Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Acta Neuropathol ; 121(3): 381-96, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21267586

ABSTRACT

Medulloblastoma is heterogeneous, being characterized by molecular subgroups that demonstrate distinct gene expression profiles. Activation of the WNT or SHH signaling pathway characterizes two of these molecular subgroups, the former associated with low-risk disease and the latter potentially targeted by novel SHH pathway inhibitors. This manuscript reports the validation of a novel diagnostic immunohistochemical method to distinguish SHH, WNT, and non-SHH/WNT tumors and details their associations with clinical, pathological and cytogenetic variables. A cohort (n = 235) of medulloblastomas from patients aged 0.4-52 years was studied for expression of four immunohistochemical markers: GAB1, ß-catenin, filamin A, and YAP1. Immunoreactivity (IR) for GAB1 characterizes only SHH tumors and nuclear IR for ß-catenin only WNT tumors. IRs for filamin A and YAP1 identify SHH and WNT tumors. SHH, WNT, and non-SHH/WNT tumors contributed 31, 14, and 55% to the series. All desmoplastic/nodular (D/N) medulloblastomas were SHH tumors, while most WNT tumors (94%) had a classic phenotype. Monosomy 6 was strongly associated with WNT tumors, while PTCH1 loss occurred almost exclusively among SHH tumors. MYC or MYCN amplification and chromosome 17 imbalance occurred predominantly among non-SHH/WNT tumors. Among patients aged 3-16 years and entered onto the SIOP PNET3 trial, outcome was significantly better for children with WNT tumors, when compared to SHH or non-SHH/WNT tumors, which showed similar survival curves. However, high-risk factors (M+ disease, LC/A pathology, MYC amplification) significantly influenced survival in both SHH and non-SHH/WNT groups. We describe a robust method for detecting SHH, WNT, and non-SHH/WNT molecular subgroups in formalin-fixed medulloblastoma samples. In corroborating other studies that indicate the value of combining clinical, pathological, and molecular variables in therapeutic stratification schemes for medulloblastoma, we also provide the first outcome data based on a clinical trial cohort and novel data on how molecular subgroups are distributed across the range of disease.


Subject(s)
Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Medulloblastoma/pathology , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Biomarkers, Tumor/metabolism , Child , Child, Preschool , Contractile Proteins/metabolism , Female , Filamins , Gene Expression Regulation, Neoplastic/physiology , Humans , Infant , Kaplan-Meier Estimate , Male , Microfilament Proteins/metabolism , Middle Aged , Phosphoproteins/metabolism , Retrospective Studies , Signal Transduction/physiology , Transcription Factors , YAP-Signaling Proteins , Young Adult , beta Catenin/metabolism
2.
Drug Metab Lett ; 4(4): 195-201, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20583968

ABSTRACT

The ATP-binding cassette transporters P-glycoprotein (ABCB1, MDR1) and multidrug resistance protein 4 (MRP4) efflux irinotecan and its active metabolite SN-38 in vitro, and thus may contribute to system clearance of these compounds. Mdr1a/b(-/-), Mrp4(-/-), and wild-type mice were administered 20 or 40 mg/kg irinotecan, and plasma samples were collected for 6 hours. Irinotecan and SN-38 lactone and carboxylate were quantitated and data were analyzed with nonlinear mixed-effects modeling. Mdr1a/b genotype was a significant covariate for the clearance of both irinotecan lactone and SN-38 lactone. Exposures to irinotecan lactone and SN-38 lactone after a 40 mg/kg dose were 1.6-fold higher in Mdr1a/b(-/-) mice compared to wild-type mice. Plasma concentrations of irinotecan lactone, irinotecan carboxylate, and SN-38 lactone in Mrp4(-/-) mice were similar to the wild-type controls. These results suggest that P-gp plays a role in irinotecan and SN-38 elimination, but Mrp4 does not affect irinotecan or SN-38 plasma pharmacokinetics.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/pharmacokinetics , Camptothecin/analogs & derivatives , Multidrug Resistance-Associated Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Antineoplastic Agents/blood , Biotransformation , Camptothecin/administration & dosage , Camptothecin/blood , Camptothecin/pharmacokinetics , Carboxylic Acids/blood , Female , Genotype , Injections, Intravenous , Irinotecan , Lactones/blood , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Multidrug Resistance-Associated Proteins/deficiency , Multidrug Resistance-Associated Proteins/genetics , Phenotype , ATP-Binding Cassette Sub-Family B Member 4
3.
Biomed Chromatogr ; 24(6): 626-31, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19810006

ABSTRACT

A method to rapidly measure dopamine (DA), dihydroxyindolphenylacetic acid, homovanillic acid, serotonin (5-HT) and 5-hydroxyindoleacetic acid concentrations in cerebrospinal fluid (CSF) has not yet been reported. A rapid, sensitive, and specific HPLC method was therefore developed using electrochemical detection. CSF was mixed with an antioxidant solution prior to freezing to prevent neurotransmitter degradation. Separation of the five analytes was obtained on an ESA MD-150 x 3.2 mm column with a flow rate of 0.37 mL/min and an acetonitrile-aqueous (5 : 95, v/v) mobile phase with 75 mM monobasic sodium phosphate buffer, 0.5 mM EDTA, 0.81 mM sodium octylsulfonate and 5% tetrahydrofuran. The optimal electrical potential settings were: guard cell +325 mV, E1 -100 mV and E2 +300 mV. Within-day and between-day precisions were <10% for all analytes and accuracies ranged from 91.0 to 106.7%. DA, 5-HT, and their metabolites were stable in CSF with antioxidant solution at 4 degrees C for 8 h in the autoinjector. This method was used to measure neurotransmitters in CSF obtained from children enrolled on an institutional medulloblastoma treatment protocol.


Subject(s)
Chromatography, High Pressure Liquid/methods , Dopamine/cerebrospinal fluid , Electrochemistry/methods , Serotonin/cerebrospinal fluid , Child , Child, Preschool , Chromatography, High Pressure Liquid/instrumentation , Dopamine/metabolism , Electrochemistry/instrumentation , Humans , Male , Serotonin/metabolism
4.
Genes Dev ; 23(23): 2729-41, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19952108

ABSTRACT

Medulloblastoma is the most common solid malignancy of childhood, with treatment side effects reducing survivors' quality of life and lethality being associated with tumor recurrence. Activation of the Sonic hedgehog (Shh) signaling pathway is implicated in human medulloblastomas. Cerebellar granule neuron precursors (CGNPs) depend on signaling by the morphogen Shh for expansion during development, and have been suggested as a cell of origin for certain medulloblastomas. Mechanisms contributing to Shh pathway-mediated proliferation and transformation remain poorly understood. We investigated interactions between Shh signaling and the recently described tumor-suppressive Hippo pathway in the developing brain and medulloblastomas. We report up-regulation of the oncogenic transcriptional coactivator yes-associated protein 1 (YAP1), which is negatively regulated by the Hippo pathway, in human medulloblastomas with aberrant Shh signaling. Consistent with conserved mechanisms between brain tumorigenesis and development, Shh induces YAP1 expression in CGNPs. Shh also promotes YAP1 nuclear localization in CGNPs, and YAP1 can drive CGNP proliferation. Furthermore, YAP1 is found in cells of the perivascular niche, where proposed tumor-repopulating cells reside. Post-irradiation, YAP1 was found in newly growing tumor cells. These findings implicate YAP1 as a new Shh effector that may be targeted by medulloblastoma therapies aimed at eliminating medulloblastoma recurrence.


Subject(s)
Cerebellar Neoplasms/metabolism , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Neurons/cytology , Neurons/metabolism , Up-Regulation , Animals , Cell Proliferation , Cells, Cultured , Cerebellar Neoplasms/pathology , Humans , Insulin Receptor Substrate Proteins/metabolism , Medulloblastoma/pathology , Mice , Protein Transport , Signal Transduction , Transcription Factors/metabolism
5.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(18-19): 1709-15, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19447687

ABSTRACT

A rapid and selective method for simultaneous determination of cyclophosphamide and its metabolite carboxyethylphosphoramide mustard (CEPM) was developed using online sample preparation and separation with tandem mass spectrometric detection. Diluted plasma was injected onto an extraction column (Cyclone MAX 0.5 mm x 50 mm, >30 microm), the sample matrix was washed with an aqueous solution, and retained analytes were transferred to an analytical column (Gemini 3 microm C18 110A, 100 mm x 2.0 mm) using a gradient mobile phase prior to detection by MS/MS. Analytes were detected in an API-3000 LC-MS/MS system using positive multiple-reaction monitoring mode (m/z 261/140 and 293/221 for CTX and CEPM, respectively). Online extraction recoveries were 76% and 72% for cyclophosphamide and CEPM. Within-day and between-day variabilities were <3.0%, and accuracies were between -6.9% and 5.2%. This method has been used to measure plasma cyclophosphamide and CEPM concentrations in an ongoing Phase II study in children with newly diagnosed medulloblastoma.


Subject(s)
Antineoplastic Agents, Alkylating/blood , Cyclophosphamide/blood , Phosphoramide Mustards/blood , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Clinical Trials, Phase II as Topic , Humans , Medulloblastoma/metabolism
6.
Curr Eye Res ; 34(5): 386-94, 2009 May.
Article in English | MEDLINE | ID: mdl-19401882

ABSTRACT

PURPOSE: To compare the expression of pregnane xenobiotic receptor and certain multi-drug resistance proteins in retinoblastoma. METHODS: Using tissue microarray analyses, we studied 62 pathology specimens for expression of pregnane xenobiotic receptor, multi-drug resistance 1/P glycoprotein, multi-drug resistance proteins 1, 2, and 4, and breast cancer resistant protein. RESULTS: Comparing tumors treated with primary enucleation with tumors treated with chemotherapy and/or radiation showed no significant differences in the expression of multi-drug resistance proteins or pregnane xenobiotic receptor. Pregnane xenobiotic receptor was correlated with multi-drug resistance protein 2 expression (p < 0.001). CONCLUSION: Our results indicate selection, rather than induction, of chemoresistant cells as a cause for treatment failure in managing retinoblastoma with primary systemic chemotherapy.


Subject(s)
Multidrug Resistance-Associated Proteins/metabolism , Receptors, Steroid/metabolism , Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Child , Cohort Studies , Drug Therapy , Eye Enucleation , Gene Expression , Humans , Microarray Analysis , Multidrug Resistance-Associated Protein 2 , Neoplasm Proteins/metabolism , Pregnane X Receptor , Radiotherapy , Retinal Neoplasms/therapy , Retinoblastoma/therapy
7.
Biomed Chromatogr ; 23(7): 707-13, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19277971

ABSTRACT

Individualization of topotecan dosing reduces inter-patient variability in topotecan exposure, presumably reducing toxicity and increasing efficacy. However, logistical limitations (e.g. requirement for plasma, intensive bedside plasma processing) have prevented widespread application of this approach to dosing topotecan. Thus, the objectives of the present study were to develop and validate an HPLC with fluorescence detection method to measure topotecan lactone in whole blood samples and to evaluate its application to individualizing topotecan dosing. Plasma samples (200 microL) were prepared using methanolic precipitation, a filtration step and then injection of 100 microL of the methanolic extract onto a Novapak C(18), 4 microm, 3.9 x 150 mm column with an isocratic mobile phase. Analytes were detected with a Shimadzu Fluorescence RF-10AXL detector with an excitation and emission wavelength of 370 and 520 nm, respectively. This method had a lower limit of quantification of 1 ng/mL (S/N >or= 5; RSD 4.9%) and was validated over a linear range of 1-100 ng/mL. Results from a 5-day validation study demonstrated good within-day and between-day precision and accuracy. Data are presented to demonstrate that the present method can be used with whole blood samples to individualize topotecan dosing in children with cancer.


Subject(s)
Chromatography, High Pressure Liquid/methods , Topotecan/blood , Drug Stability , Humans , Linear Models , Methanol/chemistry , Reproducibility of Results , Sensitivity and Specificity , Specimen Handling/methods , Spectrometry, Fluorescence/methods
8.
Am J Physiol Cell Physiol ; 295(2): C440-50, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18524938

ABSTRACT

Brain edema is an important factor leading to morbidity and mortality associated with primary brain tumors. Dexamethasone, a synthetic glucocorticoid, is routinely prescribed with antineoplastic agents to alleviate pain associated with chemotherapy and reduce intracranial pressure. We investigated whether dexamethasone treatment increased the expression and activity of multidrug resistance (MDR) transporters at the blood-brain barrier. Treatment of primary rat brain microvascular endothelial cells with submicromolar concentrations of dexamethasone induced significantly higher levels of drug efflux transporters such as breast cancer resistance protein (abcg2), P-glycoprotein (P-gp; abcb1a/abcb1b), and MDR protein 2 (Mrp2; abcc2) as indicted by protein and mRNA levels as well as by functional activity. The effect of dexamethasone on transporter function was significant within 6 h of treatment, was dose dependent, and was reversible. Dexamethasone-induced upregulation of Bcrp and P-gp expression and function was partially abrogated by the glucocorticoid receptor (GR) antagonist RU486. In contrast, RU486 had no effect on the dexamethasone-induced upregulation of Mrp2, suggesting a GR-independent regulation of Mrp2, and a GR-dependent regulation of P-gp and Bcrp. In addition to the dexamethasone-induced upregulation of MDR transporters, we measured a dose-dependent and reversible increase in the expression of the nuclear transcription factor pregnane xenobiotic receptor (PXR). Administering dexamethasone to rats caused increased expression of PXR in brain microvessels within 24 h. These results suggest that adjuvant therapy with corticosteroids such as dexamethasone in the treatment of brain tumors may increase the expression of MDR transporters at the blood-brain barrier through pathways involving GR and PXR.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Blood-Brain Barrier/metabolism , Dexamethasone/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Blotting, Western , Brain/drug effects , Brain/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression/drug effects , Glucocorticoids/pharmacology , Indenes/pharmacology , Male , Mifepristone/pharmacology , Multidrug Resistance-Associated Proteins/genetics , Pregnane X Receptor , Pregnenolone Carbonitrile/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
9.
J Pharm Sci ; 97(11): 5013-23, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18240294

ABSTRACT

A modified surgical procedure is described to implant a microdialysis probe to sample ventricular cerebrospinal fluid (vCSF) in FVB mice. Microdialysis sampling of drugs in vCSF provides insight into drug penetration into the brain across the blood brain barrier (BBB) and the blood CSF barrier (BCB); however, this method has been reported primarily in larger animal species. Implanting a microdialysis probe in the lateral ventricle of a mouse is technically very challenging. The modification consisted of changes in the stereotaxic coordinates and insertion of the cannula and ultimately the probe at a 20 degrees angle. Exact placement of the probe was confirmed using ultrasound (US), micro-computed tomography (CT), and histologic review of serial paraffin sections. Additionally, studies of topotecan CSF penetration in the FVB mouse were conducted. With this modified procedure, the ventricular CSF to plasma AUC ratio of unbound topotecan lactone was greater than that previously reported using conventional methods. We speculate this is due to changes incorporated by the modified procedure that places the probe directly into the lateral ventricle allowing sampling of that discrete compartment. Thus, we propose that this modified procedure for placement of the microdialysis probe is superior to the conventional perpendicular method previously reported.


Subject(s)
Cerebral Ventricles/surgery , Microdialysis , Animals , Antineoplastic Agents/cerebrospinal fluid , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Female , Mice , Topotecan/cerebrospinal fluid , Topotecan/pharmacokinetics
10.
Clin Cancer Res ; 13(13): 3942-50, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17606728

ABSTRACT

PURPOSE: Dysfunctional tumor vessels can be a significant barrier to effective cancer therapy. However, increasing evidence suggests that vascular endothelial growth factor (VEGF) inhibition can effect transient "normalization" of the tumor vasculature, thereby improving tumor perfusion and, consequently, delivery of systemic chemotherapy. We sought to examine temporal changes in tumor vascular function in response to the anti-VEGF antibody, bevacizumab. EXPERIMENTAL DESIGN: Established orthotopic neuroblastoma xenografts treated with bevacizumab were evaluated at serial time points for treatment-associated changes in intratumoral vascular physiology, penetration of systemically administered chemotherapy, and efficacy of combination therapy. RESULTS: After a single bevacizumab dose, a progressive decrease in tumor microvessel density to <30% of control was observed within 7 days. Assessment of the tumor microenvironment revealed a rapid, sustained decrease in both tumor vessel permeability and tumor interstitial fluid pressure, whereas intratumoral perfusion, as assessed by contrast-enhanced ultrasonography, was improved, although this latter change abated by 1 week. Intratumoral drug delivery mirrored these changes; penetration of chemotherapy was improved by as much as 81% when given 1 to 3 days after bevacizumab, compared with when both drugs were given concomitantly, or 7 days apart. Finally, administering topotecan to tumor-bearing mice 3 days after bevacizumab resulted in greater tumor growth inhibition (36% of control size) than with monotherapy (88% bevacizumab, 54% topotecan) or concomitant administration of the two drugs (44%). CONCLUSIONS: Bevacizumab-mediated VEGF blockade effects alterations in tumor vessel physiology that allow improved delivery and efficacy of chemotherapy, although careful consideration of drug scheduling is required to optimize antitumor activity.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Neuroblastoma/drug therapy , Animals , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Line, Tumor , Humans , Male , Mice , Microcirculation , Neoplasm Transplantation , Neovascularization, Pathologic , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phenotype , Pressure , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism
11.
J Clin Oncol ; 25(18): 2594-600, 2007 Jun 20.
Article in English | MEDLINE | ID: mdl-17577039

ABSTRACT

PURPOSE: To study the association between UDP-glucuronosyltransferase 1A1 (UGT1A1) genotypes and severe toxicity as well as irinotecan disposition in pediatric patients with solid tumors receiving low-dose, protracted irinotecan (15 to 75 mg/m2 daily for 5 days for 2 consecutive weeks). PATIENTS AND METHODS: Seventy-four patients on five institutional clinical trials received irinotecan (15 to 75 mg/m2) daily intravenously or orally for 5 days for 2 consecutive weeks. Genomic DNA was genotyped for UGT1A1*28, and patients were designated as 6/6, 6/7, or 7/7 depending on the number of TA repeats in the UGT1A1 promoter region. Patients were evaluated for gastrointestinal and hematologic toxicity, as well as baseline and maximal serum bilirubin levels. Toxicity and pharmacokinetic results were evaluated during courses 1 and 2 of irinotecan therapy. RESULTS: The frequencies of 6/6, 6/7, and 7/7 genotypes were 27 (36.5%), 36 (48.6%), and 9 (12.2%) of 74 patients, respectively. Patients with 7/7 genotype had a statistically greater baseline total bilirubin than patients with 6/6 or 6/7 genotype (P = .005). UGT1A1*28 genotype was not associated with grade 3 and 4 neutropenia (P = .21 for course 1; P = .23 for course 2) or diarrhea (P = .176 for course 1; P = .87 for course 2). However, patients with the 7/7 genotype tended to have higher SN-38 area under the plasma time-concentration curve (AUC) values and lower SN-38G/SN-38 AUC ratios. CONCLUSION: Severe toxicity was not increased in pediatric patients with the 7/7 genotype when treated with a low-dose protracted schedule of irinotecan. Therefore, UGT1A1 genotyping is not a useful prognostic indicator of severe toxicity for patients treated with this irinotecan dosage and schedule.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Camptothecin/analogs & derivatives , Glucuronosyltransferase/genetics , Neoplasms/drug therapy , Adolescent , Adult , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Area Under Curve , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/pharmacokinetics , Chi-Square Distribution , Child , Child, Preschool , Clinical Trials as Topic , DNA, Neoplasm/analysis , Female , Genotype , Humans , Infusions, Intravenous , Irinotecan , Male , Neoplasm Recurrence, Local , Neoplasms/enzymology , Promoter Regions, Genetic/genetics , Risk Factors
12.
J Chromatogr B Analyt Technol Biomed Life Sci ; 853(1-2): 97-104, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17446145

ABSTRACT

A sensitive method for the determination of Cloretazine (VNP40101M) and its metabolite (VNP4090CE) with an internal standard (ISTD) in human plasma was developed using high-performance liquid chromatographic separation with tandem mass spectrometric detection. Acidified plasma samples (500 microL) were prepared using solid phase extraction (SPE) columns, and 25 microL of the reconstituted sample was injected onto an Ascentis C18 HPLC column (3 microm, 5 cmx2.1 mm) with an isocratic mobile phase. Analytes were detected with an API-3000 LC-MS/MS System at unit (Q1) and low (Q3) resolution in negative multiple reaction monitoring mode: m/z 249.0 (precursor ion) to m/z 114.9 (product ion) for both Cloretazine (at 3.64 min) and VNP4090CE (at 2.91 min), and m/z 253.0 (precursor ion) to m/z 116.9 (product ion) for the ISTD. The mean recovery for Cloretazine (VNP40101M) and its metabolite (VNP4090CE) was greater than 87% with a lower limit of quantification of 1.0 ng/mL for Cloretazine (S/N=9.7, CV

Subject(s)
Chromatography, Liquid/methods , Hydrazines/blood , Spectrometry, Mass, Electrospray Ionization/methods , Sulfonamides/blood , Tandem Mass Spectrometry/methods , Humans , Hydrazines/chemistry , Hydrazines/metabolism , Reproducibility of Results , Sulfonamides/chemistry , Sulfonamides/metabolism
13.
Clin Cancer Res ; 13(5): 1511-5, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332296

ABSTRACT

PURPOSE: To report cerebrospinal fluid (CSF) penetration of erlotinib and its metabolite OSI-420. EXPERIMENTAL DESIGN: Pharmacokinetic measurements were done in plasma (days 1, 2, 3, and 8 of therapy) and, concurrently, in plasma and CSF (before and at 1, 2, 4, 8, and 24 h after dose on day 34 of therapy) in an 8-year-old patient diagnosed with glioblastoma who received local irradiation and oral erlotinib in a phase I protocol. CSF samples were collected from a ventriculoperitoneal shunt, which was externalized because of infection. Erlotinib concentrations were determined by liquid chromatography/mass spectrometry. CSF penetration of erlotinib and OSI-420 were estimated by a compartmental model and by calculating the ratio of CSF to plasma 24-h area under concentration-time curve (AUC(0-24)). RESULTS: This patient was assigned to receive erlotinib at a dose level of 70 mg/m(2), but the actual daily dose was 75 mg (78 mg/m(2)). Erlotinib and OSI-420 plasma pharmacokinetic variables on days 8 and 34 overlapped to suggest that steady state had been reached. Whereas erlotinib and OSI-420 AUC(0-24) in plasma on day 34 were 30,365 and 2,527 ng h/mL, respectively, the correspondent AUC(0-24) in the CSF were 2,129 and 240 ng h/mL, respectively. Erlotinib and OSI-420 CSF penetration were 7% and approximately 9%, respectively, using both estimate methods. The maximum steady-state CSF concentration of erlotinib was approximately 130 ng/mL (325 nmol/L). CONCLUSIONS: The plasma pharmacokinetics of erlotinib in this child overlapped with results described in adults. Oral administration of erlotinib achieves CSF concentrations comparable with those active against several cancer cell lines in preclinical models.


Subject(s)
Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Quinazolines/blood , Quinazolines/cerebrospinal fluid , Area Under Curve , Child , Erlotinib Hydrochloride , Female , Humans , Quinazolines/pharmacokinetics
14.
J Pediatr Surg ; 42(1): 160-5; discussion 165, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17208558

ABSTRACT

BACKGROUND: We have recently demonstrated that continuous delivery of interferon beta (IFN-beta) stabilizes solid tumor vasculature and improves tumor perfusion. In this study, we have further investigated the functional consequences of this effect by assessing delivery and efficacy of conventional chemotherapy against neuroblastoma xenografts when used in combination with IFN-beta. METHODS: Mice with established retroperitoneal tumors received adeno-associated virus vector encoding IFN-beta (AAV IFN-beta) or control vector. One week later, at 1 hour before sacrifice, a 1 mg/kg i.v. bolus of topotecan (TPT) was given. Intratumoral levels of TPT were measured by high-performance liquid chromatography and then standardized to plasma levels to determine tumor TPT penetration. Subsequent experiments evaluated the antitumor efficacy of topotecan alone or in combination with AAV IFN-beta. RESULTS: As observed in prior experiments, AAV IFN-beta resulted in a marked increase in tumor vessel association with stabilizing perivascular smooth muscle cells. These more "matured" vessels facilitated improved tumor TPT penetration (51.2% +/- 4.2%) compared with controls (30.8% +/- 4.7%, P = .004). In additional cohorts of mice, this resulted in an improved antitumor effect. Mice with established tumors (301.8 +/- 18.1 mm3) were treated with TPT (1 mg/kg daily for 5 days for 2 consecutive weeks) either alone or in combination with AAV IFN-beta (5 x 10(10) vector particles per mouse). Topotecan monotherapy resulted in a reduction in mean tumor volume of 12% (264.2 +/- 65.8 mm3, P = .66). However, when the same regimen was administered to mice receiving continuous IFN-beta therapy, a 61% (118.9 +/- 42.3 mm3, P = .004) reduction in mean tumor volume was achieved. CONCLUSION: Interferon beta-mediated vessel stabilization resulted in improved intratumoral delivery of systemically administered TPT, enhancing its antitumor efficacy. This approach of altering the tumor vasculature provides a strategy to help overcome solid tumor resistance to traditional cytotoxic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Vessels/drug effects , Interferon-beta/pharmacology , Neuroblastoma/metabolism , Topotecan/pharmacology , Adenoviridae , Animals , Disease Models, Animal , Genetic Vectors , Mice , Neoplasm Transplantation
15.
Cancer Res ; 66(23): 11305-13, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17145877

ABSTRACT

A potential strategy to increase the efficacy of topotecan to treat central nervous system (CNS) malignancies is modulation of the activity of ATP-binding cassette (ABC) transporters at the blood-brain and blood-cerebrospinal fluid barriers to enhance topotecan CNS penetration. This study focused on topotecan penetration into the brain extracellular fluid (ECF) and ventricular cerebrospinal fluid (CSF) in a mouse model and the effect of modulation of ABC transporters at the blood-brain and blood-cerebrospinal fluid barriers by a tyrosine kinase inhibitor (gefitinib). After 4 and 8 mg/kg topotecan i.v., the brain ECF to plasma AUC ratio of unbound topotecan lactone was 0.21 +/- 0.04 and 0.61 +/- 0.16, respectively; the ventricular CSF to plasma AUC ratio was 1.18 +/- 0.10 and 1.30 +/- 0.13, respectively. To study the effect of gefitinib on topotecan CNS penetration, 200 mg/kg gefitinib was administered orally 1 hour before 4 mg/kg topotecan i.v. The brain ECF to plasma AUC ratio of unbound topotecan lactone increased by 1.6-fold to 0.35 +/- 0.04, which was significantly different from the ratio without gefitinib (P < 0.05). The ventricular CSF to plasma AUC ratio significantly decreased to 0.98 +/- 0.05 (P < 0.05). Breast cancer resistance protein 1 (Bcrp1), an efficient topotecan transporter, was detected at the apical aspect of the choroid plexus in FVB mice. In conclusion, topotecan brain ECF penetration was lower compared with ventricular CSF penetration. Gefitinib increased topotecan brain ECF penetration but decreased the ventricular CSF penetration. These results are consistent with the possibility that expression of Bcrp1 and P-glycoprotein at the apical side of the choroid plexus facilitates an influx transport mechanism across the blood-cerebrospinal fluid barrier, resulting in high topotecan CSF penetration.


Subject(s)
Central Nervous System/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Topotecan/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Administration, Oral , Algorithms , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Blood Proteins/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Dose-Response Relationship, Drug , Extracellular Fluid/metabolism , Female , Gefitinib , Immunohistochemistry , Injections, Intravenous , Mice , Mice, Inbred C57BL , Microdialysis , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , Time Factors , Topotecan/cerebrospinal fluid , Topotecan/metabolism
16.
Invest Ophthalmol Vis Sci ; 47(4): 1269-73, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16565357

ABSTRACT

PURPOSE: To compare the expression of multidrug-resistant proteins in retinoblastoma tumors among eyes treated with primary enucleation. METHODS: A group of 18 patients with unilateral retinoblastoma with advanced intraocular disease was selected for the study. All patients had undergone primary enucleation. A histologic specimen from each patient was retrieved from the pathology archives and a tissue gene microarray was constructed (0.6 x 3-4 mm). Standard immunohistochemical techniques were used to study the tissue microarrays for the expression of the ATP-binding cassette (ABC) transporters: breast cancer resistance protein (BCRP; ABCG2), multidrug-resistant protein 1/P-glycoprotein (MDR1/Pgp; ABCB1), multidrug-resistant-associated protein 1 (MRP1; ABCC1), MRP2 (ABCC2), and MRP4 (ABCC4). RESULTS: Of the 18 specimens retrieved, 16 had adequate tissue for study. MRP1 was expressed in 8 (50%) of 16 tumors, and MRP2 was expressed in 5 (31%) of 16 tumors. MDR1/Pgp was found in 2 (12%) of 16 retinoblastomas. MRP4 and BCRP were not detected in any of the tumors studied. CONCLUSIONS: The results show that multiple ABC transporters were present in a cohort of sporadic patients with unilateral retinoblastoma who underwent primary enucleation. Studies are planned of the expression of ABC transporters in eyes treated by chemotherapy and/or radiation as a comparison with this group.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Eye Enucleation , Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , Humans , Immunoenzyme Techniques , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Retinal Neoplasms/surgery , Retinoblastoma/surgery
17.
J Chromatogr B Analyt Technol Biomed Life Sci ; 831(1-2): 169-75, 2006 Feb 02.
Article in English | MEDLINE | ID: mdl-16364699

ABSTRACT

A sensitive method for the determination of lapatinib (GW572016) in human plasma was developed using high-performance liquid chromatographic separation with tandem mass spectrometric detection. Plasma samples (100 microL) were prepared using solid phase extraction (SPE) columns, and 6.0 microL of the reconstituted eluate was injected onto a Phenomenex CuroSil-PFP 3 mu analytical column (50 mm x 2.0mm) with an isocratic mobile phase. Analytes were detected with a PE SCIEX API-365 LC-MS/MS system at unit (Q1) and low (Q3) resolution in positive multiple reaction monitoring mode (m/z 581 (precursor ion) to m/z 364 (product ion) for lapatinib). The mean recovery for lapatinib was 75% with a lower limit of quantification of 15 ng/mL (S/N=11.3, CV< or =14%). This method was validated over a linear range of 100-10,000 ng/mL, and results from a 5-day validation study demonstrated good within-day and between-day precision and accuracy. This method has been used to measure plasma lapatinib concentrations in a Phase I study in children with cancer.


