Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Invest New Drugs ; 36(4): 536-544, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29188469

RESUMEN

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.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos/farmacología , Exones/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo
2.
Invest New Drugs ; 35(3): 334-344, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28299514

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/sangre , Carboplatino/farmacología , Etopósido/farmacología , Células Neoplásicas Circulantes , Péptidos Cíclicos/farmacología , Receptores CXCR4/antagonistas & inhibidores , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carboplatino/uso terapéutico , Recuento de Células , Supervivencia sin Enfermedad , Etopósido/uso terapéutico , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Péptidos Cíclicos/uso terapéutico , Pronóstico , Receptores CXCR4/metabolismo , Carcinoma Pulmonar de Células Pequeñas/sangre , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/metabolismo
3.
Invest New Drugs ; 31(4): 833-44, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23275061

RESUMEN

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).


Asunto(s)
Indazoles/farmacología , Niacinamida/análogos & derivados , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Tetrazoles/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Disponibilidad Biológica , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Indazoles/administración & dosificación , Indazoles/química , Ratones , Mutación/genética , Niacinamida/administración & dosificación , Niacinamida/química , Niacinamida/farmacología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tetrazoles/administración & dosificación , Tetrazoles/química , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Microvasc Res ; 81(2): 216-21, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21130783

RESUMEN

Elevated circulating endothelial cell (CEC) and circulating endothelial progenitor cell (CEPC) counts may indicate vascular damage and disease status, but data on these cell populations in patients with severe sepsis are limited. This study compared CEC and CEPC counts in patients with and without severe sepsis following intensive care unit (ICU) admission. Venous blood samples were collected within 24 h, 48-72 h, and 120-144 h. Baseline demographics, 28-day mortality, ICU and hospital days, and Sequential Organ Failure Assessment (SOFA) scores were recorded. Patients with (n=18) and without (n=28) severe sepsis were balanced for mean age (63.7 and 61.3 years, respectively) and gender. There were no differences in 28-day mortality, ICU days, or hospital days. Baseline SOFA scores were higher in the sepsis group. At 48-72 h, patients with severe sepsis had significantly higher median CEC counts (51.5 vs. 28.0 cells/4 ml of blood, P=0.02). CEC values for all ICU patients were significantly (P<0.05) higher than in healthy volunteers. CEPC counts in both cohorts ranged from 0 to >21 colonies/4 ml blood (mean=1.13±2.25; median=0) without significant differences at any time point. This study demonstrates the ability to quantify CECs and CEPCs using consensus methodology. Understanding the relationship between CEC/CEPC counts and outcomes may provide insight into the mechanisms of endothelial cell changes in severe sepsis.


Asunto(s)
Células Sanguíneas/patología , Células Endoteliales/patología , Sepsis/sangre , Células Madre/patología , Adulto , Anciano , Anciano de 80 o más Años , Recuento de Células Sanguíneas , Femenino , Humanos , Unidades de Cuidados Intensivos , Tiempo de Internación/estadística & datos numéricos , Masculino , Persona de Mediana Edad , Pronóstico , Sepsis/diagnóstico , Sepsis/patología , Índice de Severidad de la Enfermedad , Choque/sangre , Choque/diagnóstico , Choque/patología , Factores de Tiempo
5.
Oncotarget ; 9(17): 13796-13806, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29568395

RESUMEN

Merestinib is an oral multi-kinase inhibitor targeting a limited number of oncokinases including MET, AXL, RON and MKNK1/2. Here, we report that merestinib inhibits neurotrophic receptor tyrosine kinases NTRK1/2/3 which are oncogenic drivers in tumors bearing NTRK fusion resulting from chromosomal rearrangements. Merestinib is shown to be a type II NTRK1 kinase inhibitor as determined by x-ray crystallography. In KM-12 cells harboring TPM3-NTRK1 fusion, merestinib exhibits potent p-NTRK1 inhibition in vitro by western blot and elicits an anti-proliferative response in two- and three-dimensional growth. Merestinib treatment demonstrated profound tumor growth inhibition in in vivo cancer models harboring either a TPM3-NTRK1 or an ETV6-NTRK3 gene fusion. To recapitulate resistance observed from type I NTRK kinase inhibitors entrectinib and larotrectinib, we generated NIH-3T3 cells exogenously expressing TPM3-NTRK1 wild-type, or acquired mutations G595R and G667C in vitro and in vivo. Merestinib blocks tumor growth of both wild-type and mutant G667C TPM3-NTRK1 expressing NIH-3T3 cell-derived tumors. These preclinical data support the clinical evaluation of merestinib, a type II NTRK kinase inhibitor (NCT02920996), both in treatment naïve patients and in patients progressed on type I NTRK kinase inhibitors with acquired secondary G667C mutation in NTRK fusion bearing tumors.

6.
Crit Care ; 9 Suppl 4: S38-45, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16168074

RESUMEN

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.


Asunto(s)
Fibrinolíticos/uso terapéutico , Deficiencia de Proteína C/tratamiento farmacológico , Proteína C/fisiología , Sepsis/tratamiento farmacológico , Animales , Antígenos/fisiología , Antígenos CD , Factores de Coagulación Sanguínea/fisiología , Receptor de Proteína C Endotelial , Glicoproteínas/fisiología , Humanos , Proteína C/uso terapéutico , Deficiencia de Proteína C/metabolismo , Deficiencia de Proteína C/fisiopatología , Receptores de Superficie Celular/fisiología , Proteínas Recombinantes/uso terapéutico , Sepsis/fisiopatología , Trombomodulina/fisiología
7.
Crit Care ; 8(2): R82-90, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15025782

RESUMEN

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.


Asunto(s)
Biomarcadores/análisis , Sepsis/diagnóstico , Infección de la Herida Quirúrgica/diagnóstico , APACHE , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/diagnóstico , Proteína C-Reactiva/análisis , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/diagnóstico , Endotelio/lesiones , Fibrina/análisis , Fibrinólisis , Humanos , Inflamación/sangre , Tiempo de Tromboplastina Parcial , Placebos , Pronóstico , Proteína C/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Sensibilidad y Especificidad , Sepsis/tratamiento farmacológico , Sepsis/mortalidad , Índice de Severidad de la Enfermedad , Procedimientos Quirúrgicos Operativos/efectos adversos , Infección de la Herida Quirúrgica/tratamiento farmacológico , Infección de la Herida Quirúrgica/mortalidad
8.
J Antimicrob Chemother ; 59(6): 1177-81, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17428810

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Actividad Bactericida de la Sangre/efectos de los fármacos , Citocinas/fisiología , Monocitos/efectos de los fármacos , Proteína C/farmacología , Ampicilina/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/inmunología , Humanos , Técnicas In Vitro , Inflamación/fisiopatología , Interleucina-1beta/farmacología , Interleucina-8/farmacología , Cinética , Levofloxacino , Ofloxacino/farmacología , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/farmacología
9.
Crit Care Med ; 32(5 Suppl): S302-8, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15118535

RESUMEN

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.


Asunto(s)
Antiinfecciosos/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Proteína C/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Recombinantes/farmacología , Células Cultivadas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Transducción de Señal/efectos de los fármacos , Quinasas Asociadas a rho
10.
Cytokine ; 27(6): 173-9, 2004 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-15304247

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea/fisiología , Citocinas/sangre , Dengue/sangre , Dengue/inmunología , Inflamación , Adolescente , Adulto , Anciano , Animales , Biomarcadores , Dengue/fisiopatología , Virus del Dengue/inmunología , Femenino , Humanos , Persona de Mediana Edad , Estudios Prospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA