epidermal-growth-factor and Carcinoma--Pancreatic-Ductal

epidermal-growth-factor has been researched along with Carcinoma--Pancreatic-Ductal* in 16 studies

Reviews

1 review(s) available for epidermal-growth-factor and Carcinoma--Pancreatic-Ductal

ArticleYear
VAV1: a new target in pancreatic cancer?
    Cancer biology & therapy, 2005, Volume: 4, Issue:5

    Pancreatic ductal adenocarcinoma (PDA) is arguably the most lethal malignancy in the United States. Despite the identification of many molecular alterations in PDA, this information has not translated into effective therapeutic strategies to date. A recent report in Cancer Cell (Fernandez-Zapico et al, Cancer Cell 2005, 7:39-49) reveals an unexpected role for the hematopoietic-specific RhoGEF VAV1 in pancreatic tumorigenesis, where ectopic expression of VAV1 as a result of promoter demethylation was identified in the majority of established cell lines and PDA tissue samples. Importantly, VAV1 expression was functionally required for optimal proliferation, transformation and survival of pancreatic cancer cell lines. This study provides the first evidence of VAV1 promoter demethylation as an event in cancer progression, suggesting that aberrant signaling pathways driven by VAV1 are potential therapeutic targets in PDA.

    Topics: Animals; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Proliferation; Cell Survival; Cell Transformation, Neoplastic; DNA Methylation; Enzyme Activation; Epidermal Growth Factor; Humans; Mice; Pancreatic Neoplasms; Promoter Regions, Genetic; Proto-Oncogene Proteins c-vav; rac1 GTP-Binding Protein; RNA Interference; RNA, Messenger; Signal Transduction

2005

Other Studies

15 other study(ies) available for epidermal-growth-factor and Carcinoma--Pancreatic-Ductal

ArticleYear
Adaptive EGF expression sensitizes pancreatic cancer cells to ionizing radiation through activation of the cyclin D1/P53/PARP pathway.
    International journal of oncology, 2019, Volume: 54, Issue:4

    It is well-known that the activation status of the P53, signal transducer and activator of transcription (Stat)3 and nuclear factor (NF)‑κB signaling pathways determines the radiosensitivity of cancer cells. However, the function of these pathways in radiosensitive vs radioresistant cancer cells remains elusive. The present study demonstrated that adaptive expression of epidermal growth factor (EGF) following exposure to ionizing radiation (IR) may induce radiosensitization of pancreatic cancer (PC) cells through induction of the cyclin D1/P53/poly(ADP‑ribose) polymerase pathway. By contrast, adaptively expressed interleukin (IL)‑6 and insulin‑like growth factor (IGF)‑1 may promote radioresistance of PC cells, likely through activation of the Stat3 and NF‑κB pathways. In addition, cyclin D1 and survivin, which are specifically expressed in the G1/S and G2/M phase of the cell cycle, respectively, are mutually exclusive in radiosensitive and radioresistant PC cells, while Bcl‑2 and Bcl‑xL expression does not differ between radiosensitive and radioresistant PC cells. Therefore, adaptively expressed EGF and IL‑6/IGF‑1 may alter these pathways to promote the radiosensitivity of PC cancers. The findings of the present study highlight potential makers for the evaluation of radiosensitivity and enable the development of effective regimens for cancer radiotherapy.

    Topics: Apoptosis; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cyclin D1; Epidermal Growth Factor; Gene Expression Regulation, Neoplastic; Humans; Pancreatic Neoplasms; Poly (ADP-Ribose) Polymerase-1; Radiation Tolerance; Signal Transduction; Tumor Suppressor Protein p53; Up-Regulation

2019
TMEM16A controls EGF-induced calcium signaling implicated in pancreatic cancer prognosis.
    Proceedings of the National Academy of Sciences of the United States of America, 2019, 06-25, Volume: 116, Issue:26

    Pancreatic cancer typically spreads rapidly and has poor survival rates. Here, we report that the calcium-activated chloride channel TMEM16A is a biomarker for pancreatic cancer with a poor prognosis. TMEM16A is up-regulated in 75% of cases of pancreatic cancer and high levels of TMEM16A expression are correlated with low patient survival probability. TMEM16A up-regulation is associated with the ligand-dependent EGFR signaling pathway. In vitro, TMEM16A is required for EGF-induced store-operated calcium entry essential for pancreatic cancer cell migration. TMEM16A also has a profound impact on phosphoproteome remodeling upon EGF stimulation. Moreover, molecular actors identified in this TMEM16A-dependent EGFR-induced calcium signaling pathway form a gene set that makes it possible not only to distinguish neuro-endocrine tumors from other forms of pancreatic cancer, but also to subdivide the latter into three clusters with distinct genetic profiles that could reflect their molecular underpinning.

    Topics: Anoctamin-1; Biomarkers, Tumor; Calcium Signaling; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Movement; Datasets as Topic; Diagnosis, Differential; Epidermal Growth Factor; ErbB Receptors; HEK293 Cells; Humans; Neoplasm Proteins; Pancreas; Pancreatic Neoplasms; Prognosis; RNA-Seq; RNA, Small Interfering; Survival Rate; Up-Regulation

2019
miR-21 promotes EGF-induced pancreatic cancer cell proliferation by targeting Spry2.
    Cell death & disease, 2018, 11-21, Volume: 9, Issue:12

    Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant cancer that lacks effective targets for therapy. Alteration of epidermal growth factor (EGF) expression has been recognized as an essential molecular event in pancreatic carcinogenesis. Accumulating studies have demonstrated that miRNAs play critical roles in EGF signaling regulation, tumor initiation, cell proliferation and apoptosis. Here, we demonstrated that miR-21 expression was induced by EGF in pancreatic cancer cells. miR-21 promoted EGF-induced proliferation, inhibited cell apoptosis and accelerated cell cycle progression. In vivo experiments confirmed the influence of miR-21 on tumor growth. Mechanistic studies revealed that miR-21 targeted MAPK/ERK and PI3K/AKT signaling pathways to modulate cell proliferation. In addition, Spry2 was proven to be a target of miR-21. Furthermore, miR-21 and Spry2 were significantly related to clinical features and may be valuable predictors of PDAC patient prognosis.

    Topics: Adenocarcinoma; Aged; Animals; Apoptosis; Carcinogenesis; Carcinoma, Pancreatic Ductal; Cell Cycle; Cell Movement; Cell Proliferation; Disease-Free Survival; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Heterografts; Humans; Intracellular Signaling Peptides and Proteins; Kaplan-Meier Estimate; Male; Membrane Proteins; Mice; MicroRNAs; Middle Aged; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; Signal Transduction

2018
Calreticulin promotes EGF-induced EMT in pancreatic cancer cells via Integrin/EGFR-ERK/MAPK signaling pathway.
    Cell death & disease, 2017, 10-26, Volume: 8, Issue:10

    Our previous study showed that Calreticulin (CRT) promoted the development of pancreatic cancer (PC) through ERK/MAPK pathway. We next investigate whether CRT promotes EGF-induced epithelial-mesenchymal transition (EMT) in PC via Integrin/EGFR-ERK/MAPK signaling, which has not been reported yet to our knowledge. EGF simultaneously induced EMT and activated Integrin/EGFR-ERK/MAPK signaling pathway in 3 PC cells. However, CRT silencing significantly inhibited EGF function, including inhibiting EGF-induced EMT-like cell morphology, EGF-enhanced cell invasion and migration, and EGF induced the decrease of E-cadherin, ZO-1, and β-catenin and the increase of the key proteins in Integrin/EGFR-ERK/MAPK signaling (pEGFR-tyr1173, Fibronectin, Integrinβ1, c-Myc and pERK). Conversely, CRT overexpression rescued the change of EMT-related proteins induced by EGF in CRT silencing PC cells. Additionally, CRT was co-stained with pEGFR1173 (with EGF), Fibronectin and Integrinβ1 by IF under confocal microscopy and was co-immunoprecipitated with Fibronectin, Integrinβ1 and c-Myc in both PC cells, all of which indicating a close interaction of CRT with Integrin/EGFR-ERK/MAPK signaling pathway in PC. In vivo, CRT silencing inhibited subcutaneous tumor growth and liver metastasis of pancreatic tumor. A positive relationship of CRT with Fibronectin, Integrinβ1, c-Myc and pERK and a negative association of CRT with E-cad was also observed in vivo and clinical samples. Meanwhile, overexpression of the above proteins was closely associated with multiple aggressive clinicopathological characteristics and the poor prognosis of PC patients. CRT promotes EGF-induced EMT in PC cells via Integrin/EGFR-ERK/MAPK signaling pathway, which would be a promising therapy target for PC.

    Topics: Animals; Calreticulin; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; Heterografts; Humans; Integrins; MAP Kinase Signaling System; Mice; Mice, Inbred BALB C; Mice, Nude; Pancreatic Neoplasms; Transfection

2017
Metformin Restrains Pancreatic Duodenal Homeobox-1 (PDX-1) Function by Inhibiting ERK Signaling in Pancreatic Ductal Adenocarcinoma.
    Current molecular medicine, 2016, Volume: 16, Issue:1

    Pancreatic ductal adenocarcinoma (PDAC) is one of the most potent and perilous diseases known, with a median survival rate of 3-5 months due to the combination of only advanced stage diagnosis and ineffective therapeutic options. Metformin (1,1-Dimethylbiguanide hydrochloride), the leading drug used for type 2 diabetes mellitus, emerges as a potential therapy for PDAC and other human cancers. Metformin exerts its anticancer action via a variety of adenosine monophosphate (AMP)-activated protein kinase (AMPK)- dependent and/or AMPK-independent mechanisms. We present data here showing that metformin downregulated pancreatic transcription factor pancreatic duodenal homeobox-1 (PDX-1), suggesting a potential novel mechanism by which metformin exerts its anticancer action. Metformin inhibited PDX-1 expression at both protein and mRNA levels and PDX-1 transactivity as well in PDAC cells. Extracellular signal-regulated kinase (ERK) was identified as a PDX-1-interacting protein by antibody array screening in GFP-PDX-1 stable HEK293 cells. Co-transfection of ERK1 with PDX-1 resulted in an enhanced PDX-1 expression in HEK293 cells in a dose-dependent manner. Immunoprecipitation/Western blotting analysis confirmed the ERK-PDX-1 interaction in PANC-1 cells stimulated by epidermal growth factor (EGF). EGF induced an enhanced PDX-1 expression in PANC-1 cells and this stimulation was inhibited by MEK inhibitor PD0325901. Metformin inhibited EGF-stimulated PDX-1 expression with an accompanied inhibition of ERK kinase activation in PANC- 1 cells. Taken together, our studies show that PDX-1 is a potential novel target for metformin in PDAC cells and that metformin may exert its anticancer action in PDAC by down-regulating PDX-1 via a mechanism involving inhibition of ERK signaling.

    Topics: Adenocarcinoma; Carcinoma, Pancreatic Ductal; Cell Line; Cell Line, Tumor; Diabetes Mellitus, Type 2; Down-Regulation; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; Genes, Homeobox; HEK293 Cells; Homeodomain Proteins; Humans; MAP Kinase Signaling System; Metformin; Pancreas; Pancreatic Neoplasms; Protein Kinase Inhibitors; Signal Transduction; Trans-Activators

2016
Assessment of different 3D culture systems to study tumor phenotype and chemosensitivity in pancreatic ductal adenocarcinoma.
    International journal of oncology, 2016, Volume: 49, Issue:1

    Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant disease with a very poor prognosis, due to the influence of the tumor stroma, which promotes tumor growth, early invasion and chemoradiation resistance. Efforts to develop models for identifying novel anticancer therapeutic compounds have been hampered by the limited ability of in vitro models to mimic these in vivo tumor-stroma interactions. This has led to the development of various three-dimensional (3D) culture platforms recapitulating the in vivo tumor-stroma crosstalk and designed to better understand basic cancer processes and screen drug action. However, a consensus for different experimental 3D platforms is still missing in PDAC. We compared four PDAC cell lines of different malignancy grown in 2D monolayers to three of the more commonly used 3D techniques (ultralow adhesion concave microwells, Matrigel inclusion and organotypic systems) and to tumors derived from their orthotopic implantation in mice. In these 3D platforms, we observed that cells grow with very different tumor morphologies and the organotypic setting most closely resembles the tumor cytoarchitecture obtained by orthotopically implanting the four cell lines in mice. We then analyzed the molecular and cellular responses of one of these cell lines to epidermal growth factor receptor (EGFR) stimulation with EGF and inhibition with erlotinib and found that only in the 3D platforms, and especially the organotypic, cells: i) responded to EGF by changing the expression of signalling components underlying cell-stroma crosstalk and tissue architecture, growth, invasion and drug resistance (E-cadherin, EGFR, ezrin, β1 integrin, NHERF1 and HIF-1α) similar to those reported in vivo; ii) had stimulated growth and increased erlotinib sensitivity in response to EGF, more faithfully mimicking their known in vivo behaviour. Altogether, these results, indicate the organotypic as the most relevant physiological 3D system to study the complex tumor stroma interactions driving progression and determining chemio-resistance.

    Topics: Adenocarcinoma; Animals; Carcinoma, Pancreatic Ductal; Cell Culture Techniques; Cell Line, Tumor; Cell Proliferation; Drug Resistance, Neoplasm; Epidermal Growth Factor; ErbB Receptors; Erlotinib Hydrochloride; Humans; Mice; Stromal Cells

2016
microRNA-10b enhances pancreatic cancer cell invasion by suppressing TIP30 expression and promoting EGF and TGF-β actions.
    Oncogene, 2014, Sep-18, Volume: 33, Issue:38

    Increased microRNA-10b (miR-10b) expression in the cancer cells in pancreatic ductal adenocarcinoma (PDAC) is a marker of disease aggressiveness. In the present study, we determined that plasma miR-10b levels are significantly increased in PDAC patients by comparison with normal controls. By gene profiling, we identified potential targets downregulated by miR-10b, including Tat-interacting protein 30 (TIP30). Immunoblotting and luciferase reporter assays confirmed that TIP30 was a direct miR-10b target. Downregulation of TIP30 by miR-10b or siRNA-mediated silencing of TIP30 enhanced epidermal growth factor (EGF)-dependent invasion. The actions of miR-10b were abrogated by expressing a modified TIP30 cDNA resistant to miR-10b. EGF-induced EGF receptor (EGFR) tyrosine phosphorylation and extracellular signal-regulated kinase phosphorylation were enhanced by miR-10b, and these effects were mimicked by TIP30 silencing. The actions of EGF in the presence of miR-10b were blocked by EGFR kinase inhibition with erlotinib and by dual inhibition of PI3K (phosphatidylinositol 3'-kinase) and MEK. Moreover, miR-10b, EGF and transforming growth factor-beta (TGF-β) combined to markedly increase cell invasion, and this effect was blocked by the combination of erlotinib and SB505124, a type I TGF-β receptor inhibitor. miR-10b also enhanced the stimulatory effects of EGF and TGF-β on cell migration and epithelial-mesenchymal transition (EMT) and decreased the expression of RAP2A, EPHB2, KLF4 and NF1. Moreover, miR-10b overexpression accelerated pancreatic cancer cell (PCC) proliferation and tumor growth in an orthotopic model. Thus, plasma miR-10b levels may serve as a diagnostic marker in PDAC, whereas intra-tumoral miR-10b promotes PCC proliferation and invasion by suppressing TIP30, which enhances EGFR signaling, facilitates EGF-TGF-β cross-talk and enhances the expression of EMT-promoting genes, whereas decreasing the expression of several metastasis-suppressing genes. Therefore, therapeutic targeting of miR-10b in PDAC may interrupt growth-promoting deleterious EGF-TGF-β interactions and antagonize the metastatic process at various levels.

    Topics: Acetyltransferases; Animals; Antineoplastic Agents; Base Sequence; Binding Sites; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Epidermal Growth Factor; Erlotinib Hydrochloride; Gene Expression; Gene Expression Regulation, Neoplastic; Humans; Kruppel-Like Factor 4; Male; Mice; Mice, Nude; MicroRNAs; Neoplasm Invasiveness; Neoplasm Transplantation; Pancreatic Neoplasms; Quinazolines; RNA Interference; Signal Transduction; Transcription Factors; Transforming Growth Factor beta

2014
Polarization of the vacuolar adenosine triphosphatase delineates a transition to high-grade pancreatic intraepithelial neoplasm lesions.
    Pancreas, 2014, Volume: 43, Issue:8

    A functional vacuolar adenosine triphosphatase (v-ATPase) complex regulates canonical Wnt/β-catenin signaling. The goal of this study was to identify the distribution of the v-ATPase in human and murine models of pancreatic intraepithelial neoplasms (PanINs) and assess its role in Wnt/β-catenin signaling.. We evaluated the immunolabeling pattern of the v-ATPase in human PanIN specimens and murine PanIN-1 and PanIN-2 lesions obtained from Ptf1a(Cre/+); LSL-Kras(G12D) mice. Wnt/β-catenin signaling was interrogated in primary PanIN cells by examining the phosphorylated levels of its surface coreceptor, low-density lipoprotein receptor-related protein-6 (LRP6), and its intracellular effector, nonphosphorylated β-catenin. The response of primary PanIN cells to epidermal growth factor (EGF) was assessed in the absence and presence of the v-ATPase inhibitor, concanamycin.. In advanced (PanIN-2), but not early (PanIN-1), lesions, the v-ATPase assumed a polarized phenotype. Blocking the v-ATPase disrupted Wnt/β-catenin signaling in primary PanIN cells despite significantly higher levels of the total and activated Wnt cell surface coreceptor, LRP6. Vacuolar adenosine triphosphatase blockade significantly decreased the total and activated levels of EGF receptor, a determinant of PanIN progression. The activation of EGF receptor and its intracellular mediator, p44/42 mitogen-activated protein kinase, was also reduced by v-ATPase blockade. This led to diminished proliferation in response to EGF ligand.. The v-ATPase regulates Wnt/β-catenin and EGF receptor signaling in PanINs.

    Topics: Adenocarcinoma in Situ; Alcian Blue; Animals; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Polarity; Disease Progression; Epidermal Growth Factor; ErbB Receptors; Humans; Islets of Langerhans; Low Density Lipoprotein Receptor-Related Protein-6; Membrane Proteins; Mice; Mice, Mutant Strains; Mitogen-Activated Protein Kinase 3; Neoplasm Grading; Neoplasm Proteins; Pancreatic Neoplasms; Protein Transport; Staining and Labeling; Vacuolar Proton-Translocating ATPases; Wnt Signaling Pathway

2014
Pancreatic ductal adenocarcinoma mice lacking mucin 1 have a profound defect in tumor growth and metastasis.
    Cancer research, 2011, Jul-01, Volume: 71, Issue:13

    MUC1 is overexpressed and aberrantly glycosylated in more than 60% of pancreatic ductal adenocarcinomas. The functional role of MUC1 in pancreatic cancer has yet to be fully elucidated due to a dearth of appropriate models. In this study, we have generated mouse models that spontaneously develop pancreatic ductal adenocarcinoma (KC), which are either Muc1-null (KCKO) or express human MUC1 (KCM). We show that KCKO mice have significantly slower tumor progression and rates of secondary metastasis, compared with both KC and KCM. Cell lines derived from KCKO tumors have significantly less tumorigenic capacity compared with cells from KCM tumors. Therefore, mice with KCKO tumors had a significant survival benefit compared with mice with KCM tumors. In vitro, KCKO cells have reduced proliferation and invasion and failed to respond to epidermal growth factor, platelet-derived growth factor, or matrix metalloproteinase 9. Further, significantly less KCKO cells entered the G(2)-M phase of the cell cycle compared with the KCM cells. Proteomics and Western blotting analysis revealed a complete loss of cdc-25c expression, phosphorylation of mitogen-activated protein kinase (MAPK), as well as a significant decrease in nestin and tubulin-α2 chain expression in KCKO cells. Treatment with a MEK1/2 inhibitor, U0126, abrogated the enhanced proliferation of the KCM cells but had minimal effect on KCKO cells, suggesting that MUC1 is necessary for MAPK activity and oncogenic signaling. This is the first study to utilize a Muc1-null PDA mouse to fully elucidate the oncogenic role of MUC1, both in vivo and in vitro.

    Topics: Animals; Butadienes; Carcinoma, Pancreatic Ductal; Cell Cycle; Cell Growth Processes; Epidermal Growth Factor; Humans; Intermediate Filament Proteins; Matrix Metalloproteinase 9; Mice; Mice, Inbred C57BL; Mice, Knockout; Mitogen-Activated Protein Kinases; Mucin-1; Neoplasm Metastasis; Nerve Tissue Proteins; Nestin; Nitriles; Pancreatic Neoplasms; Platelet-Derived Growth Factor; Protein Kinase Inhibitors; Tubulin

2011
A novel 3-dimensional culture system uncovers growth stimulatory actions by TGFβ in pancreatic cancer cells.
    Cancer biology & therapy, 2011, Aug-01, Volume: 12, Issue:3

    Transforming Growth Factor-β (TGF-β) exerts cell type-specific and context-dependent effects. Understanding the intrinsic effects of TGF-β on cancer cells in pancreatic ductal adenocarcinoma (PDAC) is a prerequisite for rationalized clinical implementation of TGF-β targeting therapies. Since the tumor microenvironment can affect how cancer cell respond to TGF-β, we employed a novel three-dimensional (3D) culturing system to recapitulate stromal and extracellular matrix interactions. We show here that TGF-β stimulates growth of human and murine pancreatic cancer cell lines (PCCs) when embedded in a 3% collagen IV/laminin-rich gelatinous medium (Matrigel™) over a solidified layer of soft agar. Moreover, in this novel 3D model, concomitant treatment with TGF-β1 and epidermal growth factor (EGF) enhanced PCC growth to a greater extent than either growth factor alone, and conferred increased chemoresistance to cytotoxic compounds. These cooperative growth-stimulatory effects were blocked by pharmacological inhibition of TGF-β type I receptor with SB431542 or the EGF receptor with erlotinib. Co-incubation with SB431542 and erlotinib enhanced the efficacy of gemcitabine and cisplatin in PCCs and in primary cell cultures established from pancreata of genetically-engineered mouse models of PDAC. These findings suggest that concomitant inhibition of TGF-β and EGF signaling may represent an effective therapeutic strategy in PDAC, and that this 3D culturing system could be utilized to test ex vivo the therapeutic response of pancreatic tumor biopsies from PDAC patients, thereby providing a functional assay to facilitate personalized targeted therapies.

    Topics: Animals; Benzamides; Carcinoma, Pancreatic Ductal; Cell Culture Techniques; Cell Line, Tumor; Cell Proliferation; Cisplatin; Culture Media; Deoxycytidine; Dioxoles; Drug Resistance, Neoplasm; Epidermal Growth Factor; ErbB Receptors; Erlotinib Hydrochloride; Extracellular Matrix; Gemcitabine; Humans; Mice; Mice, Transgenic; Pancreatic Neoplasms; Quinazolines; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Transforming Growth Factor beta1; Tumor Microenvironment

2011
Expression and prognostic value of circulating angiogenic cytokines in pancreatic cancer.
    BMC cancer, 2011, Jul-05, Volume: 11

    The utility of circulating angiogenic cytokines (CAC) as biomarkers in pancreatic cancer has not been clarified yet. We investigated the expression and prognostic associations of seven CAC in patients with pancreatic cancer.. Serum samples were collected preoperatively in patients undergoing surgery for localized pancreatic cancer (n = 74), metastatic pancreatic cancer (n = 24) or chronic pancreatitis (n = 20) and in healthy controls (n = 48). Quantitative enzyme-linked immunosorbent assays and multiplex protein arrays were used to determine circulating levels of VEGF, VEGFR-1, PlGF, PDGF-AA, PDGF-BB, Ang-1 and EGF. Multivariate analyses on cancer-specific survival were performed with a Cox proportional hazards model.. VEGF (p < 0.0001), PDGF-AA (p < 0.0001), Ang-1 (p = 0.002) and EGF (p < 0.0001) were differentially expressed in patients with pancreatic cancer compared to healthy controls. The presence of lymph node metastases was associated with increased levels of all CAC except for PlGF, whereas there were only minor associations of CAC with other clinicopathologic variables. The multivariate model including the entire angiogenic panel revealed high levels of circulating PDGF-AA (hazard ratio 4.58; 95% confidence interval 1.43 - 14.69) as predictor of poor cancer-specific survival, whereas high levels of PDGF-BB (0.15; 0.15 - 0.88), Ang-1 (0.30; 0.10 - 0.93) and VEGF (0.24; 0.09 - 0.57) were associated with a favorable prognosis.. Circulating levels of certain angiogenic cytokines correlate with patients' prognosis after resection for pancreatic cancer, if a panel of several CAC is considered simultaneously. These data should be considered in future studies evaluating angiogenic factors as prognostic biomarkers and therapeutic targets in patients with pancreatic cancer.

    Topics: Aged; Angiogenic Proteins; Angiotensin I; Becaplermin; Carcinoma, Pancreatic Ductal; Cytokines; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Humans; Kaplan-Meier Estimate; Male; Membrane Proteins; Middle Aged; Neoplasm Proteins; Pancreatic Neoplasms; Platelet-Derived Growth Factor; Prognosis; Proportional Hazards Models; Proto-Oncogene Proteins c-sis; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factor Receptor-1

2011
Cripto-1 overexpression is involved in the tumorigenesis of gastric-type and pancreatobiliary-type intraductal papillary mucinous neoplasms of the pancreas.
    Oncology reports, 2009, Volume: 21, Issue:1

    Human Cripto-1, a membrane-bound protein, plays an important role during early embryogenesis and has oncogenic properties, including cell transformation and enhancement of invasion. Cripto-1 is up-regulated in various malignant tissues and premalignant lesions. However, Cripto-1 expression in intraductal papillary mucinous neoplasms (IPMNs) has yet to be reported. This study aimed to investigate Cripto-1 expression in IPMNs and evaluate the expression patterns according to the histological grade or phenotypic subclassification. Cripto-1 expression was evaluated by immunohistochemistry using 37 IPMN tissue samples and real-time RT-PCR analysis of seven frozen samples. Cripto-1 was up-regulated in 59.5% of IPMNs. Cripto-1 was positively stained in 3 of 4 (75%) adenomas, 12 of 19 (63.2%) borderline neoplasms, 5 of 11 (45.5%) non-invasive carcinomas and 2 of 3 (66.7%) invasive carcinomas. There was no correlation between Cripto-1 overexpression and the histological grade (P>0.05). Cripto-1 expression was significantly increased in pancreatobiliary- (4/5, 80%) and gastric-type (13/19, 68.4.2%) IPMNs compared with those of the intestinal type (2/10, 20%; P<0.01). Cripto-1 mRNA expression was higher in gastric- and pancreatobiliary-type IPMNs than in intestinal ones, supporting the immunohistochemical results. It is concluded that Cripto-1 overexpression is involved in the tumorigenesis of gastric- and pancreatobiliary-type IPMNs.

    Topics: Adenocarcinoma, Mucinous; Adenocarcinoma, Papillary; Adult; Aged; Biomarkers, Tumor; Carcinoma, Pancreatic Ductal; Epidermal Growth Factor; Female; Gene Expression; GPI-Linked Proteins; Humans; Immunohistochemistry; Intercellular Signaling Peptides and Proteins; Male; Membrane Glycoproteins; Middle Aged; Neoplasm Proteins; Pancreatic Neoplasms; Reverse Transcriptase Polymerase Chain Reaction

2009
Expression of epidermal growth factors and apoptosis markers in pancreatic ductal adenocarcinoma.
    Folia histochemica et cytobiologica, 2009, Volume: 47, Issue:4

    Epidermal growth factor family members: EGF, EGFR and the c-erbB-2(HER-2/neu) are involved in the growth of pancreatic ductal carcinoma, its invasiveness and metastases. Similarly, proteins regulating apoptosis can influence the development of pancreatic cancer. The aim of our study was to assess the expressions of EGF, EGFR, c-erbB-2, Bax and Bcl-xL in comparison with anatomo-clinical parameters. We also analyzed the relationship between the epidermal growth factors and apoptosis-regulating proteins.. The levels of these proteins were determined immunohistochemically in 29 pancreatic ductal carcinoma cases.. We found no correlation of EGF, EGFR, c-erbB-2, Bax and Bcl-xL with age and gender of patients, or histological type and grade of malignancy (G). However, we observed a very strong correlation between EGF, EGFR, Bax, Bcl-xL and lymph node metastases (p=0.000, p=0.001, p=0.008, p=0.012, respectively) and between EGF, EGFR and distant metastases (p=0.002, p=0.008, respectively). Moreover, we found a correlation between Bcl-xL and c-erbB-2 (p=0.030) and between EGF and Bax (p=0.041).. These investigations seem to suggest that both epidermal growth factors (EGF, EGFR) and apoptosis-regulating proteins (Bax and Bcl-xL) play an essential role in lymph node involvement. Moreover EGF and EGFR are involved in distant metastases. The apoptosis markers appear to cooperate with epidermal growth factor proteins in the process of tumor spread.

    Topics: Adult; Aged; Apoptosis; bcl-2-Associated X Protein; bcl-X Protein; Biomarkers, Tumor; Carcinoma, Pancreatic Ductal; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Male; Middle Aged; Pancreatic Neoplasms; Proto-Oncogene Proteins c-bcl-2

2009
Neurotensin and EGF induce synergistic stimulation of DNA synthesis by increasing the duration of ERK signaling in ductal pancreatic cancer cells.
    Journal of cellular physiology, 2005, Volume: 202, Issue:3

    Neurotensin (NT) and epidermal growth factor (EGF) induced rapid extracellular-regulated protein kinase (ERK) activation through different signaling pathways in the K-Ras mutated human pancreatic carcinoma cell lines PANC-1 and MIA PaCa-2. NT stimulated ERK activation via a protein kinase C (PKC)-dependent (but EGF receptor-independent) pathway in PANC-1 and MIA PaCa-2 cells, whereas EGF promoted ERK activation through a PKC-independent pathway in these cells. Concomitant stimulation of these cells with NT and EGF induced a striking increase in the duration of ERK pathway activation as compared with that obtained in cells treated with each agonist alone. Stimulation with NT + EGF promoted synergistic stimulation of DNA synthesis and anchorage-independent growth. Addition of the MEK inhibitor U0126, either prior to stimulation with NT + EGF or 2 h after stimulation with NT + EGF prevented the synergistic increase in DNA synthesis and suppressed the sustained phase of ERK activation. Furthermore, treatment with the selective PKC inhibitor GF-1 converted the sustained ERK activation in response to NT and EGF into a transient signal and also abrogated the synergistic increase in DNA synthesis. Collectively, our results suggest that the sustained phase of ERK signaling mediates the synergistic effects of NT and EGF on DNA synthesis in pancreatic cancer cells.

    Topics: Butadienes; Carcinoma, Pancreatic Ductal; Cell Line, Tumor; Cell Proliferation; DNA Replication; Enzyme Activation; Enzyme Inhibitors; Epidermal Growth Factor; Extracellular Signal-Regulated MAP Kinases; Humans; MAP Kinase Signaling System; Neurotensin; Nitriles; Protein Kinase C

2005
Significance of cellular distribution of ezrin in pancreatic cystic neoplasms and ductal adenocarcinoma.
    Archives of surgery (Chicago, Ill. : 1960), 2005, Volume: 140, Issue:12

    Pancreatic cystic neoplasms and ductal adenocarcinoma manifest diverse clinical features and prognoses, which might be related to cellular distribution of ezrin modulated through various trophic molecules.. Laboratory investigation and retrospective analysis.. Medical school-affiliated university hospital.. Patients with solid pseudopapillary tumor (SPT) (n = 12), mucinous cystic neoplasm (MCN) (n = 18), intraductal papillary mucinous tumor (IPMT) (n = 18), and ductal adenocarcinoma (PA) (n = 73) of the pancreas were studied. Expression of epidermal growth factor (EGF) and its receptor (EGFR) and ezrin were determined using immunohistochemistry. Epidermal growth factor receptor and ezrin expression in sodium butyrate (SB)-treated PA cell line PANC-1 was determined using immunocytochemistry. Messenger RNA expression of ezrin in the PANC-1 cell line treated with SB was determined using reverse transcriptase-polymerase chain reaction. Multivariate analysis of survival of patients with PA was performed.. None of 12 SPTs displayed synchronous expression of EGF and EGFR, while all 3 malignant SPTs displayed membranous ezrin expression. One of 18 MCNs displayed synchronous expression of EGF and EGFR, while 4 of 6 borderline malignant and 8 of 8 malignant MCNs displayed membranous ezrin expression. Two of 4 borderline malignant and 11 of 11 malignant IPMTs displayed synchronous expression of EGF and EGFR, and all borderline malignant and malignant IPMTs displayed membranous ezrin expression. Less differentiated PA displayed EGF, EGFR, and membranous ezrin expression more frequently compared with more differentiated PA. Epidermal growth factor receptor expression of PANC-1 cells decreased in an SB dose dependent manner, in which PANC-1 cells became more differentiated and membranous ezrin expression of PANC-1 cells decreased correspondingly. Messenger RNA expression of ezrin in PANC-1 cells also decreased in an SB dose dependent manner. Patients with PA with membranous ezrin expression had a poorer prognosis compared with those without (P = .02).. Membranous translocation of ezrin might play a role during malignant transformation of SPT, MCN, IPMT, and PA, which are either dependent on (IPMT and PA) or independent of (SPT and MCN) the EGF-EGFR pathway. Membranous ezrin expression represents a prognostic factor for PA.

    Topics: Adenocarcinoma, Mucinous; Adolescent; Adult; Aged; Biomarkers, Tumor; Carcinoma, Pancreatic Ductal; Cytoskeletal Proteins; Disease Progression; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Immunoenzyme Techniques; Male; Middle Aged; Multivariate Analysis; Pancreatic Neoplasms; Phosphoproteins; Prognosis; Proportional Hazards Models; Retrospective Studies; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Survival Rate

2005