Subject(s)
Chromatography, High Pressure Liquid/methods , Quinazolines/blood , Spectrometry, Mass, Electrospray Ionization/methods , Child , Clinical Trials, Phase I as Topic , Drug Stability , Humans , Lapatinib , Quinazolines/pharmacokinetics
18.
J Mol Biol ; 352(1): 165-77, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16081098

ABSTRACT

Human carboxylesterase 1 (hCE1) exhibits broad substrate specificity and is involved in xenobiotic processing and endobiotic metabolism. We present and analyze crystal structures of hCE1 in complexes with the cholesterol-lowering drug mevastatin, the breast cancer drug tamoxifen, the fatty acyl ethyl ester (FAEE) analogue ethyl acetate, and the novel hCE1 inhibitor benzil. We find that mevastatin does not appear to be a substrate for hCE1, and instead acts as a partially non-competitive inhibitor of the enzyme. Similarly, we show that tamoxifen is a low micromolar, partially non-competitive inhibitor of hCE1. Further, we describe the structural basis for the inhibition of hCE1 by the nanomolar-affinity dione benzil, which acts by forming both covalent and non-covalent complexes with the enzyme. Our results provide detailed insights into the catalytic and non-catalytic processing of small molecules by hCE1, and suggest that the efficacy of clinical drugs may be modulated by targeted hCE1 inhibitors.


Subject(s)
Anticholesteremic Agents/metabolism , Antineoplastic Agents, Hormonal/metabolism , Carboxylic Ester Hydrolases , Lovastatin/analogs & derivatives , Phenylglyoxal/analogs & derivatives , Protein Structure, Quaternary , Tamoxifen/metabolism , Acetates/chemistry , Acetates/metabolism , Anticholesteremic Agents/chemistry , Antineoplastic Agents, Hormonal/chemistry , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/metabolism , Crystallography, X-Ray , Humans , Lovastatin/chemistry , Lovastatin/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Phenylglyoxal/chemistry , Phenylglyoxal/metabolism , Substrate Specificity , Tamoxifen/chemistry
20.
J Heart Lung Transplant ; 23(11): 1290-6, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15539128

ABSTRACT

BACKGROUND: The activation of poly (adenosine diphosphate) ribose synthetase (PARS) is known to be important in the cellular response to oxidative stress. Previous studies have reported that PARS inhibition confers protection in models of endotoxic shock and ischemia-reperfusion. The purpose of this study was to determine the role of PARS inhibition in lung ischemia-reperfusion injury (LIRI). METHODS: Left lungs of Long-Evans rats were rendered ischemic for 90 minutes and reperfused for up to 4 hours. Treated animals received 3 mg/kg of INO-1001 (a PARS inhibitor) intravenously 30 minutes before ischemia. Injury was quantitated in terms of tissue myeloperoxidase (MPO) content, vascular permeability ((125)I radiolabeled bovine serum albumin extravasation) and bronchoalveolar lavage (BAL) leukocyte content. BAL fluid was assessed for cytokine and chemokine content by enzyme-linked immunoassay. Further samples were processed for nuclear protein analysis by electromobility shift assay (EMSA) and cellular death by terminal deoxyribonucleotidyl transferase-mediated d-UTP biotin nick-end labeling (TUNEL) assay and caspase-3 staining. RESULTS: Lung vascular permeability was reduced in treated animals by 73% compared with positive controls (p < 0.009). The protective effects of PARS inhibition correlated with a 46% reduction in tissue MPO content (p < 0.008) and marked reductions in BAL leukocyte accumulation. This positively correlated with the diminished expression of pro-inflammatory mediators and nuclear transcription factors, as well as decreased levels of cellular death. CONCLUSIONS: The deleterious effects of LIRI are in part mediated by the formation of free radicals and superoxides, which lead to DNA single-strand breaks. This leads to activation of PARS, which causes rapid cellular energy depletion and cell death. PARS inhibition is protective against this and represents a potentially useful therapeutic tool in the prevention of LIRI.


Subject(s)
Indoles/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/physiology , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control , Animals , Male , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL