Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 114(47): 12454-12459, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109275

ABSTRACT

Recombinant factor VIIa (FVIIa) variants with increased activity offer the promise to improve the treatment of bleeding episodes in patients with inhibitor-complicated hemophilia. Here, an approach was adopted to enhance the activity of FVIIa by selectively optimizing substrate turnover at the membrane surface. Under physiological conditions, endogenous FVIIa engages its cell-localized cofactor tissue factor (TF), which stimulates activity through membrane-dependent substrate recognition and allosteric effects. To exploit these properties of TF, a covalent complex between FVIIa and the soluble ectodomain of TF (sTF) was engineered by introduction of a nonperturbing cystine bridge (FVIIa Q64C-sTF G109C) in the interface. Upon coexpression, FVIIa Q64C and sTF G109C spontaneously assembled into a covalent complex with functional properties similar to the noncovalent wild-type complex. Additional introduction of a FVIIa-M306D mutation to uncouple the sTF-mediated allosteric stimulation of FVIIa provided a final complex with FVIIa-like activity in solution, while exhibiting a two to three orders-of-magnitude increase in activity relative to FVIIa upon exposure to a procoagulant membrane. In a mouse model of hemophilia A, the complex normalized hemostasis upon vascular injury at a dose of 0.3 nmol/kg compared with 300 nmol/kg for FVIIa.


Subject(s)
Biological Therapy/methods , Factor VIIa/chemistry , Hemophilia A/therapy , Protein Engineering/methods , Thromboplastin/chemistry , Allosteric Regulation , Animals , Blood Coagulation/drug effects , Disease Models, Animal , Factor VIIa/genetics , Factor VIIa/pharmacology , Factor VIIa/therapeutic use , Female , Hemophilia A/physiopathology , Humans , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Dynamics Simulation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thromboplastin/genetics , Thromboplastin/pharmacology , Thromboplastin/therapeutic use
2.
Blood ; 118(8): 2333-41, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21700771

ABSTRACT

Current management of hemophilia B entails multiple weekly infusions of factor IX (FIX) to prevent bleeding episodes. In an attempt to make a longer acting recombinant FIX (rFIX), we have explored a new releasable protraction concept using the native N-glycans in the activation peptide as sites for attachment of polyethylene glycol (PEG). Release of the activation peptide by physiologic activators converted glycoPEGylated rFIX (N9-GP) to native rFIXa and proceeded with normal kinetics for FXIa, while the K(m) for activation by FVIIa-tissue factor (TF) was increased by 2-fold. Consistent with minimal perturbation of rFIX by the attached PEG, N9-GP retained 73%-100% specific activity in plasma and whole-blood-based assays and showed efficacy comparable with rFIX in stopping acute bleeds in hemophilia B mice. In animal models N9-GP exhibited up to 2-fold increased in vivo recovery and a markedly prolonged half-life in mini-pig (76 hours) and hemophilia B dog (113 hours) compared with rFIX (16 hours). The extended circulation time of N9-GP was reflected in prolonged correction of coagulation parameters in hemophilia B dog and duration of effect in hemophilia B mice. Collectively, these results suggest that N9-GP has the potential to offer efficacious prophylactic and acute treatment of hemophilia B patients at a reduced dosing frequency.


Subject(s)
Factor IX/chemistry , Factor IX/metabolism , Animals , Binding Sites , Disease Models, Animal , Dogs , Factor IX/genetics , Female , Half-Life , Hemophilia B/blood , Hemophilia B/drug therapy , Hemophilia B/genetics , Hemostatics/blood , Hemostatics/chemistry , Hemostatics/pharmacology , Humans , In Vitro Techniques , Kinetics , Male , Mice , Mice, Mutant Strains , Polyethylene Glycols/chemistry , Polysaccharides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Swine , Swine, Miniature
3.
Thromb Res ; 128(2): 191-5, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21429564

ABSTRACT

INTRODUCTION: Bleeding episodes in haemophilia patients with inhibitors are primarily treated with by-passing agents such as recombinant activated FVII (rFVIIa). Prophylactic treatment with rFVIIa has been shown to significantly reduce the number of bleeding episodes as compared to conventional on-demand haemostatic therapy, and a reduced dosing frequency could present an improved treatment option in inhibitor patients. MATERIALS AND METHODS: A series of glycoPEGylated rFVIIa derivatives (5-40K PEG) has been produced and their effect and pharmocokinetics have been investigated in several animal species. RESULTS: The glycoPEGylated rFVIIa derivatives exhibit significant prolongation of half-life in mice, dogs and pigs as measured by rFVIIa clot activity. The clearance of rFVIIa, rFVIIa-5K PEG, rFVIIa-10K PEG, rFVIIa-20K PEG and rFVIIa-40K PEG in minipigs were estimated to 59, 27, 22, 8.7 and 3.1 ml/h/kg, respectively. Across species a reduction in clearance as a function of the size of the attached PEG was observed. By allometric scaling, the compiled pharmacokinetics predicts a human half-life for rFVIIa-10K PEG and rFVIIa-40K PEG of approximately 7 and 12h, respectively. The rFVIIa-10K PEG and rFVIIa-40K PEG are efficacious in stopping a bleed in the haemophilia A mouse tail-bleeding model after intravenous administration. CONCLUSIONS: GlycoPEGylation of rFVIIa significantly increases the rFVIIa exposure in three animal models, glycoPEGylated rFVIIa compounds are effective in vivo and thus, represents a potential prophylactic treatment option for patients with inhibitors.


Subject(s)
Factor VIIa/pharmacokinetics , Hemophilia A/metabolism , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Animals , Disease Models, Animal , Dogs , Factor VIIa/chemistry , Factor VIIa/pharmacology , Female , Glycosylation , Half-Life , Hemorrhage/etiology , Hemorrhage/metabolism , Humans , Male , Mice , Polyethylene Glycols/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Swine , Swine, Miniature , Tail/blood supply
4.
Thromb Res ; 126(2): 144-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20542542

ABSTRACT

INTRODUCTION: There is no established laboratory method that can predict the most optimal dose of bypassing agents for treatment of haemophilia A. The objectives of the study was to develop an assay that can a) differentiate between the haemostatic capacity in blood from healthy individuals and severe and moderate haemophilia patients; b) show a dose-response correlation to rFVIIa addition; and c) show dose response differences of rFVIIa addition to plasma samples from non-inhibitor patients of different severity. MATERIALS AND METHODS: Citrated whole blood from 25 haemophilia A patients was used in four thrombelastography (TEG) assays initiated with: 1) kaolin, 2) Tissue Factor (TF, Innovin 1:42,500), 3) TF 1:42,500+1.2 nM tPA (tissue plasminogen activator) or 4) TF 1:200,000. rFVIIa was added to give a final concentration in the range of 0.02-4.8 microg/ml. RESULTS: The TEG assays showed large differences in clot formation demonstrated by prolonged clotting time (R-time), decreased maximum thrombus generation (MTG) between severe and moderate haemophilia A patients and between haemophilia patients and healthy males. The maximal amplitudes (MA) of the clot and resistance against fibrinolysis were only compromised when TF with tPA was added. CONCLUSION: In vitro addition of rFVIIa improved all TEG profiles significantly in a dose-dependent manner; but only the TEG assay containing kaolin could differentiate between the rFVIIa doses, showing that blood from severe patients need higher doses of rFVIIa to normalize the clot formation profile compared to blood from moderate patients. Kaolin seems to be the most useful TEG assay for monitoring rFVIIa treatment.


Subject(s)
Coagulants/therapeutic use , Drug Monitoring/methods , Factor VIIa/therapeutic use , Hemophilia A/diagnosis , Hemophilia A/drug therapy , Thrombelastography/methods , Adolescent , Drug Monitoring/economics , Humans , Male , Recombinant Proteins/therapeutic use , Reproducibility of Results , Thrombelastography/economics , Young Adult
5.
Thromb Haemost ; 101(5): 818-26, 2009 May.
Article in English | MEDLINE | ID: mdl-19404533

ABSTRACT

The mechanism for the elimination of factor VII (FVII) from the circulation is unknown, just as it is unclear how activation of FVII to FVIIa and subsequent complex formation with antithrombin III (AT) or alpha2-macroglobulin (alpha2M) affects clearance. The possibility that the clearance mechanism involves activation and inhibitor complex formation as obligatory intermediate reactions is examined in this study. Human and murine sera were spiked with human FVIIa in the absence and presence of heparin and analysed for complex formation. Complex formation in vivo was studied after intravenous injection of (125)I-VIIa in mice; and the pharmacokinetics (PK) of human and murine FVIIa was studied in normal mice. Furthermore, comparative PK studies were performed with FVII, FVIIa, active site blocked FVIIa and a preformed FVIIa-AT complex in normal and alpha2M-deficient mice. The data demonstrated that FVIIa-AT complexes and to a much lesser extent FVIIa-alpha2M-complexes accumulated in vivo after FVIIa administration. FVIIa-AT accounted for about 50% of total FVIIa antigen left in the circulation after 3 hours. All FVII derivatives studied including FVII, FVIIa and FVIIa-AT were cleared with similar rates suggesting an elimination kinetics which is unaffected by FVII activation and subsequent inactivation by plasma inhibitors.


Subject(s)
Antithrombin III/metabolism , Factor VII/pharmacokinetics , Factor VIIa/pharmacokinetics , Serum Globulins/metabolism , alpha-Macroglobulins/metabolism , Animals , Factor VIIa/administration & dosage , Heparin/blood , Humans , Injections, Intravenous , Iodine Radioisotopes , Mice , Mice, Inbred C57BL , Mice, Knockout , Serum Globulins/deficiency , Serum Globulins/genetics , alpha-Macroglobulins/deficiency , alpha-Macroglobulins/genetics
6.
Thromb Haemost ; 100(5): 920-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18989539

ABSTRACT

Prophylaxis with 2-4 times weekly dosing of factor (F)VIII or FIX is established as an efficacious and safe treatment in haemophilia. Although prophylaxis is not readily available for the inhibitor patient, recent studies have demonstrated a reduction in bleeding episodes in inhibitor patients treated with daily infusions of FVIIa. In order to develop a treatment option comparable to prophylaxis with FVIII or FIX we looked to PEGylation which is an established method for prolonging the circulatory half-life of proteins. However, due to the numerous interactions of FVIIa with the cell surface, TF, FIX and FX there are limited options for unspecific chemical modification of FVIIa without loss of activity. Consequently, we explored the GlycoPEGylationtrade mark technology for selective PEGylation of the two N-glycans in the FVIIa light chain and protease domain to generate seven specifically modified derivatives with PEG groups ranging from 2 to 40 kDa. These derivatives were evaluated in vitro for their ability to interact with small synthetic substrates as well as key molecules relevant to function in the coagulation pathway. The results demonstrate that modification of FVIIa using glycoPEGylation has only a very limited effect on the hydrolysis S-2288 and FX activation. However, the modification does to some extend alter the ability of FVIIa to interact with TF and more importantly, reduces the rate of ATIII inhibition by up to 50% which could allow for an extended active half-life in circulation.


Subject(s)
Coagulants/metabolism , Factor VIIa/metabolism , Animals , Antithrombin III/metabolism , Coagulants/antagonists & inhibitors , Coagulants/chemical synthesis , Factor VIIa/antagonists & inhibitors , Factor VIIa/chemical synthesis , Factor Xa/metabolism , Half-Life , Humans , Hydrolysis , Models, Molecular , Molecular Weight , Oligopeptides/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Structure-Activity Relationship , Thromboplastin/metabolism
8.
Thromb Res ; 116(1): 75-85, 2005.
Article in English | MEDLINE | ID: mdl-15850611

ABSTRACT

Tissue factor (TF) is believed to play an important role in coagulation, inflammation, angiogenesis and wound healing as well as in tumor growth and metastasis. To facilitate in vivo studies in experimental murine models, we have produced recombinant murine factor VII (FVII) and the ectodomain of murine TF, TF(1-223). Murine FVII was activated to FVIIa with human factor Xa and upon reaction with FFR-chloromethyl ketone converted into an active site-blocked TF antagonist, FFR-FVIIa. The activity of murine FVIIa was characterized in factor X activation assays as well as in clot assays with murine and human thromboplastin in murine and human plasma. In these assays murine FVIIa exhibited a specific activity equivalent to or higher than human FVIIa. Further analysis showed that murine FVIIa binds with high affinity to both murine and human TF, whereas the association of human FVIIa to murine TF is about three orders of magnitude weaker than the association to human TF. This difference was further emphasized by the effect of murine-and human FFR-FVIIa on bleeding in an in vivo mouse model. Intra-peritoneal administration of 1 mg/kg murine FFR-FVIIa significantly prolonged the tail-bleeding time, whereas no effect on bleeding was observed with a 25-times higher dose of the human FFR-FVIIa. Together, these data confirms the notion of poor species compatibility between human FVII and murine TF and emphasizes the requirement for autologous FVIIa in studies on the role of the TF in experimental in vivo pharmacology.


Subject(s)
Factor VIIa/chemical synthesis , Factor VIIa/pharmacology , Thromboplastin/metabolism , Animals , Binding Sites , Cloning, Molecular , Factor VIIa/metabolism , Hemorrhage/drug therapy , Humans , Mice , Models, Animal , Recombinant Proteins/chemical synthesis , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Species Specificity
9.
Thromb Haemost ; 93(1): 27-34, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15630487

ABSTRACT

We previously demonstrated that FVIIa bound to tissue factor (TF) induces a hyperchemotactic response towards PDGF-BB. The aim of the present study was to investigate the role of the cytoplasmic domain of TF in cell migration. Porcine aortic endothelial (PAE) cells expressing human PDGF beta-receptors (PAE/PDGFRbeta) were transfected for expression of human wildtype TF (PAE/PDGFRbeta,TF), a construct lacking the cytoplasmic domain (PAE/PDGFRbeta,TFDeltacyto), a construct with alanine replacement of serine 258 (PAE/PDGFRbeta,TFS258A), or a construct with alanine replacement of serine 253, 258 and 263 in the cytoplasmic domain (PAE/PDGFRbeta,TF3SA). All stably transfected cell lines expressed functional TF. Chemotaxis was analyzed in a modified Boyden chamber assay. PAE/PDGFRbeta,TF cells stimulated with FVIIa migrated towards a 100-fold lower concentration of PDGF-BB than in the absence of FVIIa, however, hyperchemotaxis was not seen in PAE/PDGFRbeta,TFDeltacyto cells. PAE/PDGFRbeta/TFS258A and PAE/PDGFRbeta,TF3SA cells responded to low levels of PDGF-BB, but migrated a significantly shorter distance than PAE/PDGFRbeta,TF cells. Thus, hyperchemotaxis towards PDGF-BB is likely to depend in part on phosphorylation of the cytoplasmic domain of TF. We conclude that the cytoplasmic domain of TF plays a pivotal role in modulating cellular migration response. Our results suggest that the FVIIa/TF complex mediates intracellular signaling by alternative signal transduction pathway(s).


Subject(s)
Chemotaxis , Endothelium, Vascular/cytology , Receptor, Platelet-Derived Growth Factor beta/physiology , Thromboplastin/physiology , Animals , Aorta/cytology , Cells, Cultured , Cytoplasm , Factor VIIa/pharmacology , Humans , MAP Kinase Signaling System , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Receptor, Platelet-Derived Growth Factor beta/genetics , Swine , Thromboplastin/chemistry , Thromboplastin/genetics , Transfection
10.
Blood ; 103(8): 3029-37, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15070680

ABSTRACT

Tissue factor (TF), the cellular receptor for factor VIIa (FVIIa), besides initiating blood coagulation, is believed to play an important role in tissue repair, inflammation, angiogenesis, and tumor metastasis. Like TF, the chemokine interleukin-8 (IL-8) is shown to play a critical role in these processes. To elucidate the potential mechanisms by which TF contributes to tumor invasion and metastasis, we investigated the effect of FVIIa on IL-8 expression and cell migration in a breast carcinoma cell line, MDA-MB-231, a cell line that constitutively expresses abundant TF. Expression of IL-8 mRNA in MDA-MB-231 cells was markedly up-regulated by plasma concentrations of FVII or an equivalent concentration of FVIIa (10 nM). Neither thrombin nor other proteases involved in hemostasis were effective in stimulating IL-8 in these cells. Increased transcriptional activation of the IL-8 gene is responsible for increased expression of IL-8 in FVIIa-treated cells. PAR-2-specific antibodies fully attenuated TF-FVIIa-induced IL-8 expression. Additional in vitro experiments showed that TF-FVIIa promoted tumor cell migration and invasion, active site-inactivated FVIIa, and specific antibodies against TF, PAR-2, and IL-8 inhibited TF-FVIIa-induced cell migration. In summary, the studies described herein provide insight into how TF may contribute to tumor invasion.


Subject(s)
Factor VIIa/pharmacology , Interleukin-8/genetics , Receptor, PAR-2/metabolism , Thromboplastin/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Factor VIIa/metabolism , Female , Humans , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Thromboplastin/antagonists & inhibitors , Up-Regulation/drug effects
11.
Blood ; 102(5): 1708-15, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12738672

ABSTRACT

Binding of factor VIIa (FVIIa) to its cellular receptor tissue factor (TF) was previously shown to induce various intracellular signaling events, which were thought to be responsible for TF-mediated biologic effects, including angiogenesis, tumor metastasis, and restenosis. To understand the mechanisms behind these processes, we have examined the effect of FVIIa on apoptosis. Serum deprivation-induced apoptosis of BHK(+TF) cells was characterized by apoptotic blebs, nuclei with chromatin-condensed bodies, DNA degradation, and activation of caspase 3. FVIIa markedly decreased the number of cells with apoptotic morphology and prevented the DNA degradation as measured by means of TdT-mediated dUTP nick end labeling (TUNEL). The antiapoptotic effect of FVIIa was confirmed by the observation that FVIIa attenuated caspase 3 activation. FVIIa-induced antiapoptotic effect was dependent on its proteolytic activity and TF but independent of factor Xa and thrombin. FVIIa-induced cell survival correlated with the activation of Akt and was inhibited markedly by the specific PI3-kinase inhibitor, LY294002. Blocking the activation of p44/42 mitogen-activated protein kinase (MAPK) by the specific mitogen-induced extracellular kinase (MEK) inhibitor, U0126, impaired modestly the ability of FVIIa to promote cell survival. In conclusion, FVIIa binding to TF provided protection against apoptosis induced by growth factor deprivation, primarily through activation of PI3-kinase/Akt pathway, and to a lesser extent, p44/42 MAPK pathway.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Factor VIIa/pharmacology , Protein Serine-Threonine Kinases , Animals , Blood Proteins/pharmacology , CHO Cells , Caspase 3 , Caspases/metabolism , Cell Nucleus , Cell Survival/drug effects , Cell Survival/physiology , Cricetinae , Enzyme Activation/drug effects , Factor VIIa/metabolism , Humans , Kidney/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , Signal Transduction/physiology , Thromboplastin/metabolism
12.
J Biol Chem ; 277(30): 27065-72, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-12019261

ABSTRACT

FVIIa binding to tissue factor (TF) and subsequent signal transduction have now been implicated in a variety of pathophysiological processes, including cytokine production during sepsis, tumor angiogenesis and neoangiogenesis, and leukocyte diapedesis. The molecular details, however, by which FVIIa/TF affects gene expression and cellular physiology, remain obscure. Here we show that FVIIa induces a transient phosphorylation of p70/p85(S6K) and p90(RSK) in BHK cells stably transfected with either full-length TF or with a cytoplasmic domain-truncated TF but not in wild type BHK cells. Phosphorylation of these kinases was also observed in HaCaT cells, expressing endogenous TF. Phosphorylation of p70/p85(S6K) coincided with protein kinase B and GSK-3beta phosphorylation. Activation of p70/p85(S6K) was sensitive to inhibitors of phosphatidylinositol 3-kinase and to rapamycin, whereas phosphorylation of p90(RSK) was sensitive to PD98059. FVIIa stimulation of p70/p85(S6K) and p90(RSK) correlated with phosphorylation of the eukaryotic initiation factor eIF-4E, up-regulation of protein levels of eEF1alpha and eEF2, and enhanced [(35)S]methionine incorporation. These effects were not influenced by inhibitors of thrombin or FXa generation and were strictly dependent on the presence of the extracellular domain of TF, but they did not require the intracellular portion of TF. We propose that a TF cytoplasmic domain-independent stimulation of protein synthesis via activation of S6 kinase contributes to FVIIa effects in pathophysiology.


Subject(s)
Cytoplasm/metabolism , Factor VIIa/metabolism , Protein Serine-Threonine Kinases , Ribosomal Protein S6 Kinases/metabolism , Thromboplastin/metabolism , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-4E , Flavonoids/pharmacology , Glycogen Synthase Kinase 3 , Humans , Peptide Initiation Factors/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-akt , Sirolimus/pharmacology , Thrombin/metabolism , Time Factors , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...