Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Lupus Sci Med ; 11(1)2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453421

RESUMEN

OBJECTIVE: To evaluate safety and mechanism of action of mezagitamab (TAK-079), an anti-CD38 monoclonal antibody, in patients with moderate to severe systemic lupus erythematosus (SLE). METHODS: A phase 1b double-blind, placebo-controlled, multicentre study was conducted in patients with SLE receiving standard background therapy. Eligible patients were adults who met the 2012 SLICC or ACR criteria for diagnosis, had a baseline SLE Disease Activity Index 2000 (SLEDAI-2K) score of ≥6 and were positive for anti-double-stranded DNA antibodies and/or anti-extractable nuclear antigens antibodies. Patients received 45 mg, 90 mg or 135 mg of mezagitamab or placebo every 3 weeks over 12 weeks. Primary endpoints were safety and tolerability. Secondary endpoints included pharmacokinetics and pharmacodynamics. Exploratory assessments included disease activity scales, deep immune profiling and interferon pathway analysis. RESULTS: 22 patients received at least one dose of either mezagitamab or placebo. In patients exposed to mezagitamab (n=17), drug was well tolerated. Adverse event (AEs) were balanced across treatment groups, with no treatment emergent AEs exceeding grade 2. Responder analyses for Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) and SLEDAI-2K did not reveal any observable differences across treatment groups. However, there was a trend for more profound skin responses among patients with higher CLASI scores (>10) at baseline. Pharmacodynamic analysis showed median CD38 receptor occupancy up to 88.4% on CD38+ natural killer cells with concurrent depletion of these cells up to 90% in the 135 mg group. Mean reductions in IgG and autoantibodies were less than 20% in all dose groups. Cytometry by time of flight and type 1 interferon gene analysis revealed unique fingerprints that are indicative of a broad immune landscape shift following CD38 targeting. CONCLUSIONS: Mezagitamab had a favourable safety profile in patients with moderate to severe SLE and elicited a pharmacodynamic effect consistent with CD38+ cell depletion. These findings reveal novel insights into the drug's mechanism of action and support the continued investigation of mezagitamab in autoimmune diseases.


Asunto(s)
Anticuerpos Monoclonales , Lupus Eritematoso Sistémico , Adulto , Humanos , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacología , Interferones , Lupus Eritematoso Sistémico/tratamiento farmacológico , Resultado del Tratamiento
2.
Mol Ther Oncolytics ; 23: 547-559, 2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34938855

RESUMEN

Although anti-tumor activities of type I interferons (IFNs) have been recognized for decades, the molecular mechanisms contributing to clinical response remain poorly understood. The complex functions of these pleiotropic cytokines include stimulation of innate and adaptive immune responses against tumors as well as direct inhibition of tumor cells. In high-grade, Bacillus Calmette-Guérin (BCG)-unresponsive non-muscle-invasive bladder cancer, nadofaragene firadenovec, a non-replicating adenovirus administered locally to express the IFNα2b transgene, embodies a novel approach to deploy the therapeutic activity of type I IFNs while minimizing systemic toxicities. Deciphering which functions of type I IFN are required for clinical activity will bolster efforts to maximize the efficacy of nadofaragene firadenovec and other type I IFN-based therapies, and inform strategies to address resistance. As such, we characterized the phenotypic and molecular response of human bladder cancer cell lines to IFNα delivered in multiple contexts, including adenoviral delivery. We found that constitutive activation of the type I IFN signaling pathway is a biomarker for resistance to both transcriptional response and direct cytotoxic effects of IFNα. We present several genes that discriminate between sensitive and resistant tumor cells, suggesting they should be explored for utility as biomarkers in future clinical trials of type I IFN-based anti-tumor therapies.

3.
Lab Invest ; 97(8): 962-970, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28414323

RESUMEN

PKM2 (pyruvate kinase M2), a critical regulator of glycolysis, is phosphorylated by numerous growth factor receptors and oncogenic tyrosine kinases including NPM-ALK which is expressed in a subset of aggressive T-cell non-Hodgkin lymphomas known as anaplastic large cell lymphoma, ALK-positive. Our previous work demonstrated that phosphorylation of Y105-PKM2 by NPM-ALK regulates a major metabolic shift to promote lymphomagenesis. In addition to its role in metabolism, recent studies have shown that PKM2 promotes oncogenesis by phosphorylating nuclear STAT3 (signal transducer and activator of transcription 3) and regulating transcription of genes involved in cell survival and proliferation. We hypothesized that identification of novel PKM2 interactors could provide additional insights into its expanding functional role in cancer. To this end, immunocomplexes of FLAG-tagged PKM2 were isolated from NPM-ALK-positive ALCL (anaplastic large cell lymphoma) cells and subjected to liquid chromatography tandem mass spectrometry (LC-MS/MS) which led to the identification of polypyrimidine tract-binding protein (PTBP1) as a novel interactor of PKM2. The interaction between PTBP1 and PKM2 was restricted to the nucleus and was dependent on NPM-ALK mediated Y105 phosphorylation of PKM2. Stable shRNA-mediated silencing of PTBP1 resulted in a marked decrease in pY105-PKM2 and pY705-STAT3 which led to decreased ALCL cell proliferation and colony formation. Overall, our data demonstrate that PTBP1 interacts with PKM2 and promotes ALCL oncogenesis by facilitating PKM2-dependent activation of STAT3 within the nucleus.


Asunto(s)
Carcinogénesis/metabolismo , Proteínas Portadoras/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Linfoma Anaplásico de Células Grandes/metabolismo , Proteínas de la Membrana/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Factor de Transcripción STAT3/metabolismo , Hormonas Tiroideas/metabolismo , Línea Celular Tumoral , Núcleo Celular/química , Núcleo Celular/metabolismo , Proliferación Celular , Citoplasma/química , Citoplasma/metabolismo , Humanos , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Unión a Hormona Tiroide
4.
Proc Natl Acad Sci U S A ; 112(11): 3493-8, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25733882

RESUMEN

Oncogenic c-ros oncogene1 (ROS1) fusion kinases have been identified in a variety of human cancers and are attractive targets for cancer therapy. The MET/ALK/ROS1 inhibitor crizotinib (Xalkori, PF-02341066) has demonstrated promising clinical activity in ROS1 fusion-positive non-small cell lung cancer. However, emerging clinical evidence has shown that patients can develop resistance by acquiring secondary point mutations in ROS1 kinase. In this study we characterized the ROS1 activity of PF-06463922, a novel, orally available, CNS-penetrant, ATP-competitive small-molecule inhibitor of ALK/ROS1. In vitro, PF-06463922 exhibited subnanomolar cellular potency against oncogenic ROS1 fusions and inhibited the crizotinib-refractory ROS1(G2032R) mutation and the ROS1(G2026M) gatekeeper mutation. Compared with crizotinib and the second-generation ALK/ROS1 inhibitors ceritinib and alectinib, PF-06463922 showed significantly improved inhibitory activity against ROS1 kinase. A crystal structure of the PF-06463922-ROS1 kinase complex revealed favorable interactions contributing to the high-affinity binding. In vivo, PF-06463922 showed marked antitumor activity in tumor models expressing FIG-ROS1, CD74-ROS1, and the CD74-ROS1(G2032R) mutation. Furthermore, PF-06463922 demonstrated antitumor activity in a genetically engineered mouse model of FIG-ROS1 glioblastoma. Taken together, our results indicate that PF-06463922 has potential for treating ROS1 fusion-positive cancers, including those requiring agents with CNS-penetrating properties, as well as for overcoming crizotinib resistance driven by ROS1 mutation.


Asunto(s)
Resistencia a Antineoplásicos/genética , Lactamas Macrocíclicas/farmacología , Mutación/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Pirazoles/farmacología , Piridinas/farmacología , Aminopiridinas , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Proliferación Celular/efectos de los fármacos , Crizotinib , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Glioma/patología , Humanos , Lactamas , Lactamas Macrocíclicas/química , Ratones , Modelos Moleculares , Transducción de Señal/efectos de los fármacos
5.
Blood ; 122(6): 958-68, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23814019

RESUMEN

The mechanisms underlying the pathogenesis of the constitutively active tyrosine kinase nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) expressing anaplastic large cell lymphoma are not completely understood. Here we show using an integrated phosphoproteomic and metabolomic strategy that NPM-ALK induces a metabolic shift toward aerobic glycolysis, increased lactate production, and biomass production. The metabolic shift is mediated through the anaplastic lymphoma kinase (ALK) phosphorylation of the tumor-specific isoform of pyruvate kinase (PKM2) at Y105, resulting in decreased enzymatic activity. Small molecule activation of PKM2 or expression of Y105F PKM2 mutant leads to reversal of the metabolic switch with increased oxidative phosphorylation and reduced lactate production coincident with increased cell death, decreased colony formation, and reduced tumor growth in an in vivo xenograft model. This study provides comprehensive profiling of the phosphoproteomic and metabolomic consequences of NPM-ALK expression and reveals a novel role of ALK in the regulation of multiple components of cellular metabolism. Our studies show that PKM2 is a novel substrate of ALK and plays a critical role in mediating the metabolic shift toward biomass production and tumorigenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Linfoma Anaplásico de Células Grandes/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Humanos , Metabolómica , Ratones , Ratones SCID , Trasplante de Neoplasias , Fosforilación , Proteómica , Especificidad por Sustrato , Proteínas de Unión a Hormona Tiroide
6.
J Biol Chem ; 285(34): 25880-4, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20605784

RESUMEN

The CARMA1, Bcl10, and MALT1 proteins together constitute a signaling complex (CBM signalosome) that mediates antigen-dependent activation of NF-kappaB in lymphocytes, thereby representing a cornerstone of the adaptive immune response. Although CARMA1 is restricted to cells of the immune system, the analogous CARMA3 protein has a much wider expression pattern. Emerging evidence suggests that CARMA3 can substitute for CARMA1 in non-immune cells to assemble a CARMA3-Bcl10-MALT1 signalosome and mediate G protein-coupled receptor activation of NF-kappaB. Here we show that one G protein-coupled receptor, the type 1 receptor for angiotensin II, utilizes this mechanism for activation of NF-kappaB in endothelial and vascular smooth muscle cells, thereby inducing pro-inflammatory signals within the vasculature, a key factor in atherogenesis. Further, we demonstrate that Bcl10-deficient mice are protected from developing angiotensin-dependent atherosclerosis and aortic aneurysms. By uncovering a novel vascular role for the CBM signalosome, these findings illustrate that CBM-dependent signaling has functions outside the realm of adaptive immunity and impacts pathobiology more broadly than previously known.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Angiotensina II/fisiología , Aterosclerosis/etiología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasas/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal/fisiología , Animales , Aterosclerosis/patología , Proteína 10 de la LLC-Linfoma de Células B , Vasos Sanguíneos/patología , Endotelio Vascular/patología , Inflamación/etiología , Ratones , Ratones Noqueados , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Músculo Liso Vascular/patología , FN-kappa B/metabolismo , Receptor de Angiotensina Tipo 1
7.
Collegian ; 16(2): 85-97, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19583178

RESUMEN

AIM: To explore how CNCs who provide hospital wide support after hours (AHCSs) construct their role. METHODS: This is an ethnographic study involving two AHCSs as participants. Audio visual data was collected in 2007 at a Major Metropolitan Hospital, Sydney during after hours shifts. The data was coded using the standards defined in the Nurse Practitioner (NP) competencies. RESULTS: Four hours of videotape (observed clinical practice) and 2 h of audio tape (interviews) were coded. They performed procedures (22%), gathered information to identify at risk patients (21%), conducted patient assessments (20%) and relayed information/findings to ward nurses (12%) and doctors (12%). The roles/responsibilities of AHCSs were similar to those defined for NPs. For the domain "dynamic practice" 388 activities were identified. The two participants used advanced and comprehensive assessment skills and demonstrated a high level of proficiency in performing procedures/interventions. For the domain "professional efficacy" 174 activities were coded, for "clinical Leadership" there were 135 activities. "Pro-actively identifying at risk patients in general wards" was added as a new performance indicator within the domain "clinical leadership". An analysis of the interviews corroborated the results derived from the visual data. CONCLUSION: A significant capacity for critical thinking and clinical decision making were the hallmarks of the performance of the two AHCSs; their style of practice was collaborative, flexible and autonomous. While their formal role were as CNCs the two participants operationalised their roles/responsibilities as would a Nurse Practitioner. Their practice demonstrated a new competency: "the pro-active identification of at risk patients".


Asunto(s)
Atención Posterior , Competencia Clínica/normas , Enfermeras Clínicas/normas , Enfermeras Practicantes/normas , Personal de Enfermería en Hospital/normas , Hospitales Comunitarios , Humanos , Nueva Gales del Sur , Rol de la Enfermera , Análisis y Desempeño de Tareas
8.
Am J Physiol Endocrinol Metab ; 295(5): E1142-51, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18728225

RESUMEN

c-Jun NH(2)-terminal kinase (JNK) plays an important role in insulin resistance; however, identification of pharmacologically potent and selective small molecule JNK inhibitors has been limited. Compound A has a cell IC(50) of 102 nM and is at least 100-fold selective against related kinases and 27-fold selective against glycogen synthase kinase-3beta and cyclin-dependent kinase-2. In C57BL/6 mice, compound A reduced LPS-mediated increases in both plasma cytokine levels and phosphorylated c-Jun in adipose tissue. Treatment of mice fed a high-fat diet with compound A for 3 wk resulted in a 13.1 +/- 1% decrease in body weight and a 9.3 +/- 1.5% decrease in body fat, compared with a 6.6 +/- 2.1% increase in body weight and a 6.7 +/- 2.1% increase in body fat in vehicle-treated mice. Mice pair fed to those that received compound A exhibited a body weight decrease of 7 +/- 1% and a decrease in body fat of 1.6 +/- 1.3%, suggesting that reductions in food intake could not account solely for the reductions in adiposity observed. Compound A dosed at 30 mg/kg for 13 days in high-fat fed mice resulted in a significant decrease in phosphorylated c-Jun in adipose tissue accompanied by a decrease in weight and reductions in glucose and triglycerides and increases in insulin sensitivity to levels comparable with those in lean control mice. The ability of compound A to reduce the insulin-stimulated phosphorylation of insulin receptor substrate-1 (IRS-1) von Ser307 and partially reverse the free fatty acid inhibition of glucose uptake in 3T3L1 adipocytes, suggests that enhancement of insulin signaling in addition to weight loss may contribute to the effects of compound A on insulin sensitization in vivo. Pharmacological inhibition of JNK using compound A may therefore offer an effective therapy for type 2 diabetes mediated at least in part via weight reduction.


Asunto(s)
Aminopiridinas/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Células 3T3-L1 , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Aminopiridinas/farmacocinética , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Citocinas/sangre , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Ingestión de Alimentos/efectos de los fármacos , Humanos , Insulina/sangre , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/patología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Células U937
9.
Mol Cancer Ther ; 6(12 Pt 1): 3314-22, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18089725

RESUMEN

A t(2;5) chromosomal translocation resulting in expression of an oncogenic kinase fusion protein known as nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) has been implicated in the pathogenesis of anaplastic large-cell lymphoma (ALCL). PF-2341066 was recently identified as a p.o. bioavailable, small-molecule inhibitor of the catalytic activity of c-Met kinase and the NPM-ALK fusion protein. PF-2341066 also potently inhibited NPM-ALK phosphorylation in Karpas299 or SU-DHL-1 ALCL cells (mean IC(50) value, 24 nmol/L). In biochemical and cellular screens, PF-2341066 was shown to be selective for c-Met and ALK at pharmacologically relevant concentrations across a panel of >120 diverse kinases. PF-2341066 potently inhibited cell proliferation, which was associated with G(1)-S-phase cell cycle arrest and induction of apoptosis in ALK-positive ALCL cells (IC(50) values, approximately 30 nmol/L) but not ALK-negative lymphoma cells. The induction of apoptosis was confirmed using terminal deoxyribonucleotide transferase-mediated nick-end labeling and Annexin V staining (IC(50) values, 25-50 nmol/L). P.o. administration of PF-2341066 to severe combined immunodeficient-Beige mice bearing Karpas299 ALCL tumor xenografts resulted in dose-dependent antitumor efficacy with complete regression of all tumors at the 100 mg/kg/d dose within 15 days of initial compound administration. A strong correlation was observed between antitumor response and inhibition of NPM-ALK phosphorylation and induction of apoptosis in tumor tissue. In addition, inhibition of key NPM-ALK signaling mediators, including phospholipase C-gamma, signal transducers and activators of transcription 3, extracellular signal-regulated kinases, and Akt by PF-2341066 were observed at concentrations or dose levels, which correlated with inhibition of NPM-ALK phosphorylation and function. Collectively, these data illustrate the potential clinical utility of inhibitors of NPM-ALK in treatment of patients with ALK-positive ALCL.


Asunto(s)
Antineoplásicos/farmacología , Linfoma Anaplásico de Células Grandes/patología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Piridinas/farmacología , Quinasa de Linfoma Anaplásico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Crizotinib , Ensayos de Selección de Medicamentos Antitumorales , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Ratones , Ratones SCID , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Pirazoles , Proteínas Tirosina Quinasas Receptoras
10.
Biochem Biophys Res Commun ; 361(1): 140-5, 2007 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-17659259

RESUMEN

The contribution of nutrient overload and associated inflammation to insulin resistance has highlighted several therapeutic targets including c-Jun N-terminal kinase (JNK) and S6 kinase (S6K). To investigate how a lipopolysaccharide (LPS)-mediated inflammatory response may modulate pathways implicated in insulin resistance, we characterized the LPS-induced changes in key biomarkers. Administration of 0.06-4 mg/kg LPS to C57BL/6 mice stimulated increases in plasma levels of TNFalpha, IL-12p40, IL-6 and MCP-1 and in JNK activity as measured by phosphorylated c-Jun in fat. For the first time, we show that LPS induces S6K activity by up to 6.1-fold, as measured by the phosphorylation of S6 ribosomal protein in liver, and increases by up to 1.8-fold, plasma levels of the novel pro-inflammatory cytokine osteopontin which is implicated in the pathogenesis of insulin resistance. These novel findings suggest that LPS administration may form the basis of an acute in vivo pharmacodynamic model for therapies targeting multiple pathways implicated in insulin resistance.


Asunto(s)
Modelos Animales de Enfermedad , Resistencia a la Insulina , Lipopolisacáridos/farmacología , Ratones , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Glucemia/efectos de los fármacos , Citocinas/sangre , Insulina/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones Endogámicos C57BL , Osteopontina/sangre , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
11.
Cancer Res ; 67(9): 4408-17, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17483355

RESUMEN

The c-Met receptor tyrosine kinase and its ligand, hepatocyte growth factor (HGF), have been implicated in the progression of several human cancers and are attractive therapeutic targets. PF-2341066 was identified as a potent, orally bioavailable, ATP-competitive small-molecule inhibitor of the catalytic activity of c-Met kinase. PF-2341066 was selective for c-Met (and anaplastic lymphoma kinase) compared with a panel of >120 diverse tyrosine and serine-threonine kinases. PF-2341066 potently inhibited c-Met phosphorylation and c-Met-dependent proliferation, migration, or invasion of human tumor cells in vitro (IC(50) values, 5-20 nmol/L). In addition, PF-2341066 potently inhibited HGF-stimulated endothelial cell survival or invasion and serum-stimulated tubulogenesis in vitro, suggesting that this agent also exhibits antiangiogenic properties. PF-2341066 showed efficacy at well-tolerated doses, including marked cytoreductive antitumor activity, in several tumor models that expressed activated c-Met. The antitumor efficacy of PF-2341066 was dose dependent and showed a strong correlation to inhibition of c-Met phosphorylation in vivo. Near-maximal inhibition of c-Met activity for the full dosing interval was necessary to maximize the efficacy of PF-2341066. Additional mechanism-of-action studies showed dose-dependent inhibition of c-Met-dependent signal transduction, tumor cell proliferation (Ki67), induction of apoptosis (caspase-3), and reduction of microvessel density (CD31). These results indicated that the antitumor activity of PF-2341066 may be mediated by direct effects on tumor cell growth or survival as well as antiangiogenic mechanisms. Collectively, these results show the therapeutic potential of targeting c-Met with selective small-molecule inhibitors for the treatment of human cancers.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Piperidinas/farmacología , Piridinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Neoplasias Gástricas/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Crizotinib , Perros , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/irrigación sanguínea , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA