Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Cancer ; 138(1): 206-16, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26178530

ABSTRACT

Interest has increased in the potential role of circulating tumour cells in cancer management. Most cell-based studies have been designed to determine the number of circulating tumour cells in a given volume of blood. Ability to understand the biology of the cancer cells would increase the clinical potential. The purpose of this study was to develop and validate a novel, widely applicable method for detection and characterisation of circulating tumour cells. Cells were imaged with an ImageStream(X) imaging flow cytometer which allows detection of expression of multiple biomarkers on each cell and produces high-resolution images. Depletion of haematopoietic cells was by red cell lysis, leukocyte common antigen CD45 depletion and differential centrifugation. Expression of epithelial cell adhesion molecule, cytokeratins, tumour-type-specific biomarkers and CD45 was detected by immunofluorescence. Nuclei were identified with DAPI or DRAQ5 and brightfield images of cells were collected. The method is notable for the dearth of cell damage, recoveries greater than 50%, speed and absence of reliance on the expression of a single biomarker by the tumour cells. The high-quality images obtained ensure confidence in the specificity of the method. Validation of the methodology on samples from patients with oesophageal, hepatocellular, thyroid and ovarian cancers confirms its utility and specificity. Importantly, this adaptable method is applicable to all tumour types including those of nonepithelial origin. The ability to measure simultaneously the expression of multiple biomarkers will facilitate analysis of the cancer cell biology of individual circulating tumour cells.


Subject(s)
Flow Cytometry/methods , Liver Neoplasms/diagnosis , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Ovarian Neoplasms/diagnosis , Thyroid Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Female , Humans , Immunophenotyping , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging
2.
Gynecol Oncol ; 124(1): 142-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22001143

ABSTRACT

OBJECTIVES: In the present study we explore the effects of androgens and anti-androgens on primary cultures of EOC cells. We also investigate the effects of chemotherapy on AR expression. Epithelial ovarian cancer (EOC) arises from ovarian surface epithelial cells (OSE), which express the androgen receptor (AR). Androgen stimulation of OSE cells results in increased proliferation and protection from apoptosis. Nevertheless, in clinical trials anti-androgens have had a low objective response rate in relapsed ovarian cancer. METHODS: 1. Androgen receptor (AR) expression and response to androgenic stimulation were correlated in primary ovarian cancer cells derived from ascitic fluid from patients with advanced ovarian cancer, 2. AR expression in primary epithelial ovarian cancer was investigated before and after chemotherapy using paired histological samples which had been incorporated into a tissue microarray. RESULTS: Eleven primary ovarian cancer cultures were established from ascitic fluid. There was wide variation of expression of androgen receptor mRNA between cultures. Cell division increased after dihydro-testosterone (DHT) stimulation in 6 out of 11 primary cultures. The fraction of cells in S-phase increased from 4.4% in cells grown in serum-free medium to 8.3% in cells stimulated with 100 nM of DHT (P<0.001). The increase in S-phase fraction was abrogated after treatment with the anti-androgen, bicalutamide in 4 out of 5 responsive cultures. There was a strong correlation (r(2)=0.7) between nuclear AR expression by immunohistochemistry and S-phase fraction changes in primary cultures. Paired pre- and post-chemotherapy histological samples from 29 patients were incorporated into a tissue microarray (TMA). Nuclear and cytoplasmic AR expression by immunohistochemistry (IHC) decreased significantly after chemotherapy (P<0.01). CONCLUSION: AR expression correlates with increased S-phase fraction in response to androgenic stimulation. Immunohistochemical analysis of AR expression needs to be further tested in clinical trials to select AR positive EOC for anti-androgen therapy. Anti-androgen use early in the course of ovarian cancer is more likely to be effective as these data suggest that androgen receptor expression decreases with exposure to chemotherapy and this may explain the low response rates seen in clinical trials of patients heavily pre-treated with multiple courses of chemotherapy.


Subject(s)
Androgens/pharmacology , Biomarkers, Tumor/biosynthesis , Cystadenocarcinoma, Serous/metabolism , Dihydrotestosterone/pharmacology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Hormone-Dependent/metabolism , Ovarian Neoplasms/metabolism , Receptors, Androgen/biosynthesis , Ascitic Fluid/pathology , Biomarkers, Tumor/genetics , Carcinoma, Ovarian Epithelial , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Exons , Female , Humans , Immunohistochemistry , Neoplasm Grading , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Androgen/genetics , S Phase , Stimulation, Chemical , Trinucleotide Repeats , Tumor Cells, Cultured
3.
Clin Cancer Res ; 15(4): 1241-9, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19174487

ABSTRACT

PURPOSE: High-risk neuroblastoma is characterized by poor survival rates, and the development of improved therapeutic approaches is a priority. Temozolomide and topotecan show promising clinical activity against neuroblastoma. Poly(ADP-ribose) polymerase-1 (PARP-1) promotes DNA repair and cell survival following genotoxic insult; we postulated that its inhibition may enhance the efficacy of these DNA-damaging drugs in pediatric cancers. EXPERIMENTAL DESIGN: We evaluated the chemosensitizing properties of the PARP inhibitor AG014699 (Pfizer, Inc.) in combination with temozolomide and topotecan, against human neuroblastoma cells and xenografts, alongside associated pharmacologic and toxicologic indices. RESULTS: Addition of PARP-inhibitory concentrations of AG014699 significantly potentiated growth inhibition by both topotecan (1.5- to 2.3-fold) and temozolomide (3- to 10-fold) in vitro, with equivalent effects confirmed in clonogenic assays. In two independent in vivo models (NB1691 and SHSY5Y xenografts), temozolomide caused a xenograft growth delay, which was enhanced by co-administration of AG014699, and resulted in complete and sustained tumor regression in the majority (6 of 10; 60%) of cases. Evidence of enhanced growth delay by topotecan/AG014699 co-administration was observed in NB1691 xenografts. AG014699 metabolites distributed rapidly into the plasma (Cmax, 1.2-1.9 nmol/L at 30 min) and accumulated in xenograft tissues (Cmax, 1-2 micromol/L at 120 min), associated with a sustained suppression of PARP-1 enzyme activity. Doses of AG014699 required for potentiation were not toxic per se. CONCLUSIONS: These data show enhancement of temozolomide and topotecan efficacy by PARP inhibition in neuroblastoma. Coupled with the acceptable pharmacokinetic, pharmacodynamic, and toxicity profiles of AG014699, our findings provide strong rationale for investigation of PARP inhibitors in pediatric early clinical studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dacarbazine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Neuroblastoma/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Topotecan/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Drug Synergism , Humans , Mice , Neuroblastoma/pathology , Poly (ADP-Ribose) Polymerase-1 , Temozolomide , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 8(2): 407-14, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19174557

ABSTRACT

Resistance to temozolomide and radiotherapy is a major problem for patients with glioblastoma but may be overcome using the poly(ADP-ribose) polymerase inhibitor ABT-888. Using two primary glioblastoma xenografts, the efficacy of ABT-888 combined with radiotherapy and/or temozolomide was evaluated. Treatment with ABT-888 combined with temozolomide resulted in significant survival prolongation (GBM12: 55.1%, P = 0.005; GBM22: 54.4%, P = 0.043). ABT-888 had no effect with radiotherapy alone but significantly enhanced survival in GBM12 when combined with concurrent radiotherapy/temozolomide. With multicycle therapy, ABT-888 further extended the survival benefit of temozolomide in the inherently sensitive GBM12 and GBM22 xenograft lines. However, after in vivo selection for temozolomide resistance, the derivative GBM12TMZ and GBM22TMZ lines were no longer sensitized by ABT-888 in combination with temozolomide, and a similar lack of efficacy was observed in two other temozolomide-resistant tumor lines. Thus, the sensitizing effects of ABT-888 were limited to tumor lines that have not been previously exposed to temozolomide, and these results suggest that patients with newly diagnosed glioblastoma may be more likely to respond to combined temozolomide/poly(ADP-ribose) polymerase inhibitor therapy than patients with recurrent disease.


Subject(s)
Benzimidazoles/therapeutic use , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Xenograft Model Antitumor Assays , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Cell Line, Tumor , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Glioblastoma/enzymology , Glioblastoma/radiotherapy , Humans , Mice , Mice, Nude , Poly(ADP-ribose) Polymerases , Temozolomide
5.
Clin Cancer Res ; 11(21): 7834-40, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16278406

ABSTRACT

PURPOSE: To determine the safety, maximum tolerated dose, pharmacokinetics, and toxicities associated with administration of paclitaxel poliglumex (PPX, XYOTAX, Cell Therapeutics, Inc., Bresso, Italy) given on either 3-weekly or 2-weekly schedule. EXPERIMENTAL DESIGN: Nineteen patients were investigated on the 3-weekly phase Ia study and 11 patients on the 2-weekly phase Ib study. Dose escalation starting with 100% increments and one patient per dose level was modulated in accordance with the observed toxicities. Conjugated and unconjugated paclitaxel were measured in plasma. RESULTS: Dose-limiting toxicity of neutropenia was encountered at 266 mg/m(2) (paclitaxel equivalents) in phase Ia and the maximum tolerated dose was 233 mg/m(2). Neuropathy was dose-limiting in phase Ib with a maximum tolerated dose of 177 mg/m(2). Pharmacokinetic investigations indicated a prolonged half-life of >100 hours for conjugated taxanes. Plasma concentrations of unconjugated paclitaxel were similar to those following administration of an equivalent dose of Taxol. Two partial responses were observed, one in a patient with mesothelioma at 177 mg/m(2) in phase Ia and one in a patient with gastric carcinoma at 175 mg/m(2) in phase Ib. CONCLUSION: PPX is a water-soluble paclitaxel-polymer conjugate with a prolonged half-life and limited volume of distribution. Dose-limiting toxicities were neutropenia and neuropathy. PPX showed activity in this patient population.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Paclitaxel/administration & dosage , Polyglutamic Acid/analogs & derivatives , Adult , Aged , Antineoplastic Agents, Phytogenic/pharmacokinetics , Area Under Curve , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Models, Chemical , Paclitaxel/pharmacokinetics , Polyglutamic Acid/administration & dosage , Polyglutamic Acid/pharmacokinetics , Polymers/chemistry , Time Factors
6.
Clin Cancer Res ; 11(9): 3402-9, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867241

ABSTRACT

PURPOSE: Temozolomide, a DNA methylating agent used to treat melanoma, induces DNA damage, which is repaired by O6-alkylguanine alkyltransferase (ATase) and poly(ADP-ribose) polymerase-1 (PARP-1)-dependent base excision repair. The current study was done to define the effect of temozolomide on DNA integrity and relevant repair enzymes as a prelude to a phase I trial of the combination of temozolomide with a PARP inhibitor. EXPERIMENTAL DESIGN: Temozolomide (200 mg/m2 oral administration) was given to 12 patients with metastatic malignant melanoma. Peripheral blood lymphocytes (PBL) were analyzed for PARP activity, DNA single-strand breakage, ATase levels, and DNA methylation. PARP activity was also measured in tumor biopsies from 9 of 12 patients and in PBLs from healthy volunteers. RESULTS: Temozolomide pharmacokinetics were consistent with previous reports. Temozolomide therapy caused a substantial and sustained elevation of N7-methylguanine levels, a modest and sustained reduction in ATase activity, and a modest and transient increase in DNA strand breaks and PARP activity in PBLs. PARP-1 activity in tumor homogenates was variable (828 +/- 599 pmol PAR monomer/mg protein) and was not consistently affected by temozolomide treatment. CONCLUSIONS: The effect of temozolomide reported here are consistent with those documented in previous studies with temozolomide and similar drug, dacarbazine, demonstrating that a representative patient population was investigated. Furthermore, PARP activity was not inhibited by temozolomide treatment and this newly validated pharmacodynamic assay is therefore suitable for use in a proof-of-principle phase I trial a PARP-1 inhibitor in combination with temozolomide.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Melanoma/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/pharmacokinetics , Comet Assay , DNA Damage , DNA Methylation , DNA Repair , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Dacarbazine/adverse effects , Dacarbazine/pharmacokinetics , Female , Headache/chemically induced , Humans , Lymphocytes/enzymology , Lymphocytes/metabolism , Male , Melanoma/enzymology , Melanoma/genetics , Middle Aged , Neoplasm Metastasis , Neutropenia/chemically induced , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Temozolomide , Thrombocytopenia/chemically induced , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...