Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
PLoS One ; 11(10): e0165769, 2016.
Article in English | MEDLINE | ID: mdl-27798672

ABSTRACT

Biliary tract cancers (BTC) are rare but highly aggressive malignant epithelial tumors. In order to improve the outcome in this lethal disease, novel biomarkers for diagnosis, prognosis, and therapy response prediction are urgently needed. DNA promoter methylation of PITX2 variants (PITX2ab, PITX2c) and intragenic methylation of the PITX2 adjacent non-coding RNA (PANCR) were investigated by methylations-specific qPCR assays in formalin-fixed paraffin-embedded tissue from 80 patients after resection for BTC. Results were correlated with clinicopathologic data and outcome. PITX2 variants and PANCR showed significant hypermethylation in tumor vs. normal adjacent tissue (p < 0.001 and p = 0.015), respectively. In survival analysis, dichotomized DNA methylation of variant PITX2c and PANCR were significantly associated with overall survival (OS). Patients with high tumor methylation levels of PITX2c had a shorter OS compared to patients with low methylation (12 vs. 40 months OS; HR 2.48 [1.38-4.48], p = 0.002). In contrast, PANCR hypermethylation was associated with prolonged survival (25 vs. 19 months OS; HR 0.54 [0.30-0.94], p = 0.015) and qualified as an independent prognostic factor on multivariate analysis. The biomarkers investigated in this study may help to identify BTC subpopulations at risk for worse survival. Further studies are needed to evaluate if PITX2 might be a clinically useful biomarker for an optimized and individualized treatment.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/mortality , Biliary Tract Neoplasms/genetics , Biliary Tract Neoplasms/mortality , DNA Methylation , Homeodomain Proteins/genetics , RNA, Untranslated/genetics , Transcription Factors/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Biliary Tract Neoplasms/pathology , Biliary Tract Neoplasms/surgery , Biomarkers, Tumor , Chromosomes, Human, Pair 4 , Female , Genetic Loci , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Promoter Regions, Genetic , Homeobox Protein PITX2
2.
Oncotarget ; 6(18): 16437-48, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-25999351

ABSTRACT

Although ovarian cancer is a highly chemosensitive disease, it is only infrequently cured. One of the major reasons lies in the presence of drug-resistant cancer stem-like cells, sufficient to fuel recurrence. We phenotyped cancer stem-like cells by flow cytometry and immunohistochemistry in 55 matched samples before and after taxane/platinum-based neoadjuvant chemotherapy. All used markers of stemness (ALDH1, CD24, CD117, CD133) isolated low frequencies of malignant cells. ALDH1 was the most valuable marker for tracking stemness in vivo. The enrichment of ALDH1 expression after treatment was associated with a poor response to chemotherapy, with platinum resistance and independently prognosticated unfavorable outcome. Our results suggest that increased ALDH1 expression after treatment identifies patients with aggressive tumor phenotypes.


Subject(s)
Drug Resistance, Neoplasm , Isoenzymes/metabolism , Neoadjuvant Therapy , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Retinal Dehydrogenase/metabolism , AC133 Antigen , Adult , Aged , Aldehyde Dehydrogenase 1 Family , Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Bridged-Ring Compounds/therapeutic use , CD24 Antigen/metabolism , Female , Glycoproteins/metabolism , Humans , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Peptides/metabolism , Platinum Compounds/therapeutic use , Proto-Oncogene Proteins c-kit/metabolism , Retrospective Studies , Taxoids/therapeutic use , Treatment Outcome , Tumor Stem Cell Assay
3.
Virchows Arch ; 466(2): 133-41, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25425476

ABSTRACT

Biliary tract cancers are aggressive tumors of which the incidence seems to increase. Resection with cancer-free margins is crucial for curative therapy. However, how often biliary intraepithelial neoplasia (BilIN) occurs in resection margins and what its clinical and therapeutic implications might be is largely unknown. We reexamined margins of resection specimens of adenocarcinoma of the biliary tree including the gallbladder for the presence of BilIN. When present, it was graded. The findings were correlated with clinicopathological parameters and overall survival. Complete examination of the resection margin could be performed on 55 of 78 specimens (71%). BilIN was detected in the margin in 29 specimens (53%) and was mainly low-grade (BilIN-1; N = 14 of 29; 48%). In resection specimens of extrahepatic cholangiocarcinoma, BilIN was most frequent (N = 6 of 8; 75%). BilIN was found in the resection margin more frequently in extrahepatic cholangiocarcinomas (P = 0.007) and in large primary tumors (P = 0.001) with lymphovascular (P = 0.006) and perineural invasion (P = 0.049). Patients with cancer in the resection margin (R1) had a significantly shorter overall survival than those with resection margins free of tumor (R0) irrespective of the presence of BilIN (R0 vs R1; P < 0.001) or BilIN grade (BilIN-positive vs BilIN-negative, P = 0.6, and BilIN-1 + 2 vs BilIN-3, P = 0.58). BilIN is frequently found in the surgical margin of resection specimens of adenocarcinoma of the biliary tract. Hepatopancreatobiliary surgeons will be confronted with this recently defined entity when an intraoperative frozen section of a resection margin is requested. However, this diagnosis does not require additional resection and in the intraoperative evaluation of resection, the emphasis should remain on the detection of residual invasive tumor.


Subject(s)
Adenocarcinoma/pathology , Biliary Tract Neoplasms/pathology , Carcinoma in Situ/pathology , Adenocarcinoma/mortality , Adult , Aged , Biliary Tract Neoplasms/mortality , Carcinoma in Situ/mortality , Female , Humans , Infant, Newborn , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm, Residual/mortality , Neoplasm, Residual/pathology , Proportional Hazards Models
4.
Int J Cancer ; 136(3): 699-708, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-24895230

ABSTRACT

This phase I study tested the safety, feasibility, pharmacokinetics and pharmacodynamics of cisplatin administered as hyperthermic intraoperative intraperitoneal chemoperfusion (HIPEC) in patients with platinum-sensitive recurrent epithelial ovarian cancer (EOC) undergoing secondary cytoreductive surgery followed by postoperative platinum-based intravenous chemotherapy. Twelve patients with operable, recurrent platinum-sensitive EOC (recurrence ≥6 months after first-line therapy) were included according to the classical 3+3 dose-escalation design at three dose levels-60, 80 and 100 mg/m(2). After surgical cytoreduction, a single dose of cisplatin was administered via HIPEC for 90 min at 41-43°C. Postoperatively, all patients were treated with standard intravenous platinum-based combination chemotherapy. One of six patients experienced a dose-limiting toxicity (grade 3 renal toxicity) at a dose of 100 mg/m(2). The remaining five patients treated with 100 mg/m(2) tolerated their treatment well. The recommended phase II dose was established at 100 mg/m(2). The mean peritoneal-to-plasma AUC ratio was 19·5 at the highest dose level. Cisplatin-induced DNA adducts were confirmed in tumor samples. Common postoperative grade 1-3 toxicities included fatigue, postoperative pain, nausea, and surgical site infection. The ability to administer standard intravenous platinum-based chemotherapy after HIPEC was uncompromised. Cisplatin administered as HIPEC at a dose of 100 mg/m(2) has an acceptable safety profile in selected patients undergoing secondary cytoreductive surgery for platinum-sensitive recurrent EOC. Favorable pharmacokinetic and pharmacodynamic properties of HIPEC with cisplatin were confirmed at all dose levels, especially at 100 mg/m(2). The results are encouraging to determine the efficacy of HIPEC as a complementary treatment in patients with EOC.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/administration & dosage , Cytoreduction Surgical Procedures/methods , Hyperthermia, Induced , Neoplasm Recurrence, Local/therapy , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/therapy , Adult , Aged , Carcinoma, Ovarian Epithelial , Cisplatin/adverse effects , Cisplatin/analysis , Cisplatin/pharmacokinetics , Combined Modality Therapy , DNA Adducts/analysis , Female , Humans , Middle Aged
5.
Virchows Arch ; 465(4): 419-27, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25172328

ABSTRACT

Recent studies identified unexpected expression and transcriptional complexity of the hemoprotein myoglobin (MB) in human breast cancer but its role in prostate cancer is still unclear. Expression of MB was immunohistochemically analyzed in three independent cohorts of radical prostatectomy specimens (n = 409, n = 625, and n = 237). MB expression data were correlated with clinicopathological parameters and molecular parameters of androgen and hypoxia signaling. Expression levels of novel tumor-associated MB transcript variants and the VEGF gene as a hypoxia marker were analyzed using qRT-PCR. Fifty-three percent of the prostate cancer cases were MB positive and significantly correlated with androgen receptor (AR) expression (p < 0.001). The positive correlation with CAIX (p < 0.001) and FASN (p = 0.008) as well as the paralleled increased expression of the tumor-associated MB transcript variants and VEGF suggest that hypoxia participates in MB expression regulation. Analogous to breast cancer, MB expression in prostate cancer is associated with steroid hormone signaling and markers of hypoxia. Further studies must elucidate the novel functional roles of MB in human carcinomas, which probably extend beyond its classic intramuscular function in oxygen storage.


Subject(s)
Myoglobin/biosynthesis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/biosynthesis , Aged , Biomarkers, Tumor/analysis , Cell Hypoxia/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunohistochemistry , Male , Middle Aged , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL