Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Eur Respir J ; 62(5)2023 11.
Article in English | MEDLINE | ID: mdl-37884305

ABSTRACT

BACKGROUND: COPD is an incurable disease and a leading cause of death worldwide. In mice, fibroblast growth factor (FGF)10 is essential for lung morphogenesis, and in humans, polymorphisms in the human FGF10 gene correlate with an increased susceptibility to develop COPD. METHODS: We analysed FGF10 signalling in human lung sections and isolated cells from healthy donor, smoker and COPD lungs. The development of emphysema and PH was investigated in Fgf10+/- and Fgfr2b+/- (FGF receptor 2b) mice upon chronic exposure to cigarette smoke. In addition, we overexpressed FGF10 in mice following elastase- or cigarette smoke-induced emphysema and pulmonary hypertension (PH). RESULTS: We found impaired FGF10 expression in human lung alveolar walls and in primary interstitial COPD lung fibroblasts. In contrast, FGF10 expression was increased in large pulmonary vessels in COPD lungs. Consequently, we identified impaired FGF10 signalling in alveolar walls as an integral part of the pathomechanism that leads to emphysema and PH development: mice with impaired FGF10 signalling (Fgf10+/- and Fgfr2b+/- ) spontaneously developed lung emphysema, PH and other typical pathomechanistic features that generally arise in response to cigarette smoke exposure. CONCLUSION: In a therapeutic approach, FGF10 overexpression successfully restored lung alveolar and vascular structure in mice with established cigarette smoke- and elastase-induced emphysema and PH. FGF10 treatment triggered an initial increase in the number of alveolar type 2 cells that gradually returned to the basal level when the FGF10-mediated repair process progressed. Therefore, the application of recombinant FGF10 or stimulation of the downstream signalling cascade might represent a novel therapeutic strategy in the future.


Subject(s)
Cigarette Smoking , Emphysema , Hypertension, Pulmonary , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Animals , Mice , Pulmonary Disease, Chronic Obstructive/drug therapy , Hypertension, Pulmonary/complications , Pancreatic Elastase/adverse effects , Pancreatic Elastase/metabolism , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 10/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Cigarette Smoking/adverse effects , Pulmonary Emphysema/etiology , Lung/metabolism , Emphysema/complications , Mice, Inbred C57BL
2.
In Vivo ; 37(4): 1628-1637, 2023.
Article in English | MEDLINE | ID: mdl-37369494

ABSTRACT

BACKGROUND/AIM: Increasing evidence has revealed FGFR2 as an attractive therapeutic target for cancer including cholangiocarcinoma (CCA). The present study investigated the oncogenic mechanisms by which FGF10 ligand activates FGFR2 in CCA cells and determined whether FGFR inhibitors could suppress FGF10-mediated migration of CCA cells. MATERIALS AND METHODS: Effects of FGF10 on the proliferation, migration, and invasion of KKU-M213A cells were assessed using clonogenic and transwell assays. Protein expression levels of FGFR2 and pro-angiogenic factors were determined via immunoblotting and antibody array analysis. FGFR2 knockdown using a small interfering RNA was used to validate the role of FGF10 in promoting cell migration via FGFR2. The effects of infigratinib (FGFR inhibitor) on cell viability, were determined in KKU-100, KKU-M213A, KKU-452 cells. Moreover, the efficacy of the FGFR inhibitor in suppressing migration via FGF10/FGFR2 stimulation was assessed in KKU-M213A cells. RESULTS: FGF10 significantly increased the expression of phospho-FGFR/FGFR2 and promoted the proliferation, migration, and invasion of KKU-M213A cells. FGF10 increased the expression levels of p-Akt, p-mTOR, VEGF, Slug, and pro-angiogenic proteins related to metastasis. Cell migration mediated by FGF10 was markedly decreased in FGFR2-knockdown cells. Moreover, FGF10/FGFR2 promoted the migration of cells, which was suppressed by the FGFR inhibitor. CONCLUSION: FGF10/FGFR2 activates the Akt/mTOR and VEGF/Slug pathways, which are associated with the stimulation of migration and invasion in CCA. Moreover, the FGF10/FGFR2 signaling was inhibited by an FGFR inhibitor resulting suppression of cell migration, which warrants further studies on their clinical utility for CCA treatment.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Proto-Oncogene Proteins c-akt/metabolism , Ligands , Vascular Endothelial Growth Factor A , Cell Line, Tumor , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , TOR Serine-Threonine Kinases , Protein Kinase Inhibitors/therapeutic use , Bile Ducts, Intrahepatic/metabolism , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Cell Proliferation , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Fibroblast Growth Factor 10/pharmacology , Fibroblast Growth Factor 10/therapeutic use
3.
Pathologica ; 115(2): 71-82, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37017301

ABSTRACT

Cholangiocarcinoma (CCA) is a heterogeneous group of neoplasms burdened by a dismal prognosis. Several studies have investigated the genomic profile of CCA and identified numerous druggable genetic alterations, including FGFR2 fusions/rearrangements. Approximately 5-7% of CCAs and 10-20% of intrahepatic iCCAs harbor FGFR2 fusions. With the recent advent of FGFR-targeting therapies into clinical practice, a standardization of molecular testing for FGFR2 alterations in CCA will be necessary. In this review, we describe the technical aspects and challenges related to FGFR2 testing in routine practice, focusing on the comparison between Next-Generation Sequencing (NGS) and FISH assays, the best timing to perform the test, and on the role of liquid biopsy.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/genetics , Cholangiocarcinoma/drug therapy , Mutation , Prognosis , Bile Ducts, Intrahepatic/pathology , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/drug therapy , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use
4.
J Nanobiotechnology ; 21(1): 55, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36803994

ABSTRACT

Acne is a chronic skin condition that has serious consequences for mental and social well-being because it frequently occurs on the face. Several acne treatment approaches have commonly been used but have been hampered by side effects or weak activity. Thus, the investigation of the safety and efficacy of anti-acne compounds is of considerable medical importance. Herein, an endogenous peptide (P5) derived from fibroblast growth factors 2 (FGF2) was conjugated to the polysaccharide hyaluronic acid (HA) to generate the bioconjugate nanoparticle HA-P5, which suppresses fibroblast growth factor receptors (FGFRs) to significantly rehabilitate acne lesions and reduce sebum accumulation in vivo and in vitro. Moreover, our results show that HA-P5 inhibits both fibroblast growth factor receptor 2 (FGFR2) and androgen receptor (AR) signalling in SZ95 cells, reverses the acne-prone transcriptome, and decreases sebum secretion. Furthermore, the cosuppression mechanism revealed that HA-P5 blocks FGFR2 activation, as well as the YTH N6-methyladenosine RNA binding protein F3 (YTHDF3) downstream molecules, including an N6-methyladenosine (m6A) reader that facilitates AR translation. More importantly, a significant difference between HA-P5 and the commercial FGFR inhibitor AZD4547 is that HA-P5 does not trigger the overexpression of aldo-keto reductase family 1 member C3 (AKR1C3), which blocks acne treatment by catalyzing the synthesis of testosterone. Overall, we demonstrate that a polysaccharide-conjugated and naturally derived oligopeptide HA-P5 can alleviate acne and act as an optimal FGFR2 inhibitor and reveal that YTHDF3 plays a crucial role in signalling between FGFR2 and AR.


Subject(s)
Acne Vulgaris , Receptor, Fibroblast Growth Factor, Type 2 , Humans , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Fibroblast Growth Factor 2 , Hyaluronic Acid/therapeutic use , Acne Vulgaris/drug therapy , Peptides/therapeutic use
5.
Annu Rev Med ; 74: 293-306, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36170665

ABSTRACT

Biliary tract cancer (BTC) is the second most common primary liver cancer after hepatocellular carcinoma and accounts for 2% of cancer-related deaths. BTCs are classified according to their anatomical origin into intrahepatic (iCCA), perihilar, or distal cholangiocarcinoma, as well as gall bladder carcinoma. While the mutational profiles in these anatomical BTC subtypes overlap to a large extent, iCCA is notable for the high frequency of IDH1/2 mutations (10-22%) and the nearly exclusive occurrence of FGFR2 fusions in 10-15% of patients. In recent years, FGFR2 fusions have become one of the most promising targets for precision oncology targeting BTC, with FGFR inhibitors already approved in Europe and the United States for patients with advanced, pretreated iCCA. While the therapeutic potential of nonfusion alterations is still under debate, it is expected that the field of FGFR2-directed therapies will be subject to rapid further evolution and optimization. The scope of this review is to provide an overview of oncogenic FGFR signaling in iCCA cells and highlight the pathophysiology, diagnostic testing strategies, and therapeutic promises and challenges associated with FGFR2-altered iCCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Precision Medicine , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/diagnosis , Mutation , Bile Ducts, Intrahepatic/pathology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use
6.
J Natl Compr Canc Netw ; 20(10): 1076-1079, 2022 10.
Article in English | MEDLINE | ID: mdl-36240849

ABSTRACT

Despite advances in cancer therapeutics, pancreatic ductal adenocarcinoma (PDAC) remains among the deadliest malignancies, with a poor prognosis at time of diagnosis. Research in PDAC has suggested that adaptive signaling in the tumor microenvironment may promote tumor proliferation and survival. Several FGFR fusion genes-specifically FGFR2-are involved with the creation and progression of cancer. These mutations are found in a variety of cancer types. This report presents a unique case of a young patient with stage IV PDAC with a known FGFR2 fusion. This molecular alteration afforded a remarkable response to FGFR inhibitor therapy, erdafitinib, after the patient experienced disease progression on multiple chemotherapy regimens.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pyrazoles , Quinoxalines , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Tumor Microenvironment , Pancreatic Neoplasms
7.
Gan To Kagaku Ryoho ; 49(10): 1040-1042, 2022 Oct.
Article in Japanese | MEDLINE | ID: mdl-36281591

ABSTRACT

Gemcitabine plus cisplatin is the mainstay of first-line chemotherapy for unresectable biliary tract cancer, but the standard of care for second-line has not been established. In recent years, genetic abnormalities in biliary tract cancers have been identified, and therapeutic development targeting these genetic abnormalities has been promoted. In March 2021, based on the results of the FIGHT-202 trial, pemigatinib was approved for the treatment of FGFR2 fusion gene-positive biliary tract cancer. Pemigatinib is attracting attention as new treatment option for unresectable biliary tract cancer. Clinical trial data and other information on cancer therapies targeting the FGFR2 fusion gene will be presented.


Subject(s)
Biliary Tract Neoplasms , Cisplatin , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biliary Tract Neoplasms/drug therapy , Biliary Tract Neoplasms/genetics , Cisplatin/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Clinical Trials as Topic
8.
BMC Gastroenterol ; 22(1): 360, 2022 Jul 28.
Article in English | MEDLINE | ID: mdl-35902814

