Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nat Commun ; 12(1): 3044, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031415

ABSTRACT

Unlike other malignancies, therapeutic options in pancreatic ductal adenocarcinoma (PDAC) are largely limited to cytotoxic chemotherapy without the benefit of molecular markers predicting response. Here we report tumor-cell-intrinsic chromatin accessibility patterns of treatment-naïve surgically resected PDAC tumors that were subsequently treated with (Gem)/Abraxane adjuvant chemotherapy. By ATAC-seq analyses of EpCAM+ PDAC malignant epithelial cells sorted from 54 freshly resected human tumors, we show here the discovery of a signature of 1092 chromatin loci displaying differential accessibility between patients with disease free survival (DFS) < 1 year and patients with DFS > 1 year. Analyzing transcription factor (TF) binding motifs within these loci, we identify two TFs (ZKSCAN1 and HNF1b) displaying differential nuclear localization between patients with short vs. long DFS. We further develop a chromatin accessibility microarray methodology termed "ATAC-array", an easy-to-use platform obviating the time and cost of next generation sequencing. Applying this methodology to the original ATAC-seq libraries as well as independent libraries generated from patient-derived organoids, we validate ATAC-array technology in both the original ATAC-seq cohort as well as in an independent validation cohort. We conclude that PDAC prognosis can be predicted by ATAC-array, which represents a low-cost, clinically feasible technology for assessing chromatin accessibility profiles.


Subject(s)
Chromatin Immunoprecipitation Sequencing/methods , Chromatin , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Biomarkers, Tumor , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Nucleus , Hepatocyte Nuclear Factor 1-beta/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Kruppel-Like Transcription Factors/genetics , Pancreatic Neoplasms/metabolism , Prognosis , Transcription Factors , Transcriptome , Pancreatic Neoplasms
2.
Oncogene ; 35(32): 4282-8, 2016 08 11.
Article in English | MEDLINE | ID: mdl-26592447

ABSTRACT

Pancreatic cancer is one of the most lethal malignancies, with virtually all patients eventually succumbing to their disease. Mutations in p53 have been documented in >50% of pancreatic cancers. Owing to the high incidence of p53 mutations in PanIN 3 lesions and pancreatic tumors, we interrogated the comparative ability of adult pancreatic acinar and ductal cells to respond to oncogenic Kras and mutant Tp53(R172H) using Hnf1b:CreER(T2) and Mist1:CreER(T2) mice. These studies involved co-activation of a membrane-tethered GFP lineage label, allowing for direct visualization and isolation of cells undergoing Kras and mutant p53 activation. Kras activation in Mist1(+) adult acinar cells resulted in brisk PanIN formation, whereas no evidence of pancreatic neoplasia was observed for up to 6 months following Kras activation in Hnf1beta(+) adult ductal cells. In contrast to the lack of response to oncogenic Kras alone, simultaneous activation of Kras and mutant p53 in adult ductal epithelium generated invasive PDAC in 75% of mice as early as 2.5 months after tamoxifen administration. These data demonstrate that pancreatic ductal cells, whereas exhibiting relative resistance to oncogenic Kras alone, can serve as an effective cell of origin for pancreatic ductal adenocarcinoma in the setting of gain-of-function mutations in p53.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis , Cell Line, Tumor , Cellular Senescence , Humans , Mice , Phosphoproteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
3.
Oncogene ; 34(50): 6029-39, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26364615

ABSTRACT

The crucial role of the immune system in the formation and progression of tumors has been widely accepted. On one hand, the surveillance role of the immune system plays an important role in endogenous tumor prevention, but on the other hand, in some special circumstances such as in chronic inflammation, the immune system can actually contribute to the formation and progression of tumors. In recent years, there has been an explosion of novel targeted immunotherapies for advanced cancers. In the present manuscript, we explore known and potential various types of cancer prevention strategies and focus on nonvaccine-based cancer preventive strategies targeting the immune system at the early stages of tumorigenesis.


Subject(s)
Neoplasms/immunology , Neoplasms/prevention & control , Chemoprevention , Dendritic Cells/immunology , Humans , Immunotherapy , Interleukin 1 Receptor Antagonist Protein/physiology , Interleukin-6/antagonists & inhibitors , Interleukin-8/antagonists & inhibitors , Monitoring, Immunologic , Programmed Cell Death 1 Receptor/physiology , STAT3 Transcription Factor/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
4.
Oncogene ; 34(21): 2801-6, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25065594

ABSTRACT

Somatic activation of the KRAS proto-oncogene is evident in almost all pancreatic cancers, and appears to represent an initiating event. These mutations occur primarily at codon 12 and less frequently at codons 13 and 61. Although some studies have suggested that different KRAS mutations may have variable oncogenic properties, to date there has been no comprehensive functional comparison of multiple KRAS mutations in an in vivo vertebrate tumorigenesis system. We generated a Gal4/UAS-based zebrafish model of pancreatic tumorigenesis in which the pancreatic expression of UAS-regulated oncogenes is driven by a ptf1a:Gal4-VP16 driver line. This system allowed us to rapidly compare the ability of 12 different KRAS mutations (G12A, G12C, G12D, G12F, G12R, G12S, G12V, G13C, G13D, Q61L, Q61R and A146T) to drive pancreatic tumorigenesis in vivo. Among fish injected with one of five KRAS mutations reported in other tumor types but not in human pancreatic cancer, 2/79 (2.5%) developed pancreatic tumors, with both tumors arising in fish injected with A146T. In contrast, among fish injected with one of seven KRAS mutations known to occur in human pancreatic cancer, 22/106 (20.8%) developed pancreatic cancer. All eight tumorigenic KRAS mutations were associated with downstream MAPK/ERK pathway activation in preneoplastic pancreatic epithelium, whereas nontumorigenic mutations were not. These results suggest that the spectrum of KRAS mutations observed in human pancreatic cancer reflects selection based on variable tumorigenic capacities, including the ability to activate MAPK/ERK signaling.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mutation/genetics , Pancreas/pathology , Proto-Oncogene Proteins/genetics , Vertebrates/genetics , ras Proteins/genetics , Animals , Cell Transformation, Neoplastic/pathology , Epithelium/pathology , Gene Expression Regulation, Neoplastic/genetics , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins p21(ras) , Zebrafish/genetics
5.
AJNR Am J Neuroradiol ; 29(7): 1317-23, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18417603

ABSTRACT

BACKGROUND AND PURPOSE: The occurrence of brain parenchymal signal-intensity changes within the drainage territory of developmental venous anomalies (DVAs) in the absence of cavernous malformations (CMs) has been incompletely assessed. This study was performed to evaluate the prevalence of brain parenchymal signal-intensity abnormalities subjacent to DVA, correlating with DVA morphology and location. MATERIALS AND METHODS: One hundred sixty-four patients with brain MR imaging with contrast studies performed from July 2005 through June 2006 formed the study group. The examinations were reviewed and data were collected regarding the following: location, depth, size of draining vein, associated increased signal intensity on fluid-attenuated inversion recovery and T2-weighted images, associated CMs, and associated signal intensity on gradient recalled-echo sequences. RESULTS: Of the 175 DVAs identified, 28 had associated signal-intensity abnormalities in the drainage territory. Seven of 28 DVAs with signal-intensity abnormalities were excluded because of significant adjacent white matter signal-intensity changes related to other pathology overlapping the drainage territory. Of the remaining DVAs imaged in this study, 21/168 (12.5%) had subjacent signal-intensity abnormalities. An adjusted prevalence rate of 9/115 (7.8%) was obtained by excluding patients with white matter disease more than minimal in degree. Periventricular location and older age were associated with DVA signal-intensity abnormality. CONCLUSION: Signal-intensity abnormalities detectable by standard clinical MR images were identified in association with 12.5% of consecutively identified DVAs. Excluding patients with significant underlying white matter disease, we adjusted the prevalence to 7.8%. The etiology of the signal-intensity changes is unclear but may be related to edema, gliosis, or leukoaraiosis secondary to altered hemodynamics in the drainage area.


Subject(s)
Brain Edema/diagnosis , Brain Neoplasms/diagnosis , Central Nervous System Venous Angioma/diagnosis , Cerebral Veins/abnormalities , Diffusion Magnetic Resonance Imaging , Gliosis/diagnosis , Image Processing, Computer-Assisted , Leukoaraiosis/diagnosis , Magnetic Resonance Imaging , Adult , Aged , Brain/blood supply , Brain/pathology , Cerebral Veins/pathology , Female , Humans , Male , Middle Aged , Retrospective Studies
6.
Clin Cancer Res ; 7(12): 3862-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751476

ABSTRACT

PURPOSE: Effective new markers of pancreatic carcinoma are urgently needed. In a previous analysis of gene expression in pancreatic adenocarcinoma using serial analysis of gene expression (SAGE), we found that the tag for the mesothelin mRNA transcript was present in seven of eight SAGE libraries derived from pancreatic carcinomas but not in the two SAGE libraries derived from normal pancreatic duct epithelial cells. In this study, we evaluate the potential utility of mesothelin as a tumor marker for pancreatic adenocarcinoma. EXPERIMENTAL DESIGN: Mesothelin mRNA expression was evaluated in pancreatic adenocarcinomas using reverse-transcription PCR (RT-PCR) and in situ hybridization, whereas mesothelin protein expression was evaluated by immunohistochemistry. RESULTS: Using an online SAGE database (http://www.ncbi.nlm.gov/SAGE), we found the tag for mesothelin to be consistently present in the mesothelioma, ovarian cancer, and pancreatic cancer libraries but not in normal pancreas libraries. Mesothelin mRNA expression was confirmed by in situ hybridization in 4 of 4 resected primary pancreatic adenocarcinomas and by RT-PCR in 18 of 20 pancreatic cancer cell lines, whereas mesothelin protein expression was confirmed by immunohistochemistry in all 60 resected primary pancreatic adenocarcinomas studied. The adjacent normal pancreas in these 60 cases did not label, or at most only rare benign pancreatic ducts showed weak labeling for mesothelin. CONCLUSIONS: Mesothelin is a new marker for pancreatic adenocarcinoma identified by gene expression analysis. Mesothelin overexpression in pancreatic adenocarcinoma has potential diagnostic, imaging, and therapeutic implications.


Subject(s)
Adenocarcinoma/genetics , Antigens, Neoplasm/genetics , Biomarkers, Tumor/analysis , Gene Expression Regulation, Neoplastic , Membrane Glycoproteins/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Antigens, Neoplasm/analysis , GPI-Linked Proteins , Humans , Immunohistochemistry , In Situ Hybridization , Membrane Glycoproteins/analysis , Mesothelin , Online Systems , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Cells, Cultured
7.
Cancer J ; 7(4): 242-50, 2001.
Article in English | MEDLINE | ID: mdl-11561600

ABSTRACT

The specific cell of origin responsible for generating pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma remains unknown. During development, epithelial stem cells within embryonic pancreatic epithelium give riseto mature acinar, ductal, and islet elements. Emerging evidence suggests that cells with precursor potential also exist within adult pancreas, resulting in significant developmental plasticity among both endocrine and exocrine cell types. In this review, the contribution of developmental plasticity in initiating pancreatic metaplasia and neoplasia is considered, and evidence supporting a role for epithelial stem cells in pancreatic cancer is discussed.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/pathology , Neoplasms, Multiple Primary/pathology , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Humans , Metaplasia , Neoplasms, Multiple Primary/genetics , Pancreas/embryology , Pancreas/pathology , Pancreatic Neoplasms/embryology , Pancreatic Neoplasms/genetics , Stem Cells/pathology , Time Factors
8.
Cancer Res ; 61(11): 4320-4, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389052

ABSTRACT

Serial analysis of gene expression (SAGE) can be used to quantify gene expression in human tissues. Comparison of gene expression levels in neoplastic tissues with those seen in nonneoplastic tissues can, in turn, identify novel tumor markers. Such markers are urgently needed for highly lethal cancers like pancreatic adenocarcinoma, which typically presents at an incurable, advanced stage. The results of SAGE analyses of a large number of neoplastic and nonneoplastic tissues are now available online, facilitating the rapid identification of novel tumor markers. We searched an online SAGE database to identify genes preferentially expressed in pancreatic cancers as compared with normal tissues. SAGE libraries derived from pancreatic adenocarcinomas were compared with SAGE libraries derived from nonneoplastic tissues. Three promising tags were identified. Two of these tags corresponded to genes (lipocalin and trefoil factor 2) previously shown to be overexpressed in pancreatic carcinoma, whereas the third tag corresponded to prostate stem cell antigen (PSCA), a recently discovered gene thought to be largely restricted to prostatic basal cells and prostatic adenocarcinomas. PSCA was expressed in four of the six pancreatic cancer SAGE libraries, but not in the libraries derived from normal pancreatic ductal cells. We confirmed the overexpression of the PSCA mRNA transcript in 14 of 19 pancreatic cancer cell lines by reverse transcription-PCR, and using immunohistochemistry, we demonstrated PSCA protein overexpression in 36 of 60 (60%) primary pancreatic adenocarcinomas. In 59 of 60 cases, the adjacent nonneoplastic pancreas did not label for PSCA. PSCA is a novel tumor marker for pancreatic carcinoma that has potential diagnostic and therapeutic implications. These results establish the validity of analyses of SAGE databases to identify novel tumor markers.


Subject(s)
Biomarkers, Tumor/biosynthesis , Membrane Glycoproteins/biosynthesis , Neoplasm Proteins/biosynthesis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Antigens, Neoplasm , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , GPI-Linked Proteins , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trefoil Factor-2 , Tumor Cells, Cultured
9.
Invest New Drugs ; 19(1): 21-7, 2001.
Article in English | MEDLINE | ID: mdl-11291830

ABSTRACT

Six patients with incurable malignancies were originally treated with vitamin E, 3200 IU/day for fourteen days, followed by the same dose of vitamin E daily plus LCV (20 mg/m2 i.v. bolus daily x 5) with 5FU (425 mg/m2 i.v. bolus immediately following LCV). The same schedule of LCV and 5FU was repeated 4 weeks later, then every 5 weeks indefinitely. When 3 of the first 6 had grade 3/4 toxicity, six more patients were treated on the identical drugs and schedule. Seven of twelve total patients had one or more grade 3/4 toxicities. Neutropenia, abdominal pain, and diarrhea were most common. No patient had a documented response, though seven patients did have stable disease. Though the combination of vitamin E and chemotherapy was toxic, this trial demonstrated maximal therapeutic doses of vitamin E can be combined with standard 5FU and LCV, without significantly increasing the side effects of the chemotherapy itself.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Vitamin E/therapeutic use , Abdominal Pain/chemically induced , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Diarrhea/chemically induced , Dinoprost/analogs & derivatives , Dinoprost/urine , Dose-Response Relationship, Drug , F2-Isoprostanes , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Humans , Infusions, Intravenous , Leucovorin/administration & dosage , Leucovorin/adverse effects , Leucovorin/therapeutic use , Male , Middle Aged , Neoplasms/metabolism , Neoplasms/pathology , Neutropenia/chemically induced , Vitamin E/administration & dosage , Vitamin E/adverse effects
10.
Pancreatology ; 1(6): 587-96, 2001.
Article in English | MEDLINE | ID: mdl-12120241

ABSTRACT

Exocrine pancreatic cell types comprise greater than 90% of parenchymal cell mass in the adult pancreas. However, the factors regulating differentiation of acinar and ductal epithelial cells remain incompletely characterized. Like pancreatic islet cells, acinar and ductal cells arise from pluripotent precursors within embryonic pancreatic epithelium. Recent studies have suggested that a common pool of pluripotent stem cells is responsible for generating both endocrine and exocrine cell types, and that specific signaling pathways regulate a critical balance between endocrine and exocrine lineage commitment.


Subject(s)
Pancreas/cytology , Pancreas/embryology , Animals , Cell Differentiation , Cell Lineage , Gene Expression Regulation, Developmental , Humans , Pancreatic Ducts/cytology , Pancreatic Ducts/embryology , Stem Cells/cytology , Stem Cells/physiology
11.
Am J Physiol Gastrointest Liver Physiol ; 279(4): G827-36, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11005771

ABSTRACT

The mechanisms linking acinar cell apoptosis and ductal epithelial proliferation remain unknown. To determine the relationship between these events, pancreatic duct ligation (PDL) was performed on p53(+/+) and p53(-/-) mice. In mice bearing a wild-type p53 allele, PDL resulted in upregulation of p53 protein in both acinar cells and proliferating duct-like epithelium. In contrast, upregulation of Bcl-2 occurred only in duct-like epithelium. Both p21(WAF1/CIP1) and Bax were also upregulated in duct-ligated lobes. After PDL in p53(+/+) mice, acinar cells underwent widespread apoptosis, while duct-like epithelium underwent proliferative expansion. In the absence of p53, upregulation of p53 target genes and acinar cell apoptosis did not occur. The absence of acinar cell apoptosis in p53(-/-) mice also eliminated the proliferative response to duct ligation. These data demonstrate that PDL-induced acinar cell apoptosis is a p53-dependent event and suggest a direct link between acinar cell apoptosis and proliferation of duct-like epithelium in duct-ligated pancreas.


Subject(s)
Apoptosis/physiology , Pancreas/cytology , Pancreatic Ducts/physiology , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/analysis , Epithelial Cells/cytology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Proto-Oncogene Proteins/analysis , Time Factors , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
12.
Gastrointest Endosc ; 52(3): 367-71, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10968852

ABSTRACT

BACKGROUND: Preoperative diagnosis of pancreatic adenocarcinoma can be difficult. Computed tomography (CT) is the standard, noninvasive imaging method for evaluation of suspected pancreatic adenocarcinoma, but it has limited sensitivity for diagnosis, local staging, and metastases. Endoscopic ultrasound (EUS) and fluoro-deoxyglucose/positron emission tomography (FDG-PET) are imaging methods that may improve diagnostic accuracy. METHODS: Thirty-five patients with presumed resectable pancreatic adenocarcinoma were prospectively evaluated with helical CT, EUS, and FDG-PET. RESULTS: Sensitivity for the detection of pancreatic cancer was higher for EUS (93%) and FDG-PET (87%) than for CT (53%). EUS was more sensitive than CT for local vascular invasion of the portal and superior mesenteric veins. EUS diagnosis of vascular invasion was associated with poor outcome after surgery. EUS-guided, fine-needle aspiration allowed tissue diagnosis in 14 of 21 attempts (67%). FDG-PET diagnosed 7 of 9 cases of proven metastatic disease, 4 of which were missed by CT. Two of three metastatic liver lesions suspected by CT were indeterminate for metastases. FDG-PET confirmed metastases. CONCLUSIONS: EUS and PET improve diagnostic capability in pancreatic adenocarcinoma. EUS is useful in determining local vascular invasion and obtaining tissue diagnosis. FDG-PET is useful in identifying metastatic disease. Both techniques are more sensitive than helical CT for identification of the primary tumor. (Gastrointest Endosc 2000;52:367-71).


Subject(s)
Adenocarcinoma/diagnosis , Endosonography , Pancreatic Neoplasms/diagnosis , Tomography, Emission-Computed , Tomography, X-Ray Computed , Biopsy, Needle/methods , Diagnosis, Differential , Fluorodeoxyglucose F18 , Humans , Neoplasm Invasiveness , Neoplasm Staging/methods , Prospective Studies , Sensitivity and Specificity
13.
Neoplasia ; 2(3): 261-72, 2000.
Article in English | MEDLINE | ID: mdl-10935512

ABSTRACT

Pancreatic ductal adenocarcinoma is a highly lethal malignancy that is resistant to traditional cytotoxic therapy. High rates of activating codon 12 K-Ras mutations in this disease have generated considerable interest in the therapeutic application of novel farnesyl transferase inhibitors (FTIs). However, a comprehensive analysis of the effects of FTI treatment on pancreatic cancer cells has not been performed. Treatment of five different human pancreatic cancer cell lines with FTI L-744,832 resulted in inhibition of anchorage-dependent growth, with wide variation in sensitivity among different lines. Effective growth inhibition by L-744,832 correlated with accumulation of cells with a tetraploid (4N) DNA content and high levels of cyclin B1/cdc2 kinase activity, implying cell cycle arrest downstream from the DNA damage-inducible G2/M cell cycle checkpoint. In addition, sensitive cell lines underwent apoptosis as evidenced by changes in nuclear morphology and internucleosomal DNA fragmentation. L-744,832 at a concentration of 1 microM additively enhanced the cytotoxic effect of ionizing radiation, apparently by overriding G2/M checkpoint activation. The effects of FTI treatment on cell growth and cell cycle regulation were associated with changes in posttranslational processing of H-Ras and N-Ras, but not K-Ras. The results confirm the potential therapeutic efficacy of FTI treatment in pancreatic cancer, and suggest that farnesylated proteins other than K-Ras may act as important regulators of G2/M cell cycle kinetics.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Ductal, Breast/pathology , Cyclin B/metabolism , Enzyme Inhibitors/pharmacology , Genes, ras/physiology , Methionine/analogs & derivatives , Pancreatic Neoplasms/pathology , Cyclin B1 , G2 Phase/drug effects , Humans , Methionine/pharmacology , Mitosis/drug effects
14.
Gastroenterology ; 117(6): 1416-26, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10579983

ABSTRACT

BACKGROUND & AIMS: The progenitor cells responsible for transforming growth factor (TGF)-alpha-induced pancreatic ductal metaplasia and neoplasia remain uncharacterized. During pancreatic development, differentiated cell types arise from ductal progenitor cells expressing the Pdx1 homeodomain transcription factor. The aims of this study were, first, to evaluate the role of Pdx1-expressing stem cells in MT-TGFalpha transgenic mice, and second, to further characterize cell proliferation and differentiation in this model. METHODS: To assess Pdx1 gene expression in normal and metaplastic epithelium, we performed in vivo reporter gene analysis using heterozygous Pdx1(lacZ/+) and bigenic Pdx1(lacZ/+)/MT-TGFalpha mice. RESULTS: Pdx1(lacZ/+)/MT-TGFalpha bigenics showed up-regulated Pdx1 expression in premalignant metaplastic ductal epithelium. In addition to Pdx1 gene activation, TGF-alpha-induced metaplastic epithelium demonstrated a pluripotent differentiation capacity, as evidenced by focal expression of Pax6 and initiation of islet cell neogenesis. The majority of Pdx1-positive epithelial cells showed no expression of insulin, similar to the pattern observed during embryonic development. CONCLUSIONS: Overexpression of TGF-alpha induces expansion of a Pdx1-expressing epithelium characterized by focal expression of Pax6 and initiation of islet neogenesis. These findings suggest that premalignant events induced by TGF-alpha in mouse pancreas may recapitulate a developmental program active during embryogenesis.


Subject(s)
Homeodomain Proteins , Islets of Langerhans/metabolism , Pancreatic Ducts/metabolism , Trans-Activators/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Differentiation , Cell Division , Epithelium/physiology , Metaplasia , Mice , Mice, Transgenic , Pancreatic Ducts/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
15.
J Nucl Med ; 40(11): 1784-91, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10565771

ABSTRACT

UNLABELLED: This study had two purposes: to optimize the semiquantitative interpretation of 18F-fluorodeoxyglucose (FDG) PET scans in the diagnosis of pancreatic carcinoma by analyzing different cutoff levels for the standardized uptake value (SUV), with and without correction for serum glucose level (SUV(gluc)); and to evaluate the usefulness of FDG PET when used in addition to CT for the staging and management of patients with pancreatic cancer. METHODS: Sixty-five patients who presented with suspected pancreatic carcinoma underwent whole-body FDG PET in addition to CT imaging. The PET images were analyzed visually and semiquantitatively using the SUV and SUV(gluc). The final diagnosis was obtained by pathologic (n = 56) or clinical and radiologic follow-up (n = 9). The performance of CT and PET at different cutoff levels of SUV was determined, and the impact of FDG PET in addition to CT on patient management was reviewed retrospectively. RESULTS: Fifty-two patients had proven pancreatic carcinoma, whereas 13 had benign lesions, including chronic pancreatitis (n = 10), benign biliary stricture (n = 1), pancreatic complex cyst (n = 1) and no pancreatic pathology (n = 1). Areas under receiver operating characteristic curves were not significantly different for SUV and SUV(gluc). Using a cutoff level of 3.0 for the SUV, FDG PET had higher sensitivity and specificity than CT in correctly diagnosing pancreatic carcinoma (92% and 85% versus 65% and 61%). There were 2 false-positive PET (chronic pancreatitis, also false-positive with CT) and 4 false-negative PET (all with true-positive CT, abnormal but nondiagnostic) examinations. There were 5 false-positive CT (4 chronic pancreatitis and 1 pancreatic cyst) and 18 false-negative CT (all with true-positive FDG PET scans) examinations. FDG PET clarified indeterminate hepatic lesions or identified additional distant metastases (or both) in 7 patients compared with CT. Overall, FDG PET altered the management of 28 of 65 patients (43%). CONCLUSION: FDG PET is more accurate than CT in the detection of primary tumors and in the clarification and identification of hepatic and distant metastases. The optimal cutoff value of FDG uptake to differentiate benign from malignant pancreatic lesions was 2.0. Correction for serum glucose did not significantly improve the accuracy of FDG PET. Although FDG PET cannot replace CT in defining local tumor extension, the application of FDG PET in addition to CT alters the management in up to 43% of patients with suspected pancreatic cancer.


Subject(s)
Adenocarcinoma/diagnostic imaging , Fluorodeoxyglucose F18 , Pancreatic Neoplasms/diagnostic imaging , Tomography, Emission-Computed , Adenocarcinoma/epidemiology , Blood Glucose/analysis , Female , Fluorine Radioisotopes , Humans , Male , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/epidemiology , Predictive Value of Tests , Radiopharmaceuticals , Sensitivity and Specificity , Tomography, X-Ray Computed
16.
Ann Surg Oncol ; 6(7): 651-7, 1999.
Article in English | MEDLINE | ID: mdl-10560850

ABSTRACT

BACKGROUND: Traditional teaching maintains that patients with primary colorectal adenocarcinoma require timely resection to prevent bleeding, perforation, or obstruction. The true benefits of primary tumor resection remain undocumented for patients presenting with metastatic disease, however. We postulated that resection of primary colorectal tumors could be avoided safely in a select population of asymptomatic colorectal cancer patients presenting with incurable stage IV disease. METHODS: A retrospective review of the Vanderbilt University Hospital tumor registry was performed for the years 1985 to 1997. During this period, 955 patients presented for management of primary colorectal cancer. From this group, all patients with stage IV disease at the time of diagnosis were identified. Patients who initially underwent resection of their primary lesion were included in the resection group; those who underwent initial nonoperative primary tumor management were included in the nonresection group. Data were obtained regarding age, extent of disease, nonsurgical therapy, tumor-specific complications, and palliative surgical procedures. Surgery-free survival and overall survival were analyzed using the Kaplan-Meier method. For patients with liver metastases, hepatic tumor burden was defined as either H1 (<25% parenchymal replacement), H2 (25% to 50%), or H3 (>50%) disease. RESULTS: Sixty-six patients were included in the resection group, and 23 patients with intact asymptomatic primary colorectal lesions were included in the nonresection group. Among patients with hepatic metastases, most of the patients in both groups had H1 disease. Ten patients in the resection group and 3 patients in the nonresection group presented with exclusively extrahepatic metastases. In the nonresection group, primary therapy included chemotherapy in 13 patients, external beam radiation therapy in 1 patient, and combination chemoradiation in 9 patients. The median survival in the nonresection group was 16.6 months. The 2-year actuarial survival was 18%, and the surgery-free survival was 91.3%. Only 2 of 23 patients (8.7%) managed without resection eventually developed obstruction at the primary tumor site requiring emergent diversion. There were no episodes of tumor-related hemorrhage or perforation. For the resection group, the operative morbidity was 30.3%, and the perioperative mortality rate was 4.6%. The median survival in the resection group was 14.5 months (P = 0.59, log-rank test vs. nonresection group). CONCLUSIONS: Selected patients with asymptomatic primary colorectal tumors who present with incurable metastatic disease may safely avoid resection of their primary lesions, with an anticipated low rate of hemorrhage, perforation, or obstruction before death from systemic disease. No survival advantage is gained by resection of an asymptomatic primary lesion in the setting of incurable stage IV colorectal cancer.


Subject(s)
Adenocarcinoma/therapy , Colorectal Neoplasms/therapy , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Registries , Retrospective Studies , Survival Analysis
17.
Oncology (Williston Park) ; 13(10 Suppl 5): 121-30, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10550836

ABSTRACT

Both gastric and pancreatic cancer remain leading causes of cancer death in the United States and worldwide. While surgical resection continues to be required for long-term cure of both these neoplasms, 5-year survival rates remain poor following surgery alone. For both gastric and pancreatic cancers, studies examining patterns of recurrence following apparently curative resection repeatedly demonstrate high rates of locoregional relapse. In this setting, the addition of chemoradiation delivered either before or following surgery represents a logical strategy to improve local tumor control and possibly improve survival. Data suggest that 5-fluorouracil-based chemoradiation, when given at sufficient doses, can effectively palliate patients with unresectable gastric cancer. Whether this approach improves response and survival in patients with resectable gastric cancer remains investigational. Results of ongoing and recently completed trials will provide further information on the utility of 5-fluorouracil-based regimens, as well as the use of other radiation sensitizers, in the adjuvant setting. In patients with resectable pancreatic cancer, the primary goal should be to perform a margin-negative pancreaticoduodenectomy. The use of neoadjuvant chemoradiation may increase the likelihood of achieving this goal, and this approach is being investigated.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Stomach Neoplasms/drug therapy , Stomach Neoplasms/radiotherapy , Adenocarcinoma/surgery , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy , Fluorouracil/therapeutic use , Humans , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy , Radiation-Sensitizing Agents/therapeutic use , Stomach Neoplasms/surgery
18.
J Gastrointest Surg ; 3(3): 233-43, 1999.
Article in English | MEDLINE | ID: mdl-10481116

ABSTRACT

Intraoperative assessment is inaccurate in defining the relationship of a pancreatic head neoplasm to adjacent vascular structures. We evaluated the ability of preoperative contrast-enhanced CT to predict the need for vascular resection during pancreaticoduodenectomy and examined the resected vessels for histologic evidence of tumor invasion. During a 7-year period, 63 patients underwent pancreaticoduodenectomy with en bloc resection of adjacent vascular structures for a presumed pancreatic head malignancy. Clinical, radiologic, operative, and pathologic data were reviewed and analyzed. Fifty-six patients underwent resection of the superior mesenteric-portal vein confluence, three patients required inferior vena cava resection, and the hepatic artery was resected and reconstructed in eight patients. The operative mortality rate was 1.6%, and the overall complication rate was 22%. CT predicted the need for resection of the superior mesenteric or portal veins in 84% of patients. Pathologic analysis revealed tumor invasion of the vein wall in 71% of resected specimens. Tumor invasion of vascular structures adjacent to the pancreas can be predicted with preoperative CT and should alert the surgeon that vascular resection may be required. Histologic evidence of tumor cell infiltration of vessel walls was present in the majority of the resected specimens.


Subject(s)
Pancreas/blood supply , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy , Vascular Surgical Procedures , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Carcinoma, Neuroendocrine/diagnostic imaging , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/surgery , Contrast Media , Female , Forecasting , Hepatic Artery/pathology , Hepatic Artery/surgery , Humans , Intraoperative Care , Male , Mesenteric Artery, Superior/pathology , Mesenteric Artery, Superior/surgery , Mesenteric Veins/pathology , Mesenteric Veins/surgery , Middle Aged , Neoadjuvant Therapy , Neoplasm Invasiveness , Pancreas/pathology , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Pancreaticoduodenectomy/methods , Portal Vein/pathology , Portal Vein/surgery , Radiographic Image Enhancement , Survival Rate , Tomography, X-Ray Computed , Vena Cava, Inferior/pathology , Vena Cava, Inferior/surgery
19.
Ann Surg ; 229(5): 729-37; discussion 737-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10235532

ABSTRACT

OBJECTIVE: To assess the accuracy and clinical impact of 18fluorodeoxyglucose-positron emission tomography (18FDG-PET) on the management of patients with suspected primary or recurrent pancreatic adenocarcinoma, and to assess the utility of 18FDG-PET in grading tumor response to neoadjuvant chemoradiation. SUMMARY BACKGROUND DATA: The diagnosis, staging, and treatment of pancreatic cancer remain difficult. Small primary tumors and hepatic metastases are often not well visualized by computed tomographic scanning (CT), resulting in a high incidence of nontherapeutic celiotomy and the frequent need for "blind resection." In addition, the distinction between local recurrence and nonspecific postoperative changes after resection can be difficult to ascertain on standard anatomic imaging. 18FDG-PET is a new imaging technique that takes advantage of increased glucose metabolism by tumor cells and may improve the diagnostic accuracy of preoperative studies for pancreatic adenocarcinoma. METHODS: Eighty-one 18FDG-PET scans were obtained in 70 patients undergoing evaluation for suspected primary or recurrent pancreatic adenocarcinoma. Of this group, 65 underwent evaluation for suspected primary pancreatic cancer. Nine patients underwent 18FDG-PET imaging before and after neoadjuvant chemoradiation, and in eight patients 18FDG-PET scans were performed for possible recurrent adenocarcinoma after resection. The 18FDG-PET images were analyzed visually and semiquantitatively using the standard uptake ratio (SUR). The sensitivity and specificity of 18FDG-PET and CT were determined for evaluation of the preoperative diagnosis of primary pancreatic carcinoma, and the impact of 18FDG-PET on patient management was retrospectively assessed. RESULTS: Among the 65 patients evaluated for primary tumor, 52 had proven pancreatic adenocarcinoma and 13 had benign lesions. 18FDG-PET had a higher sensitivity and specificity than CT in correctly diagnosing pancreatic carcinoma (92% and 85% vs. 65% and 62%). Eighteen patients (28%) had indeterminate or unrecognized pancreatic masses on CT clarified with 18FDG-PET. Seven patients (11%) had indeterminate or unrecognized metastatic disease clarified with 18FDG-PET. Overall, 18FDG-PET suggested potential alterations in clinical management in 28/65 patients (43%) with suspected primary pancreatic adenocarcinoma. Of the nine patients undergoing 18FDG-PET imaging before and after neoadjuvant chemoradiation, four had evidence of tumor regression by PET, three showed stable disease, and two showed tumor progression. CT was unable to detect any response to neoadjuvant therapy in this group. Eight patients had 18FDG-PET scans to evaluate suspected recurrent disease after resection. Four were noted to have new regions of 18FDG-uptake in the resection bed; four had evidence of new hepatic metastases. All proved to have metastatic pancreatic adenocarcinoma. CONCLUSIONS: These data confirm that 18FDG-PET is useful in the evaluation of patients with suspected primary or recurrent pancreatic carcinoma. 18FDG-PET is more sensitive and specific than CT in the detection of small primary tumors and in the clarification of hepatic and distant metastases. 18FDG-PET was also of benefit in assessing response to neoadjuvant chemoradiation. Although 18FDG-PET cannot replace CT in defining local tumor resectability, the application of 18FDG-PET in addition to CT may alter clinical management in a significant fraction of patients with suspected pancreatic cancer.


Subject(s)
Adenocarcinoma/diagnostic imaging , Fluorodeoxyglucose F18 , Pancreatic Neoplasms/diagnostic imaging , Radiopharmaceuticals , Tomography, Emission-Computed , Humans , Predictive Value of Tests , Reproducibility of Results , Sensitivity and Specificity
20.
Ann Surg ; 229(3): 303-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10077040

ABSTRACT

OBJECTIVE: In Western populations, long-term survival rates after curative resection of gastric cancer remain extremely poor. The lack of effective adjuvant therapy has prompted the evaluation of neoadjuvant approaches. Since 1988, we have conducted three separate phase II trials using neoadjuvant chemotherapy to treat patients with potentially resectable gastric cancer. The present study was conducted to evaluate whether response to neoadjuvant chemotherapy is predictive of survival in patients with resectable gastric cancer. METHODS: Eighty-three patients with pathologically confirmed gastric adenocarcinoma were treated with neoadjuvant chemotherapy before planned surgical resection. Response was assessed by upper gastrointestinal series, endoscopy, computed tomography scan, and pathologic examination. RESULTS: For the three phase II trials, clinical response rates ranged from 24% to 38%. Three patients (4%) had a complete pathologic response. Sixty-one patients (73%) underwent a curative resection. Median follow-up was 26 months. Univariate analysis revealed T stage, number of positive nodes, and response to chemotherapy to be significant predictors of overall survival. However, on multivariate analysis, response to chemotherapy was found to be the only independent prognostic factor. CONCLUSIONS: Response to neoadjuvant chemotherapy is the single most important predictor of overall survival after neoadjuvant chemotherapy for gastric cancer. These findings support further evaluation of neoadjuvant approaches in the treatment of this disease.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/surgery , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , Adenocarcinoma/mortality , Adult , Aged , Chemotherapy, Adjuvant , Female , Humans , Male , Middle Aged , Multivariate Analysis , Prognosis , Remission Induction , Stomach Neoplasms/mortality , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...