a similar effect of platelet stimulation may be shown in vitro.Endothelial cells were cultured from human umbilical veins in Medium 199 with 20 per cent heat-inactivated serum. Semiconfluent cultures, 100-400.000 cells per dish, were labelled with Na2Cr51O4. Twenty-four hours later human platelet suspension (final cone. 200.000 per mm3) and thrombin (final cone. 4 U/ml) were added to the medium and the culture dish shaken for 15 min. The percentage of cells detached from the culture dish and the percentage of Cr51 released from the endothelial cells into the ambient fluid during the shaking were determined and used as parameters of cell injury. Increased percentages of loosened cells and Cr51 into the ambient fluid were observed with platelet suspension and thrombin compared to controls without platelet suspension and/or thrombin. Platelet-free supernatant after reaction of thrombin with platelet suspension had a similar effect. Thrombin alone caused a moderat release of Cr51, but no increased loosening of cells. Both parameters increased with increasing dose of thrombin when the platelet count was kept constant. The percentage of loosened cells increased with increasing platelet number when the dose of thrombin was constant. A significant release of Cr51 occurred only at high concentrations of platelets. Scanning and transmission electron microscopy of endothelial cells exposed to thrombin-stimulated platelets confirmed the presence of loosening and injured cells in association with platelets. The endothelial cells were bulging, intercellular junctions opened up, the plasma membrane on the luminal side showed villi and breaks and the cytoplasm had increased electron density. Thus, platelets stimulated by thrombin may cause injury of endothelial cells also in vitro.


1999 ◽  
Vol 81 (06) ◽  
pp. 984-988 ◽  
Author(s):  
Eva Norström ◽  
Johan Sundelin ◽  
Sverker Nystedt ◽  
Anna-Karin Alm

SummaryProteolytically activated receptors define a new subclass among the G-protein coupled receptors. Proteinase activated receptor-2 (PAR-2), the second member to be identified of this growing receptor subclass, can be activated by trypsin and trypsin-like serine proteases such as mast cell tryptase. PAR-2 is expressed in endothelial cells. Here we have studied if activation of PAR-2 changes the coagulation properties of cultured human umbilical vein endothelial cells. We show that activation of PAR-2 induces rapid and transient formation of tissue factor mRNA with a maximum level 1 hour after receptor stimulation. The increased mRNA level was accompanied by an increased tissue factor activity at the endothelial cell surface, shortening coagulation time in a standard clotting assay. The level of tissue factor activity after PAR-2 activation was comparable with the effects of thrombin receptor (PAR-1) activation although neither of the two protease receptors were as strong inducers of tissue factor as tumor necrosis factor-α.


Sign in / Sign up

Export Citation Format

Share Document