Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Publication year range
1.
Cancer Sci ; 111(12): 4500-4509, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32946655

ABSTRACT

Fibroblast growth factor receptor (FGFR) is associated with proliferation, migration, and angiogenesis of carcinomas, and FGFR signaling inhibitors are considered a key drug for the treatment of solid tumors with FGFR overexpression. Amplification of FGFR2 is reportedly identified in 3%-10% of gastric cancers (GCs). The aim of this study is to clarify whether the identification of the circulating tumor cells (CTCs) with FGFR2 overexpression is useful to detect patients with FGFR2-overexpressing GC. One hundred GC patients who underwent gastrectomy were enrolled. A total volume of 8 mL of peripheral blood was collected from each patient just before gastrectomy, and mononuclear cells were enriched by Ficol density gradient centrifugation. These cells were immunostained with PI/CD45/EpCAM/FGFR2. The number of CTCs with FGFR2 expression in each sample was enumerated by FACScan. The FGFR2 expression level of the resected primary tumor was assessed by immunohistochemistry. The number of FGFR2-positive CTCs in the GC patients' peripheral blood was significantly correlated with the FGFR2 expression level of the primary GC. The relapse-free survival of the patients with FGFR2-positive CTCs (≥5 cells/10 mL blood) was significantly poorer (P = .018, log-rank) than that of the patients without FGFR2-positive CTCs (<5 cell/10 mL blood). These findings suggested that the determination of FGFR2-positive CTCs might help identify an existing tumor with FGFR2 overexpression.


Subject(s)
Neoplastic Cells, Circulating/chemistry , Neoplastic Cells, Circulating/metabolism , Receptor, Fibroblast Growth Factor, Type 2/analysis , Stomach Neoplasms/blood , Stomach Neoplasms/diagnosis , Aged , Centrifugation, Density Gradient/methods , Chi-Square Distribution , Disease-Free Survival , Female , Gastrectomy , Gene Amplification , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Prognosis , Statistics, Nonparametric , Stomach Neoplasms/mortality , Stomach Neoplasms/surgery
2.
Mod Pathol ; 29(9): 1095-103, 2016 09.
Article in English | MEDLINE | ID: mdl-27230412

ABSTRACT

FGFR2 gene amplification, and resulting FGFR2 protein overexpression, is rare in gastric cancer patients, and development of an accurate and widely available method for mass screening to identify patients who may respond to treatment with fibroblast growth factor receptor (FGFR) inhibitors is important. We first screened 312 gastric cancer patients with known copy number variations by FGFR2b immunohistochemistry using FPR2-D, an isoform-specific antibody. Next, we performed immunohistochemistry on tissue microarrays from 1574 gastric cancer patients. Selected cases were analyzed for FGFR2 amplification by FISH. In addition, FGFR2b overexpression was studied in 88 matched primary and metastatic gastric cancers. In the first cohort, FGFR2b immunohistochemistry results correlated very well with those of copy number variation (r=0.79) and FISH (r=1.0). In total, FGFR2b overexpression was identified in 73 of 1974 gastric cancers (4%). The concordance between immunohistochemistry and FISH was extremely high; all 2+ and 3+ cases identified by immunohistochemistry were FGFR2 amplified. In the matched primary and metastatic gastric cancer pairs, the positivity and percentage of positive tumor cells were significantly higher in metastatic gastric cancers than in primary gastric cancers (8% vs 3% and 75% vs 47%, respectively; P<0.001). FGFR2b overexpression was significantly more frequent in gastric cancers with diffuse subtype (P=0.01) and higher N stage (P=0.006). FGFR2b overexpression with H-score ≥150 were independent prognostic factors for overall survival with hazard ratio of 1.836 (95% confidence interval, 1.034-3.261; P=0.038). FGFR2b positivity in immunohistochemistry was strongly correlated with FGFR2 amplification. Given the low frequency of FGFR2 amplification in gastric cancers, FGFRb2 immunohistochemistry is an accurate screening tool to detect FGFR2 amplification, and both primary and metastatic gastric cancer tissues should be tested to select gastric cancer patients for treatment with FGFR2 inhibitors.


Subject(s)
Biomarkers, Tumor/analysis , Gene Amplification , Receptor, Fibroblast Growth Factor, Type 2/analysis , Stomach Neoplasms/chemistry , Biomarkers, Tumor/genetics , Female , Genetic Predisposition to Disease , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Middle Aged , Phenotype , Proportional Hazards Models , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tissue Array Analysis , Up-Regulation
3.
Anal Biochem ; 424(2): 137-9, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22387389

ABSTRACT

Here we present a highly efficient protocol for on-the-resin coupling of fluorescent dyes or other functional groups to the N-termini of synthetic peptides prior to cleavage and deprotection. The protocol avoids expensive preactivated dyes and instead employs carboxylated dyes activated by large amounts of coupling reagents. The protocol was used to label peptides with low reactivity such as long hydrophobic peptides and peptides with strong tendencies to form sterically shielding structures or aggregates in solution. In all cases, the yields far exceeded those from commercially available preactivated compounds.


Subject(s)
Peptides/chemistry , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 2/chemistry , Staining and Labeling/methods , Amino Acid Sequence , Ethylamines/chemistry , Fluorescent Dyes , Humans , Hydrophobic and Hydrophilic Interactions , Immobilized Proteins/analysis , Immobilized Proteins/chemistry , Molecular Sequence Data , Organophosphorus Compounds/chemistry , Peptides/analysis , Receptor, Fibroblast Growth Factor, Type 1/analysis , Receptor, Fibroblast Growth Factor, Type 2/analysis
4.
Mod Pathol ; 24(11): 1500-10, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21725289

ABSTRACT

Fibroblast growth factor receptor 2 (FGFR2) is a tyrosine kinase receptor involved in many biological processes such as embryogenesis, adult tissue homeostasis and cell proliferation. Mutations in FGFR2 have been reported in up to 10-12% of endometrial carcinomas identical to those found in congenital craniofacial disorders. Inhibition of FGFR2 could be a new therapeutic target in endometrial carcinoma. FGFR2 immunostaining was assessed in three tissue microarrays: one constructed from paraffin-embedded blocks of 60 samples of normal endometrium in different phases of menstrual cycle, and two tissue microarrays containing endometrial carcinoma samples (95 and 62 cases). FGFR2 expression was correlated with stage, histological type and grade as well as with immunostaining of PTEN, RASSF1A, estrogen and progesterone receptors, KI67, Cyclin D1, STAT-3 and SPRY2. FGFR2 mutations were assessed by PCR and direct sequencing, with DNA obtained from 31 paraffin-embedded endometrial carcinoma samples. In normal endometrium, FGFR2 expression was higher in the secretory than in the proliferative phase (P=0.001), with an inverse correlation with Ki67 (P=0.00032), suggesting a tumor-suppressor role for FGFR2 in normal endometrium. Cytoplasmic expression of FGFR2 was higher in endometrial carcinoma when compared with the atrophic endometrium from the same patients (P=0.0283), but was lower in comparison with normal endometrium from women in the menstrual cycle. Interestingly, nuclear staining was observed in some cases, and it was less frequent in endometrial carcinoma when compared with the adjacent atrophic endometrium (P=0.0465). There were no statistical differences when comparing superficial and myoinvasive endometrial carcinoma samples. Endometrioid endometrial carcinomas showed higher expression of FGFR2 than nonendometrioid endometrial carcinomas (fold change 2.56; P=0.0015). Grade III endometrioid endometrial carcinomas showed decreased FGFR2 expression when compared with grade II endometrioid endometrial carcinomas (P=0.0055). No differences were found regarding pathological stage. Two missense mutations of FGFR2 gene were detected in exons 6 and 11 (S252W and N549K, respectively; 6.45%). Results support the hypothesis that FGFR2 has a dual role in the endometrium, by inhibiting cell proliferation in normal endometrium during the menstrual cycle, but acting as an oncogene in endometrial carcinoma.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/chemistry , Endometrial Neoplasms/chemistry , Endometrium/chemistry , Receptor, Fibroblast Growth Factor, Type 2/analysis , Biomarkers, Tumor/genetics , Carcinoma/genetics , Carcinoma/pathology , Cell Proliferation , Cyclin D1/analysis , DNA Mutational Analysis , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/pathology , Female , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/analysis , Ki-67 Antigen/analysis , Membrane Proteins , Menstrual Cycle , Mutation, Missense , Neoplasm Staging , PTEN Phosphohydrolase/analysis , Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , STAT3 Transcription Factor/analysis , Spain , Tissue Array Analysis , Tumor Suppressor Proteins/analysis
5.
J Oral Pathol Med ; 39(7): 540-7, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20149060

ABSTRACT

Carcinoma ex pleomorphic adenoma (CXPA) is a rare malignant salivary gland tumor derived from a pre-existing pleomorphic adenoma. It is a good model to study the evolution of carcinogenesis, starting with in situ areas to frankly invasive carcinoma. Growth factors are associated with several biological and neoplastic processes by transmembrane receptors. In order to investigate, by immunohistochemistry, the expression of some growth factors and its receptors [EGF receptor, fibroblast growth factor, fibroblast growth factor receptor 1, fibroblast growth factor receptor 2, hepatocyte growth factor, c-Met, transforming growth factor (TGF) beta1, TGFbetaR-II and insulin-like growth factor receptor 1] in the progression of CXPA, we have used ten cases of CXPA in several degrees of invasion- intracapsular, minimally and frankly invasive carcinoma- with only epithelial component. Slides were qualitatively and semi-quantitatively evaluated according to the percentage of stained tumor cells from 0 to 3 (0 = less than 10%; 1 = 10-25%; 2 = 25-50%; 3 = more than 50% of cells). Malignant epithelial cells starting with in situ areas showed stronger expression than luminal cells of pleomorphic adenoma for all antibodies. Most of the intracapsular, minimally and frankly invasive CXPA presented score 3. However, score 2 was more evident in the frankly invasive one. In small nests of invasive carcinoma, negative cells were observed probably indicating that the proliferative process is replaced by the invasive mechanism. Altogether this data infers that these factors may contribute to cell proliferation during initial phases of the tumor.


Subject(s)
Adenocarcinoma/pathology , Adenoma, Pleomorphic/pathology , Intercellular Signaling Peptides and Proteins/analysis , Parotid Neoplasms/pathology , Receptors, Growth Factor/analysis , Adult , Aged , Carcinoma in Situ/pathology , Cell Proliferation , Coloring Agents , Disease Progression , Epithelial Cells/pathology , ErbB Receptors/analysis , Female , Fibroblast Growth Factors/analysis , Hepatocyte Growth Factor/analysis , Humans , Male , Middle Aged , Neoplasm Invasiveness , Protein Serine-Threonine Kinases/analysis , Proto-Oncogene Proteins c-met/analysis , Receptor, Fibroblast Growth Factor, Type 1/analysis , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, IGF Type 1/analysis , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/analysis , Submandibular Gland Neoplasms/pathology , Transforming Growth Factor beta1/analysis
6.
J Pediatr Urol ; 16(1): 41.e1-41.e10, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31718875

ABSTRACT

INTRODUCTION: Fibroblast growth factors (FGFs) play a crucial role in early embryogenesis of the genital tubercle and are involved in the development of hypospadias, affecting both endo- and ectodermally derived tissues. It was hypothesized that expression of FGFs could be qualitatively or quantitatively altered in skin of children with hypospadias. OBJECTIVE: The objective of the study was to investigate expression patterns and transcription levels of FGF8, FGF10, and FGF Receptor 2 (FGFR2) in patients with hypospadias compared to normal controls. PATIENTS AND METHODS: Skin samples from the ventro-lateral aspect of the foreskin of 32 patients with hypospadias (17 distal and 15 proximal, mean age 25 months) and 10 normal foreskin samples (mean age 77 months) were analyzed by immunohistochemistry. Staining, localization, and distribution of positive cells in epidermis and dermis were categorized independently by two researchers. Complementary DNA (cDNA) samples prepared from messenger RNA (mRNA) isolates of the same samples were analyzed by quantitative polymerase chain reaction (qPCR), comparing expressions of FGF8, FGF10, and FGFR2 with loading controls. RESULTS: Patients with hypospadias consistently showed aberrant immunohistochemical staining patterns for FGF8/FGF10/FGFR2 in epidermis and dermis compared to patients without penile malformation (p < 0.01 for all markers). qPCR displayed no difference in expression levels on mRNA level (FGFR2 p = 0.44, FGF8 p = 0.77, and FGF10 p = 0.17) comparing normal foreskin with foreskin from patients with hypospadias. Figure. DISCUSSION: The results point at an impact of FGF signaling during embryological development of hypospadias on skin, as an ectodermally derived tissue. Similar to the urethral development, this might be a result of mesothelial-epithelial interactions. The differing expression patterns in immunohistochemistry are not matched by a quantitative difference in marker expression on the mRNA level, putatively caused by post-translational modifications or alterations of the downstream pathway. FGFs, particularly FGF10 and FGFR2, are critically involved in wound healing. CONCLUSIONS: There are significant differences in localization and distribution of FGF8, FGF10, and FGFR2 in comparisons of normal foreskin to foreskin of patients with hypospadias, whereas there is no difference in the quantitative expression of these markers on the mRNA level. This confirms the notion that penile skin is affected as well by the embryological aberrations during the embryogenesis of hypospadias.


Subject(s)
Fibroblast Growth Factor 10/biosynthesis , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 8/biosynthesis , Fibroblast Growth Factor 8/genetics , Foreskin/metabolism , Hypospadias/genetics , Hypospadias/metabolism , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Transcription, Genetic , Child , Child, Preschool , Fibroblast Growth Factor 10/analysis , Fibroblast Growth Factor 8/analysis , Foreskin/chemistry , Gene Expression , Humans , Hypospadias/pathology , Immunohistochemistry , Infant , Male , Prospective Studies , Receptor, Fibroblast Growth Factor, Type 2/analysis
7.
Oncol Rep ; 21(4): 875-80, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287982

