Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 285
Filter
1.
Sci Rep ; 14(1): 9284, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38654040

ABSTRACT

Bromodomain and extra-terminal domain (BET) proteins are therapeutic targets in several cancers including the most common malignant adult brain tumor glioblastoma (GBM). Multiple small molecule inhibitors of BET proteins have been utilized in preclinical and clinical studies. Unfortunately, BET inhibitors have not shown efficacy in clinical trials enrolling GBM patients. One possible reason for this may stem from resistance mechanisms that arise after prolonged treatment within a clinical setting. However, the mechanisms and timeframe of resistance to BET inhibitors in GBM is not known. To identify the temporal order of resistance mechanisms in GBM we performed quantitative proteomics using multiplex-inhibitor bead mass spectrometry and demonstrated that intrinsic resistance to BET inhibitors in GBM treatment occurs rapidly within hours and involves the fibroblast growth factor receptor 1 (FGFR1) protein. Additionally, small molecule inhibition of BET proteins and FGFR1 simultaneously induces synergy in reducing GBM tumor growth in vitro and in vivo. Further, FGFR1 knockdown synergizes with BET inhibitor mediated reduction of GBM cell proliferation. Collectively, our studies suggest that co-targeting BET and FGFR1 may dampen resistance mechanisms to yield a clinical response in GBM.


Subject(s)
Brain Neoplasms , Bromodomain Containing Proteins , Cell Proliferation , Drug Resistance, Neoplasm , Glioblastoma , Receptor, Fibroblast Growth Factor, Type 1 , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Humans , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Mice , Xenograft Model Antitumor Assays , Proteomics/methods , Proteins/metabolism , Proteins/antagonists & inhibitors
2.
Eur J Med Chem ; 271: 116415, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38643670

ABSTRACT

Fibroblast growth factor receptor (FGFR) is an attractive target for cancer therapy, but existing FGFR inhibitors appear to hardly meet the demand for clinical application. Herein, a number of irreversible covalent FGFR inhibitors were designed and synthesized by selecting several five- and six-membered azaheterocycles as parent scaffold with different substituents to take over the hydrophobic region in the active pocket of FGFR proteins. Among the resulting target compounds, III-30 showed the most potent effect on enzyme activity inhibition and anti-proliferative activity against the tested cancer cell lines. Significantly, III-30 could inhibit the enzyme activity by achieving irreversible covalent binding with FGFR1 and FGFR4 proteins. It could also regulate FGFR-mediated signaling pathway and mitochondrial apoptotic pathway to promote cancer cell apoptosis and inhibit cancer cell invasion and metastasis. Moreover, III-30 had a good metabolic stability and showed relatively potent anti-tumor activity in the MDA-MB-231 xenograft tumor mice model.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Drug Screening Assays, Antitumor , Protein Kinase Inhibitors , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Mice , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Molecular Structure , Cell Line, Tumor , Purines/pharmacology , Purines/chemistry , Purines/chemical synthesis , Drug Discovery , Apoptosis/drug effects , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Dose-Response Relationship, Drug , Mice, Nude , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neoplasms, Experimental/metabolism , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Female
3.
Acta Pharmacol Sin ; 45(5): 988-1001, 2024 May.
Article in English | MEDLINE | ID: mdl-38279043

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a common metabolic disease that is substantially associated with obesity-induced chronic inflammation. Macrophage activation and macrophage-medicated inflammation play crucial roles in the development and progression of NAFLD. Furthermore, fibroblast growth factor receptor 1 (FGFR1) has been shown to be essentially involved in macrophage activation. This study investigated the role of FGFR1 in the NAFLD pathogenesis and indicated that a high-fat diet (HFD) increased p-FGFR1 levels in the mouse liver, which is associated with increased macrophage infiltration. In addition, macrophage-specific FGFR1 knockout or administration of FGFR1 inhibitor markedly protected the liver from HFD-induced lipid accumulation, fibrosis, and inflammatory responses. The mechanistic study showed that macrophage-specific FGFR1 knockout alleviated HFD-induced liver inflammation by suppressing the activation of MAPKs and TNF signaling pathways and reduced fat deposition in hepatocytes, thereby inhibiting the activation of hepatic stellate cells. In conclusion, the results of this research revealed that FGFR1 could protect the liver of HFD-fed mice by inhibiting MAPKs/TNF-mediated inflammatory responses in macrophages. Therefore, FGFR1 can be employed as a target to prevent the development and progression of NAFLD.


Subject(s)
Diet, High-Fat , Macrophages , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Receptor, Fibroblast Growth Factor, Type 1 , Tumor Necrosis Factor-alpha , Animals , Diet, High-Fat/adverse effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Macrophages/metabolism , Macrophages/drug effects , Mice , Male , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/etiology , Tumor Necrosis Factor-alpha/metabolism , Mice, Knockout , Liver/pathology , Liver/metabolism , Signal Transduction , Inflammation/metabolism , MAP Kinase Signaling System/drug effects
4.
J Biomol Struct Dyn ; 42(5): 2242-2256, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37211823

ABSTRACT

Developing highly potent covalent inhibitors of Fibroblast growth factor receptors 1 (FGFR1) has always been a challenging task. In the current study, various computational techniques, such as 3D-QSAR, covalent docking, fingerprinting analysis, MD simulation followed by MMGB/PBSA, and per-residue energy decomposition analysis were used to explore the binding mechanism of pyrazolo[3,4-d]pyridazinone derivatives to FGFR1. The high q2 and r2 values for the CoMFA and CoMSIA models, suggest that the constructed 3D-QSAR models could reliably predict the bioactivities of FGFR1 inhibitors. The structural requirements revealed by the model's contour maps were strategically used to computationally create an in-house library of more than 100 new FGFR1 inhibitors using the R-group exploration technique implemented in the SparkTM software. The compounds from the in-house library were also mapped in the 3D-QSAR model that predicts comparable pIC50 values with the experimental values. A comparison between 3D-QSAR generated contours and molecular docking conformation of ligands was performed to reveal the fundamentals to design potent FGFR1 covalent inhibitors. The estimated binding free energies (MMGB/PBSA) for the selected compounds were in agreement with the experimental value ranking of their binding affinities towards FGFR1. Furthermore, per-residue energy decomposition analysis has identified Arg627 and Glu531 to contribute significantly in improved binding affinity of compound W16. During ADME analysis, the majority of in-house library compounds exhibited pharmacokinetic properties superior to those of experimentally produced compounds. These new compounds may help researchers better understand FGFR1 inhibition and lead to the creation of novel, potent FGFR1 inhibitors.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Molecular Dynamics Simulation , Pyrazoles , Pyridazines , Receptor, Fibroblast Growth Factor, Type 1 , Antineoplastic Agents/pharmacology , Molecular Docking Simulation , Protein Binding , Quantitative Structure-Activity Relationship , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridazines/chemistry , Pyridazines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
5.
J Biol Chem ; 299(1): 102729, 2023 01.
Article in English | MEDLINE | ID: mdl-36410439

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) is a receptor tyrosine kinase that plays a major role in developmental processes and metabolism. The dysregulation of FGFR1 through genetic aberrations leads to skeletal and metabolic diseases as well as cancer. For this reason, FGFR1 is a promising therapeutic target, yet a very challenging one due to potential on-target toxicity. More puzzling is that both agonistic and antagonistic FGFR1 antibodies are reported to exhibit similar toxicity profiles in vivo, namely weight loss. In this study, we aimed to assess and compare the mechanism of action of these molecules to better understand this apparent contradiction. By systematically comparing the binding of these antibodies and the activation or the inhibition of the major FGFR1 signaling events, we demonstrated that the molecules displayed similar properties and can behave either as an agonist or antagonist depending on the presence or the absence of the endogenous ligand. We further demonstrated that these findings translated in xenografts mice models. In addition, using time-resolved FRET and mass spectrometry analysis, we showed a functionally distinct FGFR1 active conformation in the presence of an antibody that preferentially activates the FGFR substrate 2 (FRS2)-dependent signaling pathway, demonstrating that modulating the geometry of a FGFR1 dimer can effectively change the signaling outputs and ultimately the activity of the molecule in preclinical studies. Altogether, our results highlighted how bivalent antibodies can exhibit both agonistic and antagonistic activities and have implications for targeting other receptor tyrosine kinases with antibodies.


Subject(s)
Antibodies, Monoclonal , Receptor, Fibroblast Growth Factor, Type 1 , Signal Transduction , Animals , Humans , Mice , Neoplasms , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology
6.
s.l; CONETEC; 30 oct. 2022.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-1400072

ABSTRACT

INTRODUCCIÓN: Los colangiocarcinomas abarcan todos los tumores que se originan en el epitelio de la vía biliar. Más del 90 % son adenocarcinomas y generalmente se clasifican como intrahepáticos o extrahepático. La mediana de edad en el momento del diagnóstico es de 65 años, y tasas de sobrevida relativa a los 5 años son cercanas al 10%. La resección completa con márgenes negativos es el único tratamiento potencialmente curativo para pacientes con enfermedad resecable. Sin embargo, la mayoría de los pacientes son diagnosticados con enfermedad no resecable, localmente avanzada o metastásica debido a la ausencia de síntomas hasta un momento posterior del curso de la enfermedad. Para los pacientes con estadio avanzado o irresecable y de buen estado general, el tratamiento de primera línea estándar es gemcitabina y cisplatino. En el entorno de segunda línea una vez que la enfermedad ha progresado el esquema de quimioterapia mFOLFOX (ácido folínico o leucovorina, fluorouracilo y oxaliplatino) es el régimen de tratamiento preferido. El perfil molecular de los colangiocarcinomas desempeña un papel cada vez más importante en la determinación del pronóstico y la selección de tratamientos. Recientemente se han identificado alteraciones genéticas del receptor del factor de crecimiento fibroblástico (FGFR, su sigla del inglés Fibroblast Growth Factor Receptor) en aproximadamente en un 10-16% de los colangiocarcinomas intrahepáticos y se han denominado indistintamente traslocaciones, fusiones o reordenamientos. Estas fusiones o reordenamientos rara vez se producen en los colangiocarcinomas extra-hepáticos. TECNOLOGÍA: Pemigatinib (Pemazyre®) es un inhibidor de la cinasa de FGFR1, 2 y 3 que inhibe la fosforilación y la señalización de FGFR y disminuye la viabilidad celular en las células que expresan alteraciones genéticas de FGFR, incluyendo mutaciones puntuales, amplificaciones y fusiones o reordenamientos. OBJETIVO: El objetivo del presente informe es evaluar rápidamente los parámetros de eficacia, seguridad, costos y recomendaciones disponibles acerca del empleo de pemigatinib en el tratamiento de pacientes con colangiocarcinoma. MÉTODOS: Se realizó una búsqueda bibliográfica en las principales bases de datos tales como PUBMED, LILACS, BRISA, COCHRANE, SCIELO, EMBASE, TRIPDATABASE como así también en sociedades científicas, agencias reguladoras, financiadores de salud y agencias de evaluación de tecnologías sanitarias. Se priorizó la inclusión de revisiones sistemáticas, ensayos clínicos controlados aleatorizados, evaluación de tecnología sanitaria y guías de práctica clínica de alta calidad metodológica. En PubMed se utilizó la estrategia de búsqueda que se detalla en el Anexo I. La fecha de búsqueda de información fue hasta el 24 de octubre de 2022. Para la búsqueda en Pubmed se utilizó la siguiente estrategia de búsqueda: (pemigatinib [Supplementary Concept] OR pemigatinib [tiab] OR Pemazyre [tiab] OR INCB054828 [tiab]) AND (Cholangiocarcinoma [MESH] OR cholangiocarcinoma [tiab]). RECOMENDACIONES: Las guías de la Red Nacional de Centros para el Tratamiento Integral del cáncer (NCCN, su sigla del inglés National Comprehensive Cancer Network) de los Estados Unidos, menciona al pemigatinib como una de las opciones de tratamiento para el colangiocarcinoma irresecable o metastásico con fusiones o reordenamientos de FGFR2, después de la progresión de la enfermedad a una primera línea de tratamento. La Sociedad Española de Oncología Médica (SEOM) recomienda que los pacientes candidatos al tratamiento deben enrolarse en ensayos clínicos. Añade que cuando esté disponible el fármaco en el país, se debería realizar una caracterización molecular exhaustiva del tumor en aquellos pacientes con estadios avanzado; ya que ciertas alteraciones genéticas, como las mutaciones de IDH1, las fusiones de FGFR2, mutaciones del gen BRAF, la inestabilidad de los microsatélites y los reordenamientos de NTRK, entre otros, pueden beneficiarse de terapias dirigidas específicas. La valoración del beneficio clínico según la de la Sociedad Europea de Oncología Médica (ESMO, su sigla del inglés European Society for Medical Oncology) para considerar la relevancia del beneficio clínico de forma sistematizada para este fármaco en la indicación evaluada es de 3 puntos. Según esta escala las calificaciones más altas en el ámbito no curativo son 5 y 4, que indican una magnitud sustancial del beneficio clínico. En Argentina, la Asociación de Oncología Clínica (AAOC) en sus recomendaciones actuales para el tratamiento oncológico del cáncer de vía biliar no menciona su uso dentro de las opciones terapêuticas. CONCLUSIONES: La eficacia y seguridad de pemigatinib frente a otros fármacos disponibles en la indicación evaluada no pudo ser establecida debido a que no se encontraron estudios comparativos. La evidencia que sustenta la aprobación de comercialización de pemigatinib para el tratamiento de adultos con colangiocarcinoma avanzado progresados a una primera línea de tratamiento y con alteraciones moleculares en el receptor del factor de crecimiento de fibroblastos 2, se basa en un único ensayo clínico no aleatorizado en pocos participantes. Este estudio mostró que aquellos adultos que utilizaron el tratamiento tendrían una tasa de respuesta cercanas al 37% al mediano plazo. Las agencias regulatorias relevadas han autorizado su comercialización de forma acelerada, bajo la denominación de medicamento huérfano, y sujeta a un ensayo de confirmación por parte del productor de la tecnología. Una guía de la Asociación Argentina de Oncología Clínica actualizada no lo menciona dentro de las opciones actuales de tratamiento, mientras que el resto de las recomendaciones provenientes de países de altos ingresos relevados lo mencionan como una opción terapéutica. Reino Unido brinda cobertura en la indicación evaluada sujeta a un acuerdo de comercialización con un descuento en el precio de venta. Otros países, como España y Canadá, no han autorizado su cobertura debido a que consideran que la evidencia que sustenta su utilización es insuficiente y existe incertidumbre sobre su beneficio en comparación con otras alternativas terapéuticas disponibles. No se hallaron evaluaciones económicas publicadas, aunque el costo del fármaco es muy elevado.


Subject(s)
Humans , Cholangiocarcinoma/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Argentina , Efficacy , Cost-Benefit Analysis
7.
Cancer Discov ; 12(2): 402-415, 2022 02.
Article in English | MEDLINE | ID: mdl-34551969

ABSTRACT

Futibatinib, a highly selective, irreversible FGFR1-4 inhibitor, was evaluated in a large multihistology phase I dose-expansion trial that enrolled 197 patients with advanced solid tumors. Futibatinib demonstrated an objective response rate (ORR) of 13.7%, with responses in a broad spectrum of tumors (cholangiocarcinoma and gastric, urothelial, central nervous system, head and neck, and breast cancer) bearing both known and previously uncharacterized FGFR1-3 aberrations. The greatest activity was observed in FGFR2 fusion/rearrangement-positive intrahepatic cholangiocarcinoma (ORR, 25.4%). Some patients with acquired resistance to a prior FGFR inhibitor also experienced responses with futibatinib. Futibatinib demonstrated a manageable safety profile. The most common treatment-emergent adverse events were hyperphosphatemia (81.2%), diarrhea (33.5%), and nausea (30.4%). These results formed the basis for ongoing futibatinib phase II/III trials and demonstrate the potential of genomically selected early-phase trials to help identify molecular subsets likely to benefit from targeted therapy. SIGNIFICANCE: This phase I dose-expansion trial demonstrated clinical activity and tolerability of the irreversible FGFR1-4 inhibitor futibatinib across a broad spectrum of FGFR-aberrant tumors. These results formed the rationale for ongoing phase II/III futibatinib trials in cholangiocarcinoma, breast cancer, gastroesophageal cancer, and a genomically selected disease-agnostic population.This article is highlighted in the In This Issue feature, p. 275.


Subject(s)
Antineoplastic Agents/therapeutic use , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Bile Duct Neoplasms/mortality , Cholangiocarcinoma/mortality , Disease-Free Survival , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrroles/administration & dosage , Pyrroles/adverse effects , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
8.
Bioorg Chem ; 119: 105504, 2022 02.
Article in English | MEDLINE | ID: mdl-34836644

ABSTRACT

This work deals with the design and synthesis of a series of new substituted 2-arylbenzothiazole compounds attached to 4-oxothiazolidin-2-ylidene ring 2-12 and chain elongation with different amino acids and their corresponding ester derivatives 13-18. All prepared derivatives were screened for their in vitro cytotoxicity activities against two cancer cell lines (HepG-2 and MCF-7) in comparison with doxorubicin; in addition to their safety towards thenormal cell line. Furthermore, all compounds 2-18 were evaluated as FGFR-1 inhibitors using AZD4547 as a reference. The 4-oxothiazolidin-2-ylidene derivatives 3 and 8 exhibited the highest cytotoxic activity (IC50 HepG-2 = 2.06, 2.21 µM and IC50 MCF-7 = 0.73, 0.77 µM, respectively) through their promising FGFR-1 suppression effects (IC50 = 16.31 and 18.08 nM, respectively) in comparison to AZD4547 (IC50 = 21.45 nM). Cell cycle and apoptosis analysis indicated that compounds 3 and 8 induce pronounced increase in the cell percentages at pre-G1 and G2/M phase compared to the untreated MCF-7 cancer cells, in addition to their up regulation of caspase-3/7/9. The molecular docking simulation was created to elucidate the binding modes of benzothiazole derivatives 1-18 bearing various scaffolds within the ATP-binding pocket of FGFR-1 enzyme compared with AZD4547.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Structure-Activity Relationship
9.
Cells ; 10(12)2021 11 30.
Article in English | MEDLINE | ID: mdl-34943871

ABSTRACT

FGFR signalling is one of the most prominent pathways involved in cell growth and development as well as cancer progression. FGFR1 amplification occurs in approximately 20% of all squamous cell lung carcinomas (SCC), a predominant subtype of non-small cell lung carcinoma (NSCLC), indicating FGFR as a potential target for the new anti-cancer treatment. However, acquired resistance to this type of therapies remains a serious clinical challenge. Here, we investigated the NSCLC cell lines response and potential mechanism of acquired resistance to novel selective FGFR inhibitor CPL304110. We found that despite significant genomic differences between CPL304110-sensitive cell lines, their resistant variants were characterised by upregulated p38 expression/phosphorylation, as well as enhanced expression of genes involved in MAPK signalling. We revealed that p38 inhibition restored sensitivity to CPL304110 in these cells. Moreover, the overexpression of this kinase in parental cells led to impaired response to FGFR inhibition, thus confirming that p38 MAPK is a driver of resistance to a novel FGFR inhibitor. Taken together, our results provide an insight into the potential direction for NSCLC targeted therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Humans , Receptor, Fibroblast Growth Factor, Type 1/metabolism
10.
Cells ; 10(10)2021 10 13.
Article in English | MEDLINE | ID: mdl-34685716

ABSTRACT

Mitochondria, abundant organelles in high energy demand cells such as cardiomyocytes, can determine cell death or survival by regulating the opening of mitochondrial permeability transition pore, mPTP. We addressed the hypothesis that the growth factor FGF2, known to reside in intracellular locations, can directly influence mitochondrial susceptibility to mPTP opening. Rat cardiac subsarcolemmal (SSM) or interfibrillar (IFM) mitochondrial suspensions exposed directly to rat 18 kDa low molecular weight (Lo-) FGF2 isoform displayed increased resistance to calcium overload-induced mPTP, measured spectrophotometrically as "swelling", or as cytochrome c release from mitochondria. Inhibition of mitochondrial protein kinase C epsilon abrogated direct Lo-FGF2 mito-protection. Exposure to the rat 23 kDa high molecular weight (Hi) FGF2 isoform promoted cytochrome c release from SSM and IFM under nonstressed conditions. The effect of Hi-FGF2 was prevented by mPTP inhibitors, pre-exposure to Lo-FGF2, and okadaic acid, a serine/threonine phosphatase inhibitor. Western blotting and immunoelectron microscopy pointed to the presence of immunoreactive FGFR1 in cardiac mitochondria in situ. The direct mito-protective effect of Lo-FGF2, as well as the deleterious effect of Hi-FGF2, were prevented by FGFR1 inhibitors and FGFR1 neutralizing antibodies. We propose that intracellular FGF2 isoforms can modulate mPTP opening by interacting with mito-FGFR1 and relaying isoform-specific intramitochondrial signal transduction.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Calcium/metabolism , Cytochromes c/metabolism , Male , Mitochondria, Heart/ultrastructure , Protein Isoforms/metabolism , Protein Kinase C-epsilon/metabolism , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
11.
Lancet Gastroenterol Hepatol ; 6(10): 803-815, 2021 10.
Article in English | MEDLINE | ID: mdl-34358484

ABSTRACT

BACKGROUND: Treatment options are sparse for patients with advanced cholangiocarcinoma after progression on first-line gemcitabine-based therapy. FGFR2 fusions or rearrangements occur in 10-16% of patients with intrahepatic cholangiocarcinoma. Infigratinib is a selective, ATP-competitive inhibitor of fibroblast growth factor receptors. We aimed to evaluate the antitumour activity of infigratinib in patients with locally advanced or metastatic cholangiocarcinoma, FGFR2 alterations, and previous gemcitabine-based treatment. METHODS: This multicentre, open-label, single-arm, phase 2 study recruited patients from 18 academic centres and hospitals in the USA, Belgium, Spain, Germany, Singapore, Taiwan, and Thailand. Eligible participants were aged 18 years or older, had histologically or cytologically confirmed, locally advanced or metastatic cholangiocarcinoma and FGFR2 fusions or rearrangements, and were previously treated with at least one gemcitabine-containing regimen. Patients received 125 mg of oral infigratinib once daily for 21 days of 28-day cycles until disease progression, intolerance, withdrawal of consent, or death. Radiological tumour evaluation was done at baseline and every 8 weeks until disease progression via CT or MRI of the chest, abdomen, and pelvis. The primary endpoint was objective response rate, defined as the proportion of patients with a best overall response of a confirmed complete or partial response, as assessed by blinded independent central review (BICR) according to Response Evaluation Criteria in Solid Tumors, version 1.1. The primary outcome and safety were analysed in the full analysis set, which comprised all patients who received at least one dose of infigratinib. This trial is registered with ClinicalTrials.gov, NCT02150967, and is ongoing. FINDINGS: Between June 23, 2014, and March 31, 2020, 122 patients were enrolled into our study, of whom 108 with FGFR2 fusions or rearrangements received at least one dose of infigratinib and comprised the full analysis set. After a median follow-up of 10·6 months (IQR 6·2-15·6), the BICR-assessed objective response rate was 23·1% (95% CI 15·6-32·2; 25 of 108 patients), with one confirmed complete response in a patient who only had non-target lesions identified at baseline and 24 partial responses. The most common treatment-emergent adverse events of any grade were hyperphosphataemia (n=83), stomatitis (n=59), fatigue (n=43), and alopecia (n=41). The most common ocular toxicity was dry eyes (n=37). Central serous retinopathy-like and retinal pigment epithelial detachment-like events occurred in 18 (17%) patients, of which ten (9%) were grade 1, seven (6%) were grade 2, and one (1%) was grade 3. There were no treatment-related deaths. INTERPRETATION: Infigratinib has promising clinical activity and a manageable adverse event profile in previously treated patients with locally advanced or metastatic cholangiocarcinoma harbouring FGFR2 gene fusions or rearrangements, and so represents a potential new therapeutic option in this setting. FUNDING: QED Therapeutics and Novartis.


Subject(s)
Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Neoplasm Metastasis/drug therapy , Phenylurea Compounds/therapeutic use , Pyrimidines/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Alopecia/chemically induced , Alopecia/epidemiology , Central Serous Chorioretinopathy/chemically induced , Central Serous Chorioretinopathy/epidemiology , Cholangiocarcinoma/secondary , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Disease Progression , Dry Eye Syndromes/chemically induced , Dry Eye Syndromes/epidemiology , Fatigue/chemically induced , Fatigue/epidemiology , Female , Humans , Hyperphosphatemia/chemically induced , Hyperphosphatemia/epidemiology , Male , Middle Aged , Neoplasm Metastasis/pathology , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/genetics , Retinal Detachment/chemically induced , Retinal Detachment/epidemiology , Safety , Stomatitis/chemically induced , Stomatitis/epidemiology , Treatment Outcome , Gemcitabine
12.
J Med Chem ; 64(15): 10666-10679, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34269576

ABSTRACT

Aberrant activation of FGFR has been linked to the pathogenesis of many tumor types. Selective inhibition of FGFR has emerged as a promising approach for cancer treatment. Herein, we describe the discovery of compound 38 (INCB054828, pemigatinib), a highly potent and selective inhibitor of FGFR1, FGFR2, and FGFR3 with excellent physiochemical properties and pharmacokinetic profiles. Pemigatinib has received accelerated approval from the U.S. Food and Drug Administration for the treatment of adults with previously treated, unresectable locally advanced or metastatic cholangiocarcinoma with a FGFR2 fusion or other rearrangement. Additional clinical trials are ongoing to evaluate pemigatinib in patients with FGFR alterations.


Subject(s)
Drug Discovery , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Structure , Morpholines/chemical synthesis , Morpholines/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Structure-Activity Relationship , United States , United States Food and Drug Administration
13.
Bioorg Med Chem Lett ; 48: 128258, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34246754

ABSTRACT

The present work represents the design and synthesis of some azaheterocyclic coumarin derivatives which are evaluated as anti-lung cancer agents. Ten out of the twenty azaheterocyclic compounds showed superior activity than the standard drug staurosporine against non-small cell lung cancer (A549). Representing the four different azaheterocyclic series, compounds 4a, 5d, 6e, and 7d, which demonstrated IC50s of 2.38, 2.39, 1.05 and 3.98 µM, respectively, each exhibiting the best cytotoxicity in its group, were selected for further assessment of their toxicity on normal lung cells (WI-38). Compound 4a was selected for further investigations because it remarkably revealed less cytotoxicity (IC50 = 53.76 µM) than 7d (IC50 = 19.95 µM) on (WI-38) compared to staurosporine (IC50 = 24.41 µM). 4a was assessed for its ability to inhibit the angiokinases VEGFR-2, PDGFR, FGFR and the growth factor EGFR, remarkably it showed better VEGFR-2, PDGFR, FGFR inhibition than the reference drugs used and exhibited as well noticeable EGFR inhibition. Going further, 4a was capable of arresting the cell cycle at pre-G1 phase and S phase and inducing apoptosis. Moreover, the capability of the target 4a to interact with the key amino acids of VEGFR-2 binding site was detected by molecular docking. Finally, the in silico physicochemical properties of 4a were studied.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Growth Inhibitors/pharmacology , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Growth Inhibitors/chemical synthesis , Growth Inhibitors/chemistry , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Structure-Activity Relationship , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
J Cancer Res Clin Oncol ; 147(10): 2955-2968, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34156519

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC), the most common manifestation of liver cancer, is one of the leading causes of cancer-related mortality worldwide with limited treatment options. Infigratinib, a pan-FGFR inhibitor, has shown a potent antitumour effect in HCC. However, drug resistance is often observed in long-term treatment. In this study, we examined the potential feedback mechanism(s) leading to infigratinib and explored a combination therapy to overcome resistance in HCC. METHODS: Patient-derived xenograft (PDX) tumours were subcutaneously implanted into SCID mice and were subsequently treated with infigratinib. Tumour growth was monitored over time, and tumour samples were subjected to immunohistochemistry and Western blotting. For drug combination studies, mice were treated with infigratinib and/or varlitinib. Gene overexpression and knockdown studies were conducted to investigate the relationship between EZH2 and ErbB activity in infigratinib resistance. RESULTS: Infigratinib-resistant tumours exhibited higher levels of p-ErbB2 and p-ErbB3, concomitant with an increase in EZH2 expression. Gene overexpression and knockdown studies revealed that EZH2 directly regulates the levels of p-ErbB2 and p-ErbB3 in acquired resistance to infigratinib. The addition of varlitinib effectively overcame infigratinib resistance and prolonged the antitumour response, with minimal toxicity. CONCLUSION: The upregulation of the ErbB family by EZH2 appears to contribute to infigratinib resistance. The combination of infigratinib and varlitinib showed a potent antitumour effect and did not result in additional toxicity, warranting further clinical investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm/drug effects , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Receptor, ErbB-2/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Receptor, ErbB-2/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
J Med Chem ; 64(13): 9078-9099, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34129329

ABSTRACT

Fibroblast growth factor receptors (FGFRs) have become promising therapeutic targets in various types of cancers. In fact, several selective irreversible inhibitors capable of covalently reacting with the conserved cysteine of FGFRs are currently being evaluated in clinical trials. In this article, we optimized and discovered a novel lead compound 36 with remarkable inhibitory effects against FGFR (1-3), which is a derivative of 2H-pyrazolo[3,4-d]pyrimidine. The irreversible binding to FGFRs was characterized by LC-MS. This compound has been shown to exhibit significant anti-proliferation effects against NCI-H1581 and SNU-16 cancer cell lines both in vitro and in vivo. Compound 36 has also demonstrated a low toxicity profile and adequate pharmacokinetic properties and is currently under validation as a potential drug candidate.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
16.
Cancer Med ; 10(14): 4874-4884, 2021 07.
Article in English | MEDLINE | ID: mdl-34114373

ABSTRACT

Abnormallyactivated FGFR1 has been validated as a therapeutic target for differentcancers. Although a variety of FGFR inhibitors have shown benefit in manyclinical patients with FGFR1 aberration, FGFR1 mutant resistance such as V561Mmutation, has been reported. To date however, no FGFR inhibitors have beenapproved to treat patients with FGFR mutant resistance. Herein, we report that GZD824, athird generation ABL inhibitor (Phase II, China), overcomes FGFR1-V561F/M mutant resistance in vitro and in vivo. GZD824potently suppresses FGFR1/2/3 with an IC50 value of 4.14 ± 0.96, 2.77 ± 0.082, and 8.10 ± 0.15 nmol/L. It effectively overcomes FGFR1-V561F/M and other mutantresistance in Ba/F3 stable cells (IC50 :8.1-55.0 nM), and effectively inhibits the growth of Ba/F3-FGFR1-V561F/M mutantxenograft tumors in vivo (TGI=73.4%, 49.8% at20mg/kg, p.o, q2d). GZD824may be considered to be an effective drug to treat patients with FGFR1 abnormalactivation or mutant resistance in clinical trials.


Subject(s)
Benzamides/pharmacology , Drug Resistance, Neoplasm/drug effects , Mutation , Pyrazoles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, SCID , Morpholines/pharmacology , Neoplasm Transplantation , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Pyrimidines/pharmacology , Pyrroles/pharmacology , Quinoxalines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors
17.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946884

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive breast tumor subtype characterized by poor clinical outcome. In recent years, numerous advancements have been made to better understand the biological landscape of TNBC, though appropriate targets still remain to be determined. In the present study, we have determined that the expression levels of FGF2 and S100A4 are higher in TNBC with respect to non-TNBC patients when analyzing "The Invasive Breast Cancer Cohort of The Cancer Genome Atlas" (TCGA) dataset. In addition, we have found that the gene expression of FGF2 is positively correlated with S100A4 in TNBC samples. Performing quantitative PCR, Western blot, CRISPR/Cas9 genome editing, promoter studies, immunofluorescence analysis, subcellular fractionation studies, and ChIP assays, we have also demonstrated that FGF2 induces in TNBC cells the upregulation and secretion of S100A4 via FGFR1, along with the ERK1/2-AKT-c-Rel transduction signaling. Using conditioned medium from TNBC cells stimulated with FGF2, we have also ascertained that the paracrine activation of the S100A4/RAGE pathway triggers angiogenic effects in vascular endothelial cells (HUVECs) and promotes the migration of cancer-associated fibroblasts (CAFs). Collectively, our data provide novel insights into the action of the FGF2/FGFR1 axis through S100A4 toward stimulatory effects elicited in TNBC cells.


Subject(s)
Fibroblast Growth Factor 2/physiology , Neoplasm Proteins/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , S100 Calcium-Binding Protein A4/physiology , Signal Transduction/physiology , Triple Negative Breast Neoplasms/physiopathology , Antigens, Neoplasm/physiology , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/physiology , Human Umbilical Vein Endothelial Cells , Humans , Mitogen-Activated Protein Kinases/physiology , Neovascularization, Pathologic/physiopathology , Paracrine Communication , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-rel/physiology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/blood supply , Tumor Cells, Cultured
18.
Mol Med ; 27(1): 46, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33962559

ABSTRACT

BACKGROUND: Antibody drug conjugates (ADCs) represent one of the most promising approaches in the current immuno-oncology research. The precise delivery of cytotoxic drugs to the cancer cells using ADCs specific for tumor-associated antigens enables sparing the healthy cells and thereby reduces unwanted side effects. Overexpression of fibroblast growth factor receptor 1 (FGFR1) has been demonstrated in numerous tumors and thereby constitutes a convenient molecular target for selective cancer treatment. We have recently engineered tetravalent anti-FGFR1 antibody, T-Fc, and have demonstrated that it displays extremely efficient internalization into FGFR1 producing cells, a feature highly desirable in the ADC approach. We have revealed that T-Fc mediates clustering of FGFR1, largely enhancing the uptake of FGFR1-T-Fc complexes by induction of clathrin-independent endocytic routes. The aim of this study was to obtain highly internalizing cytotoxic conjugate of the T-Fc for specific delivery of drugs into FGFR1-positive cancer cells. METHODS: Conjugation of the T-Fc to a cytotoxic payload, vcMMAE, was carried out via maleimide chemistry, yielding the T-Fc-vcMMAE. The specific binding of the T-Fc-vcMMAE conjugate to FGFR1 was confirmed in vitro with BLI technique. Confocal microscopy and flow cytometry were applied to determine FGFR1-dependence of the T-Fc-vcMMAE internalization. Western blot analyses of FGFR1-dependent signaling were conducted to assess the impact of the T-Fc-vcMMAE on FGFR1 activation and initiation of downstream signaling cascades. Finally, using FGFR1-negative and FGFR1-possitive cell lines, the cytotoxic potential of the T-Fc-vcMMAE was evaluated. RESULTS: We have performed the efficient conjugation of the tetravalent engineered antibody with a cytotoxic drug and generated FGFR1-specific ADC molecule, T-Fc-vcMMAE. We have demonstrated that T-Fc-vcMMAE conjugate exhibits high selectivity and affinity for FGFR1, similarly to T-Fc. Furthermore, we have shown that T-Fc constitutes an effective drug delivery vehicle as T-Fc-vcMMAE was efficiently and selectively internalized by FGFR1-producing cells leading to their death. Interestingly, we show that the efficiency of the uptake of T-Fc-vcMMAE corresponds well with the cytotoxicity of the conjugate, but doesn't correlate with the FGFR1expression level. CONCLUSION: Our results show that T-Fc-vcMMAE fulfills the key criteria for the successful cytotoxic drug carrier in a targeted approach against FGFR1-positive cancer cells. Furthermore, our data implicate that not solely expression level of the receptor, but rather its cellular trafficking should be taken into account for selection of suitable molecular targets and cancer models for successful ADC approach.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Immunoconjugates/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antineoplastic Agents, Immunological/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Fluorescent Antibody Technique , Gene Expression , Genetic Engineering , Humans , Immunoconjugates/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
19.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34042967

ABSTRACT

Regeneration as an adult developmental process is in many aspects similar to embryonic development. Although many studies point out similarities and differences, no large-scale, direct and functional comparative analyses between development and regeneration of a specific cell type or structure in one animal exist. Here, we use the brittle star Amphiura filiformis to characterise the role of the FGF signalling pathway during skeletal development in embryos and arm regeneration. In both processes, we find ligands expressed in ectodermal cells that flank underlying skeletal mesenchymal cells, which express the receptors. Perturbation of FGF signalling showed inhibited skeleton formation in both embryogenesis and regeneration, without affecting other key developmental processes. Differential transcriptome analysis finds mostly differentiation genes rather than transcription factors to be downregulated in both contexts. Moreover, comparative gene analysis allowed us to discover brittle star-specific differentiation genes. In conclusion, our results show that the FGF pathway is crucial for skeletogenesis in the brittle star, as in other deuterostomes, and provide evidence for the re-deployment of a developmental gene regulatory module during regeneration.


Subject(s)
Bone Development/physiology , Bone Regeneration/physiology , Bone and Bones/embryology , Fibroblast Growth Factors/metabolism , Starfish/embryology , Animals , Bone and Bones/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Embryonic Development/genetics , Extremities/growth & development , Mesoderm/cytology , Mesoderm/metabolism , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Signal Transduction/physiology , Starfish/genetics , Starfish/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
Eur J Cancer ; 151: 136-149, 2021 07.
Article in English | MEDLINE | ID: mdl-33984662

ABSTRACT

Amplification of fibroblast growth factor receptor 1 (FGFR1) in non-small cell lung cancer (NSCLC) has been considered as an actionable drug target. However, pan-FGFR tyrosine kinase inhibitors did not demonstrate convincing clinical efficacy in FGFR1-amplified NSCLC patients. This study aimed to characterise the molecular context of FGFR1 expression and to define biomarkers predictive of FGFR1 inhibitor response. In this study, 635 NSCLC samples were characterised for FGFR1 protein expression by immunohistochemistry and copy number gain (CNG) by in situ hybridisation (n = 298) or DNA microarray (n = 189). FGFR1 gene expression (n = 369) and immune cell profiles (n = 309) were also examined. Furthermore, gene expression, methylation and microRNA data from The Cancer Genome Atlas (TCGA) were compared. A panel of FGFR1-amplified NSCLC patient-derived xenograft (PDX) models were tested for response to the selective FGFR1 antagonist M6123. A minority of patients demonstrated FGFR1 CNG (10.5%) or increased FGFR1 mRNA (8.7%) and protein expression (4.4%). FGFR1 CNG correlated weakly with FGFR1 gene and protein expression. Tumours overexpressing FGFR1 protein were typically devoid of driver alterations (e.g. EGFR, KRAS) and showed reduced infiltration of T-lymphocytes and lower PD-L1 expression. Promoter methylation and microRNA were identified as regulators of FGFR1 expression in NSCLC and other cancers. Finally, NSCLC PDX models demonstrating FGFR1 amplification and FGFR1 protein overexpression were sensitive to M6123. The unique molecular and immune features of tumours with high FGFR1 expression provide a rationale to stratify patients in future clinical trials of FGFR1 pathway-targeting agents.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , DNA Methylation , Epigenesis, Genetic , Lung Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Antineoplastic Agents/pharmacology , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Female , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Targeted Therapy , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...