ABSTRACT

BACKGROUND: Gastric carcinosarcoma is most frequently diagnosed at an advanced stage when the tumor is generally large with invasion into other organs, lymph node metastasis, and distant metastasis. Standard chemotherapy has not been established, and surgery is the only curative treatment. Here, we present a case of postoperative recurrence of gastric carcinosarcoma under long-term tumor control with pazopanib. CASE PRESENTATION: A 77-year-old man was referred to our hospital because of nausea and vomiting. Computed tomography and upper gastrointestinal endoscopy revealed a type 1 tumor arising from the gastric antrum and extending into the duodenal bulb. He underwent distal gastrectomy (D2) with Roux-en-Y reconstruction. Histopathologically, the tumor had mixed adenocarcinoma and sarcoma components. According to the tumor-node-metastasis classification, the diagnosis was primary gastric carcinosarcoma pT1bN1M0 stage IB. Liver metastasis was detected 2 months after surgery; multiple lung metastases were detected 17 month after surgery. A genomic profiling test was performed using liver specimens as the patient became refractory to chemotherapy commonly used for gastric cancer, and the test revealed FGFR2 amplification along with TP53 R209*, AKT3 N127D, NOTCH1 A2036T, and POLD1 M161I. The patient was treated with pazopanib (800 mg/daily), and the tumor growth was controlled for 11 months. CONCLUSIONS: We report a case of postoperative recurrence of gastric carcinosarcoma under long-term tumor control with pazopanib. This case suggested that pazopanib may be effective in treating gastric carcinosarcoma.


Subject(s)
Carcinosarcoma , Stomach Neoplasms , Aged , Carcinosarcoma/drug therapy , Carcinosarcoma/genetics , Carcinosarcoma/pathology , Gastrectomy/methods , Humans , Indazoles/therapeutic use , Male , Pyrimidines , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Sulfonamides
9.
Cancer Genet ; 266-267: 39-43, 2022 08.
Article in English | MEDLINE | ID: mdl-35749865

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is a fatal bile duct cancer with dismal prognosis and limited therapeutic options. FGFR family fusion have been identified in many diseases, and FGFR2 fusion is a validated oncogenic driver in ICC. At present, a variety of fusion forms have been reported, including gene-gene, gene-intergenic, and intergenic-intergenic fusion. Here, by performing RNA- and DNA-sequencing analysis, FGFR2 fusions were found in 10.1% of ICC, including 4 gene-intergenic fusions. We confirmed that the non-canonical rearrangements can generate chimeric transcripts, and used conventional splicing mechanism to explain the event. Our study provides possible target therapy for these 4 patients and possibility analysis scheme for similar situation.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , DNA, Intergenic , Humans , Pyrimidines/adverse effects , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use
10.
Int J Colorectal Dis ; 37(6): 1393-1402, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35585358

ABSTRACT

PURPOSE: Fibroblast growth factor receptor 2 (FGFR2) and human epidermal growth factor receptor 2 (HER2) proteins are both molecular targets for cancer therapy. The objective of this study was to evaluate the expression status of FGFR2 and HER2 in patients with gastric cancer (GC) or colorectal cancer (CRC). METHODS: Archived tumor tissue samples from patients with histologically-confirmed GC or CRC suitable for chemotherapy were analyzed for FGFR2 and HER2 expression using immunohistochemistry and fluorescence in situ hybridization (HER2 in CRC only). RESULTS: A total of 176 GC patients and 389 CRC patients were enrolled. Among patients with GC, 25.6% were FGFR2-positive and 26.1% were HER2-positive. Among patients with CRC, 2.9% were FGFR2-positive and 16.2% were HER2-positive. No clear relationship was found between FGFR2 and HER2 status in either GC or CRC. In GC, FGFR2 and HER2 statuses did not differ between different primary cancer locations, whereas there were some differences between histological types. Based on FGFR2- and/or HER2-positive status, 117 patients were identified as potentially suitable for inclusion in clinical trials of therapeutic agents targeting the relevant protein (GC = 45, CRC = 72; FGFR = 56, HER2 = 62), of whom 7 were eventually enrolled into such clinical trials. CONCLUSIONS: This study indicated the prevalence of FGFR2 and HER2 in GC and CRC in the Japanese population. The screening performed in this study could be useful for identifying eligible patients for future clinical trials of agents targeting these proteins. TRIAL REGISTRATION: Clinical trial registration Japic CTI No.: JapicCTI-163380.  https://www. CLINICALTRIALS: jp/cti-user/trial/ShowDirect.jsp?directLink=RNlzx1PPCuT.PrVNPxPRwA .


Subject(s)
Colorectal Neoplasms , Stomach Neoplasms , Colorectal Neoplasms/genetics , Humans , In Situ Hybridization, Fluorescence , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Stomach Neoplasms/genetics
11.
Cancer Treat Res Commun ; 31: 100568, 2022.
Article in English | MEDLINE | ID: mdl-35477128

ABSTRACT

According to previous reports,10-16% of patients with clinically advanced cholangiocarcinoma develop FGFR2 fusions or rearrangements. Treatment with FGFR2-specific inhibitors (tyrosine kinase inhibitors, TKIs) has proven effective for patients with cholangiocarcinoma. In this study, we report a case of advanced cholangiocarcinoma, in which the patient was unable to tolerate the adverse effects of standard first-line chemotherapy. Genetic testing suggested the presence of a novel variant resulting from FDFT1/FGFR2 rearrangement. Owing to poor accessibility and high price, only a limited number of patients with advanced cholangiocarcinoma have access to TKIs and precision targeted therapy in China. Anlotinib is a novel small-molecule multi-target TKI developed independently in China. It has a broad target spectrum, including FGFR, and can effectively inhibit tumor angiogenesis and tumor cell proliferation, thereby achieving an anti-tumor effect. Here, the patient was prescribed anlotinib. After treatment, the tumor size continued to shrink, and no significant adverse effects were reported. The finding suggested that anlotinib may be effective in patients with FDFT1/FGFR2 rearrangement and could serve as a novel treatment option for affected patients in future.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Quinolines , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Humans , Indoles , Quinolines/pharmacology , Quinolines/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use
12.
Trends Cancer ; 8(2): 83-86, 2022 02.
Article in English | MEDLINE | ID: mdl-34840108

ABSTRACT

Fibroblast growth factor receptor 2 (FGFR2) fusion proteins (FFs) are oncogenic drivers in 10-15% of intrahepatic cholangiocarcinoma (iCCA). FGFR-specific inhibitors provide temporary benefit in FF+ unresectable patients. Recent work with mouse iCCA models has documented the necessary role of RAS-ERK downstream to FFs and provided examples of preclinical experimentation aimed at improving FF targeting in iCCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Animals , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use
13.
MAbs ; 13(1): 1981202, 2021.
Article in English | MEDLINE | ID: mdl-34719330

ABSTRACT

Bemarituzumab (FPA144) is a first-in-class, humanized, afucosylated immunoglobulin G1 monoclonal antibody (mAb) directed against fibroblast growth factor receptor 2b (FGFR2b) with two mechanisms of action against FGFR2b-overexpressing tumors: inhibition of FGFR2b signaling and enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). Bemarituzumab is being developed as a cancer therapeutic, and we summarize here the key nonclinical data that supported moving it into clinical trials. Bemarituzumab displayed sub-nanomolar cross-species affinity for FGFR2b receptors, with >20-fold enhanced binding affinity to human Fc gamma receptor IIIa compared with the fucosylated version. In vitro, bemarituzumab induced potent ADCC against FGFR2b-expressing tumor cells, and inhibited FGFR2 phosphorylation and proliferation of SNU-16 gastric cancer cells in a concentration-dependent manner. In vivo, bemarituzumab inhibited tumor growth through inhibition of the FGFR2b pathway and/or ADCC in mouse models. Bemarituzumab demonstrated enhanced anti-tumor activity in combination with chemotherapy, and due to bemarituzumab-induced natural killer cell-dependent increase in programmed death-ligand 1, also resulted in enhanced anti-tumor activity when combined with an anti-programmed death-1 antibody. Repeat-dose toxicity studies established the highest non-severely-toxic dose at 1 and 100 mg/kg in rats and cynomolgus monkeys, respectively. In pharmacokinetic (PK) studies, bemarituzumab exposure increase was greater than dose-proportional, with the linear clearance in the expected dose range for a mAb. The PK data in cynomolgus monkeys were used to project bemarituzumab linear PK in humans, which were consistent with the observed human Phase 1 data. These key nonclinical studies facilitated the successful advancement of bemarituzumab into the clinic.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 2 , Stomach Neoplasms , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Mice , Rats , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology
14.
Int J Radiat Oncol Biol Phys ; 105(2): 410-422, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31255687

ABSTRACT

PURPOSE: Fibroblast growth factor receptor 2 (FGFR2) has been previously reported to be overexpressed in several types of cancer, whereas the expression in normal tissue is considered to be moderate to low. Thus, FGFR2 is regarded as an attractive tumor antigen for targeted alpha therapy. This study reports the evaluation of an FGFR2-targeted thorium-227 conjugate (FGFR2-TTC, BAY 2304058) comprising an anti-FGFR2 antibody, a chelator moiety covalently conjugated to the antibody, and the alpha particle-emitting radionuclide thorium-227. FGFR2-TTC was assessed as a monotherapy and in combination with the DNA damage response inhibitor ATRi BAY 1895344. METHODS AND MATERIALS: The in vitro cytotoxicity and mechanism of action were evaluated by determining cell viability, the DNA damage response marker γH2A.X, and cell cycle analyses. The in vivo efficacy was determined using human tumor xenograft models in nude mice. RESULTS: In vitro mechanistic assays demonstrated upregulation of γH2A.X and induction of cell cycle arrest in several FGFR2-expressing cancer cell lines after treatment with FGFR2-TTC. In vivo, FGFR2-TTC significantly inhibited tumor growth at a dose of 500 kBq/kg in the xenograft models NCI-H716, SNU-16, and MFM-223. By combining FGFR2-TTC with the ATR inhibitor BAY 1895344, an increased potency was observed in vitro, as were elevated levels of γH2A.X and inhibition of FGFR2-TTC-mediated cell cycle arrest. In the MFM-223 tumor xenograft model, combination of the ATRi BAY 1895344 with FGFR2-TTC resulted in significant tumor growth inhibition at doses at which the single agents had no effect. CONCLUSIONS: The data provide a mechanism-based rationale for combining the FGFR2-TTC with the ATRi BAY 1895344 as a new therapeutic approach for treatment of FGFR2-positive tumors from different cancer indications.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Breast Neoplasms/radiotherapy , Protein Kinase Inhibitors/therapeutic use , Radioimmunotherapy/methods , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Thorium/therapeutic use , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chelating Agents/therapeutic use , DNA Damage , Drug Combinations , Drug Synergism , G2 Phase Cell Cycle Checkpoints/radiation effects , Histones/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Thorium/pharmacokinetics , Thorium Compounds/therapeutic use , Up-Regulation , Xenograft Model Antitumor Assays
15.
Int J Biol Macromol ; 133: 58-66, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-30981773

ABSTRACT

Nanoparticle-based pulmonary delivery of protein therapeutics provides a promising approach for improving protein bioavailability to treat either local or systemic diseases, however high-efficient nanocarrier is a great challenge. Here, biomimetic phosphorylcholine-chitosan nanoparticles (PCCs-NPs) taking advantages of both zwitterionic phosphorylcholine and chitosan were developed as a pulmonary protein delivery platform. msFGFR2c, a potential therapeutic protein for lung fibrosis as model was loaded into PCCs-NPs via ionic gelation. The obtained msFGFR2c/PCCs-NPs inhibited α-SMA expression in fibroblasts induced by TGF-ß1, slightly more effective than naked msFGFR2c. After orotracheal administration to bleomycin-induced pulmonary fibrosis model rats, msFGFR2c/PCCs-NPs resulted in a significant antifibrotic efficacy, with reduction in inflammatory cytokines and α-SMA expression, remarkable attenuation of lung fibrosis score and collagen deposition, and significant increase in survival rate, while naked msFGFR2c exhibited a poor efficacy. The in vitro and in vivo results strongly indicated that PCCs-NPs may be a promising nanocarrier for pulmonary protein delivery.


Subject(s)
Bleomycin/adverse effects , Chitosan/chemistry , Lung/metabolism , Nanoparticles/chemistry , Peptide Fragments/chemistry , Peptide Fragments/therapeutic use , Pulmonary Fibrosis/drug therapy , Receptor, Fibroblast Growth Factor, Type 2/chemistry , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Animals , Cell Line , Drug Carriers/chemistry , Female , Humans , Lung/drug effects , Phosphorylcholine/chemistry , Pulmonary Fibrosis/chemically induced , Rats , Rats, Wistar
16.
Biol Pharm Bull ; 35(5): 731-6, 2012.
Article in English | MEDLINE | ID: mdl-22687409

ABSTRACT

We have developed a strong inhibitor (S252W mutant soluble ectodomain of fibroblast growth factor recptor-2 IIIc, msFGFR2) that binds FGFs strongly and blocks the activation of FGFRs. In vitro, msFGFR2 could inhibit the promoting effect of transforming growth factor (TGF)-ß1 on the proliferation of primary lung fibroblasts. In vivo, msFGFR2 alleviated lung fibrosis through inhibiting the expression of α-smooth muscle actin (SMA) and collagen deposit. In Western blotting of the right lung tissues and immunohistochemical assay, we found the level of p-FGFRs, p-mitogen activated protein kinase (MAPK) and p-Smad3 in the mice of bleomycin (BLM) group treated with msFGFR2 was down dramatically compared with the mice of BLM group, which suggested the activations of FGF and TGF-ß signals were blocked meanwhile. In summary, msFGFR2 attenuated BLM-induced fibrosis and is an attractive therapeutic candidate for human pulmonary fibrosis.


Subject(s)
Fibroblast Growth Factors/metabolism , Fibroblasts/drug effects , Lung/drug effects , Peptide Fragments/therapeutic use , Pulmonary Fibrosis/prevention & control , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/therapeutic use , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Animals , Bleomycin , Blotting, Western , Cell Proliferation/drug effects , Collagen/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Peptide Fragments/pharmacology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Receptor, Fibroblast Growth Factor, Type 2/pharmacology , Recombinant Proteins/pharmacology , Smad3 Protein/metabolism
17.
Mol Cancer Ther ; 10(9): 1656-66, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21750221

ABSTRACT

The fibroblast growth factor (FGF) signaling pathway is a recognized target of cancer therapy. We have developed a strong inhibitor (S252W mutant soluble ectodomain of FGF recptor-2 IIIc, msFGFR2) that binds FGFs and blocks the activation of FGFRs. Thermodynamic binding studies indicated that msFGFR2 bound FGF-2 16.9 times as strongly as wild-type soluble FGFR2IIIc ectodomain (wsFGFR2). It successfully suppressed the growth, angiogenesis, and metastasis of two tumor cell lines in vitro and in vivo, and it potently inhibited cancer cell proliferation but not normal cell proliferation. Therefore, msFGFR2 is a useful probe for FGF-dependent signaling pathways and a potential broad-spectrum antitumor agent.


Subject(s)
Antineoplastic Agents/pharmacology , Peptide Fragments/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/pharmacology , Recombinant Proteins/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Kaplan-Meier Estimate , Ligands , Mice , Mice, Inbred BALB C , Mice, Nude , Mutant Proteins/genetics , Mutant Proteins/pharmacology , Mutant Proteins/therapeutic use , NIH 3T3 Cells , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasms/drug therapy , Neoplasms/mortality , Neoplasms/pathology , Neovascularization, Physiologic/drug effects , Peptide Fragments/chemistry , Peptide Fragments/therapeutic use , Protein Binding , Receptor, Fibroblast Growth Factor, Type 2/chemistry , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...