ABSTRACT

Keratinocyte growth factor (KGF) from gastric fibroblasts have been reported to stimulate proliferation of scirrhous gastric cancer cells with K-samII amplification in a paracrine manner. The aim of this study was to evaluate the clinical significance of the co-expression of K-sam and KGF in gastric carcinomas. A total of 136 primary gastric tumors were investigated by staining with antibodies against K-sam and KGF. K-sam expression on cancer cells and KGF expression on fibroblasts was estimated. The relationship between the K-sam and/or KGF expression and the clinicopathological characteristics were analyzed. K-sam expression was positive in 42 (31%) of 136 gastric carcinomas. K-sam expression was positively correlated with scirrhous cancer (p<0.001), diffuse type (p=0.031), invasion depth (p=0.018) and infiltration type (p<0.001). Prognosis of K-sam positive patients was significantly poorer than that of K-sam negative patients (p<0.001). The prognosis of patients with both K-sam and KGF positive tumors was significantly worse in comparison to either negative tumors (p<0.001). In 94 patients with a curative resection, a multivariate analysis revealed the co-expression of K-sam and KGF to be an independent prognostic factor (p=0.029). In conclusion, the co-expression of K-sam and KGF in gastric cancer might be a useful prognostic factor.


Subject(s)
Fibroblast Growth Factor 7/analysis , Receptor, Fibroblast Growth Factor, Type 2/analysis , Stomach Neoplasms/chemistry , Aged , Animals , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Middle Aged , Prognosis , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate
8.
Int J Immunopathol Pharmacol ; 22(3): 627-38, 2009.
Article in English | MEDLINE | ID: mdl-19822079

ABSTRACT

Basic fibroblast growth factor (bFGF) is a heparin-binding cationic protein involved in a variety of pathological conditions including angiogenesis and solid tumour growth. The basic fibroblast growth factor receptor (FGFR) family comprises at least 4 high affinity tyrosine kinase receptors that require syndecans for their function. Mounting evidence indicates that syndecans, that bind both bFGF and their FGFRs, will act as stimulators, whereas syndecans that only bind bFGF will act as inhibitors of signaling by sequestering the growth factor. Recent findings have highlighted the importance of syndecans in urological cancers. The aim of this study is to investigate the expression of bFGF, its receptors (R1 and R2) and syndecans (1-4) in invasive urothelial carcinoma and normal-looking urothelium by Western blotting, RT-PCR, and immunohistochemistry analyses. Interestingly, bFGF, FGFR1 and FGFR2 protein levels statistically increased in bladder cancer tissues. mRNA of FGFR1 and syndecans (1-4), showed a statistically significant increase while an mRNA increase in the other molecules analysed was not significant. bFGF, its receptors and syndecan immunostaining were mainly present in the urothelium both in normal-looking tissues and urothelial neoplastic cells. In conclusion, our data report that the bFGF, FGFR and syndecan expressions are altered in bladder tumours.


Subject(s)
Carcinoma/chemistry , Fibroblast Growth Factor 2/analysis , Receptor, Fibroblast Growth Factor, Type 2/analysis , Syndecans/analysis , Urinary Bladder Neoplasms/chemistry , Aged , Blotting, Western , Carcinoma/genetics , Carcinoma/pathology , Carcinoma/surgery , Cystectomy , Female , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/analysis , Receptor, Fibroblast Growth Factor, Type 1/analysis , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Syndecan-1/analysis , Syndecan-2/analysis , Syndecan-3/analysis , Syndecan-4/analysis , Syndecans/genetics , Up-Regulation , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery , Urothelium/chemistry
9.
Theriogenology ; 71(6): 1005-10, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19157529

ABSTRACT

Neurotrophins and basic fibroblast growth factor are ligands of tyrosine kinase receptors, though they bind to different tyrosine kinase receptor classes. Neurotrophins bind to receptor tyrosine kinase class VII, Trk receptor family, while basic fibroblast growth factor binds to receptor tyrosine kinase class IV, FGF receptor family. The mammalian uterine tract immunolocalizes neurotrophins and bFGF; therefore their cognate receptors might exert a role during embryonic development. Using RT-PCR, we found mRNA for p75(NTR) TrkA, TrkC and FGFr2 throughout the early bovine embryonic development in vitro. Immunofluorescent staining, assessed by confocal microscopy, showed the expression of TrkA and TrkC proteins in oocytes and all embryonic stages analyzed. We have provided a novel description of TrkA and TrkC proteins, and TrkA, TrkC, p75(NTR) and FGFr2 mRNA expression throughout mammalian embryonic development. This work may help to design future research with neurotrophins in bovine embryo culture and embryonic stem cells.


Subject(s)
Cattle/embryology , Embryo Culture Techniques/veterinary , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, trkA/analysis , Receptor, trkC/analysis , Animals , Blastocyst/chemistry , Blotting, Western , Embryonic Development , Fluorescent Antibody Technique , Immunohistochemistry , Microscopy, Confocal , Morula/chemistry , Oocytes/chemistry , RNA, Messenger/analysis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, trkA/genetics , Receptor, trkC/genetics , Reverse Transcriptase Polymerase Chain Reaction , Zygote/chemistry
10.
Clin Cancer Res ; 13(10): 3051-7, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17505008

ABSTRACT

PURPOSE: AZD2171 is an oral, highly potent, and selective vascular endothelial growth factor signaling inhibitor that inhibits all vascular endothelial growth factor receptor tyrosine kinases. The purpose of this study was to investigate the activity of AZD2171 in gastric cancer. EXPERIMENTAL DESIGN: We examined the antitumor effect of AZD2171 on the eight gastric cancer cell lines in vitro and in vivo. RESULTS: AZD2171 directly inhibited the growth of two gastric cancer cell lines (KATO-III and OCUM2M), with an IC(50) of 0.15 and 0.37 micromol/L, respectively, more potently than the epidermal growth factor receptor tyrosine kinase inhibitor gefitinib. Reverse transcription-PCR experiments and immunoblotting revealed that sensitive cell lines dominantly expressed COOH terminus-truncated fibroblast growth factor receptor 2 (FGFR2) splicing variants that were constitutively phosphorylated and spontaneously dimerized. AZD2171 completely inhibited the phosphorylation of FGFR2 and downstream signaling proteins (FRS2, AKT, and mitogen-activated protein kinase) in sensitive cell lines at a 10-fold lower concentration (0.1 micromol/L) than in the other cell lines. An in vitro kinase assay showed that AZD2171 inhibited kinase activity of immunoprecipitated FGFR2 with submicromolar K(i) values ( approximately 0.05 micromol/L). Finally, we assessed the antitumor activity of AZD2171 in human gastric tumor xenograft models in mice. Oral administration of AZD2171 (1.5 or 6 mg/kg/d) significantly and dose-dependently inhibited tumor growth in mice bearing KATO-III and OCUM2M tumor xenografts. CONCLUSIONS: AZD2171 exerted potent antitumor activity against gastric cancer xenografts overexpressing FGFR2. The results of these preclinical studies indicate that AZD2171 may provide clinical benefit in patients with certain types of gastric cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Quinazolines/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Dimerization , Gefitinib , Humans , Mice , Mice, Inbred Strains , Quinazolines/administration & dosage , Quinazolines/pharmacology , RNA, Small Interfering/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Stomach Neoplasms/enzymology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
11.
Hum Pathol ; 76: 100-109, 2018 06.
Article in English | MEDLINE | ID: mdl-29514108

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is a subtype of primary liver cancer rarely curable by surgery that is increasing rapidly in incidence. Chromosomal translocations and amplifications of the fibroblast growth factor receptor 2 (FGFR2) locus are present in several kinds of tumors including ICC, but their incidence has not been assessed in Chinese patients. Using break-apart probes and by determining the ratios of FGFR2/chromosome enumeration probe (CEP) 10 double-color probes, we evaluated 122 ICCs for the presence of FGFR2 translocations and amplifications, respectively, by fluorescence in situ hybridization. We further determined FGFR2 protein expression by immunohistochemistry and analyzed the clinicopathologic records of the patients. Eight tumors (6.6%) had FGFR2 translocations, whereas 15 (12.3%) had low-level FGFR2 amplification. Interestingly, the tumors that showed both translocation and low-level amplification frequently were of the mass-forming type. Compared with the ICCs with normal FGFR2s, tumors with amplifications secreted less mucus (P = .017) and typically were accompanied by hepatitis B virus infection (P = .004). Tumors with low-level amplification generally were of lower stage (P = .013) and associated with better overall survival (P = .017). As tumors with FGFR2 amplification exhibit different biology from lesions with a normal gene, low-level amplification of FGFR2 may play an important role in tumor progression and may be a marker for targeted therapy.


Subject(s)
Bile Duct Neoplasms/genetics , Biomarkers, Tumor/genetics , Cholangiocarcinoma/genetics , Gene Amplification , Receptor, Fibroblast Growth Factor, Type 2/genetics , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/chemistry , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/virology , Biomarkers, Tumor/analysis , China , Cholangiocarcinoma/chemistry , Cholangiocarcinoma/pathology , Cholangiocarcinoma/virology , Female , Genetic Predisposition to Disease , Hepatitis B virus/isolation & purification , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Staging , Phenotype , Prognosis , Receptor, Fibroblast Growth Factor, Type 2/analysis , Translocation, Genetic
12.
Anticancer Res ; 38(5): 3105-3110, 2018 05.
Article in English | MEDLINE | ID: mdl-29715147

ABSTRACT

BACKGROUND/AIM: The expression of IL-8 and FGFR has been related to prognosis and pathological features in renal cell carcinoma. We investigated the relationship between IL-8 and FGFR and the outcome in metastatic renal cell carcinoma (mRCC) patients. MATERIALS AND METHODS: Clinical data and histological samples of patients affected by mRCC and treated with targeted agents were reviewed. The expression of proteins was assessed using immunohistochemistry. RESULTS: FGFR1, FGFR2, and IL-8 were found to be expressed in 16%, 30%, and 50% of cases, respectively. Significant correlations were found between selected proteins. A lack of expression of FGFR2 and IL8 was found to be correlated with increased progression-free survival (PFS). The survival rate at 24 months was 44%, 38%, and 79% of those expressing both, one, or none of the evaluated proteins, respectively (p=0.047). CONCLUSION: This analysis found a relationship between the expression of IL-8 and FGFR2 in mRCC patients treated with targeted agents.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Aged , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/mortality , Disease-Free Survival , Female , Humans , Interleukin-8/analysis , Interleukin-8/biosynthesis , Kidney Neoplasms/drug therapy , Kidney Neoplasms/mortality , Male , Middle Aged , Prognosis , Receptor, Fibroblast Growth Factor, Type 1/analysis , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Retrospective Studies
13.
Breast Cancer Res ; 9(6): 113, 2007.
Article in English | MEDLINE | ID: mdl-18036273

ABSTRACT

Recently, genome-wide association studies of breast cancer revealed single nucleotide polymorphisms (SNPs) in five genes with novel association to susceptibility. While there is little doubt that the novel susceptibility markers produced from such highly powered studies are true, the mechanisms by which they cause the susceptibility remain undetermined. We have looked at the expression levels of the identified genes in tumours and found that they are highly significantly differentially expressed between the five established breast cancer subtypes. Also, a significant association between SNPs in these genes and their expression in tumours was seen as well as a significantly different frequency of the SNPs between the subtypes. This suggests that the observed genes are associated with different breast cancer subtypes, and may exert their effect through their expression in the tumours. Thus, future studies stratifying patients by their molecular subtypes may give much more power to classic case control studies, and genes of no or borderline significance may appear to be high-penetrant for certain subtypes and, therefore, be identifiable.


Subject(s)
Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Polymorphism, Single Nucleotide , Analysis of Variance , Apoptosis Regulatory Proteins , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , High Mobility Group Proteins , Humans , MAP Kinase Kinase Kinase 1/analysis , MAP Kinase Kinase Kinase 1/genetics , Microfilament Proteins/analysis , Microfilament Proteins/genetics , RNA, Long Noncoding , RNA, Untranslated/analysis , RNA, Untranslated/genetics , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptors, Progesterone/analysis , Receptors, Progesterone/genetics , Trans-Activators , Up-Regulation
14.
Arch Dermatol Res ; 299(7): 315-25, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17643254

ABSTRACT

Pseudoepitheliomatous hyperplasia (PEH) is an exuberant proliferation of the epidermis. The underlying mechanism(s) that lead to PEH have not been completely elucidated. Here, we characterize PEH during the healing stages of cutaneous leishmanial ulcers in mice. During experimental cutaneous leishmaniasis (CL) C57BL/6 mice produce PEH, and BALB/c do not. A series of immunohistochemical and immunological studies were performed to identify the secretory products of PEH regulation. We observed that the distribution of TNF-alpha and IFN-gamma under PEH had a stripe-like diffuse pattern and localized in the upper part of the papillary dermis directly under the proliferating epidermis. Macrophages were identified as the major source of TNF-alpha (56.3%). The importance of IFN-gamma and TNF-alpha in PEH development was proven by the initiation of PEH after three intralesional injections of TNF-alpha and IFN-gamma every three days in infected BALB/c mice. In C57BL/6 mice, keratinocyte growth factor (KGF) expressing cells were found immediately under the basal membrane of the hyperplastic epidermis in comparison with sporadic KGF positive cells deep in the dermis of BALB/c mice. Quantitative RT-PCR analysis demonstrated increased KGF and KGF receptor expression in uninfected C57BL/6 mice as compared to BALB/c mice. These data indicate that Th1 cytokines and KGF play a critical role in PEH initiation during CL.


Subject(s)
Epidermis/pathology , Fibroblast Growth Factor 7/physiology , Interferon-gamma/physiology , Leishmaniasis, Cutaneous/pathology , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Collagen/analysis , Epithelial Cells/pathology , Female , Fibroblast Growth Factor 7/analysis , Fibroblast Growth Factor 7/genetics , Hyperplasia , Mice , Mice, Inbred BALB C , Receptor, Fibroblast Growth Factor, Type 2/analysis
15.
Int J Oncol ; 28(2): 307-14, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16391783

ABSTRACT

The keratinocyte growth factor receptor, also known as KGFR/FGFR2 IIIb, is mainly localized in epithelial cells, and participates in the proliferation of these cells. In contrast, a recent study has revealed that the overexpression of KGFR in salivary adenocarcinoma induces growth inhibition, cell differentiation and apoptosis. We attempted to clarify the expression and role of KGFR in normal and cancerous human gastric tissues and cancer cell lines. Reverse-transcription polymerase chain reaction and Western blot analyses showed KGFR mRNA and its protein expression in NUGC-4, KATO-III and MKN-7 gastric cancer cell lines, but not in the NS-8 cell line. Immunohistochemically, KGFR immunoreactivity was weakly detected in the luminal surface of normal gastric epithelial cells. In addition, KGFR immunoreactivity was strongly detected in the nucleus and cytoplasm of many parietal cells. In gastric cancer tissue, KGFR was expressed in the cell membrane and cytoplasm of cancer cells in 46 of 126 (36.5%) cases. KGFR expression in gastric cancer cells was significantly associated with early-type macroscopic findings, shallow invasion of the gastric wall and expansive growth type. KGFR expression tended to correlate with a good prognosis in gastric cancer. These findings indicate that KGFR expression plays important roles in the differentiation of normal gastric epithelial cells and parietal cell functions. Furthermore, a decreased expression level or the non-expression of KGFR in gastric cancer cells may be associated with the proliferation and invasion of gastric cancer cells and a poor prognosis for the patient.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 2/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Cell Differentiation , Cell Line, Tumor , Epithelial Cells/metabolism , Female , Gastric Mucosa/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Invasiveness , Parietal Cells, Gastric/metabolism , Prognosis , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, Fibroblast Growth Factor, Type 2/genetics
16.
Eur Cell Mater ; 11: 27-33; discussion 34, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16447140

ABSTRACT

Tissue engineering of the small intestine offers an alternative to long-term intravenous nutrition and transplantation in patients with intestinal failure. Initial work, although encouraging, is limited by the volume of neonatal tissue required to produce a small neomucosal cyst. Our novel approach is to implant tubular poly-lactide-co-glycolide (PGLA) foam scaffolds subcutaneously. The aim of this study was to investigate whether these scaffolds would support growth of intestinal neomucosa. PGLA scaffolds were implanted subcutaneously into 8 Lewis rats; after 5 weeks, 'organoid units' were injected into the lumens. Tissue was assessed histologically after harvesting and quantitative immunohistochemistry was performed using antibodies against vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor 2 (VEGF-R2), fibroblast growth factor basic (bFGF) and fibroblast growth factor receptor 2 (FGF-R2). At 4 weeks post organoid unit implantation, clearly recognisable mucosa and submucosa was present on the luminal surface of the scaffold. Densities of VEGF and VEGF-R2 positive cells increased with time post organoid unit implantation. This pilot study demonstrates that it is possible to tissue engineer small intestinal neomucosa using subcutaneously implanted PLGA scaffolds. The yield of the process compares favourably to the published literature. Further work is required to optimise the technique.


Subject(s)
Implants, Experimental , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Lactic Acid , Polyglycolic Acid , Polymers , Tissue Engineering/methods , Animals , Cell Proliferation , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/physiology , Immunohistochemistry , Intestinal Mucosa/chemistry , Male , Materials Testing , Neovascularization, Physiologic , Organoids/cytology , Organoids/growth & development , Organoids/physiology , Pilot Projects , Polylactic Acid-Polyglycolic Acid Copolymer , Prostheses and Implants , Rats , Rats, Inbred Lew , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, Fibroblast Growth Factor, Type 2/physiology , Receptors, Vascular Endothelial Growth Factor/analysis , Receptors, Vascular Endothelial Growth Factor/physiology , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/physiology
17.
Cancer Res ; 76(21): 6331-6339, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27543601

ABSTRACT

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.


Subject(s)
Aminobenzoates/therapeutic use , Antibodies, Monoclonal/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Oligopeptides/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/analysis , Animals , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Receptor, Fibroblast Growth Factor, Type 2/immunology , Xenograft Model Antitumor Assays
18.
Oncotarget ; 7(16): 22234-44, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26993773

ABSTRACT

BACKGROUND: Malignant peripheral nerve sheath tumors (MPNST) are rare, highly malignant, and poorly understood sarcomas. The often poor outcome of MPNST highlights the necessity of identifying prognostic predictors for this aggressive sarcoma. Here, we investigate the role of fibroblast growth factor receptor (FGFR) family members in human MPNSTs. RESULTS: aCGH and bioinformatics analysis identified frequent amplification of the FGFR1 gene. FISH analysis revealed that 26.9% MPNST samples had amplification of FGFR1, with both focal and polysomy patterns observed. IHC identified that FGFR1 protein expression was positively correlated with FGFR1 gene amplification. High expression of FGFR1 protein was associated with better overall survival (OS) and was an independent prognostic predictor for OS of MPNST patients. Additionally, combined expression of FGFR1 and FGFR2 protein characterized a subtype of MPNST with better OS. FGFR4 protein was expressed 82.3% of MPNST samples, and was associated with poor disease-free survival. MATERIALS AND METHODS: We performed microarray-based comparative genomic hybridization (aCGH) profiling of two cohorts of primary MPNST tissue samples including 25 patients treated at The University of Texas MD Anderson Cancer Center and 26 patients from Tianjin Medical University Cancer Institute and Hospital. Fluorescence in situ hybridization (FISH) was used to validate the gene amplification detected by aCGH analysis. Another cohort of 63 formalin-fixed paraffin-embedded MPNST samples (including 52 samples for FISH assay) was obtained to explore FGFR1, 2, 3, and 4 protein expression by immunohistochemical (IHC) analysis. CONCLUSIONS: Our integrated genomic and molecular studies provide evidence that FGFRs play different prognostic roles in MPNST.


Subject(s)
Biomarkers, Tumor/analysis , Neurilemmoma/pathology , Receptors, Fibroblast Growth Factor/biosynthesis , Adult , Aged , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neurilemmoma/mortality , Prognosis , Proportional Hazards Models , Receptor, Fibroblast Growth Factor, Type 1/analysis , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Receptor, Fibroblast Growth Factor, Type 3/analysis , Receptor, Fibroblast Growth Factor, Type 3/biosynthesis , Receptor, Fibroblast Growth Factor, Type 4/analysis , Receptor, Fibroblast Growth Factor, Type 4/biosynthesis , Receptors, Fibroblast Growth Factor/analysis , Young Adult
19.
Gut Liver ; 10(5): 731-8, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27114414

ABSTRACT

BACKGROUND/AIMS: Pregnancy-associated gastric cancer is a rare condition. This case-control study was performed to identify the clinicopathological features and prognostic factors of pregnancy-associated gastric cancer. METHODS: All consecutive patients who presented to our tertiary referral hospital with pregnancy-associated gastric cancer from 1991 to 2012 were identified. Two age-, sex-, and stagematched controls for each case were also identified from the records. Clinicopathological, gynecological, and oncological outcomes were recorded. Immunohistochemical staining was performed for estrogen receptor, progesterone receptor, epidermal growth factor receptor, human epidermal growth factor receptor, and E-cadherin. Fluorescence in situ hybridization was performed for fibroblast growth factor receptor 2. RESULTS: The median overall survival rates of the pregnancyassociated gastric cancer and control groups were 7.0 months and 15.0 months, respectively (p=0.189). Poor prognostic factors included advanced stage and tumor location in the corpus or the entire stomach but not pregnancy status or loss of E-cadherin. Pregnancy-associated gastric cancer was associated with a longer time from diagnosis to treatment (21 days vs 7 days, p=0.021). The two groups did not differ in the expression of the receptors or E-cadherin. CONCLUSIONS: The dismal prognosis of pregnancy-associated gastric cancer may related to the tumor stage and location rather than to pregnancy itself.


Subject(s)
Pregnancy Complications, Neoplastic/mortality , Stomach Neoplasms/mortality , Adult , Biomarkers, Tumor/analysis , Cadherins/analysis , Case-Control Studies , ErbB Receptors/analysis , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Neoplasm Staging , Pregnancy , Pregnancy Complications, Neoplastic/pathology , Prognosis , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Stomach/pathology , Stomach Neoplasms/chemistry , Stomach Neoplasms/pathology , Survival Rate , Time-to-Treatment , Young Adult
20.
Am J Clin Pathol ; 143(6): 865-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25972329

ABSTRACT

OBJECTIVES: Fibroblast growth factor receptor 2 (FGFR2) amplification has been reported to be a target for treatment in gastric cancer. However, an optimal tissue source and method for evaluating FGFR2 have yet to be established. METHODS: Copy numbers were compared by quantitative polymerase chain reaction (qPCR) using frozen vs formalin-fixed, paraffin-embedded (FFPE) tissue and biopsy vs surgical specimens. We correlated the results of qPCR and immunohistochemistry (IHC) with fluorescence in situ hybridization (FISH) using stage IV gastric cancer biopsy specimens and validated the results in surgical specimens. RESULTS: FFPE tissues were suitable for qPCR, and biopsy specimens were equivalent to or better than surgical specimens. qPCR and IHC results exhibited an excellent correlation with FISH at eight or more copies by qPCR in any kind of tissue, 5% or more by IHC in biopsy specimens, and 10% or more by IHC in surgical specimens. FGFR2 amplification was 6.6% in stage IV gastric cancers, and 42% of these showed heterogeneous amplification and overexpression. IHC indicated a good correlation with FISH even in the heterogeneous cases. CONCLUSIONS: FFPE biopsy tissues are an adequate source for FGFR2 evaluation in gastric carcinomas, and a qPCR-based copy number assay can be used for screening. IHC is also a valid and practical method for evaluating FGFR2, considering frequent heterogeneity.


Subject(s)
Adenocarcinoma/genetics , Receptor, Fibroblast Growth Factor, Type 2/analysis , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stomach Neoplasms/genetics , Aged , Female , Gene Dosage , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL