Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Dis Esophagus ; 17(1): 38-43, 2004.
Article in English | MEDLINE | ID: mdl-15209739

ABSTRACT

Pathologic complete response in the resected esophagus can be achieved in approximately 30% of patients with locally advanced esophageal or gastroesophageal junction carcinoma after preoperative chemoradiation therapy. These patients tend to have a longer survival than those who have less than pathologic complete response. Post-chemoradiation esophageal biopsy (PCEB) is used to check for the presence of residual tumor before a definitive resection is performed, but the clinical significance of PCEB findings is not clear due to the possibility of sampling bias and the superficial nature of the specimen obtained. We evaluated the use of PCEB (defined as biopsy taken within 30 days before esophagectomy) in predicting residual cancer in post-treatment esophagectomy specimens. PCEB was performed in 65 of 183 (36%) patients with locally advanced esophageal or gastroesophageal junction carcinoma, who received preoperative chemoradiation therapy. The cancer status in PCEB was correlated with the residual cancer in the esophagectomy specimens. PCEB had no cancer in 80% (52 of 65) of patients (Bx-negative) and cancer in 20% (13 of 65) of patients (Bx-positive). There was no difference in the presence of residual cancer (either in esophagus or lymph node) in esophagectomy specimens between Bx-negative patients (77%, 40 of 52) or Bx-positive patients (92%, 12 of 13), P = 0.44. The positive predictive value of biopsy was 92% (12 of 13), negative predictive value 23% (12 of 52), sensitivity 23% (12 of 52) and specificity 92% (12 of 13). There was no difference in the residual cancer staging in the esophagectomy specimen between Bx-positive and Bx-negative patients. In contrast, residual metastatic carcinoma in lymph nodes was more frequent in Bx-positive patients (69.2%, 9 of 13) than in Bx-negative patients (28.8%, 15 of 52), P = 0.01. Our data suggest that PCEB is a specific but not a sensitive predictor of residual cancer following esophagectomy. Bx-positive patients tend to have more frequent residual tumor in lymph nodes. The utility of PCEB in predicting residual cancer in the lymph nodes needs to be explored further along with molecular predictors of response to preoperative therapy.


Subject(s)
Biopsy, Needle , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophagogastric Junction/pathology , Neoplasm, Residual/pathology , Chemotherapy, Adjuvant , Combined Modality Therapy , Esophageal Neoplasms/mortality , Esophagectomy/methods , Esophagectomy/mortality , Esophagogastric Junction/surgery , Female , Humans , Immunohistochemistry , Male , Neoplasm Staging , Postoperative Care , Predictive Value of Tests , Preoperative Care/methods , Probability , Prognosis , Radiotherapy, Adjuvant , Retrospective Studies , Sensitivity and Specificity , Survival Analysis
2.
Int J Oncol ; 19(4): 681-5, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562741

ABSTRACT

A histopathology study of 22 pancreatic adenocarcinoma cases revealed that 13 of the patients presented with hyperplastic lesions (atypical and non-atypical hyperplasia, mucous cell hypertrophy, focal epithelial hyperplasia, and ductal papillary hyperplasia), and 9 exhibited fibrosis adjacent to the carcinoma. All lesions expressed high levels of epidermal growth factor receptor (EGF-R) (p<0.0001 and p=0.0008, respectively) as compared with normal ductal epithelium. Non-atypical and atypical hyperplastic lesions also had a higher proliferating cell nuclear antigen (PCNA) labeling index (p<0.001 and p=0.0008, respectively) than normal ductal epithelium. A gradient in PCNA+ nuclei was found in acinar cells adjacent to the tumors. In 16 cases with marked fibrosis, we observed a significant increase of PCNA+ nuclei in stromal fibroblasts (p=0.0041) and significant upregulation of basic fibroblast growth factor (bFGF) mRNA expression in adjacent tumor cells (p=0.0213). These data suggest that the production of bFGF by pancreatic cancer cells induces ductal and stromal hyperplasia of the pancreas.


Subject(s)
Adenocarcinoma/metabolism , Fibroblast Growth Factor 2/metabolism , Pancreatic Ducts/pathology , Pancreatic Neoplasms/metabolism , Stromal Cells/pathology , Adenocarcinoma/surgery , DNA Primers/chemistry , DNA Probes , Fibroblast Growth Factor 2/genetics , Fibrosis , Humans , Hyperplasia , In Situ Hybridization , Mitotic Index , Neoplasm Staging , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/surgery , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/metabolism , Stromal Cells/metabolism , Tumor Cells, Cultured/pathology
3.
Ann Surg Oncol ; 8(2): 123-32, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11258776

ABSTRACT

BACKGROUND: For patients with potentially resectable pancreatic cancer, the poor outcome associated with resection alone and the survival advantage demonstrated for combined-modality therapy have stimulated interest in preoperative chemoradiotherapy. The goal of this study was to analyze the effects of different preoperative chemoradiotherapy schedules, intraoperative radiation therapy, patient factors. and histopathologic variables on survival duration and patterns of treatment failure in patients who underwent pancreaticoduodenectomy for adenocarcinoma of the pancreatic head. METHODS: Data on 132 consecutive patients who received preoperative chemoradiation followed by pancreaticoduodenectomy for adenocarcinoma of the pancreatic head between June 1990 and June 1999 were retrieved from a prospective pancreatic tumor database. Patients received either 45.0 or 50.4 Gy radiation at 1.8 Gy per fraction in 28 fractions or 30.0 Gy at 3.0 Gy per fraction in 10 fractions with concomitant infusional chemotherapy (5-fluorouracil, paclitaxel, or gemcitabine). If restaging studies demonstrated no evidence of disease progression, patients underwent pancreaticoduodenectomy. All patients were evaluated with serial postoperative computed tomography scans to document first sites of tumor recurrence. RESULTS: The overall median survival from the time of tissue diagnosis was 21 months (range 19-26, 95%CI). At last follow-up, 41 patients (31%) were alive with no clinical or radiographic evidence of disease. The survival duration was superior for women (P = .04) and for patients with no evidence of lymph node metastasis (P = .03). There was no difference in survival duration associated with patient age, dose of preoperative radiation therapy, the delivery of intraoperative radiotherapy, tumor grade, tumor size, retroperitoneal margin status, or the histologic grade of chemoradiation treatment effect. CONCLUSION: This analysis supports prior studies which suggest that the survival duration of patients with potentially resectable pancreatic cancer is maximized by the combination of chemoradiation and pancreaticoduodenectomy. Furthermore, there was no difference in survival duration between patients who received the less toxic rapid-fractionation chemoradiotherapy schedule (30 Gy, 2 weeks) and those who received standard-fractionation chemoradiotherapy (50.4 Gy, 5.5 weeks). Short-course rapid-fractionation preoperative chemoradiotherapy combined with pancreaticoduodenectomy, when performed on accurately staged patients, maximizes survival duration and is associated with a low incidence of local tumor recurrence.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Adenocarcinoma/mortality , Adenocarcinoma/surgery , Antineoplastic Agents/therapeutic use , Chemotherapy, Adjuvant , Combined Modality Therapy/methods , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Fluorouracil/administration & dosage , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/epidemiology , Paclitaxel/administration & dosage , Pancreatectomy/adverse effects , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Radiotherapy Dosage , Radiotherapy, Adjuvant , Survival Analysis , Treatment Outcome , Gemcitabine
4.
J Clin Oncol ; 18(20): 3529-34, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11032595

ABSTRACT

PURPOSE: Preoperative combined-modality therapy for rectal cancer may allow for sphincter preservation, while decreasing recurrence rates and improving the overall prognosis. Oral chemotherapy with uracil and tegafur (UFT) plus leucovorin (LV) may reduce costs and complications associated with protracted infusions of fluorouracil. Our goal was to evaluate the safety of UFT plus LV combined with preoperative radiation and determine the maximum-tolerated dose (MTD) and dose-limiting toxicity (DLT) of UFT plus LV in this setting. PATIENTS AND METHODS: Patients with tumor-node-metastasis stage II or III rectal cancer received escalating doses of UFT (starting at 250mg/m(2)/d, with 50-mg/m(2)/d increments between consecutive cohorts) and fixed doses of LV (90 mg/d). The UFT and LV combination was given 5 days per week concurrently with a 5-week course of preoperative radiation totaling 45 Gy (1.8 Gy/fraction). Surgery was performed 4 to 6 weeks after radiation and was followed by four 35-day cycles of fixed doses of UFT and LV (28 days of therapy each cycle). RESULTS: Fifteen patients were treated, and 13 received the full preoperative chemotherapy. All planned radiation was delivered successfully. The MTD of UFT with radiation was 350 mg/m(2)/d with 90 mg/d of LV. Diarrhea was the DLT. Sphincter-preserving surgery was performed in 12 of 14 patients. One patient had progressive disease before surgery. Pathologic evaluation of 14 resected specimens showed a complete response in three cases. CONCLUSION: Preoperative chemoradiation with oral UFT plus LV is feasible and well tolerated and should be further investigated.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Rectal Neoplasms/therapy , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Combined Modality Therapy , Drug Administration Schedule , Female , Humans , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Middle Aged , Postoperative Care , Preoperative Care , Rectal Neoplasms/drug therapy , Rectal Neoplasms/radiotherapy , Rectal Neoplasms/surgery , Surgical Procedures, Operative , Tegafur/administration & dosage , Tegafur/adverse effects , Uracil/administration & dosage , Uracil/adverse effects
5.
Cancer ; 89(6): 1252-8, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11002220

ABSTRACT

BACKGROUND: Human pancreatic ribonuclease (RNase 1) is a pancreatic enzyme that is present at high levels in the serum of most patients with pancreatic adenocarcinoma. For this reason, the authors studied its patterns of expression at the single-cell level in pancreatic adenocarcinoma tissues by immunohistochemical analysis and in situ hybridization (ISH). METHODS: Immunohistochemical analysis with polyclonal antibodies against RNase 1 and by ISH with digoxigenin-labeled RNase 1 probe were used to detect RNase 1 in the neoplastic cells of ductal type pancreatic adenocarcinomas. RESULTS: Fifteen of 18 carcinoma samples were positive for RNase 1, demonstrating that the expression of ribonuclease that the authors observed previously in human pancreatic adenocarcinoma cell lines was not an artifact of cell culture. The authors also found RNase 1 in some of the metaplastic ducts and atrophic islets in 4 of 6 chronic pancreatitis samples, and they observed RNase 1 immunostaining in hyperplastic ducts adjacent to one of the well-differentiated adenocarcinomas. CONCLUSIONS: The expression levels of RNase 1 by tumor cells from pancreatic adenocarcinomas are consistent with the high RNase 1 levels found in the serum of most patients with pancreatic adenocarcinoma. This expression of RNase 1, which is an acinar protein, demonstrates that the patterns of gene expression in pancreatic adenocarcinoma are distinct from those of normal pancreatic duct cells. Conversely, RNase 1 expression levels in altered ductal cells from some chronic pancreatitis tissues and hyperplastic ducts from carcinoma tissues suggest that abnormal expression levels may be an early event in pancreatic tumorigenesis.


Subject(s)
Adenocarcinoma/enzymology , Carcinoma, Pancreatic Ductal/enzymology , Pancreatic Neoplasms/enzymology , Ribonuclease, Pancreatic/metabolism , Adenocarcinoma/pathology , Adult , Aged , Antibodies , Carcinoma, Pancreatic Ductal/pathology , Chronic Disease , Female , Humans , Immunohistochemistry , In Situ Hybridization , Liver Neoplasms/enzymology , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Middle Aged , Pancreas/enzymology , Pancreatic Neoplasms/pathology , Pancreatitis/enzymology , Ribonuclease, Pancreatic/biosynthesis , Ribonuclease, Pancreatic/immunology , Risk Factors
6.
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
7.
J Gastrointest Surg ; 3(3): 263-77, 1999.
Article in English | MEDLINE | ID: mdl-10481119

ABSTRACT

The p53 tumor suppressor gene and the Bcl-2 proto-oncogene regulate cell cycle progression and apoptosis. We evaluated the expression of these molecular markers with standard pathologic prognostic variables in patients who received multimodality therapy for resectable adenocarcinoma of the pancreas to study the effect of p53 and Bcl-2 on survival duration. Immunohistochemical staining of archival material was performed to determine levels of expression of p53 and Bcl-2 proteins in 70 patients with adenocarcinoma of pancreatic origin. All patients underwent a potentially curative pancreaticoduodenectomy and standardized pathologic analysis of resected specimens. Potential pathologic and molecular prognostic variables were assessed for their effect on survival duration. Nuclear staining for p53 was observed in 33 (47%) of 70 specimens. Immunostaining for Bcl-2 was observed in 23 specimens (33%). A trend toward improved survival duration was seen in patients whose tumors stained positive for either p53 or Bcl-2. Negative staining for both markers predicted short survival (P = 0.01). By univariate and multivariate analyses, no single pathologic factor was associated with survival duration. Immunohistochemical staging using both p53 and Bcl-2 significantly predicted survival duration by univariate and multivariate analysis; patients whose tumors stained positively for p53 and/or overexpressed Bcl-2 had a significantly longer survival than those whose tumors stained negative for both proteins.


Subject(s)
Adenocarcinoma/surgery , Biomarkers, Tumor/analysis , Pancreatic Neoplasms/surgery , Proto-Oncogene Proteins c-bcl-2/analysis , Tumor Suppressor Protein p53/analysis , Adenocarcinoma/pathology , Analysis of Variance , Apoptosis/genetics , Cell Cycle/genetics , Cell Nucleus/ultrastructure , Coloring Agents , Combined Modality Therapy , Female , Forecasting , Gene Expression Regulation, Neoplastic/genetics , Genes, bcl-2/genetics , Genes, p53/genetics , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Pancreatic Neoplasms/pathology , Pancreaticoduodenectomy , Prognosis , Prospective Studies , Proto-Oncogene Mas , Survival Rate
8.
Gastroenterology ; 117(2): 350-8, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10419916

ABSTRACT

BACKGROUND & AIMS: Cyclooxygenase (COX) enzymes catalyze the conversion of arachidonic acid to prostaglandins. Evidence suggests that nonsteroidal anti-inflammatory drugs reduce the risk of colorectal cancer (CRC) and that this effect is mediated through COX inhibition. We analyzed and compared expression of the inducible COX-2 isoform in colorectal neoplasms from patients with hereditary nonpolyposis colorectal cancer (HNPCC), familial adenomatous polyposis (FAP), and sporadic CRC. Given that COX-2 is induced by transforming growth factor (TGF)-beta and that TGF-beta type II receptor (RII) mutations are found in HNPCCs, we determined the relationship between RII status and COX-2 expression. METHODS: COX-2 protein expression was determined in colorectal epithelia using immunohistochemistry and Western blotting. Patients with HNPCC had known mutations in hMLH1 or hMSH2 genes and/or met the Amsterdam criteria. In CRCs from HNPCC cases, mutations were sought in the coding region of the RII gene using the polymerase chain reaction. RESULTS: COX-2 was detected in adenomas from 2 of 3 HNPCC, 6 of 7 FAP, and 5 of 8 sporadic cases. In CRCs, COX-2 staining was found in 16 of 24 (67%) HNPCC vs. 24 of 26 (92%) sporadic cases (P = 0.035) and in 2 of 2 FAP cases. Staining intensity was reduced in HNPCCs compared with sporadic CRCs (P = 0.035). Staining localized to the cytoplasm of neoplastic cells; normal epithelial cells were negative for COX-2. Overexpression of COX-2 in CRCs relative to normal mucosa was confirmed by Western blotting. TGF-beta RII mutations were detected in 12 of 14 HNPCCs examined, including 3 of 4 COX-2-negative and 9 of 10 COX-2-positive cancers. CONCLUSIONS: The frequency and intensity of COX-2 expression was significantly reduced in HNPCCs relative to sporadic CRCs, and was not a consequence of RII mutations. Given that many HNPCCs express COX-2, inhibition of this enzyme may be an important strategy to prevent CRC in these patients.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/enzymology , Isoenzymes/analysis , Prostaglandin-Endoperoxide Synthases/analysis , Adenomatous Polyposis Coli/enzymology , Blotting, Western , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Humans , Immunohistochemistry , Isoenzymes/drug effects , Isoenzymes/immunology , Membrane Proteins , Mutation , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/immunology , Receptors, Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
9.
Int J Radiat Oncol Biol Phys ; 44(5): 1027-38, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10421535

ABSTRACT

PURPOSE: To evaluate the rates of tumor downstaging after preoperative chemoradiation for locally advanced rectal cancer. MATERIALS AND METHODS: Preoperative chemoradiotherapy (CTX/XRT) that delivered 45 Gy in 25 fractions over 5 weeks with continuous infusion 5-fluorouracil (300 mg/m2/day) was given to 117 patients. The pretreatment stage distribution, as determined by endorectal ultrasound (u), included uT2N0 in 2%, uT3N0 in 47%, uT3N1 in 49%, and uT4N0 in 2% of cases; endorectal ultrasound was not performed in 13% of cases (15 patients). Approximately 6 weeks after completion of CTX/XRT, surgery was performed. RESULTS: The pathological tumor stages were Tis-2N0 in 26%, T2N1 in 5%, T3N0 in 21%, T3N1 in 15%, T4N0 in 5%, and T4NI in 1%; a complete response (CR) to preoperative CTX/XRT was pathologically confirmed in 32 (27%) of patients. Tumor downstaging occurred in 72 (62%) cases. Only 3% of cases had pathologic evidence of progressive disease. Pretreatment tumor size (< 5 cm vs. > or = 5 cm) was the only factor predictive of tumor downstaging (p < 0.04). A decrease of > 1 T-stage level was accomplished in 45% of those downstaged. Overall, a sphincter-saving (SP) procedure was possible in 59% of patients and an abdominoperineal resection (APR) was required in 41 % of cases. Factors predictive of SP included downstaging (p < 0.03), age > 40 years (p < 0.007), pretreatment tumor distance, 3 to 6 cm from the anal verge (p < 0.00001), tumor size <6 cm (p < 0.02), mobility (p < 0.004), tumor stage 6 cm from the anal verge, SP was performed in 14 of the 15 (93%) patients with a CR and 32 of 33 (97%) of patients with residual disease (p < 0.00004). CONCLUSIONS: Significant tumor downstaging results from preoperative chemoradiation allowing sphincter sparing surgery in over 40% of patients whose tumors were located < 6 cm from the anal verge and who otherwise would have required colostomy.


Subject(s)
Anal Canal , Neoplasm Staging , Rectal Neoplasms/drug therapy , Rectal Neoplasms/radiotherapy , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/therapeutic use , Combined Modality Therapy , Disease-Free Survival , Female , Fluorouracil/therapeutic use , Humans , Lymphatic Metastasis/pathology , Male , Middle Aged , Neoplasm, Residual , Radiotherapy Dosage , Rectal Neoplasms/mortality , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery
10.
J Invest Dermatol ; 112(5): 802-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10233775

ABSTRACT

The cytokine interferon-beta is a regulator of cell replication and function, including invasion and induction of angiogenesis. The goal of this study was to determine whether the expression of interferon-beta by cells in the epidermis correlated with terminal differentiation. In situ hybridization analysis and immunohistochemical staining of formalin-fixed, paraffin-embedded specimens of normal human and murine epidermis and human and murine skin tumors of epithelial origin revealed that only differentiated, nondividing cells of the epidermis expressed interferon-beta protein. Keratinocyte cultures established from the epidermis of 3 d old mice were maintained under conditions permitting continuous cell division or induction of differentiation. Continuously dividing cells did not produce interferon-beta whereas nondividing differentiated cells expressing keratin 1 did. Growth-arrested, undifferentiated keratinocytes also expressed interferon-beta protein. Neutralizing interferon-beta in the culture medium inhibited differentiation, but the addition of exogenous interferon-beta did not stimulate differentiation. These data indicate that interferon-beta is produced by growth-arrested, terminally differentiated keratinocytes.


Subject(s)
Epidermal Cells , Epidermis/metabolism , Interferon-beta/biosynthesis , Animals , Antibodies/pharmacology , Calcium/physiology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Cell Division/physiology , Cells, Cultured , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Interferon-beta/immunology , Interferon-beta/pharmacology , Interferon-beta/physiology , Keratin-14 , Keratinocytes/cytology , Keratinocytes/metabolism , Keratins/biosynthesis , Membrane Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Transgenic , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Time Factors
11.
Oncol Rep ; 6(3): 497-505, 1999.
Article in English | MEDLINE | ID: mdl-10203581

ABSTRACT

Most hereditary non-polyposis colorectal cancer (HNPCC) is due to germline mutations in DNA mismatch repair genes. Tumors arising as a result of these mutations display instability in microsatellites, which are short tandem repeats of DNA that are distributed throughout the genome. Although a subset of sporadic colorectal carcinomas also have microsatellite instability (MSI), the phenotype is a useful screening test in identifying patients with HNPCC caused by mutations in mismatch repair (MMR) genes. Studies have shown that some microsatellite markers are more efficient than others in identifying tumors with MSI. Furthermore, the frequency of instability can be assessed by categorizing patients into high (MSI-H, >/= 30-40% positive markers), low (MSI-L), and microsatellite stable (MSS) groups. Using a panel of 28 microsatellite markers, tumor and normal DNA from 10 HNPCC patients was used to identify the five most efficient markers for detecting MSI (BAT26, D2S123, FGA, D18S35, and TP53-DI). Each of the five markers detected MSI in 80-100% of the cases examined. We then expanded the sample size to 17 tumors from HNPCC patients. Each case had evidence for a mutation in either hMSH2 or hMLH1. We compared the efficiency of our panel of five best markers with another panel of five markers (BAT25, BAT26, D2S123, D17S250, and D5S346) identified as being efficient markers for detection of MSI at a recent NCI workshop. Our five selected markers were more efficient (85% vs. 79%) in detecting MSI. However, using either panel, 100% of the cases fell into the MSI-H category and the probability of misclassifying an MSI-H case as MSI-L is very low (0.002-0.008). We also examined four cases meeting the Amsterdam criteria for HNPCC, but with no evidence for mutation in either the hMSH2 or hMLH1 gene. With our panel, three were classified as MSI-H, while only two were classified as such with the NCI reference panel. The probability of misclassifying an MSI-L case as an MSI-H, using a panel of five markers is high (0.263).


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Microsatellite Repeats , Basic Helix-Loop-Helix Transcription Factors , DNA-Binding Proteins/genetics , Fluorescent Dyes , Genetic Markers , Germ-Line Mutation , Humans , Immunohistochemistry , MutS Homolog 2 Protein , Polymerase Chain Reaction/methods , Proto-Oncogene Proteins/genetics
12.
Ann Surg Oncol ; 6(1): 26-32, 1999.
Article in English | MEDLINE | ID: mdl-10030412

ABSTRACT

BACKGROUND: Local excision of rectal cancer preserves anal continence, bladder function, and normal sexual function. However, local recurrence after excision remains a significant problem. To further define the indications for local excision, we analyzed possible factors predictive of recurrence after local excision of rectal cancer. METHODS: The charts of all patients undergoing local excision of adenocarcinoma of the rectum between 1985 and 1995 at a single institution were reviewed. Patients with metastatic disease at the time of excision and patients treated preoperatively with chemoradiation therapy were excluded. All available slides were reviewed by a single pathologist, who assessed the depth of invasion; the presence or absence of vascular invasion, lymphatic invasion, perineural invasion, and lymphocytic infiltrate; the mucinous status; and the degree of differentiation. Using the log-rank test and Cox proportional hazards model, univariate and multivariate analyses were performed to identify predictors of recurrence. RESULTS: Ninety patients underwent local excision, 46 transanally and 44 using a Kraske approach. The breakdown of patients by tumor stage was as follows: Tis, 13%; T1, 41%; T2, 30%; T3, 15%; and Tx, 1%. Sixty-eight percent of patients with T1 tumors were treated with postoperative radiotherapy; all patients with T2 or T3 tumors were treated postoperatively with or without 5-fluorouracil. The median duration of follow-up was 51 months. The median tumor diameter was 2.5 cm (range, 0.4 to 7 cm), and the median distance of the tumor from the anal verge was 4.5 cm (range, 1 to 10 cm). The 4-year actuarial local disease-free survival rate broken down by tumor stage was as follows: Tis, 100%; T1, 95%; T2, 80%; and T3, 73%. The median time to local recurrence was 23 months (range, 7 to 61 months). Multivariate analysis showed that only tumor stage and margin status were predictors of local recurrence. CONCLUSIONS: Local excision and postoperative radiotherapy result in adequate local control of early stage (Tis and T1) adenocarcinoma of the rectum. Higher rates of recurrence were seen in patients with T2 and T3 tumors, especially in those with positive margins.


Subject(s)
Adenocarcinoma/diagnosis , Neoplasm Recurrence, Local/diagnosis , Postoperative Care , Rectal Neoplasms/diagnosis , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Radiotherapy, Adjuvant , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery , Risk Factors , Time Factors
13.
Clin Cancer Res ; 5(1): 25-33, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9918199

ABSTRACT

We examined the expression level of several genes that regulate distinct steps of metastasis in formalin-fixed, paraffin-embedded, archival specimens of primary human pancreatic carcinomas from patients undergoing curative surgery. The expression of epidermal growth factor receptor, E-cadherin, type IV collagenase [matrix metalloproteinase (MMP) 2 and MMP-9), basic fibroblast growth factor, vascular endothelial growth factor/vascular permeability factor, and interleukin 8 was examined by a colorimetric in situ mRNA hybridization technique. Down-regulation of E-cadherin and up-regulation of type IV collagenase (MMP-9 and MMP-2) at the periphery of the neoplasms (P = 0.0167, 0.0102, and 0.0349, respectively) had significant prognostic value. The ratio of type IV collagenase expression (mean of the expression of MMP-2 and MMP-9) to E-cadherin expression (MMP:E-cadherin ratio) at the periphery of the tumors was significantly higher in patients with recurrent disease (4.7 +/- 2.1) than in patients who were disease free (2.3 +/- 1.7; P = 0.0008). Death from pancreatic cancer was significantly associated with a high MMP:E-cadherin ratio (>3.0) by overall survival analysis (P < 0.0002), whereas a low MMP:E-cadherin ratio (<3.0) was found in seven of eight patients alive 28-64 months after surgery. Multivariate analysis of overall survival showed that the MMP:E-cadherin ratio was a significant independent prognostic factor, whereas stage, nodal metastasis, and histological type were not. These data show that multiparametric analysis for several metastasis-related genes may allow physicians to assess the metastatic potential and hence predict the clinical outcome of individual patients with resectable pancreatic carcinoma.


Subject(s)
Adenocarcinoma/genetics , Cadherins/biosynthesis , Collagenases/biosynthesis , Pancreatic Neoplasms/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cadherins/genetics , Collagenases/genetics , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization , Matrix Metalloproteinase 9 , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Predictive Value of Tests , Prognosis
14.
Clin Cancer Res ; 5(1): 119-27, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9918209

ABSTRACT

Pancreatic adenocarcinoma is a leading cause of adult cancer mortality in the United States. Recent studies have revealed that point mutation of the K-ras oncogene is a common event in pancreatic cancer, and oncogenesis mediated by Ras may also involve activation of Rel/nuclear factor (NF)-kappa B transcription factors. Furthermore, the c-rel member of Rel/NF-kappa B transcription factor family was first identified as a cellular homologue of the v-rel oncogene, suggesting that other members of the Rel/NF-kappa B family are potentially oncogenes. We therefore investigated the possibility that Rel/NF-kappa B transcription factors are activated in pancreatic cancer. Immunohistochemical analysis, Western blot and Northern blot analysis, electrophoretic mobility shift assays, and chloramphenicol acetyltransferase assays were performed to determine RelA activity in human pancreatic adenocarcinomas and normal tissues and nontumorigenic or tumorigenic cell lines. RelA, the p65 subunit of NF-kappa B, was constitutively activated in approximately 67% (16 of 24) of pancreatic adenocarcinomas but not in normal pancreatic tissues. Constitutive RelA activity was also detected in 9 of 11 human pancreatic tumor cell lines but not in nontumorigenic Syrian golden hamster cell lines. I kappa B alpha, a previously identified NF-kappa B-inducible gene, was overexpressed in human pancreatic tumor tissues and cell lines, and RelA activation could be inhibited by curcumin and dominant-negative mutants of I kappa B alpha, raf, and MEKK1. This is the first report demonstrating constitutive activation of RelA in nonlymphoid human cancer. These data are consistent with the possibility that RelA is constitutively activated by the upstream signaling pathway involving Ras and mitogen-activated protein kinases in pancreatic tumor cells. Constitutive RelA activity may play a key role in pancreatic tumorigenesis through activation of its downstream target genes.


Subject(s)
Adenocarcinoma/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adult , Amino Acid Sequence , Animals , Cricetinae , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Immunohistochemistry , Mesocricetus , Molecular Sequence Data , Pancreatic Neoplasms/genetics , Point Mutation , Transcription Factor RelA , Tumor Cells, Cultured
15.
Endocr Pract ; 5(1): 43-5, 1999.
Article in English | MEDLINE | ID: mdl-15251702

ABSTRACT

OBJECTIVE: To describe a rare case of sarcoidosis diagnosed after surgical resection of an adenoma in a patient with Cushing's disease. METHODS: We present a case report and discuss the only other similar case identified from the published medical literature. RESULTS: In a 33-year-old woman with symptoms of cortisol excess (including progressive weight gain, proximal muscle weakness, ecchymoses, and amenorrhea), laboratory studies showed serum cortisol values consistent with Cushing's syndrome. After inferior petrosal sinus sampling, the patient underwent surgical resection of an adenoma in the right aspect of the anterior pituitary gland. Maintenance corticosteroid therapy was implemented, and the signs and symptoms of Cushing's disease began to resolve. Five months postoperatively, multiple erythematous lesions developed on the patient's arms and legs. Skin biopsy specimens demonstrated noncaseating granulomas, and sarcoidosis was diagnosed. Two months later, the lesions resolved spontaneously without therapy other than continued corticosteroid replacement. CONCLUSION: Physicians should be aware that unusual postoperative symptoms in a patient who has undergone successful treatment of Cushing's syndrome may be attributable to the emergence of another corticosteroid-responsive disease such as sarcoidosis.

16.
J Clin Oncol ; 16(12): 3843-50, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9850029

ABSTRACT

PURPOSE: To evaluate the toxicities, radiographic and pathologic responses, and event-free outcomes with combined modality treatment that involves preoperative rapid-fractionation chemoradiation, pancreaticoduodenectomy, and electron-beam intraoperative radiation therapy (EB-IORT) for patients with resectable pancreatic adenocarcinoma. PATIENTS AND METHODS: Patients with radiographically resectable localized adenocarcinoma of the pancreatic head were entered onto a preoperative protocol that consisted of a 2-week course of fluorouracil (5-FU) 300 mg/m2 daily 5 days per week and concomitant rapid-fractionation radiation 30 Gy, 3 Gy daily 5 days per week. Radiographic restaging was performed 4 weeks after chemoradiation, and patients with localized disease underwent pancreaticoduodenectomy with EB-IORT 10 to 15 Gy. RESULTS: Thirty-five patients were entered onto the study and completed chemoradiation, 34 (97%) as outpatients. Three patients (9%) experienced grade 3 nausea and vomiting; no other grade 3 or 4 toxicities were observed. Of the 27 patients taken to surgery, 20 patients (74%) underwent pancreaticoduodenectomy with EB-IORT. All patients had a less than grade III pathologic response to preoperative chemoradiation. At a median follow-up of 37 months, the 3-year survival rate in patients who underwent combined modality therapy was 23%. CONCLUSION: Combined modality treatment with preoperative rapid-fractionation chemoradiation, pancreaticoduodenectomy, and EB-IORT is associated with minimal toxicity and excellent locoregional control. This represents one approach to maximize the proportion of patients who receive all components of combined modality therapy and avoids the toxicity of pancreaticoduodenectomy in patients found to have metastatic disease at the time of restaging.


Subject(s)
Adenocarcinoma/therapy , Antimetabolites, Antineoplastic/therapeutic use , Fluorouracil/therapeutic use , Pancreatic Neoplasms/therapy , Pancreaticoduodenectomy , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Adenocarcinoma/surgery , Adult , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Combined Modality Therapy , Dose Fractionation, Radiation , Electrons/therapeutic use , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Male , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Pancreatic Neoplasms/surgery , Radiotherapy, Adjuvant/adverse effects , Radiotherapy, Adjuvant/methods , Survival Analysis , Treatment Outcome
17.
Telemed J ; 4(2): 167-75, 1998.
Article in English | MEDLINE | ID: mdl-9710649

ABSTRACT

OBJECTIVE: The ISIS Center at Georgetown University received a grant from the U.S. Army to act as systems integrator for a project to design, develop, and implement a commercial off-the-shelf teleradiology system to support the U.S. troops in Bosnia-Herzegovina. The goal of the project was to minimize troop movement while providing primary diagnosis to military personnel. This paper focuses on Digital Imaging Communications in Medicine (DICOM) 3.0 related issues that arose from this type of teleradiology implementation. The objective is to show that using the DICOM standard provides a good starting point for systems integration but is not a plug-and-play operation. METHODS: Systems were purchased that were based on the DICOM 3.0 standard. The modalities implemented in this effort were computed radiography (CR), computed tomography (CT), film digitization (FD), and ultrasonography (US). Dry laser printing and multiple-display workstations were critical components of this network. The modalities and output devices were integrated using the DICOM 3.0 standard. All image acquisition from the modalities is directly to a workstation. The workstation distributes the images to other local and remote workstations, to the dry laser printer, and to other vendors' workstations using the DICOM 3.0 standard. All systems were integrated and tested prior to deployment or purchase. Local and wide area networking were also tested prior to implementation of the deployable radiology network. RESULTS: The results of the integration of the multivendor network were positive. Eventually, all vendors' systems did communicate. Software configuration and operational changes were made to many systems in order to facilitate this communication. Often, software fixes or patches were provided by a vendor to modify their DICOM 3.0 implementation to allow better communications with another vendor's system. All systems were commercially available, and any modifications or changes provided became part of the vendor's commercially available package. CONCLUSION: Seven DICOM interfaces were implemented for this project, and none was achieved without modification of configuration files, changes or patches in vendor software, or operational changes. Some of the problems encountered included missing or ignored required data elements, padding of data values, unique study identifiers (UID), and the use of application entity titles. The difficulties with multivendor connectivity lie in the understanding and interpretation of standards such as DICOM 3.0. The success of this network proves that these problems can be overcome and a clinically successful network implemented utilizing multiple vendors' systems.


Subject(s)
Teleradiology/methods , Computer Communication Networks , Computer Systems , Data Display , Humans , Lasers , Military Medicine , Printing , Radiography , Radiology Information Systems , Software , Systems Integration , Teleradiology/instrumentation , Tomography, X-Ray Computed , Ultrasonography , United States
18.
Br J Surg ; 85(5): 611-7, 1998 May.
Article in English | MEDLINE | ID: mdl-9635805

ABSTRACT

BACKGROUND: The survival of patients who underwent pancreaticoduodenectomy with or without en bloc resection of the superior mesenteric-portal vein (SMPV) confluence for adenocarcinoma of the pancreatic head was compared. METHODS: To be considered for surgery, patients were required to fulfil the following computed tomography criteria for resectability: (1) absence of extrapancreatic disease, (2) no evidence of tumour extension to the superior mesenteric artery (SMA) or coeliac axis, and (3) a patent SMPV confluence. Tumour adherence to the superior mesenteric vein (SMV) or SMPV confluence was assessed at operation and en bloc venous resection was performed when necessary to achieve complete tumour extirpation. RESULTS: Seventy-five consecutive patients underwent pancreaticoduodenectomy, 44 without venous resection and 31 with en bloc resection of the SMPV confluence. There were no perioperative deaths in either group; late (more than 6 months) occlusion of the reconstructed SMPV confluence contributed to the death of two patients. Median survival in the 31 patients who required venous resection at the time of pancreaticoduodenectomy was 22 months, and that for the 44 control patients was 20 months (P = 0.25). CONCLUSION: Patients with adenocarcinoma of the pancreatic head who require venous resection during pancreaticoduodenectomy for isolated tumour extension to the SMV or SMPV confluence (in the absence of tumour extension to the SMA or coeliac axis) have a duration of survival no different from that of patients who undergo standard pancreaticoduodenectomy. These data suggest that venous involvement is a function of tumour location rather than an indicator of aggressive tumour biology.


Subject(s)
Mesenteric Veins/surgery , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy/methods , Portal Vein/surgery , Adult , Aged , Aged, 80 and over , Anastomosis, Surgical , Blood Loss, Surgical , Blood Vessel Prosthesis Implantation , Female , Follow-Up Studies , Humans , Intraoperative Care , Length of Stay , Male , Middle Aged , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/radiotherapy , Pancreaticoduodenectomy/mortality , Surgical Flaps , Survival Analysis , Survival Rate
19.
Eur J Cancer ; 34(3): 337-40, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9640218

ABSTRACT

Angiogenesis is essential for growth and metastasis of solid malignancies. In several tumours, tumour vessel count and expression of vascular endothelial growth factor (VEGF), a potent angiogenic factor, have been associated with prognosis. To determine if vessel count and VEGF expression are prognostic factors in pancreatic cancer, we examined these parameters in resected tumour specimens from 22 patients who did not receive pre-operative therapy. Paraffin-embedded tumour specimens were immunohistochemically stained for factor VIII (surrogate for vessels) and VEGF. Vessel counts and VEGF expression were evaluated without knowledge of patient outcome. The median follow-up for the entire group had not been reached as of 23.1 months (range 10-69 months). The mean vessel count and VEGF expression were no different between those patients who had recurrences and those who did not. By linear regression analysis, the correlation of VEGF expression with vessel count did not reach statistical significance (P = 0.0685). Survival and time to recurrence were similar in patients with high and low vessel counts and VEGF expression of 1, 2 or 3. Tumour differentiation or lymph node positivity had no effect on either VEGF expression or vessel count. Our data suggest that, in contrast to findings in other solid malignancies, vessel count and VEGF expression are not predictors of survival or recurrence in patients with resectable adenocarcinoma of the pancreas.


Subject(s)
Adenocarcinoma/blood supply , Endothelial Growth Factors/metabolism , Pancreatic Neoplasms/blood supply , Follow-Up Studies , Humans , Immunohistochemistry , Neoplasm Recurrence, Local , Neovascularization, Pathologic , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Survival Analysis
20.
Clin Cancer Res ; 4(2): 429-34, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9516932

ABSTRACT

We have previously shown that platelet-derived endothelial cell growth factor (PD-ECGF) is associated with angiogenesis of human colon cancer; this factor is expressed at high levels in vascular tumors that express low levels of vascular endothelial growth factor (VEGF). In these colon cancers, the major source of PD-ECGF is the infiltrating cells. In this study, we examined the role of PD-ECGF in the angiogenesis of human gastric cancer. Immunostaining for PD-ECGF was done on 93 gastric cancers previously stained for VEGF, basic fibroblast growth factor, and factor VIII-related antigen (specific for endothelial cells). To determine the cell type expressing PD-ECGF, double staining was done using antibodies to both PD-ECGF and CD68 (specific for macrophages). PD-ECGF was expressed more frequently in infiltrating cells (positive CD68 staining; 53.8%) than in tumor epithelium (9.7%; P < 0.0001). Infiltrating cells simultaneously stained positive for both PD-ECGF and CD68. An association between PD-ECGF expression in infiltrating cells, VEGF expression in tumor epithelium, and vessel count was observed in intestinal-type gastric cancer but not in diffuse-type gastric cancer. Vessel count was greater in tumors with high expression of both PD-ECGF and VEGF than in those with high expression of either factor alone (P = 0.002). Multiple angiogenic factors expressed by both tumor cells and infiltrating cells may play a role in the regulation of angiogenesis in intestinal-type gastric cancer.


Subject(s)
Neovascularization, Pathologic/metabolism , Stomach Neoplasms/blood supply , Thymidine Phosphorylase/biosynthesis , Adult , Aged , Endothelial Growth Factors/biosynthesis , Female , Humans , Immunohistochemistry , Lymphokines/biosynthesis , Male , Middle Aged , Neoplasm Staging , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...