Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Article in English | MEDLINE | ID: mdl-38935270

ABSTRACT

PURPOSE OF REVIEW: The burdens of pancreatic ductal adenocarcinoma (PDAC) and acute pancreatitis are increasing globally. We reviewed current literature on whether acute pancreatitis is a causal factor for PDAC and examined clinical manifestations of PDAC-associated acute pancreatitis. RECENT FINDINGS: Recent findings detail the timing of acute pancreatitis before and after PDAC occurrence, further solidifying the evidence for PDAC-associated acute pancreatitis and for acute pancreatitis as a causal risk factor for PDAC. The risk of PDAC remains elevated above the general population in patients with distant history of acute pancreatitis. PDAC risk also increases with recurrent acute pancreatitis episodes, independent of smoking and alcohol. Mechanisms linking acute pancreatitis to PDAC include inflammation and neutrophil infiltration, which can be attenuated by suppressing inflammation and/or epigenetic modulation, thus slowing the progression of acinar-to-ductal metaplasia. Clinical presentation and management of acute pancreatitis in the context of PDAC are discussed, including challenges acute pancreatitis poses in the diagnosis and treatment of PDAC, and novel interventions for PDAC-associated acute pancreatitis. SUMMARY: PDAC risk may be reduced with improved acute pancreatitis prevention and treatment, such as antiinflammatories or epigenetic modulators. Increased acute pancreatitis and PDAC burden warrant more research on better diagnosis and management of PDAC-associated acute pancreatitis.

2.
Int J Cancer ; 141(5): 905-915, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28542740

ABSTRACT

Noninvasive biomarkers for early pancreatic ductal adenocarcinoma (PDAC) diagnosis and disease risk stratification are greatly needed. We conducted a nested case-control study within the Prospective Investigation into Cancer and Nutrition (EPIC) cohort to evaluate prediagnostic microRNAs (miRs) as biomarkers of subsequent PDAC risk. A panel of eight miRs (miR-10a, -10b, -21-3p, -21-5p, -30c, -106b, -155 and -212) based on previous evidence from our group was evaluated in 225 microscopically confirmed PDAC cases and 225 controls matched on center, sex, fasting status and age/date/time of blood collection. MiR levels in prediagnostic plasma samples were determined by quantitative RT-PCR. Logistic regression was used to model levels and PDAC risk, adjusting for covariates and to estimate area under the receiver operating characteristic curves (AUC). Plasma miR-10b, -21-5p, -30c and -106b levels were significantly higher in cases diagnosed within 2 years of blood collection compared to matched controls (all p-values <0.04). Based on adjusted logistic regression models, levels for six miRs (miR-10a, -10b, -21-5p, -30c, -155 and -212) overall, and for four miRs (-10a, -10b, -21-5p and -30c) at shorter follow-up time between blood collection and diagnosis (≤5 yr, ≤2 yr), were statistically significantly associated with risk. A score based on the panel showed a linear dose-response trend with risk (p-value = 0.0006). For shorter follow-up (≤5 yr), AUC for the score was 0.73, and for individual miRs ranged from 0.73 (miR-212) to 0.79 (miR-21-5p).


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , MicroRNAs/blood , Pancreatic Neoplasms/genetics , Adult , Aged , Area Under Curve , Biomarkers, Tumor/blood , Carcinoma, Pancreatic Ductal/blood , Case-Control Studies , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/blood , Polymerase Chain Reaction , Prospective Studies , ROC Curve , Sensitivity and Specificity
3.
Am J Epidemiol ; 186(8): 944-951, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28541521

ABSTRACT

Early-detection tests for pancreatic ductal adenocarcinoma (PDAC) are needed. Since a hypothetical screening test would be applied during antecedent clinical encounters, we sought to define the variability in health-care utilization leading up to PDAC diagnosis. This was a retrospective cohort study that included patients diagnosed with PDAC in the Indianapolis, Indiana, area between 1999 and 2013 with at least 1 health-care encounter during the antecedent 36-month period (n = 1,023). Patients were classified by unique patterns of health-care utilization using a group-based trajectory model. The prevalences of PDAC signals, such as diabetes mellitus (DM) and chronic pancreatitis, were compared. Four distinct trajectories were identified, the most common (42.0%) being having few clinical encounters more than 6 months prior to PDAC diagnosis (late acceleration). In all cases, a minority of persons had DM (30.6%, with 9.5% <1.5 years before PDAC) or any pancreatic disorder (39.9%); these were least common in the late-acceleration group (DM, 14.7%; any pancreatic disorder, 32.1% (P < 0.001)). The most common pattern of antecedent care was having few clinical encounters until shortly before PDAC diagnosis. Since the majority of patients diagnosed with PDAC do not have an antecedent PDAC signal, early-detection strategies limited to these groups may not apply to the majority of cases.


Subject(s)
Carcinoma, Pancreatic Ductal , Early Detection of Cancer/methods , Pancreatic Neoplasms , Patient Acceptance of Health Care/statistics & numerical data , Aged , Carcinoma, Pancreatic Ductal/complications , Diabetes Mellitus/epidemiology , Diabetes Mellitus/etiology , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/complications , Pancreatitis, Chronic/epidemiology , Pancreatitis, Chronic/etiology , Retrospective Studies , Pancreatic Neoplasms
4.
Biomacromolecules ; 17(12): 3872-3882, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27936722

ABSTRACT

Photopolymerized biomimetic hydrogels with adaptable properties have been widely used for cell and tissue engineering applications. As a widely adopted gel cross-linking method, photopolymerization provides experimenters on-demand and spatial-temporal controls in gelation kinetics. Long wavelength ultraviolet (UV) light initiated photopolymerization is among the most popular methods in the fabrication of cell-laden hydrogels owing to its rapid and relatively mild gelation conditions. The use of UV light, however, still causes concerns regarding its potential negative impacts on cells. Alternatively, visible light based photopolymerization can be used to cross-link cell-laden hydrogels. The majority of visible light based gelation schemes involve photoinitiator, co-initiator, and comonomer. This multicomponent initiation system creates added challenges for optimizing hydrogel formulations. Here, we report a co-initiator/comonomer-free visible light initiated thiol-norbornene photopolymerization scheme to prepare modular biomimetic hydrogels suitable for in situ cell encapsulation. Eosin-Y was used as the sole initiator to initiate modular gelation between synthetic macromers (e.g., thiolated poly(vinyl alcohol) or poly(ethylene glycol)) and functionalized extracellular matrices (ECMs) including norbornene-functionalized gelatin (GelNB) or thiolated hyaluronic acid (THA). These components are modularly cross-linked to afford bioinert (i.e., purely synthetic), bioactive (i.e., using gelatin), and biomimetic (i.e., using gelatin and hyaluronic acid) hydrogels. The stiffness of the hydrogels can be easily tuned without affecting the contents of the bioactive components. Furthermore, the use of naturally derived biomacromolecules (e.g., gelatin and HA) renders these hydrogels susceptible to enzyme-mediated degradation. In addition to demonstrating efficient and tunable visible light mediated gelation, we also utilized this biomimetic modular gelation system to formulate artificial tumor niche and to study the effects of cell density and gel modulus on the formation of pancreatic ductal adenocarcinoma (PDAC) spheroids.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cross-Linking Reagents/chemistry , Hydrogels/chemistry , Norbornanes/chemistry , Pancreatic Neoplasms/pathology , Spheroids, Cellular/pathology , Sulfhydryl Compounds/chemistry , Cell Count , Cell Survival , Humans , Light , Polymerization , Tissue Engineering , Tumor Cells, Cultured
5.
Am J Pathol ; 184(3): 674-85, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412093

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is associated with a high incidence of hepatic metastases, as well as occasional pulmonary metastases. To delineate the potential role of cancer stem cells (CSCs) in PDAC metastasis, human PDAC cells were injected into the spleen of mice. The characteristics and expression of markers associated with CSC and epithelial-mesenchymal transition (EMT) of metastatic cells that developed in the liver and lung were then compared with parental cells. The metastatic cells were polygonal, and larger than parental cells. Metastatic cells also exhibited decreased proliferation and increased adhesion to extracellular matrices, as well as enhanced migration and invasion in vitro and increased metastatic capacity in vivo. The CSC markers ALDH1A1, ABCG2, and nestin were expressed at high levels in metastatic cells and exhibited changes consistent with EMT (eg, decreased E-cadherin expression). Moreover, metastatic cells readily formed spheres in culture and exhibited an increased side population by flow analysis. Nestin and ABCG2 were also expressed at high levels in metastatic lesions from PDAC patients, and silencing nestin with shRNA in PDAC cells derived from lung metastases resulted in a marked decrease in the capacity of the cells to form spheres and to yield pulmonary or hepatic metastases. Thus, the metastatic potential of human PDAC cells correlates with CSCs and with EMT characteristics and is dependent on nestin expression.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/pathology , Neoplastic Stem Cells/pathology , Nestin/genetics , Pancreatic Neoplasms/pathology , Animals , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Nestin/metabolism , RNA, Small Interfering/metabolism
6.
Nano Lett ; 14(12): 6955-63, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25379951

ABSTRACT

MicroRNAs (miRs) are small noncoding RNAs that regulate mRNA stability and/or translation. Because of their release into the circulation and their remarkable stability, miR levels in plasma and other biological fluids can serve as diagnostic and prognostic disease biomarkers. However, quantifying miRs in the circulation is challenging due to issues with sensitivity and specificity. This Letter describes for the first time the design and characterization of a regenerative, solid-state localized surface plasmon resonance (LSPR) sensor based on highly sensitive nanostructures (gold nanoprisms) that obviates the need for labels or amplification of the miRs. Our direct hybridization approach has enabled the detection of subfemtomolar concentration of miR-X (X = 21 and 10b) in human plasma in pancreatic cancer patients. Our LSPR-based measurements showed that the miR levels measured directly in patient plasma were at least 2-fold higher than following RNA extraction and quantification by reverse transcriptase-polymerase chain reaction. Through LSPR-based measurements we have shown nearly 4-fold higher concentrations of miR-10b than miR-21 in plasma of pancreatic cancer patients. We propose that our highly sensitive and selective detection approach for assaying miRs in plasma can be applied to many cancer types and disease states and should allow a rational approach for testing the utility of miRs as markers for early disease diagnosis and prognosis, which could allow for the design of effective individualized therapeutic approaches.


Subject(s)
Biomarkers, Tumor/blood , Biosensing Techniques/instrumentation , MicroRNAs/blood , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/metabolism , Surface Plasmon Resonance/instrumentation , Equipment Design , Equipment Failure Analysis , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Microchemistry/instrumentation , Pancreatic Neoplasms/genetics , Reproducibility of Results , Sensitivity and Specificity
7.
Am J Gastroenterol ; 109(12): 1942-52, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25350767

ABSTRACT

OBJECTIVES: Accurate peripheral markers for the diagnosis of pancreatic ductal adenocarcinoma (PDAC) are lacking. We measured the differential expression of select microRNAs (miRNAs) in plasma and bile among patients with PDAC, chronic pancreatitis (CP), and controls. METHODS: We identified patients (n=215) with treatment-naive PDAC (n=77), CP with bile/pancreatic duct pathology (n=67), and controls (n=71) who had been prospectively enrolled in a Pancreatobiliary Biorepository at the time of endoscopic retrograde cholangiopancreatography or endoscopic ultrasound. Controls were patients with choledocholithiasis but normal pancreata. The sample was separated into training (n=95) and validation (n=120) cohorts to establish and then test the performance of PDAC Signature Panels in diagnosing PDAC. The training cohort (n=95) included age-matched patients with PDAC, CP, and controls. Panels were derived from the differential expression of 10 candidate miRNAs in plasma or bile. We selected miRNAs having excellent accuracy for inclusion in regression models. RESULTS: Using the training cohort, we confirmed the differential expression of 9/10 miRNAs in plasma (miR-10b, -30c, -106b, -132, -155, -181a, -181b, -196a, and -212) and 7/10 in bile (excluding miR-21, -132, and -181b). Of these, five (miR-10b, -155, -106b, -30c, and -212) had excellent accuracy for distinguishing PDAC. In the training and validation cohorts, the sensitivity/specificity for a PDAC Panel derived from plasma was 95/100% and 100/100%, respectively; in bile, these were 96/100% and 100/100%. CONCLUSIONS: Increased expression of miRNA-10b, -155, and -106b in plasma appears highly accurate in diagnosing PDAC. Additional studies are needed to confirm this Panel and explore its value as a prognostic test.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Aged , Bile/chemistry , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/metabolism , Case-Control Studies , Female , Humans , Male , MicroRNAs/blood , MicroRNAs/metabolism , Middle Aged , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/metabolism , Pilot Projects , Sensitivity and Specificity
8.
J Cell Physiol ; 228(3): 556-562, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22806240

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most frequently diagnosed cancers and the fourth leading cause of cancer-related death in the United States, suggesting that there is an urgent need to design novel strategies for achieving better treatment outcome of patients diagnosed with PDAC. Our previous study has shown that activation of Notch and NF-κB play a critical role in the development of PDAC in the compound K-Ras(G12D) and Ink4a/Arf deficient transgenic mice. However, the exact molecular mechanism by which mutated K-Ras and Ink4a/Arf deficiency contribute to progression of PDAC remains largely elusive. In the present study, we used multiple methods, such as real-time RT-PCR, Western blotting assay, and immunohistochemistry to gain further mechanistic insight. We found that the deletion of Ink4a/Arf in K-Ras(G12D) expressing mice led to high expression of PDGF-D signaling pathway in the tumor and tumor-derived cell line (RInk-1 cells). Furthermore, PDGF-D knock-down in RInk-1 cells resulted in the inhibition of pancreatosphere formation and down-regulation of EZH2, CD44, EpCAM, and vimentin. Moreover, we demonstrated that epithelial-mesenchymal transition (EMT) was induced in the compound mice, which is linked with aggressiveness of PDAC. In addition, we demonstrated that tumors from compound transgenic mice have higher expression of cancer stem cell (CSC) markers. These results suggest that the acquisition of EMT phenotype and induction of CSC characteristics could be linked with the aggressiveness of PDAC mediated in part through the activation of PDGF-D, signaling.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/deficiency , Pancreatic Neoplasms/etiology , Proto-Oncogene Proteins p21(ras)/deficiency , Animals , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , Epithelial-Mesenchymal Transition , Genes, ras , Humans , Lymphokines/deficiency , Lymphokines/genetics , Mice , Mice, Transgenic , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenotype , Platelet-Derived Growth Factor/deficiency , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction
9.
Am J Pathol ; 180(5): 1928-41, 2012 May.
Article in English | MEDLINE | ID: mdl-22440254

ABSTRACT

In pancreatic ductal adenocarcinoma (PDAC), the fibroblast growth factor receptor 1 (FGFR-1) IIIb isoform correlates with the inhibition of cancer cell proliferation, migration, and invasion, whereas FGFR-1 IIIc enhances cancer cell proliferation. The FGFR-2 IIIb isoform is expressed in PDAC, and its expression correlates with increased venous invasion. We examined the role of FGFR-2 IIIc in PDAC. FGFR-2 IIIc was expressed in all six pancreatic cancer cell lines examined and was highest in PANC-1 cells. FGFR-2 IIIc was abundant in the cancer cells from 83 of 117 PDAC cases, which correlated with decreased duration to development of liver metastasis after surgery. FGFR-2 IIIc-transfected cells exhibited increased proliferation in vitro and formed larger subcutaneous and orthotopic tumors, the latter producing more liver metastases. Moreover, FGF-2 exerted a more rapid stimulatory effect on the levels of phosphorylated extracellular signal-regulated kinase (p-ERK) in FGFR-2 IIIc stably transfected PANC-1 cells, compared with control cells. FGFR-2 IIIc-transfected cells also formed more spheres and contained more side population cells. Suppression of FGFR-2 IIIc expression inhibited the proliferation of PANC-1 cells, whereas an anti-FGFR-2 IIIc antibody inhibited the proliferation and migration of PANC-1 cells. Thus, high FGFR-2 IIIc levels in PDAC contribute to disease aggressiveness and confer to pancreatic cancer cells features suggestive of cancer stem cells, indicating that FGFR-2 IIIc may be a novel and important therapeutic target in PDAC.


Subject(s)
Adenocarcinoma/metabolism , Pancreatic Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/secondary , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Real-Time Polymerase Chain Reaction/methods , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/physiology , Transfection , Transplantation, Heterologous
10.
Clin Cancer Res ; 29(13): 2348-2350, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37099035

ABSTRACT

Several approaches for overcoming immunotherapy resistance in pancreatic and colorectal cancer syngeneic models were assessed using heparin and immunotherapy. Beneficial responses were attributed to heparin-induced vascular normalization, ensuing CD8+ T-cell infiltration, and M1 macrophage polarization, suggesting the potential for heparin-anchored therapies in cold tumors such as pancreatic cancer. See related article by Wei et al., p. 2525.


Subject(s)
Heparin , Pancreatic Neoplasms , Humans , Heparin/therapeutic use , Anticoagulants , Microcirculation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Immunotherapy , CD8-Positive T-Lymphocytes/immunology
11.
J Cell Physiol ; 227(10): 3373-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22213426

ABSTRACT

Human pancreatic cancer (PC) is an aggressive disease, which has been recapitulated in transgenic animal model that provides unique opportunity for mechanistic understanding of disease progression and also for testing the efficacy of novel therapeutics. Emerging evidence suggests deregulated expression of microRNAs (miRNAs) in human PC, and thus we investigated the expression of miRNAs in pancreas tissues obtained from transgenic mouse models of K-Ras (K), Pdx1-Cre (C), K-Ras;Pdx1-Cre (KC), and K-Ras;Pdx1-Cre;INK4a/Arf (KCI), initially from pooled RNA samples using miRNA profiling, and further confirmed in individual specimens by quantitative RT-PCR. We found over-expression of miR-21, miR-221, miR-27a, miR-27b, and miR-155, and down-regulation of miR-216a, miR-216b, miR-217, and miR-146a expression in tumors derived from KC and KCI mouse model, which was consistent with data from KCI-derived RInk-1 cells. Mechanistic investigations revealed a significant induction of EGFR, K-Ras, and MT1-MMP protein expression in tissues from both KC and KCI mouse compared to tissues from K or C, and these results were consistent with similar findings in RInk-1 cells compared to human MIAPaCa-2 cells. Furthermore, miR-155 knock-down in RInk-1 cells resulted in the inhibition of cell growth and colony formation consistent with down-regulation of EGFR, MT1-MMP, and K-Ras expression. In addition, miR-216b which target Ras, and forced re-expression of miR-216b in RInk-1 cells showed inhibition of cell proliferation and colony formation, which was correlated with reduced expression of Ras, EGFR, and MT1-MMP. These findings suggest that these models would be useful for preclinical evaluation of novel miRNA-targeted agents for designing personalized therapy for PC.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation/genetics , Genes, ras , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Animals , Cell Line , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , MicroRNAs/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Stem Cells/metabolism
12.
Gastroenterology ; 141(3): 1091-101, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21699781

ABSTRACT

BACKGROUND & AIMS: Rb1 encodes a cell-cycle regulator that is functionally disrupted in most human cancers. Pancreatic ductal adenocarcinomas (PDACs) have a high frequency of mutations in KRAS and INK4A/CDKN2A that might allow cells to bypass the regulatory actions of retinoblastoma (RB). To determine the role of loss of RB function in PDAC progression, we investigated the effects of Rb disruption during pancreatic malignant transformation initiated by oncogenic Kras. METHODS: We generated mice with pancreas-specific disruption of Rb, in the absence or presence of oncogenic Kras, to examine the role of RB in pancreatic carcinogenesis. RESULTS: In the presence of oncogenic Kras, loss of Rb from the pancreatic epithelium accelerated formation of pancreatic intraepithelial neoplasia (PanIN), increased the frequency of cystic neoplasms, and promoted rapid progression toward PDAC. Early stage cancers were characterized by acute pancreatic inflammation, associated with up-regulation of proinflammatory cytokines within the pancreas. Despite the presence of markers associated with oncogene-induced senescence, low-grade PanIN were highly proliferative and expressed high levels of p53. Pancreatic cancer cell lines derived from these mice expressed high levels of cytokines, and transcriptional activity of p53 was impaired. CONCLUSIONS: Rb encodes a tumor suppressor that attenuates progression of oncogenic Kras-induced carcinogenesis in the pancreas by mediating the senescence response and promoting activity of the tumor suppressor p53.


Subject(s)
Carcinoma in Situ/physiopathology , Cellular Senescence/physiology , Gene Deletion , Pancreatic Neoplasms/physiopathology , Precancerous Conditions/physiopathology , Proto-Oncogene Proteins p21(ras)/physiology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Animals , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/physiopathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cytokines/physiology , Disease Models, Animal , Disease Progression , Mice , Mice, Mutant Strains , Pancreatic Neoplasms/pathology , Precancerous Conditions/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/physiology
13.
Mol Carcinog ; 51(10): 771-82, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22965423

ABSTRACT

Pancreatic cancer is the fourth leading cause of cancer death in the United States because most patients are diagnosed too late in the course of the disease to be treated effectively. Thus, there is a pressing need to more clearly understand how gene expression is regulated in cancer cells and to identify new biomarkers and therapeutic targets. Translational regulation is thought to occur primarily through non-SMAD directed signaling pathways. We tested the hypothesis that SMAD4-dependent signaling does play a role in the regulation of mRNA entry into polysomes and that novel candidate genes in pancreatic cancer could be identified using polysome RNA from the human pancreatic cancer cell line BxPC3 with or without a functional SMAD4 gene. We found that (i) differentially expressed whole cell and cytoplasm RNA levels are both poor predictors of polysome RNA levels; (ii) for a majority of RNAs, differential RNA levels are regulated independently in the nucleus, cytoplasm, and polysomes; (iii) for most of the remaining polysome RNA, levels are regulated via a "tagging" of the RNAs in the nucleus for rapid entry into the polysomes; (iv) a SMAD4-dependent pathway appears to indeed play a role in regulating mRNA entry into polysomes; and (v) a gene list derived from differentially expressed polysome RNA in BxPC3 cells generated new candidate genes and cell pathways potentially related to pancreatic cancer.


Subject(s)
Pancreatic Neoplasms/metabolism , Polyribosomes/metabolism , RNA/metabolism , Smad4 Protein/metabolism , Cell Nucleus/genetics , Cytoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms/genetics , Polyribosomes/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
14.
Pharmacol Ther ; 236: 108111, 2022 08.
Article in English | MEDLINE | ID: mdl-35016920

ABSTRACT

Immune checkpoint inhibitors (ICIs) have transformed the treatment paradigm for several malignancies. While the use of single-agent or combined ICIs has achieved acceptable disease control rates in a variety of solid tumors, such approaches have yet to show substantial therapeutic efficacy in select difficult-to-treat cancer types. Recently, select chemotherapy regimens are emerging as extensive modifiers of the tumor microenvironment, leading to the reprogramming of local immune responses. Accordingly, data is now emerging to suggest that certain anti-neoplastic agents modulate various immune cell processes, most notably the cross-presentation of tumor antigens, leukocyte trafficking, and cytokine biosynthesis. As such, the combination of ICIs and cytotoxic chemotherapy are beginning to show promise in many cancers that have long been considered poorly responsive to ICI-based immunotherapy. Here, we discuss past and present attempts to advance chemo-immunotherapy in these difficult-to-treat cancer histologies, mechanisms through which select chemotherapies modify tumor immunogenicity, as well as important considerations when designing such approaches to maximize efficacy and improve therapeutic response rates.


Subject(s)
Antineoplastic Agents , Neoplasms , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Immunologic Factors/pharmacology , Immunotherapy , Neoplasms/drug therapy , Tumor Microenvironment
15.
Pancreas ; 51(6): 568-574, 2022 07 01.
Article in English | MEDLINE | ID: mdl-36206460

ABSTRACT

ABSTRACT: Acute pancreatitis (AP) is a disease characterized by an acute inflammatory phase followed by a convalescent phase. Diabetes mellitus (DM) was historically felt to be a transient phenomenon related to acute inflammation; however, it is increasingly recognized as an important late and chronic complication. There are several challenges that have prevented precisely determining the incidence rate of DM after AP and understanding the underlying mechanisms. The DREAM (Diabetes RElated to Acute Pancreatitis and its Mechanisms) Study is a prospective cohort study designed to address these and other knowledge gaps to provide the evidence needed to screen for, prevent, and treat DM after AP. In the following article, we summarize literature regarding the epidemiology of DM after AP and provide the rationale and an overview of the DREAM study.


Subject(s)
Diabetes Mellitus, Type 1 , Pancreatitis , Acute Disease , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/epidemiology , Humans , Incidence , Pancreatitis/complications , Pancreatitis/epidemiology , Prospective Studies
16.
Crit Rev Eukaryot Gene Expr ; 21(2): 115-29, 2011.
Article in English | MEDLINE | ID: mdl-22077151

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy characterized by a plethora of molecular alterations that include major and minor driving mutations, the presence of intense desmoplasia exhibiting numerous proliferating pancreatic stellate cells (PSC) and cancer-associated fibroblasts that produce fibronectin and collagens, and foci of inflammatory cells that produce mitogenic cytokines. This review will focus on signaling by tyrosine kinase receptors, and the role of transforming growth factor beta in this malignancy is described briefly. Potential for therapeutic interventions will be discussed in relation to specific pathways.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Signal Transduction , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
17.
J Clin Invest ; 118(1): 89-99, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18064304

ABSTRACT

Cells isolated from many types of human cancers express heparin-binding growth factors (HBGFs) that drive tumor growth, metastasis, and angiogenesis. The heparan sulfate proteoglycan glypican-1 (GPC1) is a coreceptor for HBGFs. Here we show that both cancer cell-derived and host-derived GPC1 are crucial for efficient growth, metastasis, and angiogenesis of human and mouse cancer cells. Thus downregulation of GPC1 in the human pancreatic cancer cell line PANC-1, using antisense approaches, resulted in prolonged doubling times and decreased anchorage-independent growth in vitro as well as attenuated tumor growth, angiogenesis, and metastasis when these cells were transplanted into athymic mice. Moreover, athymic mice that lacked GPC1 exhibited decreased tumor angiogenesis and metastasis following intrapancreatic implantation with either PANC-1 or T3M4 human pancreatic cancer cells and fewer pulmonary metastases following intravenous injection of murine B16-F10 melanoma cells. In addition, hepatic endothelial cells isolated from these mice exhibited an attenuated mitogenic response to VEGF-A. These data indicate that cancer cell- and host-derived GPC1 are crucial for full mitogenic, angiogenic, and metastatic potential of cancer cells. Thus targeting GPC1 might provide new avenues for cancer therapy and for the prevention of cancer metastasis.


Subject(s)
Glypicans/metabolism , Lung Neoplasms/metabolism , Melanoma/metabolism , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/metabolism , Animals , COS Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Cytokines/genetics , Cytokines/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Glypicans/genetics , Lung Neoplasms/genetics , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma/genetics , Melanoma/pathology , Melanoma/prevention & control , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/prevention & control , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
18.
RNA ; 15(10): 1917-28, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19703940

ABSTRACT

With no known exceptions, every published microarray study to determine differential mRNA levels in eukaryotes used RNA extracted from whole cells. It is assumed that the use of whole cell RNA in microarray gene expression analysis provides a legitimate profile of steady-state mRNA. Standard labeling methods and the prevailing dogma that mRNA resides almost exclusively in the cytoplasm has led to the long-standing belief that the nuclear RNA contribution is negligible. We report that unadulterated cytoplasmic RNA uncovers differentially expressed mRNAs that otherwise would not have been detected when using whole cell RNA and that the inclusion of nuclear RNA has a large impact on whole cell gene expression microarray results by distorting the mRNA profile to the extent that a substantial number of false positives are generated. We conclude that to produce a valid profile of the steady-state mRNA population, the nuclear component must be excluded, and to arrive at a more realistic view of a cell's gene expression profile, the nuclear and cytoplasmic RNA fractions should be analyzed separately.


Subject(s)
Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Animals , Blotting, Western , Cell Line , Gene Expression Profiling , Humans , Mice , Polymerase Chain Reaction
19.
Cancer Lett ; 504: 1-14, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33549709

ABSTRACT

Immunotherapy has revolutionized cancer treatment in the last decade, and strategies to re-activate cytotoxic immunity are now standard of care in several malignancies. Despite rapid advances in immunotherapy for most solid cancers, progress in immunotherapy against pancreatic ductal adenocarcinoma (PDAC) has been exceptionally difficult. This is true for several approaches, most notably immune checkpoint inhibitors (ICIs) and GM-CSF cell-based vaccines (GVAX). Though many immunotherapies have been explored in clinical trials, few have shown significant therapeutic efficacy. Further, many have shown high rates of serious adverse effects and dose-limiting toxicities, and to date, immunotherapy regimens have not been successfully implemented in PDAC. Here, we provide a comprehensive summary of the key clinical trials exploring immunotherapy in PDAC, followed by a brief discussion of emerging molecular mechanisms that may explain the relative failure of immunotherapy in pancreas cancer thus far.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Immunotherapy , Pancreatic Neoplasms/therapy , Carcinoma, Pancreatic Ductal/radiotherapy , Combined Modality Therapy , Humans , Pancreatic Neoplasms/radiotherapy
20.
Front Oncol ; 11: 688377, 2021.
Article in English | MEDLINE | ID: mdl-34336673

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, with a median survival time of 10-12 months. Clinically, these poor outcomes are attributed to several factors, including late stage at the time of diagnosis impeding resectability, as well as multi-drug resistance. Despite the high prevalence of drug-resistant phenotypes, nearly all patients are offered chemotherapy leading to modest improvements in postoperative survival. However, chemotherapy is all too often associated with toxicity, and many patients elect for palliative care. In cases of inoperable disease, cytotoxic therapies are less efficacious but still carry the same risk of serious adverse effects, and clinical outcomes remain particularly poor. Here we discuss the current state of pancreatic cancer therapy, both surgical and medical, and emerging factors limiting the efficacy of both. Combined, this review highlights an unmet clinical need to improve our understanding of the mechanisms underlying the poor therapeutic responses seen in patients with PDAC, in hopes of increasing drug efficacy, extending patient survival, and improving quality of life.

SELECTION OF CITATIONS
SEARCH DETAIL