Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Immunol ; 202(10): 3065-3075, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30979816

RESUMEN

Although multidisciplinary treatment is widely applied in colorectal cancer (CRC), the prognosis of patients with advanced CRC remains poor. Immunotherapy blocking of programmed cell death ligand 1 (PD-L1) is a promising approach. Binding of the transmembrane protein PD-L1 expressed by tumor cells or tumor microenvironment cells to its receptor programmed cell death 1 (PD-1) induces immunosuppressive signals and reduces the proliferation of T cells, which is an important mechanism of tumor immune escape and a key issue in immunotherapy. However, the regulation of PD-L1 expression is poorly understood in CRC. Fibroblast growth factor (FGF) receptor (FGFR) 2 causes the tyrosine kinase domains to initiate a cascade of intracellular signals by binding to FGFs and dimerization (pairing of receptors), which is involved in tumorigenesis and progression. In this study, we showed that PD-L1 and FGFR2 were frequently overexpressed in CRC, and FGFR2 expression was significantly associated with lymph node metastasis, clinical stage, and poor survival. In the current study, PD-L1 expression was positively correlated with FGFR2 expression in CRC. Tumor-derived-activated FGFR2 induced PD-L1 expression via the JAK/STAT3 signaling pathway in human CRC cells (SW480 and NCI-H716), which induced the apoptosis of Jurkat T cells. FGFR2 also promoted the expression of PD-L1 in a xenograft mouse model of CRC. The results of our study reveal a novel mechanism of PD-L1 expression in CRC, thus providing a theoretical basis for reversing the immune tolerance of FGFR2 overexpression in CRC.


Asunto(s)
Antígeno B7-H1/inmunología , Neoplasias Colorrectales/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Quinasas Janus/inmunología , Proteínas de Neoplasias/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Factor de Transcripción STAT3/inmunología , Transducción de Señal/inmunología , Antígeno B7-H1/genética , Células CACO-2 , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Células HCT116 , Humanos , Quinasas Janus/genética , Células Jurkat , Metástasis Linfática , Proteínas de Neoplasias/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/genética
2.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799514

RESUMEN

Treatment of patients with urothelial carcinoma (UC) of the bladder or renal cancer has changed significantly during recent years and efforts towards biomarker-directed therapy are being investigated. Immune checkpoint inhibition (ICI) or fibroblast growth factor receptor (FGFR) directed therapy are being evaluated for non-muscle invasive bladder cancer (NMIBC) patients, as well as muscle-invasive bladder cancer (MIBC) patients. Meanwhile, efforts to predict tumor response to neoadjuvant chemotherapy (NAC) are still ongoing, and genomic biomarkers are being evaluated in prospective clinical trials. Currently, patients with metastatic UC (mUC) are usually treated with second-line ICI, while cisplatin-ineligible patients with programmed death-ligand 1 (PD-L1) positive tumors can benefit from first-line ICI. Platinum-relapsed UC patients harboring FGFR2/3 mutations can be treated with erdafitinib, while enfortumab vedotin has emerged as a novel third-line treatment option for mUC. In metastatic (clear cell) renal cell carcinoma (RCC), ICI was first introduced as second-line treatment after vascular endothelial growth factor receptor-tyrosine kinase inhibition (VEGFR-TKI). Currently, ICIs have also been introduced as first-line treatment in metastatic RCC. Although there is no evidence up to now for beneficial adjuvant treatment after surgery with VEGFR-TKIs in high-risk non-metastatic RCC, several trials are underway investigating the potential beneficial effect of ICIs in this setting.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Biomarcadores de Tumor/inmunología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Cisplatino/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Metástasis Linfática , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Quinoxalinas/uso terapéutico , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/inmunología , Recurrencia , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/patología
3.
Eur J Immunol ; 45(9): 2517-28, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26081170

RESUMEN

Chronic skin inflammation resulting from a defective epidermal barrier is a hallmark of atopic dermatitis (AD). We previously demonstrated that mice lacking FGF receptors 1 and 2 in keratinocytes (K5-R1/R2 mice) develop an AD-like chronic dermatitis as a result of an impaired epidermal barrier. Here, we show that γδ T cells, which rapidly respond to various insults, accumulate in the epidermis of K5-R1/R2 mice before the development of histological abnormalities. Their number and activation further increase as the phenotype progresses, most likely as a consequence of increased expression of Il-2 and Il-7 and the stress-induced proteins Rae-1, H60c, Mult1, PlexinB2, and Skint1. To determine the role of γδ T cells in the skin phenotype, we generated quadruple mutant K5-R1/-R2 mice lacking γδ T cells. Surprisingly, loss of γδ T cells did not or only marginally affect keratinocyte proliferation, epidermal thickness, epidermal barrier function, and accumulation and activation of different immune cells in the skin of K5-R1/R2 mice, possibly due to partial compensation by αß T cells. These results demonstrate that γδ T cells do not contribute to the development or maintenance of chronic inflammation in response to a defect in the epidermal barrier.


Asunto(s)
Dermatitis/inmunología , Queratinocitos/inmunología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Subgrupos de Linfocitos T/inmunología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Proliferación Celular , Enfermedad Crónica , Dermatitis/genética , Dermatitis/patología , Modelos Animales de Enfermedad , Epidermis/inmunología , Epidermis/patología , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulinas/genética , Inmunoglobulinas/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-7/genética , Interleucina-7/inmunología , Queratinocitos/patología , Activación de Linfocitos , Depleción Linfocítica , Proteínas de la Membrana , Ratones , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/inmunología , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/inmunología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Transducción de Señal , Subgrupos de Linfocitos T/patología
4.
Eur J Haematol ; 96(4): 425-34, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26115424

RESUMEN

BACKGROUND: Multipotent mesenchymal stromal cells (MSCs) are used for prophylaxis of acute graft-versus-host disease (aGvHD) after allogeneic hematopoietic cell transplantation (allo-HCT). Not all samples of MSC are efficient for aGvHD prevention. The suitability of MSCs for aGvHD prophylaxis was studied. METHODS: MSCs were derived from the bone marrow (BM) of HCT donor and cultivated for no more than three passages. The characteristics of donor BM samples including colony-forming unit fibroblast (CFU-F) concentration, growth parameters of MSCs, and the relative expression levels (REL) of different genes were analyzed. MSCs were injected intravenously precisely at the moment of blood cell reconstitution. RESULTS: MSCs infusion induced a significant threefold decrease in aGvHD development and improved overall survival compared with the standard prophylaxis group. In ineffective MSC samples (9.4%), a significant decrease in total cell production and the REL of CSF1, FGFR1, and PDGFRB was observed. In all studied BM samples, the cumulative MSC production and CFU-F concentrations decreased with age. The expression levels of FGFR2, PPARG, and VEGF differed by age. CONCLUSIONS: A universal single indicator for the prediction of MSC eligibility for aGvHD prophylaxis was not identified. A multiparameter mathematical model for selecting MSC samples effective for the prevention of aGvHD was proposed.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Agonistas Mieloablativos/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Adolescente , Adulto , Femenino , Expresión Génica , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/mortalidad , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/patología , Síndromes Mielodisplásicos/terapia , PPAR gamma/genética , PPAR gamma/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/inmunología , Análisis de Supervivencia , Trasplante Homólogo
5.
J Biol Chem ; 286(48): 41230-41245, 2011 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-21990352

RESUMEN

Previous studies indicate that astrocytes are the brain cells that express acidic fibroblast growth factor (aFGF) and that the expression is increased upon activation. However, there has been no study investigating the significance of this phenomenon. Here we report that aFGF treatment of IFNγ-stimulated human astrocytes, and LPS/IFNγ-stimulated human microglia, enhances their secretion of inflammatory cytokines and other materials toxic to human neuroblastoma SH-SY5Y cells. The mechanism of aFGF enhancement involves stimulation of the receptor FGFR2 IIIb. We show by RT-PCR that this receptor, but not other FGF receptors, is robustly expressed by astrocytes and microglia. We establish by Western blotting, and immunohistochemistry on postmortem human brain tissue that the FGFR2 IIIb protein is expressed by both of these glial cell types. We blocked the inflammatory stimulant action of aFGF by transfecting microglia and astrocytes with a small inhibitory RNA (siRNA) to FGFR2 IIIb as well as by removal of aFGF using an anti-aFGF antibody. Treatment with bFGF in combination with the stimulants was without effect, but together with aFGF, it partially counteracted the action of aFGF, indicating that it may be a weak antagonist of FGFR2 IIIb. The inflammatory effect was also attenuated by treatment with inhibitors of protein kinase C, Src tyrosine kinase, and MEK-1/2 indicating the involvement of these intracellular pathways. Our data suggest that inhibition of expression or release of aFGF could have therapeutic potential by inhibiting inflammation in neurodegenerative diseases such as Alzheimer disease where many neuroinflammatory molecules are prominently expressed.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/enzimología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Antivirales/farmacología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación Enzimológica de la Expresión Génica , Humanos , Interferón gamma/farmacología , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Proteínas del Tejido Nervioso/genética , Neuroglía/patología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética
6.
Cancer Chemother Pharmacol ; 88(5): 899-910, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34383128

RESUMEN

PURPOSE: A population pharmacokinetic (PK) analysis of the anti-fibroblast growth factor receptor 2b antibody, bemarituzumab, was performed to evaluate the impact of covariates on the PK and assess whether dose adjustment is necessary for a future phase 3 trial. METHODS: Serum concentration data were obtained from three clinical trials, with 1552 bemarituzumab serum samples from 173 patients, and were analyzed using nonlinear mixed-effects modeling. RESULTS: A two-compartment model with parallel linear and nonlinear (Michaelis-Menten) elimination from the central compartment best described the bemarituzumab serum concentration data. The final model estimated a typical linear clearance (CL) of 0.311 L/day, volume of distribution in the central compartment (Vc) of 3.58 L, distribution clearance (Q) of 0.952 L/day, volume of distribution in the peripheral compartment (Vp) of 2.71 L, maximum drug elimination by nonlinear clearance (Vmax) of 2.80 µg/day, and Michaelis-Menten constant (Km) of 4.45 µg/mL. Baseline body weight, baseline albumin, gender, and chemotherapy were identified as statistically significant covariates on the PK of bemarituzumab. Given the low interindividual variability of bemarituzumab key PK parameters (CL and Vc) and the small or modest effect of all statistically significant covariates on bemarituzumab exposure at steady-state, no covariate is expected to have clinically meaningful effects on bemarituzumab exposure. CONCLUSION: No covariate had a clinically meaningful impact on bemarituzumab exposure. These results indicate that dose adjustment of bemarituzumab is not necessary, based on the aforementioned covariates, for a future phase 3 trial in gastric and gastroesophageal junction adenocarcinoma population with FGFR2b overexpression in combination with mFOLFOX6.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacocinética , Modelos Teóricos , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/farmacocinética , Femenino , Humanos , Infusiones Intravenosas , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología
7.
Cells ; 10(4)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33924474

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease of the central nervous system (CNS) affecting more than two million people worldwide. In MS, oligodendrocytes and myelin sheaths are destroyed by autoimmune-mediated inflammation, while remyelination is impaired. Recent investigations of post-mortem tissue suggest that Fibroblast growth factor (FGF) signaling may regulate inflammation and myelination in MS. FGF2 expression seems to correlate positively with macrophages/microglia and negatively with myelination; FGF1 was suggested to promote remyelination. In myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis (EAE), systemic deletion of FGF2 suggested that FGF2 may promote remyelination. Specific deletion of FGF receptors (FGFRs) in oligodendrocytes in this EAE model resulted in a decrease of lymphocyte and macrophage/microglia infiltration as well as myelin and axon degeneration. These effects were mediated by ERK/Akt phosphorylation, a brain-derived neurotrophic factor, and downregulation of inhibitors of remyelination. In the first part of this review, the most important pharmacotherapeutic principles for MS will be illustrated, and then we will review recent advances made on FGF signaling in MS. Thus, we will suggest application of FGFR inhibitors, which are currently used in Phase II and III cancer trials, as a therapeutic option to reduce inflammation and induce remyelination in EAE and eventually MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Microglía/inmunología , Esclerosis Múltiple/genética , Vaina de Mielina/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Factor 2 de Crecimiento de Fibroblastos/deficiencia , Regulación de la Expresión Génica , Humanos , Factores Inmunológicos/uso terapéutico , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/patología , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Oligodendroglía/efectos de los fármacos , Oligodendroglía/inmunología , Oligodendroglía/patología , Fragmentos de Péptidos/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Remielinización/efectos de los fármacos , Remielinización/genética , Remielinización/inmunología , Transducción de Señal
8.
J Clin Oncol ; 38(21): 2418-2426, 2020 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-32167861

RESUMEN

PURPOSE: To evaluate the safety, pharmacokinetics, and preliminary activity of bemarituzumab in patients with FGFR2b-overexpressing gastric and gastroesophageal junction adenocarcinoma (GEA). PATIENTS AND METHODS: FPA144-001 was a phase I, open-label, multicenter trial consisting of the following 3 parts: part 1a involved dose escalation in patients with recurrent solid tumors at doses ranging from 0.3 to 15 mg/kg; part 1b involved dose escalation in patients with advanced-stage GEA; and part 2 involved dose expansion in patients with advanced-stage GEA that overexpressed FGFR2b at various levels (4 cohorts; high, medium, low, and no FGFR2b overexpression) and 1 cohort of patients with FGFR2b-overexpressing advanced-stage bladder cancer. RESULTS: Seventy-nine patients were enrolled; 19 were enrolled in part 1a, 8 in part 1b, and 52 in part 2. No dose-limiting toxicities were reported, and the recommended dose was identified as 15 mg/kg every 2 weeks based on safety, tolerability, pharmacokinetic parameters, and clinical activity. The most frequent treatment-related adverse events (TRAEs) were fatigue (17.7%), nausea (11.4%), and dry eye (10.1%). Grade 3 TRAEs included nausea (2 patients) and anemia, neutropenia, increased AST, increased alkaline phosphatase, vomiting, and an infusion reaction (1 patient each). Three (10.7%) of 28 patients assigned to a cohort receiving a dose of ≥ 10 mg/kg every 2 weeks for ≥ 70 days reported reversible grade 2 corneal TRAEs. No TRAEs of grade ≥ 4 were reported. Five (17.9%; 95% CI, 6.1% to 36.9%) of 28 patients with high FGFR2b-overexpressing GEA had a confirmed partial response. CONCLUSION: Overall, bemarituzumab seems to be well tolerated and demonstrated single-agent activity as late-line therapy in patients with advanced-stage GEA. Bemarituzumab is currently being evaluated in combination with chemotherapy in a phase III trial as front-line therapy for patients with high FGFR2b-overexpressing advanced-stage GEA.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/farmacocinética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
9.
Target Oncol ; 14(5): 591-601, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31502117

RESUMEN

BACKGROUND: Fibroblast growth factor receptor (FGFR) 2 is overexpressed in several tumor types, including triple-negative breast cancer and gastric cancer, both of which have a high unmet medical need. Aprutumab ixadotin (BAY 1187982) is the first antibody-drug conjugate (ADC) to target FGFR2 and the first to use a novel auristatin-based payload. OBJECTIVE: This first-in-human trial was conducted to determine the safety, tolerability, and maximum tolerated dose (MTD) of aprutumab ixadotin in patients with advanced solid tumors from cancer indications known to be FGFR2-positive. PATIENTS AND METHODS: In this open-label, multicenter, phase I dose-escalation trial (NCT02368951), patients with advanced solid tumors received escalating doses of aprutumab ixadotin (starting at 0.1 mg/kg body weight), administered intravenously on day 1 of every 21-day cycle. Primary endpoints included safety, tolerability, and the MTD of aprutumab ixadotin; secondary endpoints were pharmacokinetic evaluation and tumor response to aprutumab ixadotin. RESULTS: Twenty patients received aprutumab ixadotin across five cohorts, at doses of 0.1-1.3 mg/kg. The most common grade ≥ 3 drug-related adverse events were anemia, aspartate aminotransferase increase, proteinuria, and thrombocytopenia. Dose-limiting toxicities were thrombocytopenia, proteinuria, and corneal epithelial microcysts, and were only seen in the two highest dosing cohorts. The MTD was determined to be 0.2 mg/kg due to lack of quantitative data following discontinuations at 0.4 and 0.8 mg/kg doses. One patient had stable disease; no responses were reported. CONCLUSIONS: Aprutumab ixadotin was poorly tolerated, with an MTD found to be below the therapeutic threshold estimated preclinically; therefore, the trial was terminated early. CLINICALTRIALS. GOV IDENTIFIER: NCT02368951.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Colangiocarcinoma/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Inmunoconjugados/uso terapéutico , Oligopéptidos/uso terapéutico , Adulto , Anciano , Terminación Anticipada de los Ensayos Clínicos , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Insuficiencia del Tratamiento , Adulto Joven
10.
Acta Otolaryngol ; 128(4): 360-4, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18368565

RESUMEN

CONCLUSION: Distribution of the receptor for epidermal growth factor (EGF-R) and of the receptor for the keratinocyte growth factor (KGF-R) in cholesteatoma was found to differ in analogy with other epithelial tissues and accordingly to epidermal differentiation and intensity of paracrine stimulation. Moreover, both EGF-R and KGF-R expression was increased, suggesting a fair correlation with aggressiveness and recurrence rate of this pathology. OBJECTIVES: To obtain information on the biological behaviour of cholesteatoma by assessing the expression and localization of EGF-R and KGF-R and correlating their tissue distribution with that of cytokeratins as a marker of differentiation. MATERIALS AND METHODS: Cholesteatoma tissue was taken during tympanoplasty surgery and processed for indirect immunofluorescence. Murine monoclonal antibodies were tested for the different growth factor receptors and pancytokeratins analysed. Fluorescence intensity signal was measured on randomly captured digital images, using FISH 2000/HI software, with a pseudocolours generation module. RESULTS: EGF-R was mostly expressed at the level of keratinocytes of the basal layer, while KGF-R signal was mainly distributed on the spinous and granular suprabasal layers that were also highly positive for cytokeratins. Significant correlation between the immunofluorescence signals was found for KGF-R and cytokeratins only, demonstrating that KGF-R expression is increased in more differentiated areas of the cholesteatoma tissue, while EGF-R is associated with proliferative and migratory portions of the lesion.


Asunto(s)
Colesteatoma del Oído Medio/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Anticuerpos Monoclonales , Biomarcadores/metabolismo , Colesteatoma del Oído Medio/patología , Receptores ErbB/biosíntesis , Receptores ErbB/inmunología , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Queratinocitos/metabolismo , Microscopía Fluorescente , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología
11.
In Vivo ; 21(4): 623-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17708356

RESUMEN

BACKGROUND: Increased expression of fibroblast growth factors and their receptors (FGFRs) has recently been described in oral squamous cell carcinoma. In addition, we have previously described a molecular basis for an association between oral cancer and diabetes. The expression of FGFR-2 and FGFR-3 investigated in an experimental model of chemically induced carcinogenesis in normal and diabetic (type I) rats. MATERIALS AND METHODS: Tissue sections ranging from normal mucosa to moderately-differentiated oral squamous cell carcinoma were studied using monoclonal antibodies against FGFR-2 and FGFR-3 proteins. RESULTS: A similar pattern of elevated FGFR-2 and FGFR-3 expression was observed in the initial stages of oncogenesis for both diabetic and non-diabetic animals. In the last stages of oral oncogenesis, the expression of both proteins remained relatively stable. CONCLUSION: It seems that diabetes does not affect the FGFR-2 and FGFR-3 pattern of expression throughout the various stages of oral oncogenesis.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Neoplasias de la Boca/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Anticuerpos Monoclonales , Carcinoma de Células Escamosas/patología , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Neoplasias de la Boca/patología , Ratas , Ratas Sprague-Dawley , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/inmunología
12.
Cancer Res ; 76(21): 6331-6339, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27543601

RESUMEN

The fibroblast growth factor receptor FGFR2 is overexpressed in a variety of solid tumors, including breast, gastric, and ovarian tumors, where it offers a potential therapeutic target. In this study, we present evidence of the preclinical efficacy of BAY 1187982, a novel antibody-drug conjugate (ADC). It consists of a fully human FGFR2 monoclonal antibody (mAb BAY 1179470), which binds to the FGFR2 isoforms FGFR2-IIIb and FGFR2-IIIc, conjugated through a noncleavable linker to a novel derivative of the microtubule-disrupting cytotoxic drug auristatin (FGFR2-ADC). In FGFR2-expressing cancer cell lines, this FGFR2-ADC exhibited potency in the low nanomolar to subnanomolar range and was more than 100-fold selective against FGFR2-negative cell lines. High expression levels of FGFR2 in cells correlated with efficient internalization, efficacy, and cytotoxic effects in vitro Pharmacokinetic analyses in mice bearing FGFR2-positive NCI-H716 tumors indicated that the toxophore metabolite of FGFR2-ADC was enriched more than 30-fold in tumors compared with healthy tissues. Efficacy studies demonstrated that FGFR2-ADC treatment leads to a significant tumor growth inhibition or tumor regression of cell line-based or patient-derived xenograft models of human gastric or breast cancer. Furthermore, FGFR2 amplification or mRNA overexpression predicted high efficacy in both of these types of in vivo model systems. Taken together, our results strongly support the clinical evaluation of BAY 1187982 in cancer patients and a phase I study (NCT02368951) has been initiated. Cancer Res; 76(21); 6331-9. ©2016 AACR.


Asunto(s)
Aminobenzoatos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Inmunoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/análisis , Animales , Anticuerpos Monoclonales Humanizados , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Nutr Diabetes ; 6(11): e233, 2016 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-27892934

RESUMEN

Expression of ß-Kotho, fibroblast growth factor receptor (FGFR)-1c and 2c, which bind FGF21, is decreased in the white adipose tissue of obese mice. The aim of the present study was to determine the role of FGFR2c in the development of obesity and diabetes in KKAy mice. Treatment with mouse monoclonal FGFR2-IIIc antibody (0.5 mg kg-1) significantly suppressed body weight gain and epididymal white adipose tissue weight in individually housed KKAy mice while having no effect on daily food intake. In addition, treatment with FGFR2-IIIc antibody significantly increased plasma-free fatty acid levels while having no effect on blood glucose or plasma FGF21 levels. Moreover, treatment with FGFR2-IIIc antibody had no significant effect on the expression of uncoupling protein-1, uncoupling protein-2 or peroxisome proliferator-activated receptor-γ coactivator 1α in the epididymal white adipose tissue. The treatment with FGFR2-IIIc antibody had no significant effects on daily food intake and body weight gain in individually housed KK mice. These findings suggest that FGFR2-IIIc upregulates the adiposity induced by social isolation in KKAy mice, and that decreased expression and/or function of FGFR2c might be a compensatory response to enhanced adiposity. Inhibition of FGFR2-IIIc function might be a novel therapeutic approach for obesity.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Anticuerpos Monoclonales/farmacología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Aumento de Peso/efectos de los fármacos , Animales , Anticuerpos Monoclonales/administración & dosificación , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos , Ratones Obesos
14.
Cytokine Growth Factor Rev ; 28: 63-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26559461

RESUMEN

The FGF family comprises 22 members with diverse functions in development and health. FGF10 specifically activates FGFR2b in a paracrine manner with heparan sulfate as a co-factor. FGF10and FGFR2b are preferentially expressed in the mesenchyme and epithelium, respectively. FGF10 is a mesenchymal signaling molecule in the epithelium. FGF10 knockout mice die shortly after birth due to the complete absence of lungs as well as fore- and hindlimbs. FGF10 is also essential for the development of multiple organs. The phenotypes of Fgf10 knockout mice are very similar to those of FGFR2b knockout mice, indicating that FGF10 acts as a ligand that is specific to FGFR2b in mouse multi-organ development. FGF10 also plays roles in epithelial-mesenchymal transition, the repair of tissue injury, and embryonic stem cell differentiation. In humans, FGF10 loss-of-function mutations result in inherited diseases including aplasia of lacrimal and salivary gland, lacrimo-auriculo-dento-digital syndrome, and chronic obstructive pulmonary disease. FGF10 is also involved in the oncogenicity of pancreatic and breast cancers. Single nucleotide polymorphisms in FGF10 are also potential risk factors for limb deficiencies, cleft lip and palate, and extreme myopia. These findings indicate that FGF10 is a crucial paracrine signal from the mesenchyme to epithelium for development, health, and disease.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Mesodermo/metabolismo , Animales , Diferenciación Celular/fisiología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Humanos , Mesodermo/citología , Ratones , Ratones Noqueados , Comunicación Paracrina , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología
15.
Mol Cancer Ther ; 14(10): 2270-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26269606

RESUMEN

FGF receptors (FGFR) are attractive candidate targets for cancer therapy because they are dysregulated in several human malignancies. FGFR2 and FGFR3 can be inhibited potentially without disrupting adult tissue homeostasis. In contrast, blocking the closely related FGFR1 and FGFR4, which regulate specific metabolic functions, carries a greater safety risk. An anti-FGFR3 antibody was redesigned here to create function-blocking antibodies that bind with dual specificity to FGFR3 and FGFR2 but spare FGFR1 and FGFR4. R3Mab, a previously developed monospecific anti-FGFR3 antibody, was modified via structure-guided phage display and acquired additional binding to FGFR2. The initial variant was trispecific, binding tightly to FGFR3 and FGFR2 and moderately to FGFR4, while sparing FGFR1. The X-ray crystallographic structure indicated that the antibody variant was bound to a similar epitope on FGFR2 as R3Mab on FGFR3. The antibody was further engineered to decrease FGFR4-binding affinity while retaining affinity for FGFR3 and FGFR2. The resulting dual-specific antibodies blocked FGF binding to FGFR3 and FGFR2 and inhibited downstream signaling. Moreover, they displayed efficacy in mice against human tumor xenografts overexpressing FGFR3 or FGFR2. Thus, a monospecific antibody can be exquisitely tailored to confer or remove binding to closely related targets to expand and refine therapeutic potential.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/inmunología , Animales , Anticuerpos Monoclonales/química , Especificidad de Anticuerpos , Antineoplásicos/química , Línea Celular Tumoral , Cristalografía por Rayos X , Diseño de Fármacos , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones SCID , Simulación del Acoplamiento Molecular , Unión Proteica , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/química , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Mol Biotechnol ; 56(10): 939-52, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24899248

RESUMEN

KGFR is involved in the pathogenesis of several human cancers. In this study, we generated and characterized a monoclonal antibody specific to KGFR (SC-101 mAb) and evaluated its potential use in basic research and as a diagnostic and prognostic tool. The specificity and biological activity of the SC-101 mAb were evaluated by Western blotting, immunofluorescence, and immunoprecipitation analyses on various cell lines. KGFR expression in breast, pancreatic, and thyroid carcinoma was assessed by immunohistochemistry (IHC) with SC-101 mAb. KGFR expression levels revealed by SC-101 mAb resulted to increase proportionally with tumor grade in breast and pancreatic cancer. In addition, SC-101 mAb was able to detect KGFR down-modulation in thyroid cancer. SC-101 mAb might represent a useful tool for basic research applications, and it could also contribute to improve the accuracy of diagnosis and prognosis of epithelial tumors.


Asunto(s)
Anticuerpos Monoclonales , Neoplasias/diagnóstico , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Especificidad de Anticuerpos/inmunología , Línea Celular Tumoral , Factor 7 de Crecimiento de Fibroblastos/farmacología , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/inmunología , Pronóstico , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo
17.
Int J Clin Exp Pathol ; 5(6): 569-80, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22949940

RESUMEN

BACKGROUND: Epithelial cell (EC)-derived Interleukin-7 (IL-7) plays a crucial role in control of neighboring intestinal intraepithelial lymphocytes (IEL) development and homeostasis, and IEL derived keratinocyte growth factor (KGF) promotes intestinal epithelial growth, which was regulated by EC-derived IL-7. On this basis, we hypothesize that there is a crosstalk between IELs and ECs, and KGF could regulate the EC-derived IL-7 expression, which should be associated with the protective effects by KGF on intestinal injury. METHODS: Histological evaluation was performed in small intestine tissues of patients with intestinal obstruction and IL-7 expression was detected by immunofluorescence. Intestinal epithelial cells (LoVo) and adult C57BL/6J mice undergoing ischemia/reperfusion injury were treated with recombinant KGF. KGF, KGF receptor(KGFR) and IL-7 expressions were measured with western blot and immunofluorescence analysis. RESULTS: IL-7 expression increased in the mild ischemia while decreased in severe ischemia small intestinal tissues of patients with intestinal obstruction. KGF expression significantly decreased while IL-7 expression increased early after acute intestinal I/R administration in a mouse model. KGF treatment significantly increased the IL-7 expression both in vitro and in vivo, while when the KGFR was blocked, the findings above were absent. In addition, our results showed changes of IL-7 expression at different stages after acute intestinal I/R administration, KGF treatment significantly attenuated the decreasing of IL-7 expression caused by acute intestinal I/R. CONCLUSION: KGF could up-regulate the IL-7 expression both in vitro and in vivo through KGFR pathway, which should have associated with the protective effects of KGF in intestinal injury.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos/farmacología , Interleucina-7/metabolismo , Mucosa Intestinal/efectos de los fármacos , Obstrucción Intestinal/metabolismo , Isquemia/metabolismo , Daño por Reperfusión/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/patología , Obstrucción Intestinal/patología , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/farmacología , Daño por Reperfusión/patología , Regulación hacia Arriba
18.
Mol Cancer Ther ; 11(9): 2010-20, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22778155

RESUMEN

A high percentage of colorectal carcinomas overexpress a lot of growth factors and their receptors, including fibroblast growth factor (FGF) and FGF receptor (FGFR). We previously reported that FGFR2 overexpression was associated with distant metastasis and that FGFR2 inhibition suppressed cell growth, migration, and invasion. The FGFR2 splicing isoform FGFR2IIIb is associated with well-differentiated histologic type, tumor angiogenesis, and adhesion to extracellular matrices. Another isoform, FGFR2IIIc, correlates with the aggressiveness of various types of cancer. In the present study, we examined the expression and roles of FGFR2IIIc in colorectal carcinoma to determine the effectiveness of FGFR2IIIc-targeting therapy. In normal colorectal tissues, FGFR2IIIc expression was weakly detected in superficial colorectal epithelial cells and was not detected in proliferative zone cells. FGFR2IIIc-positive cells were detected by immunohistochemistry in the following lesions, listed in the order of increasing percentage: hyperplastic polyps < low-grade adenomas < high-grade adenomas < carcinomas. FGFR2IIIc immunoreactivity was expressed in 27% of colorectal carcinoma cases, and this expression correlated with distant metastasis and poor prognosis. FGFR2IIIc-transfected colorectal carcinoma cells showed increased cell growth, soft agar colony formation, migration, and invasion, as well as decreased adhesion to extracellular matrices. Furthermore, FGFR2IIIc-transfected colorectal carcinoma cells formed larger tumors in subcutaneous tissues and the cecum of nude mice. Fully human anti-FGFR2IIIc monoclonal antibody inhibited the growth and migration of colorectal carcinoma cells through alterations in cell migration, cell death, and development-related genes. In conclusion, FGFR2IIIc plays an important role in colorectal carcinogenesis and tumor progression. Monoclonal antibody against FGFR2IIIc has promising potential in colorectal carcinoma therapy.


Asunto(s)
Adenocarcinoma/metabolismo , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Neoplasias Colorrectales/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Adenoma/tratamiento farmacológico , Adenoma/metabolismo , Adenoma/patología , Anciano , Animales , Adhesión Celular , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transcriptoma/efectos de los fármacos
19.
Cancer Lett ; 309(2): 209-19, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21745712

RESUMEN

Fibroblast growth factor receptor 2 (FGFR2) is considered a novel therapeutic target for various cancer. We used a silencing strategy to clarify the effect of reduced FGFR2 expression in human colorectal cancer (CRC) cells. The invasive front of cancer cells exhibited stronger FGFR2 expression than the surface area of the cancers. FGFR2 shRNA-transfected LoVo cells inhibited cell migration, invasion and tumor growth in vitro and in vivo. Thus, FGFR2 plays important roles in CRC progression in association with tumor cell migration, invasion and growth, and FGFR2 might be a novel therapeutic target for CRC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/terapia , ARN Interferente Pequeño/farmacología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Anciano , Animales , Anticuerpos Monoclonales , Adhesión Celular , Movimiento Celular/genética , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Matriz Extracelular , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Transducción de Señal
20.
Cancer Res ; 70(19): 7630-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20709759

RESUMEN

Dysregulated fibroblast growth factor (FGF) signaling has been implicated in the pathogenesis of human cancers. Aberrant activation of FGF receptor 2 (FGFR2) signaling, through overexpression of FGFR2 and/or its ligands, mutations, and receptor amplification, has been found in a variety of human tumors. We generated monoclonal antibodies against the extracellular ligand-binding domain of FGFR2 to address the role of FGFR2 in tumorigenesis and to explore the potential of FGFR2 as a novel therapeutic target. We surveyed a broad panel of human cancer cell lines for the dysregulation of FGFR2 signaling and discovered that breast and gastric cancer cell lines harboring FGFR2 amplification predominantly express the IIIb isoform of the receptor. Therefore, we used an FGFR2-IIIb-specific antibody, GP369, to investigate the importance of FGFR2 signaling in vitro and in vivo. GP369 specifically and potently suppressed ligand-induced phosphorylation of FGFR2-IIIb and downstream signaling, as well as FGFR2-driven proliferation in vitro. The administration of GP369 in mice significantly inhibited the growth of human cancer xenografts harboring activated FGFR2 signaling. Our findings support the hypothesis that dysregulated FGFR2 signaling is one of the critical oncogenic pathways involved in the initiation and/or maintenance of tumors. Cancer patients with aberrantly activated/amplified FGFR2 signaling could potentially benefit from therapeutic intervention with FGFR2-targeting antibodies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias/terapia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Amplificación de Genes , Humanos , Ratones , Ratones SCID , Datos de Secuencia Molecular , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/inmunología , Fosforilación/efectos de los fármacos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA