Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Bioconjug Chem ; 30(3): 775-784, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30676028

RESUMO

A nonvolatile fluorine-18 aldehyde prosthetic group was developed from [18F]SFB, and used for site-specific labeling of active site inhibited factor VII (FVIIai). FVIIai has a high affinity for tissue factor (TF), a transmembrane protein involved in angiogenesis, proliferation, cell migration, and survival of cancer cells. A hydroxylamine N-glycan modified FVIIai (FVIIai-ONH2) was used for oxime coupling with the aldehyde [18F]2 under mild and optimized conditions in an isolated RCY of 4.7 ± 0.9%, and a synthesis time of 267 ± 5 min (from EOB). Retained binding and specificity of the resulting [18F]FVIIai to TF was shown in vitro. TF-expression imaging capability was evaluated by in vivo PET/CT imaging in a pancreatic human xenograft cancer mouse model. The conjugate showed exceptional stability in plasma (>95% at 4 h) and a binding fraction of 90%. In vivo PET/CT imaging showed a mean tumor uptake of 3.8 ± 0.2% ID/g at 4 h post-injection, a comparable uptake in liver and kidneys, and low uptake in normal tissues. In conclusion, FVIIai was labeled with fluorine-18 at the N-glycan chain without affecting TF binding. In vitro specificity and a good in vivo imaging contrast at 4 h postinjection was demonstrated.


Assuntos
Aldeídos/química , Fator VII/antagonistas & inibidores , Radioisótopos de Flúor/química , Oximas/química , Animais , Sítios de Ligação , Domínio Catalítico , Ciclização , Camundongos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tromboplastina/metabolismo , Distribuição Tecidual , Água
2.
Blood ; 130(14): 1661-1670, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28729433

RESUMO

Safe and effective antithrombotic therapy requires understanding of mechanisms that contribute to pathological thrombosis but have a lesser impact on hemostasis. We found that the extrinsic tissue factor (TF) coagulation initiation complex can selectively activate the antihemophilic cofactor, FVIII, triggering the hemostatic intrinsic coagulation pathway independently of thrombin feedback loops. In a mouse model with a relatively mild thrombogenic lesion, TF-dependent FVIII activation sets the threshold for thrombus formation through contact phase-generated FIXa. In vitro, FXa stably associated with TF-FVIIa activates FVIII, but not FV. Moreover, nascent FXa product of TF-FVIIa can transiently escape the slow kinetics of Kunitz-type inhibition by TF pathway inhibitor and preferentially activates FVIII over FV. Thus, TF synergistically primes FIXa-dependent thrombin generation independently of cofactor activation by thrombin. Accordingly, FVIIa mutants deficient in direct TF-dependent thrombin generation, but preserving FVIIIa generation by nascent FXa, can support intrinsic pathway coagulation. In ex vivo flowing blood, a TF-FVIIa mutant complex with impaired free FXa generation but activating both FVIII and FIX supports efficient FVIII-dependent thrombus formation. Thus, a previously unrecognized TF-initiated pathway directly yielding FVIIIa-FIXa intrinsic tenase complex may be prohemostatic before further coagulation amplification by thrombin-dependent feedback loops enhances the risk of thrombosis.


Assuntos
Coagulação Sanguínea , Fator VIII/metabolismo , Fator VIIa/metabolismo , Fator Xa/metabolismo , Tromboplastina/metabolismo , Fator VIIIa/metabolismo , Humanos , Trombina/metabolismo
3.
Bioconjug Chem ; 29(1): 117-125, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29206443

RESUMO

A method for site-specific radiolabeling of the serine protease active site inhibited factor seven (FVIIai) with 64Cu has been applied using a biorthogonal click reaction. FVIIai binds to tissue factor (TF), a trans-membrane protein involved in hemostasis, angiogenesis, proliferation, cell migration, and survival of cancer cells. First a single azide moiety was introduced in the active site of this 50 kDa protease. Then a NOTA moiety was introduced via a strain promoted azide-alkyne reaction and the corresponding conjugate was labeled with 64Cu. Binding to TF and the stability was evaluated in vitro. TF targeting capability of the radiolabeled conjugate was tested in vivo by positron emission tomography (PET) imaging in pancreatic human xenograft cancer mouse models with various TF expressions. The conjugate showed good stability (>91% at 16 h), an immunoreactivity of 93.5%, and a mean tumor uptake of 2.1 ± 0.2%ID/g at 15 h post injection. In conclusion, FVIIai was radiolabeled with 64Cu in single well-defined position of the protein. This method can be utilized to prepare conjugates from serine proteases with the label at a specific position.


Assuntos
Azidas/química , Química Click/métodos , Radioisótopos de Cobre/química , Fator VII/química , Neoplasias Pancreáticas/diagnóstico por imagem , Serina Proteases/química , Tromboplastina/análise , Animais , Domínio Catalítico , Linhagem Celular Tumoral , Fator VII/farmacologia , Feminino , Compostos Heterocíclicos/química , Compostos Heterocíclicos com 1 Anel , Humanos , Marcação por Isótopo/métodos , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Serina Proteases/farmacologia
4.
Nature ; 483(7391): 627-31, 2012 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-22407318

RESUMO

The gut microbiota is a complex ecosystem that has coevolved with host physiology. Colonization of germ-free (GF) mice with a microbiota promotes increased vessel density in the small intestine, but little is known about the mechanisms involved. Tissue factor (TF) is the membrane receptor that initiates the extrinsic coagulation pathway, and it promotes developmental and tumour angiogenesis. Here we show that the gut microbiota promotes TF glycosylation associated with localization of TF on the cell surface, the activation of coagulation proteases, and phosphorylation of the TF cytoplasmic domain in the small intestine. Anti-TF treatment of colonized GF mice decreased microbiota-induced vascular remodelling and expression of the proangiogenic factor angiopoietin-1 (Ang-1) in the small intestine. Mice with a genetic deletion of the TF cytoplasmic domain or with hypomorphic TF (F3) alleles had a decreased intestinal vessel density. Coagulation proteases downstream of TF activate protease-activated receptor (PAR) signalling implicated in angiogenesis. Vessel density and phosphorylation of the cytoplasmic domain of TF were decreased in small intestine from PAR1-deficient (F2r(-/-)) but not PAR2-deficient (F2rl1(-/-)) mice, and inhibition of thrombin showed that thrombin-PAR1 signalling was upstream of TF phosphorylation. Thus, the microbiota-induced extravascular TF-PAR1 signalling loop is a novel pathway that may be modulated to influence vascular remodelling in the small intestine.


Assuntos
Intestino Delgado/irrigação sanguínea , Intestino Delgado/microbiologia , Neovascularização Fisiológica , Receptor PAR-1/metabolismo , Tromboplastina/metabolismo , Alelos , Angiopoietina-1/metabolismo , Animais , Enterócitos/metabolismo , Enterócitos/microbiologia , Feminino , Glicosilação , Intestino Delgado/citologia , Camundongos , Fosforilação , Estrutura Terciária de Proteína/genética , Receptor PAR-1/deficiência , Receptor PAR-1/genética , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Transdução de Sinais , Trombina/metabolismo , Tromboplastina/química , Tromboplastina/genética
5.
Proc Natl Acad Sci U S A ; 109(10): 3927-31, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22355108

RESUMO

Tissue factor pathway inhibitor (TFPI) blocks thrombin generation via the extrinsic blood coagulation pathway. Because the severe bleeding in patients with hemophilia occurs from deficiency of intrinsic blood coagulation pathway factor VIII or IX, pharmacological agents that inactivate TFPI and, therefore, restore thrombin generation via the extrinsic pathway, are being developed for treatment of hemophilia. Murine models of combined TFPI and factor VIII deficiency were used to examine the impact of TFPI deficiency on bleeding and clotting in hemophilia. In breeding studies, Factor VIII null (F8(-/-)) did not rescue the embryonic death of TFPI null (Tfpi(-/-)) mice. Tfpi(+/-) did not alter the bleeding phenotype of F8(-/-) mice. However, total inhibition of intravascular TFPI through injection of anti-TFPI antibody mitigated tail vein bleeding. Interestingly, tail blood loss progressively decreased at doses greater than needed to totally inhibit plasma TFPI, suggesting that inhibition of a sequestered pool of TFPI released at the injury site mitigates bleeding. Because TFPI is sequestered within platelets and released following their activation, the function of platelet TFPI was examined in F8(-/-) mice lacking hematopoietic cell TFPI that was generated by fetal liver transplantation. Blood loss after tail transection significantly decreased in Tfpi(+/-);F8(-/-) mice with hematopoietic Tfpi(-/-) cells compared with Tfpi(+/-);F8(-/-) mice with Tfpi(+/+) hematopoietic cells. Additionally, following femoral vein injury, Tfpi(+/-);F8(-/-) mice with Tfpi(-/-) hematopoietic cells had increased fibrin deposition compared with identical-genotype mice with Tfpi(+/+) hematopoietic cells. These findings implicate platelet TFPI as a primary physiological regulator of bleeding in hemophilia.


Assuntos
Hemofilia A/metabolismo , Lipoproteínas/genética , Lipoproteínas/metabolismo , Animais , Coagulação Sanguínea , Modelos Animais de Doenças , Feminino , Teste de Complementação Genética , Genótipo , Células-Tronco Hematopoéticas/citologia , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Hemorragia , Fígado/embriologia , Transplante de Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo
6.
J Labelled Comp Radiopharm ; 58(5): 196-201, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25820758

RESUMO

Activated factor VII blocked in the active site with Phe-Phe-Arg-chloromethyl ketone (active site inhibited factor VII (ASIS)) is a 50-kDa protein that binds with high affinity to its receptor, tissue factor (TF). TF is a transmembrane glycoprotein that plays an important role in, for example, thrombosis, metastasis, tumor growth, and tumor angiogenesis. The aim of this study was to develop an (18)F-labeled ASIS derivative to assess TF expression in tumors. Active site inhibited factor VII was labeled using N-succinimidyl-4-[(18)F]fluorobenzoate, and the [(18)F]ASIS was purified on a PD-10 desalting column. The radiochemical yield was 25 ± 6%, the radiochemical purity was >97%, and the pseudospecific radioactivity was 35 ± 9 GBq/µmol. The binding efficacy was evaluated in pull-down experiments, which monitored the binding of unlabeled ASIS and [(18)F]ASIS to TF and to a specific anti-factor VII antibody (F1A2-mAb). No significant difference in binding efficacy between [(18)F]ASIS and ASIS could be detected. Furthermore, [(18)F]ASIS was relatively stable in vitro and in vivo in mice. In conclusion, [(18)F]ASIS has for the first time been successfully synthesized as a possible positron emission tomography tracer to image TF expression levels. In vivo positron emission tomography studies to evaluate the full potential of [(18)F]ASIS are in progress.


Assuntos
Clorometilcetonas de Aminoácidos/química , Fator VII/química , Compostos Radiofarmacêuticos/síntese química , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Domínio Catalítico , Fator VII/antagonistas & inibidores , Radioisótopos de Flúor/química , Camundongos , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
7.
J Biol Chem ; 286(7): 5756-67, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21149441

RESUMO

Protease-activated receptor (PAR) signaling is closely linked to the cellular activation of the pro- and anticoagulant pathways. The endothelial protein C receptor (EPCR) is crucial for signaling by activated protein C through PAR1, but EPCR may have additional roles by interacting with the 4-carboxyglutamic acid domains of procoagulant coagulation factors VII (FVII) and X (FX). Here we show that soluble EPCR regulates the interaction of FX with human or mouse tissue factor (TF)-FVIIa complexes. Mutagenesis of the FVIIa 4-carboxyglutamic acid domain and dose titrations with FX showed that EPCR interacted primarily with FX to attenuate FX activation in lipid-free assay systems. In human cell models of TF signaling, antibody inhibition of EPCR selectively blocked PAR activation by the ternary TF-FVIIa-FXa complex but not by the non-coagulant TF-FVIIa binary complex. Heterologous expression of EPCR promoted PAR1 and PAR2 cleavage by FXa in the ternary complex but did not alter PAR2 cleavage by TF-FVIIa. In murine smooth muscle cells that constitutively express EPCR and TF, thrombin and FVIIa/FX but not FVIIa alone induced PAR1-dependent signaling. Although thrombin signaling was unchanged, cells with genetically reduced levels of EPCR no longer showed a signaling response to the ternary complex. These results demonstrate that EPCR interacts with the ternary TF coagulation initiation complex to enable PAR signaling and suggest that EPCR may play a role in regulating the biology of TF-expressing extravascular and vessel wall cells that are exposed to limited concentrations of FVIIa and FX provided by ectopic synthesis or vascular leakage.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Tromboplastina/metabolismo , Animais , Antígenos CD/genética , Células Cultivadas , Receptor de Proteína C Endotelial , Fator VIIa/genética , Fator VIIa/metabolismo , Fator X/genética , Fator X/metabolismo , Glicoproteínas/genética , Humanos , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Miócitos de Músculo Liso/citologia , Proteína C/genética , Proteína C/metabolismo , Estrutura Terciária de Proteína , Receptor PAR-1/genética , Receptor PAR-2/genética , Receptores de Superfície Celular/genética , Trombina/genética , Trombina/metabolismo , Tromboplastina/genética
8.
Blood ; 116(26): 6106-13, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20861457

RESUMO

Constitutive expression of tissue factor (TF) by cancer cells triggers local activation of the coagulation cascade and promotes breast cancer progression through cell signaling involving protease activated receptor (PAR)2. In human breast cancer, TF and PAR2 are up-regulated and TF cytoplasmic domain phosphorylation is correlated with relapse. Here we show that cancer cell PAR2 signaling promotes angiogenesis independent of PAR2 phosphorylation at the recognized ß-arrestin recruitment site. Similar to PAR2(-/-) mice, TF cytoplasmic domain-deleted (TF(ΔCT)) mice have delayed spontaneous breast cancer development in the polyoma middle T model. Simultaneous deletion of PAR2 in TF(ΔCT) mice did not further delay tumor appearance, consistent with overlapping roles of TF and PAR2 in promoting the angiogenic switch in early stages of breast cancer. In advanced carcinomas, tumor-associated macrophages were reduced in TF(ΔCT) and TF(ΔCT)/PAR2(-/-) mice, and increased tumor vessel diameters of TF(ΔCT) mice were partially reversed by PAR2-deficiency, indicating that the TF cytoplasmic domain has additional roles that are interdependent with PAR2 signaling in regulating host angiogenic responses. These experiments demonstrate a crosstalk of tumor cell TF cytoplasmic domain and PAR2 signaling and provide a possible mechanism for the close correlation between TF phosphorylation and cancer recurrence of TF and PAR2-positive clinical breast cancer.


Assuntos
Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/patologia , Neovascularização Patológica , Receptor PAR-2/fisiologia , Tromboplastina/fisiologia , Animais , Feminino , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
9.
J Biol Chem ; 285(26): 19959-66, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20388709

RESUMO

The complex of factor VIIa (FVIIa) with tissue factor (TF) triggers coagulation by recognizing its macromolecular substrate factors IX (FIX) and X (FX) predominantly through extended exosite interactions. In addition, TF mediates unique cell-signaling properties in cancer, angiogenesis, and inflammation that involve proteolytic cleavage of protease-activated receptor 2 (PAR2). PAR2 is cleaved by FVIIa in the binary TF.FVIIa complex and by FXa in the ternary TF.FVIIa.FXa complex, but physiological roles of these signaling complexes are incompletely understood. In a screen of FVIIa protease domain mutants, three variants (Q40A, Q143N, and T151S) activated macromolecular coagulation substrates and supported signaling of the ternary TF.FVIIa-Xa complex normally but were severely impaired in binary TF.FVIIa.PAR2 signaling. The residues identified were located in the model-predicted S2' pocket of FVIIa, and complementary PAR2 P2' Leu-38 replacements demonstrated that the P2' side chain was indeed crucial for PAR2 cleavage by TF.FVIIa. In addition, PAR2 was activated more efficiently by FVIIa T99Y, consistent with further contributions from the S2 subsite. The P2 residue preference of FVIIa and FXa predicted additional PAR2 mutants that were efficiently activated by TF.FVIIa but resistant to cleavage by the alternative PAR2 activator FXa. Thus, contrary to the paradigm of exosite-assisted cleavage of PAR1 by thrombin, the cofactor-associated protease FVIIa recognizes PAR2 predominantly by catalytic cleft interactions. Furthermore, the delineated molecular details of this substrate interaction enabled protein engineering of protease-selective PAR2 receptors that will aid further studies to dissect the roles of TF signaling complexes in vivo.


Assuntos
Fator VIIa/metabolismo , Receptor PAR-2/metabolismo , Transdução de Sinais , Tromboplastina/metabolismo , Substituição de Aminoácidos , Sítios de Ligação/genética , Linhagem Celular , Linhagem Celular Tumoral , Fator VIIa/química , Fator VIIa/genética , Humanos , Substâncias Macromoleculares/metabolismo , Modelos Moleculares , Mutação , Ligação Proteica , Engenharia de Proteínas , Estrutura Terciária de Proteína , Receptor PAR-2/genética , Especificidade por Substrato , Tromboplastina/genética , Transfecção
10.
Br J Haematol ; 152(1): 99-107, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21083658

RESUMO

NN1731 is a recombinant activated factor VII (rFVIIa) analogue with increased intrinsic activity. This also applies to its reactivity towards antithrombin (AT), the role of which was investigated in a pharmacokinetic (PK) study. NN1731 or rFVIIa was administered to normal and haemophilia A dogs and elimination was measured by FVIIa clot activity, FVIIa- and FVIIa-AT antigen. In vitro AT complex formation was studied in canine plasma spiked with NN1731 or rFVIIa. Based on FVIIa antigen concentrations, PK profiles in normal and haemophilia A dogs were similar for NN1731 and rFVIIa with antigen half lives, t(½) ≈1·8 h. In contrast, PK profiles based on activity measurements were distinctly different. NN1731 induced a strong, short lasting (t(½) ≈0·5 h) pro-coagulant response, whereas rFVIIa induced a lower, longer lasting (t(½) ≈1·1 h) response. Western Blot and FVIIa-AT antigen analysis demonstrated in vivo AT complex formation that accounted for these divergences. AT complex formation with FVIIa or NN1731 in vitro in canine plasma was considerably slower than the in vivo reaction. The results suggest that in vivo inhibition by AT contributes significantly to define drug duration in haemophilia treatment with rFVIIa and in particular with the NN1731 analogue.


Assuntos
Proteínas Antitrombina/fisiologia , Coagulantes/farmacocinética , Fator VII/farmacocinética , Hemofilia A/sangue , Animais , Coagulação Sanguínea/efeitos dos fármacos , Inibidores dos Fatores de Coagulação Sanguínea/fisiologia , Coagulantes/antagonistas & inibidores , Modelos Animais de Doenças , Cães , Fator VII/antagonistas & inibidores , Fator VIIa/antagonistas & inibidores , Meia-Vida , Masculino , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/sangue , Tromboelastografia/métodos
11.
Crit Care Med ; 37(6): 1874-80, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19384216

RESUMO

OBJECTIVE: To evaluate the safety and efficacy of active site inactivated recombinant factor VIIa (FFR-rFVIIa) in patients with acute respiratory distress syndrome. DESIGN: Phase II, randomized, double-blind, placebo-controlled, dose-escalation trial. SETTING: Forty-six intensive care units (ICU) in ten countries. PATIENTS: All adult (>or=18 years), mechanically ventilated patients with acute onset (within 48 hour) of acute lung injury/acute respiratory distress syndrome were included. INTERVENTIONS: Four sequential (in ascending order) treatment groups (cohorts) in single and multiple dosing strategies. Subjects were randomized in a 2:1 ratio to either FFR-rFVIIa or placebo within each cohort. MEASUREMENTS AND RESULTS: Data were collected daily for 7 days and on days 14 and 28 for efficacy variables including hematology and coagulation parameters, plasma d-dimer levels, plasma interleukin-6 levels, vital signs, ventilator parameters, lung injury score, and Sequential Organ Failure Assessment score. The study was discontinued prematurely by the Safety Committee based on statistical analysis of the mortality in cohort 3 (4 x 400 microg/kg), which suggested that, after adjusting for prognostic covariates, 28-day mortality was significantly higher in this cohort than in the placebo group and time to death was significantly shorter. A total of 214 patients (147 male) were included in the trial, mean age 59 years (range, 24-85 years). Overall, there were no significant differences in mortality rates in treated and placebo patients (36/144 deaths FFR-rFVIIa, 15/70 placebo). There was no treatment effect of FFR-rFVIIa on vital signs, blood chemistry parameters, hematology parameters, or amount of transfusion products required. There was a trend to increased bleeding with increasing FFR-rFVIIa dose. CONCLUSION: In this randomized double-blind, placebo- controlled, dose-escalation trial, FFR-rFVIIa had no beneficial effects on morbidity or outcome overall. The cohort of patients receiving 4 x 400 g/kg of FFR-rFVIIa had increased mortality rates compared with placebo-treated patients, and there was a trend to increased risk of serious bleeding with increasing doses.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Fator VIIa/administração & dosagem , Síndrome do Desconforto Respiratório/tratamento farmacológico , Lesão Pulmonar Aguda/complicações , Adulto , Idoso , Idoso de 80 Anos ou mais , Método Duplo-Cego , Fator VIIa/efeitos adversos , Fator VIIa/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Síndrome do Desconforto Respiratório/complicações , Adulto Jovem
12.
Thromb Haemost ; 101(5): 818-26, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19404533

RESUMO

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.


Assuntos
Antitrombina III/metabolismo , Fator VII/farmacocinética , Fator VIIa/farmacocinética , Soroglobulinas/metabolismo , alfa-Macroglobulinas/metabolismo , Animais , Fator VIIa/administração & dosagem , Heparina/sangue , Humanos , Injeções Intravenosas , Radioisótopos do Iodo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Soroglobulinas/deficiência , Soroglobulinas/genética , alfa-Macroglobulinas/deficiência , alfa-Macroglobulinas/genética
13.
J Surg Res ; 153(2): 239-45, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19062044

RESUMO

BACKGROUND: Expression of the principal initiator of coagulation, tissue factor (TF), by colorectal cancer (CRC) cells is involved in tumoral angiogenesis and metastasis progression, after binding of factor VIIa (FVIIa) to TF and generation of TF-FVIIa activity. We thus hypothesized that inhibition of the TF pathway by active site-blocked FVIIa (FFR-FVIIa) may prevent the development of hepatic metastasis in CRC. METHODS: Rat tumoral cells (DHDK12 proB cells) expressing high levels of TF were injected in the portal vein in syngenic BDIX rats. Rats received intraperitoneal injection of either FFR-FVIIa, from d 3 to d 7 (adjuvant treatment) (n = 19), or solvent buffer (n = 18) (control group). Additionally, cancer cells were infused subcutaneously in 20 other rats, which were assigned to FFR-FVIIa adjuvant treatment (n = 10), or buffer treatment (n = 10). Macroscopic and histological analysis was performed at d 14. RESULTS: In the control group, infusion of cancer cells resulted in development of macroscopic hepatic tumors in 17/18 rats. In the adjuvant FFR-FVIIa group, macroscopic hepatic tumors were visible on the liver surface in 3/19 rats (P = 0.002 versus control). All rats with subcutaneous injection of proB cells exhibited macroscopic tumors, with no significant difference between the control and the treated ones. CONCLUSION: Inhibition of the proteolytic activity of TF-FVIIa complex blunted hematogenous hepatic metastasis, suggesting that TF-FVIIa is a relevant target for the prevention of hepatic metastasis in CRC. TF-blocking agents should be investigated as adjuvant treatment in this setting.


Assuntos
Carcinoma/metabolismo , Neoplasias Colorretais/metabolismo , Fator VIIa/metabolismo , Neoplasias Hepáticas/metabolismo , Tromboplastina/metabolismo , Animais , Carcinoma/secundário , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Fator VIIa/antagonistas & inibidores , Neoplasias Hepáticas/secundário , Masculino , Metástase Neoplásica , Neoplasias Experimentais/metabolismo , Ratos , Tromboplastina/antagonistas & inibidores
14.
Br J Haematol ; 143(4): 559-69, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18950467

RESUMO

The haemostatic effect of recombinant activated factor VII (rFVIIa;NovoSeven) in thrombocytopenic patients has been a matter of controversy. Haemostasis by rFVIIa occurs via FVIIa-mediated thrombin generation in a platelet-dependent manner and may therefore be suboptimal in patients without functional platelets. Under such conditions, a clot-stabilizing agent, such as factor XIII (FXIII), may supplement the effect ofrFVIIa and improve haemostasis. Recombinant factor XIII (rFXIII-A2) is produced as an A2 homodimer of the FXIII A subunit and is equivalent to cellular FXIII normally found in platelets. The combined effects of rFVIIa andrFXIII-A2 were evaluated in clot lysis assays using factor XIII-deficient plasma and by whole blood thrombelastography (TEG) analysis from normal donors and thrombocytopenic stem cell transplantation patients. Clotting time was shortened by rFVIIa (0.6-10 microg/ml). rFVIIa only modestly improved anti-fibrinolysis,whereas rFXIII-A2 (0-20 microg/ml) enhanced anti-fibrinolysis without effect on clotting time. TEG analysis showed rFVIIa shortened the clotting time, and enhanced clot development, maximal mechanical strength and resistance to fibrinolysis, whereas, rFXIII-A2 enhanced clot development,maximal mechanical strength and markedly enhanced resistance to fibrinolysis. These data illustrate that rFVIIa and rFXIII-A2 contribute to clot formation and stability by different mechanisms suggesting enhanced haemostatic efficacy by combining these agents.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator VIIa/farmacologia , Fator XIIIa/farmacologia , Trombocitopenia/sangue , Contagem de Células Sanguíneas , Relação Dose-Resposta a Droga , Fibrinólise/efeitos dos fármacos , Humanos , Proteínas Recombinantes/farmacologia , Transplante de Células-Tronco , Tromboelastografia/métodos , Trombocitopenia/terapia
15.
J Nucl Med ; 57(7): 1112-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27013699

RESUMO

UNLABELLED: Tissue factor (TF) is the main initiator of the extrinsic coagulation cascade. However, TF also plays an important role in cancer. TF expression has been reported in 53%-89% of all pancreatic adenocarcinomas, and the expression level of TF has in clinical studies correlated with advanced stage, increased microvessel density, metastasis, and poor overall survival. Imaging of TF expression is of clinical relevance as a prognostic biomarker and as a companion diagnostic for TF-directed therapies currently under clinical development. Factor VII (FVII) is the natural ligand to TF. The purpose of this study was to investigate the possibility of using active site-inhibited FVII (FVIIai) labeled with (64)Cu for PET imaging of TF expression. METHODS: FVIIai was conjugated to 2-S-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA) and labeled with (64)Cu ((64)Cu-NOTA-FVIIai). Longitudinal in vivo PET imaging was performed at 1, 4, 15, and 36 h after injection of (64)Cu-NOTA-FVIIai in mice with pancreatic adenocarcinomas (BxPC-3). The specificity of TF imaging with (64)Cu-NOTA-FVIIai was investigated in subcutaneous pancreatic tumor models with different levels of TF expression and in a competition experiment. In addition, imaging of orthotopic pancreatic tumors was performed using (64)Cu-NOTA-FVIIai and PET/MRI. In vivo imaging data were supported by ex vivo biodistribution, flow cytometry, and immunohistochemistry. RESULTS: Longitudinal PET imaging with (64)Cu-NOTA-FVIIai showed a tumor uptake of 2.3 ± 0.2, 3.7 ± 0.3, 3.4 ± 0.3, and 2.4 ± 0.3 percentage injected dose per gram at 1, 4, 15, and 36 h after injection, respectively. An increase in tumor-to-normal-tissue contrast was observed over the imaging time course. Competition with unlabeled FVIIai significantly (P < 0.001) reduced the tumor uptake. The tumor uptake observed in models with different TF expression levels was significantly different from each other (P < 0.001) and was in agreement with the TF level evaluated by TF immunohistochemistry staining. Orthotopic tumors were clearly visible on the PET/MR images, and the uptake of (64)Cu-NOTA-FVIIai was colocalized with viable tumor tissue. CONCLUSION: (64)Cu-NOTA-FVIIai is well suited for PET imaging of tumor TF expression, and imaging is capable of distinguishing the TF expression level of various pancreatic tumor models.


Assuntos
Fator VII/metabolismo , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/metabolismo , Tromboplastina/metabolismo , Animais , Linhagem Celular Tumoral , Radioisótopos de Cobre , Humanos , Marcação por Isótopo , Imageamento por Ressonância Magnética , Camundongos , Imagem Multimodal , Transplante de Neoplasias , Tomografia por Emissão de Pósitrons , Radiometria , Compostos Radiofarmacêuticos , Distribuição Tecidual
16.
J Nucl Med ; 57(1): 89-95, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26383146

RESUMO

UNLABELLED: Tissue factor (TF) is upregulated in many solid tumors, and its expression is linked to tumor angiogenesis, invasion, metastasis, and prognosis. A noninvasive assessment of tumor TF expression status is therefore of obvious clinical relevance. Factor VII is the natural ligand to TF. Here we report the development of a new PET tracer for specific imaging of TF using an (18)F-labeled derivative of factor VII. METHODS: Active site-inhibited factor VIIa (FVIIai) was obtained by inactivation with phenylalanine-phenylalanine-arginine-chloromethyl ketone. FVIIai was radiolabeled with N-succinimidyl 4-(18)F-fluorobenzoate and purified. The corresponding product, (18)F-FVIIai, was injected into nude mice with subcutaneous human pancreatic xenograft tumors (BxPC-3) and investigated using small-animal PET/CT imaging 1, 2, and 4 h after injection. Ex vivo biodistribution was performed after the last imaging session, and tumor tissue was preserved for molecular analysis. A blocking experiment was performed in a second set of mice. The expression pattern of TF in the tumors was visualized by immunohistochemistry and the amount of TF in tumor homogenates was measured by enzyme-linked immunosorbent assay and correlated with the uptake of (18)F-FVIIai in the tumors measured in vivo by PET imaging. RESULTS: The PET images showed high uptake of (18)F-FVIIai in the tumor regions, with a mean uptake of 2.5 ± 0.3 percentage injected dose per gram (%ID/g) (mean ± SEM) 4 h after injection of 7.3-9.3 MBq of (18)F-FVIIai and with an average maximum uptake in the tumors of 7.1 ± 0.7 %ID/g at 4 h. In comparison, the muscle uptake was 0.2 ± 0.01 %ID/g at 4 h. At 4 h, the tumors had the highest uptake of any organ. Blocking with FVIIai significantly reduced the uptake of (18)F-FVIIai from 2.9 ± 0.1 to 1.4 ± 0.1 %ID/g (P < 0.001). The uptake of (18)F-FVIIai measured in vivo by PET imaging correlated (r = 0.72, P < 0.02) with TF protein level measured ex vivo. CONCLUSION: (18)F-FVIIai is a promising PET tracer for specific and noninvasive imaging of tumor TF expression. The tracer merits further development and clinical translation, with potential to become a companion diagnostics for emerging TF-targeted therapies.


Assuntos
Domínio Catalítico , Fator VII/metabolismo , Radioisótopos de Flúor , Regulação Neoplásica da Expressão Gênica , Tomografia por Emissão de Pósitrons/métodos , Tromboplastina/metabolismo , Animais , Ligação Competitiva , Linhagem Celular Tumoral , Feminino , Humanos , Ligantes , Camundongos , Radioquímica , Distribuição Tecidual , Tomografia Computadorizada por Raios X
17.
Thromb Haemost ; 93(1): 27-34, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15630487

RESUMO

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).


Assuntos
Quimiotaxia , Endotélio Vascular/citologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Tromboplastina/fisiologia , Animais , Aorta/citologia , Células Cultivadas , Citoplasma , Fator VIIa/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Suínos , Tromboplastina/química , Tromboplastina/genética , Transfecção
18.
Thromb Res ; 116(1): 75-85, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15850611

RESUMO

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.


Assuntos
Fator VIIa/síntese química , Fator VIIa/farmacologia , Tromboplastina/metabolismo , Animais , Sítios de Ligação , Clonagem Molecular , Fator VIIa/metabolismo , Hemorragia/tratamento farmacológico , Humanos , Camundongos , Modelos Animais , Proteínas Recombinantes/síntese química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Especificidade da Espécie
19.
Arterioscler Thromb Vasc Biol ; 22(9): 1421-6, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12231560

RESUMO

OBJECTIVE: The plasminogen system has been proposed to participate in vascular remodeling and angiogenesis. Although plasmin-mediated proteolysis could contribute these processes, proteolytic targets for plasmin and their downstream effector molecules are yet to be fully defined. The aim of the present study was to elucidate potential mechanisms by which plasmin affects various cellular processes. METHODS AND RESULTS: Plasmin upregulated the expression of Cyr61, a growth factor-like gene that has been implicated in cell proliferation, adhesion, and migration. Plasmin-induced gene expression is dependent on its proteolytic activity and requires its binding to cells. Studies that used wild-type fibroblasts and fibroblasts derived from PAR-1- and PAR-2-deficient mice showed that plasmin induced Cyr61 gene expression in wild-type fibroblasts and PAR-2-deficient cells but not in PAR-1-deficient cells. Consistent with this, plasmin induced the activation of p44/42 mitogen-activated protein kinase in wild-type, PAR-2 -/- cells but not in PAR-1 -/- cells. In contrast with thrombin, plasmin failed to induce Ca2+ signaling in fibroblasts. CONCLUSIONS: Plasmin induced an angiogenic and wound-healing promoter, Cyr61, in fibroblasts through activation of PAR-1. Plasmin-induced Cyr61 expression is mediated via the p44/42 mitogen-activated protein kinase pathway independent of Ca2+ signaling.


Assuntos
Proteínas de Caenorhabditis elegans , Cálcio/metabolismo , Fibrinolisina/fisiologia , Fibroblastos/química , Fibroblastos/metabolismo , Regulação da Expressão Gênica/genética , Substâncias de Crescimento/genética , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular , Sistema de Sinalização das MAP Quinases/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/genética , Receptores de Trombina/fisiologia , Animais , Butadienos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Linhagem Celular , Proteína Rica em Cisteína 61 , Citoplasma/química , Citoplasma/enzimologia , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Flavonoides/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Substâncias de Crescimento/fisiologia , Humanos , Proteínas Imediatamente Precoces/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Nitrilas/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Receptor PAR-1 , Receptor PAR-2 , Receptores de Trombina/antagonistas & inibidores , Receptores de Trombina/deficiência , Receptores de Trombina/genética , Trombina/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Cicatrização/genética , Cicatrização/fisiologia
20.
J Drug Target ; 23(3): 257-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25510254

RESUMO

We have developed a specific technique for imaging cancer in vivo using Cy5.5-labeled factor VIIa (fVIIa), clotting-deficient FFRck-fVIIa, paclitaxel-FFRck-fVIIa, and anti-tissue factor (TF) antibody. FVIIa is the natural ligand for TF. We took advantage of the fact that vascular endothelial cells (VECs) in cancer, but not normal tissue, aberrantly express TF due to its induction by vascular endothelial growth factor (VEGF). Under physiological conditions, TF is expressed by stromal cells and outer blood vessel layers (smooth muscle and adventitia), but not by VECs. We hypothesized that labeled fVIIa or anti-TF antibodies could be used to image the tumor vasculature in vivo. To test this, Cy5.5-labeled fVIIa, FFRck-fVIIa, paclitaxel-FFRck-fVIIa, and anti-TF antibody were developed and administered to athymic nude mice carrying xenografts including glioma U87EGFRviii, pancreatic cancer ASPC-1 and Mia PaCa-2, and squamous cell carcinoma KB-V1. Cy5.5 labeled with these targeting proteins specifically localized to the tumor xenografts for at least 14 days but unconjugated Cy5.5 did not localize to any xenografts or organs. This method of imaging TF in the tumor VECs may be useful in detecting primary tumors and metastases as well as monitoring in vivo therapeutic responses.


Assuntos
Carbocianinas/análise , Fator VIIa/análise , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Imagem Óptica/métodos , Tromboplastina/imunologia , Clorometilcetonas de Aminoácidos/química , Animais , Carbocianinas/química , Células Cultivadas , Fator VIIa/química , Xenoenxertos/imunologia , Humanos , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Paclitaxel/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA