Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Invest New Drugs ; 36(4): 536-544, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29188469

RESUMO

Purpose Approximately 3% of lung cancer bears mutations leading to MET exon 14 skipping, an oncogenic driver which is further evidenced by case reports of patient response to MET kinase inhibitor treatment. Approximately 15% of tumors harboring MET exon14 skipping have concurrent MET amplification. Experimental Design Merestinib is a type II MET kinase inhibitor. Emibetuzumab, a bivalent anti-MET antibody, internalizes MET receptor. Each single agent and the combination were evaluated in the Hs746t gastric cancer line bearing MET exon14 skipping and MET amplification. Results Merestinib inhibited Hs746t cell proliferation (IC50=34 nM) and totally eliminated pMET at 100nM. Emibetuzumab showed little anti-proliferative activity against Hs746t cells (IC50>100nM), did not reduce pMET, and slightly reduced cell surface MET. In the Hs746t xenograft model, dose dependent differences in durability of response were seen with merestinib including durable tumor regression (91.8%) at 12 mg/kg qd. Emibetuzumab treatment (10mg/kg qw) provided transient tumor regression (37.7%), but tumors re-grew while on treatment. Concurrent combination of merestinib (6 mg/kg qd) and emibetuzumab resulted in 85% tumor regression, while a sequential combination (initiating merestinib first) resulted in longer duration of treatment response. Conclusions Data in this study support a clinical evaluation of merestinib in patients with MET exon 14 skipping (NCT02920996). As a type II MET kinase inhibitor, merestinib may provide a therapeutic option to treatment naïve patients or to patients who progress on type I MET inhibitor treatment. Data also support clinical evaluation of the sequential combination of merestinib with emibetuzumab when patients progress on single agent merestinib.


Assuntos
Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos/farmacologia , Éxons/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo
2.
Invest New Drugs ; 35(3): 334-344, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28299514

RESUMO

Background Circulating tumor cells (CTCs) and chemokine (C-X-C motif) receptor 4 (CXCR4) expression in CTCs and tumor tissue were evaluated as prognostic or predictive markers of CXCR4 peptide antagonist LY2510924 plus carboplatin-etoposide (CE) versus CE in extensive-stage disease small cell lung cancer (ED-SCLC). Methods This exploratory analysis of a phase II study evaluated CXCR4 expression in baseline tumor tissue and peripheral blood CTCs and in post-treatment CTCs. Optimum cutoff values were determined for CTC counts and CXCR4 expression in tumors and CTCs as predictors of survival outcome. Kaplan-Meier estimates and hazard ratios were used to determine biomarker prognostic and predictive values. Results There was weak positive correlation at baseline between CXCR4 expression in tumor tissue and CTCs. Optimum cutoff values were H-score ≥ 210 for CXCR4+ tumor, ≥7% CTCs with CXCR4 expression (CXCR4+ CTCs), and ≥6 CTCs/7.5 mL blood. Baseline H-score for CXCR4+ tumor was not prognostic of progression-free survival (PFS) or overall survival (OS). Baseline CXCR4+ CTCs ≥7% was prognostic of shorter PFS. CTCs ≥6 at baseline and cycle 2, day 1 were prognostic of shorter PFS and OS. None of the biomarkers at their respective optimum cutoffs was predictive of treatment response of LY2510924 plus CE versus CE. Conclusions In patients with ED-SCLC, baseline CXCR4 expression in tumor tissue was not prognostic of survival or predictive of LY2510924 treatment response. Baseline CXCR4+ CTCs ≥7% was prognostic of shorter PFS. CTC count ≥6 at baseline and after 1 cycle of treatment were prognostic of shorter PFS and OS.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Biomarcadores Tumorais/sangue , Carboplatina/farmacologia , Etoposídeo/farmacologia , Células Neoplásicas Circulantes , Peptídeos Cíclicos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carboplatina/uso terapêutico , Contagem de Células , Intervalo Livre de Doença , Etoposídeo/uso terapêutico , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Peptídeos Cíclicos/uso terapêutico , Prognóstico , Receptores CXCR4/metabolismo , Carcinoma de Pequenas Células do Pulmão/sangue , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/metabolismo
3.
Invest New Drugs ; 31(4): 833-44, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23275061

RESUMO

The HGF/MET signaling pathway regulates a wide variety of normal cellular functions that can be subverted to support neoplasia, including cell proliferation, survival, apoptosis, scattering and motility, invasion, and angiogenesis. MET over-expression (with or without gene amplification), aberrant autocrine or paracrine ligand production, and missense MET mutations are mechanisms that lead to activation of the MET pathway in tumors and are associated with poor prognostic outcome. We report here preclinical development of a potent, orally bioavailable, small-molecule inhibitor LY2801653 targeting MET kinase. LY2801653 is a type-II ATP competitive, slow-off inhibitor of MET tyrosine kinase with a dissociation constant (Ki) of 2 nM, a pharmacodynamic residence time (Koff) of 0.00132 min(-1) and t1/2 of 525 min. LY2801653 demonstrated in vitro effects on MET pathway-dependent cell scattering and cell proliferation; in vivo anti-tumor effects in MET amplified (MKN45), MET autocrine (U-87MG, and KP4) and MET over-expressed (H441) xenograft models; and in vivo vessel normalization effects. LY2801653 also maintained potency against 13 MET variants, each bearing a single-point mutation. In subsequent nonclinical characterization, LY2801653 was found to have potent activity against several other receptor tyrosine oncokinases including MST1R, FLT3, AXL, MERTK, TEK, ROS1, DDR1/2 and against the serine/threonine kinases MKNK1/2. The potential value of MET and other inhibited targets within a number of malignancies (such as colon, bile ducts, and lung) is discussed. LY2801653 is currently in phase 1 clinical testing in patients with advanced cancer (trial I3O-MC-JSBA, NCT01285037).


Assuntos
Indazóis/farmacologia , Niacinamida/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Tetrazóis/farmacologia , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacologia , Disponibilidade Biológica , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Indazóis/administração & dosagem , Indazóis/química , Camundongos , Mutação/genética , Niacinamida/administração & dosagem , Niacinamida/química , Niacinamida/farmacologia , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Tetrazóis/administração & dosagem , Tetrazóis/química , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Am Coll Cardiol ; 46(1): 21-8, 2005 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15992630

RESUMO

OBJECTIVES: We investigated coagulation abnormalities in out-of-hospital cardiac arrest (OHCA) patients, with special attention to the protein C anticoagulant pathway. BACKGROUND: Successfully resuscitated cardiac arrest is followed by a systemic inflammatory response and by activation of coagulation, both of which may contribute to organ failure and neurological dysfunction. METHODS: Coagulation parameters were measured in all patients admitted after successfully resuscitated OHCA. RESULTS: At admission, 67 patients had a systemic inflammatory response with increased interleukin-6 and coagulation activity (thrombin-antithrombin complex), reduced anticoagulation (antithrombin, protein C, and protein S), activated fibrinolysis (plasmin-antiplasmin complex), and, in some cases, inhibited fibrinolysis (increased plasminogen activator inhibitor-1 with a peak on day 1). These abnormalities were more severe in patients who died within two days (50 of 67, 75%) and were most severe in patients dying from early refractory shock. Protein C and S levels were low compared to healthy volunteers and discriminated OHCA survivors from nonsurvivors. Furthermore, a subgroup of patients had a transient increase in plasma-activated protein C at admission followed by undetectable levels. This, along with an increase in soluble thrombomodulin over time, suggests secondary endothelial injury and dysfunction of the protein C anticoagulant pathway similar to that observed in severe sepsis. CONCLUSIONS: Major coagulation abnormalities were found after successful resuscitation of cardiac arrest. These abnormalities are consistent with secondary down-regulation of the thrombomodulin-endothelial protein C receptor pathway.


Assuntos
Reanimação Cardiopulmonar , Parada Cardíaca/sangue , Parada Cardíaca/terapia , Proteína C/metabolismo , Idoso , Antitrombinas/metabolismo , Fatores de Coagulação Sanguínea/metabolismo , Estudos de Casos e Controles , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Tempo de Tromboplastina Parcial , Proteína S/metabolismo , Tempo de Trombina , Trombomodulina/sangue
5.
Crit Care ; 9 Suppl 4: S38-45, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16168074

RESUMO

It has been hypothesized that the protein C pathway is a pivotal link between the inflammation and coagulation cascades. The demonstration that a survival benefit is associated with administration of drotrecogin alfa (activated) (recombinant human activated protein C [APC]) in severe sepsis patients has provided new insights into the protein C pathway. APC was originally identified based on its antithrombotic properties, which result from the inhibition of activated Factors V and VIII. In the early 1990s, any potential anti-inflammatory properties of APC were thought to relate primarily to its inhibition of thrombin generation. However, the mid-1990s saw the identification of the endothelial protein C receptor (EPCR), which has subsequently been shown to be neither endothelial specific nor protein C specific, but has a primary function as a cofactor for enhancing the generation of APC or behaving as an APC receptor. Thus, the potential biologic activities of APC can be classed into two categories related either to the limiting of thrombin generation or to cellular effects initiated by binding to the EPCR. Intracellular signaling initiated by binding of APC to its receptor appears to be mediated by interaction with an adjacent protease-activated receptor (PAR), or by indirect activation of the sphingosine 1-phosphate pathway. Based mostly on in vitro studies, binding of APC to its receptor on endothelial cells leads to a decrease in thrombin-induced endothelial permeability injury, while such binding on blood cells, epithelial cells, and neurons has been shown to inhibit chemotaxis, be anti-apoptotic, and be neuroprotective, respectively. In the Recombinant Human Activated Protein C Worldwide Evaluation in Severe Sepsis (PROWESS) study, drotrecogin alfa (activated) was associated with improved cardiovascular function, respiratory function, and a prevention of hematologic dysfunction. This article discusses the way in which the interactions of APC may alter the microcirculation.


Assuntos
Fibrinolíticos/uso terapêutico , Deficiência de Proteína C/tratamento farmacológico , Proteína C/fisiologia , Sepse/tratamento farmacológico , Animais , Antígenos/fisiologia , Antígenos CD , Fatores de Coagulação Sanguínea/fisiologia , Receptor de Proteína C Endotelial , Glicoproteínas/fisiologia , Humanos , Proteína C/uso terapêutico , Deficiência de Proteína C/metabolismo , Deficiência de Proteína C/fisiopatologia , Receptores de Superfície Celular/fisiologia , Proteínas Recombinantes/uso terapêutico , Sepse/fisiopatologia , Trombomodulina/fisiologia
6.
Crit Care ; 9(4): R331-43, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16137345

RESUMO

INTRODUCTION: We report data from adult and pediatric patients with severe sepsis from studies evaluating drotrecogin alfa (activated) (DrotAA) and presenting with purpura fulminans (PF), meningitis (MEN), or meningococcal disease (MD) (PF/MEN/MD). Such conditions may be associated with an increased bleeding risk but occur in a relatively small proportion of patients presenting with severe sepsis; pooling data across clinical trials provides an opportunity for improving the characterization of outcomes. METHODS: A retrospective analysis of placebo-controlled, open-label, and compassionate-use trials was conducted. Adult patients received infusions of either DrotAA or placebo. All pediatric patients (<18 years old) received DrotAA. 189 adult and 121 pediatric patients presented with PF/MEN/MD. RESULTS: Fewer adult patients with PF/MEN/MD met cardiovascular (68.3% versus 78.8%) or respiratory (57.8% versus 80.5%) organ dysfunction entry criteria than those without. DrotAA-treated adult patients with PF/MEN/MD (n = 163) had an observed 28-day mortality rate of 19.0%, a 28-day serious bleeding event (SBE) rate of 6.1%, and an intracranial hemorrhage (ICH) rate of 4.3%. Six of the seven ICHs occurred in patients with MEN (three of whom were more than 65 years old with a history of hypertension). DrotAA-treated adult patients without PF/MEN/MD (n = 3,088) had an observed 28-day mortality rate of 25.5%, a 28-day SBE rate of 5.8%, and an ICH rate of 1.0%. In contrast, a greater number of pediatric patients with PF/MEN/MD met the cardiovascular organ dysfunction entry criterion (93.5% versus 82.5%) than those without. DrotAA-treated PF/MEN/MD pediatric patients (n = 119) had a 14-day mortality rate of 10.1%, an SBE rate of 5.9%, and an ICH rate of 2.5%. DrotAA-treated pediatric patients without PF/MEN/MD (n = 142) had a 14-day mortality rate of 14.1%, an SBE rate of 9.2%, and an ICH rate of 3.5%. CONCLUSION: DrotAA-treated adult patients with severe sepsis presenting with PF/MEN/MD had a similar SBE rate, a lower observed 28-day mortality rate, and a higher observed rate of ICH than DrotAA-treated patients without PF/MEN/MD. DrotAA-treated pediatric patients with severe sepsis with PF/MEN/MD may differ from adults, because all three outcome rates (SBE, mortality, and ICH) were lower in pediatric patients with PF/MEN/MD.


Assuntos
Anti-Infecciosos/uso terapêutico , Vasculite por IgA/epidemiologia , Meningites Bacterianas/epidemiologia , Infecções Meningocócicas/epidemiologia , Proteína C/uso terapêutico , Sepse/tratamento farmacológico , Sepse/epidemiologia , Adulto , Idoso , Criança , Pré-Escolar , Comorbidade , Feminino , Hemorragia/epidemiologia , Humanos , Hemorragias Intracranianas/epidemiologia , Masculino , Pessoa de Meia-Idade , Razão de Chances , Proteínas Recombinantes/uso terapêutico , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento
7.
Clin Infect Dis ; 37(1): 50-8, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12830408

RESUMO

Clinical trials with novel therapeutic agents for severe sepsis have suggested that patients might respond differently depending on causative microorganism. Data from a large, placebo-controlled trial of recombinant human drotrecogin alfa (activated) (DrotAA) were analyzed by type of causative microorganism for treatment-associated differences in mortality, coagulopathy, and inflammatory response. Compared with placebo, mortality rates associated with DrotAA were consistently reduced for each microorganism group (gram-positive bacteria, gram-negative bacteria, mixed bacteria, fungi, other, and unknown microbial etiology), with a stratified relative risk (RR) of 0.80 (95% confidence interval [CI], 0.69-0.94). The greatest reduction in the mortality rate was for Streptococcus pneumoniae infection (RR, 0.56; 95% CI, 0.35-0.88). Levels of coagulation and inflammation biomarkers varied with different pathogens at study entry. Results demonstrate that DrotAA, administered as an adjunct to standard anti-infective therapy, can improve the rate of survival for patients who develop severe sepsis regardless of causative microorganism.


Assuntos
Anti-Infecciosos/uso terapêutico , Proteína C/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Sepse/tratamento farmacológico , Antibacterianos , Bactérias/classificação , Biomarcadores , Coagulação Sanguínea/efeitos dos fármacos , Método Duplo-Cego , Fungos/classificação , Humanos , Inflamação/etiologia , Estudos Prospectivos , Proteína C/metabolismo , Sepse/microbiologia , Sepse/mortalidade , Análise de Sobrevida
8.
Thromb Haemost ; 90(4): 642-53, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14515185

RESUMO

Drotrecogin alfa (activated) improved survival in patients with severe sepsis in PROWESS, a double-blind, study of 1690 adult patients randomized to drotrecogin alfa (activated) at 24 microg/kg/h (N=850) or placebo (N=840) infused for 96 hours. Pharmacodynamic effects of drotrecogin alfa (activated) were assessed with 15 prospectively defined systemic biomarkers of hemostasis, inflammation and endothelial injury. The last-observation-carried-forward (LOCF) method of imputation for missing observations was the prospectively defined statistical method. The results were also analyzed with only the observed values without imputation for missing data (repeated measures analysis). With both statistical methods, drotrecogin alfa (activated)-treated patients demonstrated antithrombotic (reduced markers of thrombin generation and accelerated normalization of anticoagulant factor, protein C and fibrinolytic factors) and anticoagulant (prolonged PT and APTT) effects compared with placebo. A profibrinolytic (reduction in plasminogen activator inhibitor-1) effect was significant only with the LOCF imputation method in observed case and percent change from baseline analyses. An anti-inflammatory (reduction in interleukin-6) effect was significant only with the LOCF imputation method in change from baseline and percent change from baseline analyses. Drotrecogin alfa (activated) is a new and promising agent for treatment of patients with severe sepsis. The extensive analysis of systemic biomarkers confirms the previously published antithrombotic effects. However, the present results using different statistical methods do not provide a strong basis for systemic anti-inflammatory or pro-fibrinolytic effects. These latter two effects may occur at the local or cellular level. The systemic biomarkers reported here might not be the most appropriate approach to demonstrate these potential effects of drotrecogin alfa (activated).


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Proteína C/farmacologia , Proteínas Recombinantes/farmacologia , Sepse/tratamento farmacológico , Anti-Inflamatórios não Esteroides/farmacologia , Biomarcadores/sangue , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Fibrinólise , Fibrinolíticos/farmacologia , Humanos , Inflamação/sangue , Cinética , Inibidor 1 de Ativador de Plasminogênio/sangue , Sepse/sangue , Sepse/mortalidade , Trombose/sangue
9.
Crit Care ; 8(2): R82-90, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15025782

RESUMO

INTRODUCTION: PROWESS (Recombinant Human Activated Protein C Worldwide Evaluation in Severe Sepsis) was a phase III, randomized, double blind, placebo controlled, multicenter trial conducted in patients with severe sepsis from 164 medical centers. Here we report data collected at study entry for 1690 patients and over the following 7 days for the 840 patients who received placebo (in addition to usual standard of care). METHODS: Nineteen biomarkers of coagulation activation, anticoagulation, fibrinolysis, endothelial injury, and inflammation were analyzed to determine the relationships between baseline values and their change over time, with 28-day survival, and type of infecting causative micro-organism. RESULTS: Levels of 13 of the 19 biomarkers at baseline correlated with Acute Physiology and Chronic Health Evaluation II scores, and nearly all patients exhibited coagulopathy, endothelial injury, and inflammation at baseline. At study entry, elevated D-dimer, thrombin-antithrombin complexes, IL-6, and prolonged prothrombin time were present in 99.7%, 95.5%, 98.5%, and 93.4% of patients, respectively. Markers of endothelial injury (soluble thrombomodulin) and deficient protein C, protein S, and antithrombin were apparent in 72%, 87.6%, 77.8%, and 81.7%, respectively. Impaired fibrinolysis (elevated plasminogen activator inhibitor-1) was observed in 44% of patients. During the first 7 days, increased prothrombin time (which is readily measurable in most clinical settings) was highly evident among patients who were not alive at 28 days. CONCLUSION: Abnormalities in biomarkers of inflammation and coagulation were related to disease severity and mortality outcome in patients with severe sepsis. Coagulopathy and inflammation were universal host responses to infection in patients with severe sepsis, which were similar across causative micro-organism groups.


Assuntos
Biomarcadores/análise , Sepse/diagnóstico , Infecção da Ferida Cirúrgica/diagnóstico , APACHE , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/diagnóstico , Proteína C-Reativa/análise , Coagulação Intravascular Disseminada/sangue , Coagulação Intravascular Disseminada/diagnóstico , Endotélio/lesões , Fibrina/análise , Fibrinólise , Humanos , Inflamação/sangue , Tempo de Tromboplastina Parcial , Placebos , Prognóstico , Proteína C/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Sensibilidade e Especificidade , Sepse/tratamento farmacológico , Sepse/mortalidade , Índice de Gravidade de Doença , Procedimentos Cirúrgicos Operatórios/efeitos adversos , Infecção da Ferida Cirúrgica/tratamento farmacológico , Infecção da Ferida Cirúrgica/mortalidade
10.
Clin Cancer Res ; 20(23): 6059-70, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25231402

RESUMO

PURPOSE: MET, the receptor for hepatocyte growth factor (HGF), has been implicated in driving tumor proliferation and metastasis. High MET expression is correlated with poor prognosis in multiple cancers. Activation of MET can be induced either by HGF-independent mechanisms such as gene amplification, specific genetic mutations, and transcriptional upregulation or by HGF-dependent autocrine or paracrine mechanisms. EXPERIMENTAL DESIGN/RESULTS: Here, we report on LY2875358, a novel humanized bivalent anti-MET antibody that has high neutralization and internalization activities, resulting in inhibition of both HGF-dependent and HGF-independent MET pathway activation and tumor growth. In contrast to other bivalent MET antibodies, LY2875358 exhibits no functional agonist activity and does not stimulate biologic activities such as cell proliferation, scattering, invasion, tubulogenesis, or apoptosis protection in various HGF-responsive cells and no evidence of inducing proliferation in vivo in a monkey toxicity study. LY2875358 blocks HGF binding to MET and HGF-induced MET phosphorylation and cell proliferation. In contrast to the humanized one-armed 5D5 anti-MET antibody, LY2875358 induces internalization and degradation of MET that inhibits cell proliferation and tumor growth in models where MET is constitutively activated. Moreover, LY2875358 has potent antitumor activity in both HGF-dependent and HGF-independent (MET-amplified) xenograft tumor models. Together, these findings indicate that the mechanism of action of LY2875358 is different from that of the one-armed MET antibody. CONCLUSIONS: LY2875358 may provide a promising therapeutic strategy for patients whose tumors are driven by both HGF-dependent and HGF-independent MET activation. LY2875358 is currently being investigated in multiple clinical studies.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fosforilação , Transporte Proteico , Proteólise , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Clin Cancer Res ; 19(20): 5699-710, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23989980

RESUMO

PURPOSE: Lung cancer is the leading cause of cancer-related death worldwide. Sustained activation, overexpression, or mutation of the MET pathway is associated with a poor prognosis in a variety of tumors, including non-small cell lung cancer (NSCLC), implicating the MET pathway as a potential therapeutic target for lung cancer. Previously, we reported on the development of LY2801653: a novel, orally bioavailable oncokinase inhibitor with MET as one of its targets. Here, we discuss the evaluation of LY2801653 in both preclinical in vitro and in vivo NSCLC models. Experimental Design/ RESULTS: Treatment with LY2801653 showed tumor growth inhibition in tumor cell lines and patient-derived tumor xenograft models as a single agent (37.4%-90.0% inhibition) or when used in combination with cisplatin, gemcitabine, or erlotinib (66.5%-86.3% inhibition). Mechanistic studies showed that treatment with LY2801653 inhibited the constitutive activation of MET pathway signaling and resulted in inhibition of NCI-H441 cell proliferation, anchorage-independent growth, migration, and invasion. These in vitro findings were confirmed in the H441 orthotopic model where LY2801653 treatment significantly inhibited both primary tumor growth (87.9% inhibition) and metastasis (64.5% inhibition of lymph node and 67.7% inhibition of chest wall). Tumor-bearing animals treated with LY2801653 had a significantly greater survival time (87% increase compared with the vehicle-treated mice). In the MET-independent NCI-H1299 orthotopic model, treatment with LY2801653 showed a significant inhibition of primary tumor growth but not metastasis. CONCLUSIONS: Collectively, these results support clinical evaluation of LY2801653 in NSCLCs and suggest that differences in the MET activation of tumors may be predictive of response.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Indazóis/farmacologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Niacinamida/análogos & derivados , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Expressão Gênica , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Indazóis/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Camundongos , Metástase Neoplásica , Niacinamida/administração & dosagem , Niacinamida/farmacologia , Proteínas Oncogênicas/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Crit Care ; 25(2): 270-5, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20149590

RESUMO

PURPOSE: The purpose of this retrospective study was to evaluate cardiac troponin-I (cTnI) as a 28-day mortality prognosticator and predictor for a drotrecogin alfa (activated) (DrotAA) survival benefit in recombinant human activated Protein C Worldwide Evaluation in Severe Sepsis patients. METHODS: Cardiac troponin-I was measured using the Access AccuTnI Troponin I assay (Beckman Coulter, Fullerton, CA). There were 598 patients (305 DrotAA, 293 placebo) with baseline cTnI data (cTnI negative [<0.06 ng/mL], n = 147; cTnI positive [>or=0.06 ng/mL], n = 451). RESULTS: Cardiac troponin-I-positive patients were older (mean age, 61 vs 56 years; P = .002), were sicker (mean Acute Physiology and Chronic Health Evaluation II, 26.1 vs 22.3; P < .001), had lower baseline protein C levels (mean level, 49% vs 56%; P = .017), and had higher 28-day mortality (32% vs 14%, P < .0001) than cTnI-negative patients. Elevated cTnI was an independent prognosticator of mortality (odds ratio, 2.020; 95% confidence interval, 1.153-3.541) after adjusting for other significant variables. Breslow-Day interaction test between cTnI levels and treatment was not significant (P = .65). CONCLUSION: This is the largest severe sepsis study reporting an association between elevated cTnI and higher mortality. Cardiac troponin-I elevation was not predictive of a survival benefit with DrotAA treatment.


Assuntos
Anti-Infecciosos/uso terapêutico , Miocárdio/metabolismo , Proteína C/uso terapêutico , Sepse/mortalidade , Troponina I/sangue , APACHE , Fatores Etários , Biomarcadores/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Ensaios Clínicos Controlados Aleatórios como Assunto , Proteínas Recombinantes/uso terapêutico , Estudos Retrospectivos , Sepse/sangue , Sepse/tratamento farmacológico , Índice de Gravidade de Doença
13.
J Antimicrob Chemother ; 59(6): 1177-81, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17428810

RESUMO

OBJECTIVES: To determine the effects of recombinant human activated protein C (rhAPC) on the antimicrobial activity and cytokine production of normal human monocyte-derived macrophages (MDMs) in the presence and absence of Escherichia coli infection, with and without treatment with levofloxacin or ampicillin. METHODS: MDM monolayers were infected with E. coli ATCC 25922 and treated with levofloxacin or ampicillin in the presence or absence of rhAPC. Antimicrobial activity and cytokine (TNF-alpha, IL-1beta, IL-6 and IL-8) concentrations in the supernatants were measured. RESULTS: When low concentrations of levofloxacin were used, a therapeutic concentration of rhAPC enhanced intracellular antibacterial activity at all time points. With ampicillin, antibacterial activity increased, was unaffected or diminished depending upon the drug concentration and assay time. Without antibiotics, rhAPC had no antibacterial effect. E. coli caused cytokine production to increase many fold. This increase was significantly greater with antibiotics (P < 0.01). Without antibiotics, rhAPC decreased production of TNF-alpha, IL-1beta and IL-6, but not IL-8. At high levofloxacin concentrations, rhAPC was associated with further increases in the concentrations of these cytokines. Cytokine concentrations at 24 h were unaffected by rhAPC in the presence of ampicillin and E. coli. CONCLUSIONS: rhAPC can affect the bactericidal activity and cytokine production of human MDM in the presence of infection and antibiotic therapy. Importantly, factors such as type and concentration of antibiotics, presence of bacteria and timing must be taken into consideration when evaluating cytokine data from septic patients.


Assuntos
Antibacterianos/farmacologia , Atividade Bactericida do Sangue/efeitos dos fármacos , Citocinas/fisiologia , Monócitos/efeitos dos fármacos , Proteína C/farmacologia , Ampicilina/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/imunologia , Humanos , Técnicas In Vitro , Inflamação/fisiopatologia , Interleucina-1beta/farmacologia , Interleucina-8/farmacologia , Cinética , Levofloxacino , Ofloxacino/farmacologia , Proteínas Recombinantes/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
14.
J Infect Dis ; 196 Suppl 2: S390-9, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17940975

RESUMO

BACKGROUND: Infection of primates with Zaire ebolavirus (ZEBOV) leads to hypotension, coagulation disorders, and an impaired immune response and, in many ways, resembles severe sepsis. Rapid decreases in plasma levels of protein C are a prominent feature of severe sepsis and ZEBOV hemorrhagic fever (ZHF). Currently, recombinant human activated protein C (rhAPC [Xigris; Eli Lilly]) is licensed for treating human patients with severe sepsis who are at high risk of death. The aim of this study was to test the efficacy of rhAPC as a potential treatment for ZHF. METHODS: Fourteen rhesus macaques were challenged with a uniformly lethal dose of ZEBOV; 11 of these monkeys were treated by intravenous infusion with rhAPC beginning 30-60 min after challenge and continuing for 7 days. Three control monkeys received sterile saline in parallel. RESULTS: All 3 control monkeys died on day 8, whereas 2 of the 11 rhAPC-treated monkeys survived. The mean time to death for the rhAPC-treated monkeys that did not survive ZEBOV challenge was 12.6 days. The difference in survival was significant when the rhAPC-treated monkeys were compared with historical controls. CONCLUSIONS: The experimental findings provide evidence that ZHF and severe sepsis share underlying mechanisms and may respond to the same therapies.


Assuntos
Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/tratamento farmacológico , Proteína C/uso terapêutico , Animais , Cateterismo Venoso Central , Modelos Animais de Doenças , Macaca mulatta , Oligopeptídeos/uso terapêutico , Primatas , Proteína C/administração & dosagem , Proteína C/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico
15.
Crit Care Med ; 32(5 Suppl): S239-46, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15118525

RESUMO

OBJECTIVE: Coagulation activation is part of the acute innate host response to infection that, when uncontrolled, may contribute to organ dysfunction and death. Activated protein C limits excessive coagulation activation by inactivating factors Va and VIIIa. The factor V Leiden mutation (R506Q), a prothrombotic gene polymorphism, disrupts the activity of this natural anticoagulant by rendering factor Va partially resistant to inactivation by activated protein C. Previous findings in the mouse factor V Leiden endotoxemia model and in patients with severe sepsis suggest that factor V Leiden constitutes a rare example of a balanced gene polymorphism that may provide a survival advantage for heterozygous carriers with severe sepsis. We sought to confirm that carriers of this prothrombotic factor V Leiden mutation do not have an increased risk of developing severe sepsis and that carriers with severe sepsis derive similar treatment benefit from recombinant human activated protein C (drotrecogin alfa [activated]) as non-factor V Leiden carriers. DESIGN: Prospective collection of factor V Leiden status from two clinical studies of severe sepsis (PROWESS and ENHANCE). SETTING: : A total of 447 clinical sites across 25 countries. PATIENTS: A total of 3894 adult patients with severe sepsis. INTERVENTION: Either 24 microg x kg x hr drotrecogin alfa (activated) (n = 3063) or placebo (n = 800) for 96 hrs or no exposure to the study drug (n = 31). MAIN RESULTS: The effect of the factor V Leiden carrier status in severe sepsis in the PROWESS study has been previously reported. The combined data on factor V Leiden status from 3894 adult patients with severe sepsis from the PROWESS and ENHANCE (a single-arm, open-label study of drotrecogin alfa [activated]) studies are reported here. At study entry, 3.9% of patients (150/3894) presenting with severe sepsis were heterozygous carriers. No homozygous factor V Leiden carriers were identified. The proportion of factor V Leiden carriers in patients with severe sepsis differs slightly from that predicted (allelic frequency of 2.5%) by the Hardy-Weinberg equation for the general white population (p =.05). There was no significant difference in baseline disease severity (Acute Physiology and Chronic Health Evaluation II score or number of organ dysfunctions) between heterozygous carriers and non-Leiden carriers. There was no significant difference in serious bleeding or thrombotic event rates with drotrecogin alfa (activated) treatment between heterozygous carriers and non-Leiden carriers. The 28-day mortality rates for heterozygous carriers and non-Leiden carriers with drotrecogin alfa (activated) treatment were 20.3% and 24.9%, respectively (risk ratio, 0.82; 95% confidence interval, 0.57-1.17). CONCLUSIONS: : Compared with non-Leiden carriers, factor V Leiden heterozygous carriers may have a slightly decreased risk of developing severe sepsis from infection, do not seem to have increased mortality in severe sepsis, and derive similar benefit and risk profiles from drotrecogin alfa (activated) treatment. Therefore, factor V Leiden carriers should not be excluded from this new sepsis therapy.


Assuntos
Anti-Infecciosos/uso terapêutico , Fator V/genética , Polimorfismo Genético , Proteína C/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Sepse/tratamento farmacológico , Sepse/genética , Ensaios Clínicos como Assunto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Índice de Gravidade de Doença
16.
Crit Care Med ; 32(5 Suppl): S194-201, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15118517

RESUMO

OBJECTIVE: To review the results from clinical trials of treatments for severe sepsis involving the protein C/activated protein C pathway. DATA SOURCE: Published research and review articles (PubMed, from 1985 to 2003) relating to clinical trials of compounds involving the protein C pathway. DATA EXTRACTION AND SYNTHESIS: Protein C is converted to activated protein C when thrombin complexes with thrombomodulin. Sepsis is associated with rapid depletion of protein C and blunted endogenous protein C activation. Treatment with protein C concentrate is followed by increased activated protein C plasma levels and a dose-dependent decrease in d-dimer levels in children with purpura fulminans. This supplementation is safe. A phase III trial of recombinant human activated protein C (drotrecogin alfa [activated]) in severe sepsis demonstrated a 6.1% absolute reduction in 28-day mortality compared with placebo. The short- and long-term survival rates associated with drotrecogin alfa (activated) were better in patients at high risk of death associated with a better cost/effectiveness ratio. Treatment with drotrecogin alfa (activated) was associated with an increased risk of serious bleeding compared with placebo during the 28-day study period (3.5% vs. 2.0%). CONCLUSIONS: Treatment with protein C concentrate is followed by an improvement of the coagulopathy and is safe in children with purpura fulminans; however, a large trial involving a high dose is required to determine its effect on mortality and morbidity. Treatment with drotrecogin alfa (activated) leads to substantial reduction in mortality and has an acceptable risk/benefit ratio in septic patients at high risk of death.


Assuntos
Proteína C/uso terapêutico , Sepse/tratamento farmacológico , Ensaios Clínicos como Assunto , Humanos , Proteína C/fisiologia , Proteínas Recombinantes/uso terapêutico , Sepse/imunologia , Índice de Gravidade de Doença
17.
Crit Care Med ; 30(5 Suppl): S318-24, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12004254

RESUMO

OBJECTIVE: To review the physiologic and biochemical mechanisms and the rationale for the use of soluble thrombomodulin, plasma-derived protein C, and recombinant human activated protein C in sepsis. DATA SOURCES AND STUDY SELECTION: Research and review articles related to the protein C pathway published in English from 1960 to present. DATA EXTRACTION AND SYNTHESIS: The protein C anticoagulant pathway plays a major role in controlling microvascular coagulation and inflammation. Protein C is the zymogen of the vitamin K-dependent serine protease activated protein C. Protein C is converted to activated protein C when thrombin complexes with thrombomodulin, an endothelial surface transmembrane glycoprotein. Activated protein C inactivates factors Va and VIIIa and effectively limits further thrombin generation. This protein also enhances endogenous fibrinolytic activity and modulates the inflammatory response. A rapid depletion of protein C occurs in sepsis, which contributes to sepsis-induced coagulopathy and correlates with a poor prognosis. The decrease in tissue levels of thrombomodulin in patients with meningococcemia suggests that the ability to convert protein C to activated protein C may also be compromised. The ability of soluble thrombomodulin to block fibrinogen clotting and cell activation, to activate protein C, and to promote thrombin inhibition in different animal models suggests that soluble thrombomodulin could be a useful therapeutic agent in sepsis. However, soluble thrombomodulin is less effective in blocking fibrinogen and platelet activation and in promoting thrombin inhibition than endothelial surface membrane-bound thrombomodulin. Only activated protein C, and not protein C, has clearly shown a reduction in mortality in experimental animal models of sepsis and in humans. CONCLUSIONS: The multipotent pharmacodynamic effects (antithrombotic, profibrinolytic, and anti-inflammatory) of activated protein C may explain why recombinantly derived human activated protein C is the first experimental agent to demonstrate a significant survival benefit in patients with severe sepsis.


Assuntos
Coagulação Intravascular Disseminada/fisiopatologia , Fibrinolíticos/uso terapêutico , Proteína C/fisiologia , Proteína C/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Sepse , Trombomodulina/fisiologia , Coagulação Intravascular Disseminada/metabolismo , Humanos , Proteína C/metabolismo , Sepse/tratamento farmacológico , Sepse/metabolismo , Sepse/fisiopatologia , Trombomodulina/metabolismo
18.
Crit Care Med ; 32(5 Suppl): S302-8, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15118535

RESUMO

OBJECTIVE: To explore whether the improvement in organ function and the vasoactive effect observed in the clinical studies of drotrecogin alfa (activated) (recombinant human activated protein C, rhAPC) in sepsis are a result of rhAPC's effect on endothelial cell (EC) permeability and modulation of the intracellular cytoskeleton via the Rho kinase signaling pathway. DESIGN: Findings regarding dose and duration of exposure to the drug with sequential addition of rhAPC and mediators (thrombin, histamine, interleukin-1 beta). SETTING: Research laboratory in a pharmaceutical company. SUBJECTS: Cultured primary human EC from different tissues and vascular beds. INTERVENTIONS: A monolayer of EC was incubated with either rhAPC, thrombin, histamine, or interleukin-1 beta alone or with rhAPC in combination with thrombin or interleukin-beta. The effect of rhAPC and mediators on EC permeability was monitored with measurement of electrical resistance. The effect on Rho kinase pathway signaling was monitored by the levels of phosphorylated myosin light chain and blockage with the Rho kinase specific inhibitor, Y27632. MEASUREMENTS AND MAIN RESULTS: Thrombin alone induced an early, concentration-dependent, and transient leakiness of EC. Interleukin-1 beta (0.5 ng/mL) induced an early, irreversible leakiness of EC. rhAPC (0.05-0.2 microg/mL, approximate median therapeutic blood levels) alone had no effect on EC permeability. rhAPC at > or=1 microg/mL induced an early EC leakage. rhAPC (0.19 microg/mL) attenuated the leakage induced by 0.5 ng/mL interleukin-1beta on microvascular EC derived from lung and skin and partially attenuated the leakage induced by 0.25 nM thrombin on human coronary arterial ECs. Levels of phosphorylated myosin light chain increased rapidly in human coronary arterial ECs when stimulated with thrombin or rhAPC (about 100-fold less potent) in a concentration-dependent manner via the Rho kinase signaling pathway. Short (5 mins) preconditioning of human coronary arterial ECs with 0.19 microg/mL rhAPC partially blocked the increase in phosphorylated myosin light chain levels induced by thrombin (0.06-0.2 nM). CONCLUSIONS: At concentrations exceeding physiologic and therapeutic levels, rhAPC increases EC permeability, an effect not seen at lower concentrations. The data suggest that interpretation of published in vitro and in vivo data of rhAPC and EC permeability should take into consideration the concentrations of rhAPC used or achieved. Other preliminary novel observations suggest that studying the effects of rhAPC on EC permeability and intracellular cytoskeletal organization may provide understanding of the effect of rhAPC on EC function.


Assuntos
Anti-Infecciosos/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Proteína C/farmacologia , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Recombinantes/farmacologia , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Transdução de Sinais/efeitos dos fármacos , Quinases Associadas a rho
19.
Cytokine ; 27(6): 173-9, 2004 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-15304247

RESUMO

BACKGROUND: Dengue virus infection has been rising in tropical countries. Clinical manifestations range from fever and general malaise to hemorrhagic manifestations and death. The role of endothelial damage and cytokines has not been well established for dengue infection. OBJECTIVE: Determine the profile of the pro-inflammatory cytokines and several markers of coagulopathy of dengue infection. METHODS: Patients admitted between September 2000 and April 2001, who met the WHO dengue diagnosis criteria, were enrolled. Blood samples were collected at 0, 6, 12, 24, 48, 72 h and 5 and 7 days after hospitalization. Profile of pro-inflammatory cytokines, markers of coagulopathy, protein C, protein S, d-dimer, prothrombin time, activated partial thromboplastin time, fibrinogen and activated protein C levels were determined. RESULTS: Thirty-three patients were enrolled. Median (range) age was 31 (13-70) years; 51.5% (17/33) were female. Ten of 33 (30%) presented with hemorrhagic manifestations. Patients were classified: Grade 1: 23/33 (70%), Grade II: 10/33 (30%). At study entry IL-6 was the most elevated, followed by IL-8 and TNF alpha. IL-10 was not elevated. No significant differences (P < 0.05) were demonstrated in the levels of any of the haemostatic or cytokine markers by disease severity (Grade I versus Grade II patients). CONCLUSION: The systemic host inflammatory and coagulation activation response occurs early in patients with dengue viral infection in the absence of severe hemorrhagic manifestations, and provides the basis for considering future clinical study in the use of recombinant human activated protein C to treat patients with severe sepsis from dengue infection.


Assuntos
Coagulação Sanguínea/fisiologia , Citocinas/sangue , Dengue/sangue , Dengue/imunologia , Inflamação , Adolescente , Adulto , Idoso , Animais , Biomarcadores , Dengue/fisiopatologia , Vírus da Dengue/imunologia , Feminino , Humanos , Pessoa de Meia-Idade , Estudos Prospectivos
20.
Blood ; 102(9): 3085-92, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12869495

RESUMO

Sepsis is associated with systemic inflammation, coagulopathy, and disrupted protein C (PC) pathway function. The effect of prothrombotic polymorphism, factor V Leiden (Arg506Gln; FV Leiden), was examined in a large clinical trial (PROWESS) of severe sepsis and a mouse endotoxemia model. In PROWESS, 4.1% (n = 65) of patients were heterozygous FV Leiden (VL+/-) carriers. The 28-day mortality was lower in VL+/- (13.9%) than in non-FV Leiden (VL-/-; 27.9%) patients (P =.013). The mortality benefit of recombinant human activated PC (rhAPC) treatment was similar in VL+/- (placebo, 15.6%; rhAPC,12.1%) and VL-/- patients (placebo, 31.0%; rhAPC, 24.7%; interaction P =.981). VL+/- status did not appear to influence baseline biomarkers of coagulopathy and inflammation or disease severity, with the exception that vasopressor usage was less in VL+/- patients (46.2% versus 63.0%; P =.009). In a median lethal dose (40 mg/kg) endotoxin mouse model, VL+/- mice had lower mortality than wild-type mice (19% versus 57%; P =.008), whereas the mortality of homozygous (VL+/+) mice was almost identical to that of wild-type mice (65% versus 57%; P =.76). The findings suggest that FV Leiden constitutes a rare example of a balanced gene polymorphism that maintains the FV Leiden mutation in the general gene pool due to a survival advantage of VL+/- in severe sepsis.


Assuntos
Endotoxemia/genética , Fator V/fisiologia , Sepse/genética , Idoso , Animais , Biomarcadores/sangue , Coagulação Sanguínea , Endotoxemia/mortalidade , Feminino , Heterozigoto , Humanos , Inflamação , Masculino , Camundongos , Camundongos Mutantes , Pessoa de Meia-Idade , Mutação Puntual , Proteína C/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Estudos Retrospectivos , Sepse/tratamento farmacológico , Sepse/mortalidade , Taxa de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA