Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Affiliation country
Publication year range
1.
J Allergy Clin Immunol ; 137(6): 1717-1726.e13, 2016 06.
Article in English | MEDLINE | ID: mdl-26688517

ABSTRACT

BACKGROUND: Increase of bronchial smooth muscle (BSM) mass is a crucial feature of asthma remodeling. The mechanisms of such an increased BSM mass are complex but involve enhanced mitochondrial biogenesis, leading to increased proliferation of BSM cells in asthmatic patients. The major tumor suppressor protein p53 is a key cell regulator involved in cell proliferation and has also been implicated in mitochondrial biogenesis. However, the role of p53 in BSM cell proliferation and mitochondrial biogenesis has not been investigated thus far. OBJECTIVE: We sought to evaluate the role of p53 in proliferation of BSM cells in asthmatic patients and mitochondrial biogenesis. METHODS: The expression of p53 was assessed both in vitro by using flow cytometry and Western blotting and ex vivo by using RT-PCR after laser microdissection. The role of p53 was assessed with small hairpin RNA lentivirus in both asthmatic patients and control subjects with BSM cell proliferation by using 5-bromo-2'-deoxyuridine and cell counting and in the expression of p21, BCL2-associated X protein, mitochondrial transcription factor A (TFAM), and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α). RESULTS: Twenty-nine patients with moderate-to-severe asthma and 26 control subjects were enrolled in the study. p53 expression was increased in BSM from asthmatic patients both ex vivo and in vitro, with a decreased interaction with mouse double minute 2 homolog (Mdm2) and an increased phosphorylation of serine 20. p53 did not inhibit the transcription of both TFAM and PGC-1α in BSM cells from asthmatic patients. As a consequence, p53 is unable to slow the increased mitochondrial biogenesis and hence the subsequent increased proliferation of BSM cells in asthmatic patients. CONCLUSION: This study suggests that p53 might act as a new potential therapeutic target against BSM remodeling in asthmatic patients.


Subject(s)
Asthma/metabolism , Bronchi/metabolism , Muscle, Smooth/metabolism , Organelle Biogenesis , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Anti-Asthmatic Agents/therapeutic use , Asthma/diagnosis , Asthma/drug therapy , Case-Control Studies , Cell Proliferation , Female , Gene Expression , Humans , Male , Middle Aged , Respiratory Function Tests , Risk Factors , Tumor Suppressor Protein p53/genetics
2.
Onkologie ; 36(11): 670-3, 2013.
Article in English | MEDLINE | ID: mdl-24192772

ABSTRACT

BACKGROUND: Treatment of osteosarcoma of the extremities consists of surgical resection preceded and followed by chemotherapy, including high-dose methotrexate or adriamycin-based protocols. When distant relapse occurs, therapeutic options are scarce. Trabectedin, a DNA-binding agent, is indicated for the treatment of patients with advanced soft tissue sarcomas after failure of anthracyclines and ifosfamide. In this indication, the 6-month progression-free survival is about 35-40%. Recent reports showed that some specific single nucleotide polymorphisms (SNPs) from DNA repair genes could be associated with sensitivity to trabectedin in soft tissue sarcomas. CASE REPORTS: We report our experience of 2 metastatic, heavily pre-treated osteosarcoma patients who were treated with trabectedin. Pyrosequencing analyses of tumors from both patients for several SNPs of the ERCC1, ERCC5 and BRAC1 genes were performed. Both patients showed major response to trabectedin, which was interestingly related with homozygoty of the common guanine allele of ERCC5 (G/G genotype; Asp/Asp) after pyrosenquencing analysis of tumors from both patients. This polymorphism was previously shown to be associated with better outcome in soft tissue sarcoma patients treated with trabectedin. CONCLUSION: Homozygoty for the wild-type Asp1104 SNP of the ERCC5 gene was found in 2 cases of relapsed osteosarcoma, who responded to trabectedin.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , DNA-Binding Proteins/genetics , Dioxoles/therapeutic use , Endonucleases/genetics , Nuclear Proteins/genetics , Osteosarcoma/drug therapy , Osteosarcoma/secondary , Tetrahydroisoquinolines/therapeutic use , Transcription Factors/genetics , Adolescent , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Female , Genetic Markers/genetics , Humans , Male , Osteosarcoma/genetics , Polymorphism, Single Nucleotide/genetics , Trabectedin , Treatment Outcome , Young Adult
3.
Carcinogenesis ; 33(5): 962-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22345163

ABSTRACT

ERCC2 [Xeroderma pigmentosum (XP) group D] belongs to the nucleotide excision repair pathway. It is also part of the TFIIH transcription complex and is required for the association of the cyclin-dependent kinase (CDK)-activating kinase (CAK) subcomplex with TFIIH. Using the NCI-60 panel of human tumor cell lines, we had shown that the ERCC2 gene variant Gln(751) was significantly associated to increased taxanes sensitivity and decreased ERCC2 gene expression. Since TFIIH is involved in both DNA repair and cell cycle progression, we hypothesized that quantitative or qualitative ERCC2 alterations might cause CAK liberation, allowing its activation of the G(2)/M transition. Enhancing mitosis entry would lead to hypersensitivity to spindle poisons, explaining the effect of ERCC2 polymorphisms on taxane sensitivity. Starting from ERCC2-deficient XP6BE, we generated several isogenic clones differing only by the Lys751Gln variation. Wild-type and variant ERCC2-expressing clones recovered ultraviolet radiation and cisplatin resistance but presented similar sensitivity to paclitaxel, demonstrating that the amino acid change was not involved in paclitaxel differential sensitivity in the NCI-60 panel. Using small interfering RNA approach, we knocked down ERCC2 expression and observed a block in the G(2)/M phase, with a consistent increase in paclitaxel sensitivity and no change in cisplatin sensitivity. We observed in addition an increase in CDK1 activity, as evaluated by histone H1 phosphorylation. We evaluated messenger RNA (mRNA) half-life in the isogenic lines and observed a more rapid degradation in cells bearing the variant construct. We concluded that the increased paclitaxel sensitivity of ERCC2 variant cell lines is a consequence of lower gene expression, likely due to decreased stability of the variant ERCC2 mRNA.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Xeroderma Pigmentosum Group D Protein/genetics , Adult , Cell Division/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Cyclin-Dependent Kinases/genetics , DNA Repair , DNA-Binding Proteins/genetics , Drug Screening Assays, Antitumor/methods , Female , Fibroblasts/metabolism , G2 Phase/genetics , Gene Expression , Histones/metabolism , Humans , Paclitaxel/pharmacology , Phosphorylation , Polymorphism, Genetic , RNA, Messenger/genetics , Taxoids/pharmacology , Transcription Factor TFIIH/genetics , Young Adult , Cyclin-Dependent Kinase-Activating Kinase
4.
Bull Cancer ; 109(1): 58-64, 2022 Jan.
Article in French | MEDLINE | ID: mdl-34903368

ABSTRACT

Three-dimensional (3D) culture of organoids from primary cells (wild type) or tumoroids from tumor cells, is used to study the physiological mechanisms in vivo, in order to model normal or tumor tissues more accurately than conventional two-dimensional (2D) culture. The features of this 3D culture, such as the three-dimensional structure, the self-renewal capacity and differentiation are preserved and appropriate to cancer study since their cellular characteristics are very similar to in vivo models. Here, we summarize the recent advances in the rapidly evolving field of organoids and their applications to cancer biology, clinical research and personalized medicine.


Subject(s)
Biomedical Research , Cell Culture Techniques, Three Dimensional/methods , Neoplasms/pathology , Organoids/pathology , Bronchi/anatomy & histology , Carcinogenesis/genetics , Cell Differentiation , Cell Self Renewal , Humans , Lung Neoplasms/pathology , Neoplasms/genetics , Precision Medicine , Tumor Cells, Cultured/pathology
5.
Blood Adv ; 5(23): 5372-5386, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34555842

ABSTRACT

Non-Hodgkin B-cell lymphomas (B-NHL) mainly develop within lymph nodes as aggregates of tumor cells densely packed with their surrounding microenvironment, creating a tumor niche specific to each lymphoma subtypes. In vitro preclinical models mimicking biomechanical forces, cellular microenvironment, and 3D organization of B-cell lymphomas remain scarce, while all these parameters are key determinants of lymphomagenesis and drug resistance. Using a microfluidic method based on cell encapsulation inside permeable, elastic, and hollow alginate microspheres, we developed a new tunable 3D model incorporating lymphoma B cells, extracellular matrix (ECM), and/or tonsil stromal cells (TSC). Under 3D confinement, lymphoma B cells were able to form cohesive spheroids resulting from overexpression of ECM components. Moreover, lymphoma B cells and TSC dynamically formed self-organized 3D spheroids favoring tumor cell growth. 3D culture induced resistance to the classical chemotherapeutic agent doxorubicin, but not to the BCL2 inhibitor ABT-199, identifying this approach as a relevant in vitro model to assess the activity of therapeutic agents in B-NHL. RNA-sequence analysis highlighted the synergy of 3D, ECM, and TSC in upregulating similar pathways in malignant B cells in vitro than those overexpressed in primary lymphoma B cells in situ. Finally, our 3D model including ECM and TSC allowed long-term in vitro survival of primary follicular lymphoma B cells. In conclusion, we propose a new high-throughput 3D model mimicking lymphoma tumor niche and making it possible to study the dynamic relationship between lymphoma B cells and their microenvironment and to screen new anti-cancer drugs.


Subject(s)
Antineoplastic Agents , Lymphoma, B-Cell , Lymphoma, Non-Hodgkin , B-Lymphocytes , Cell Proliferation , Humans , Lymphoma, B-Cell/drug therapy , Tumor Microenvironment
6.
Clin Pharmacol Ther ; 109(5): 1244-1255, 2021 05.
Article in English | MEDLINE | ID: mdl-33047329

ABSTRACT

Variations in clinical response to tamoxifen (TAM) may be related to polymorphic cytochromes P450 (CYPs) involved in forming its active metabolite endoxifen (ENDO). We developed a population pharmacokinetic (PopPK) model for tamoxifen and six metabolites to determine clinically relevant factors of ENDO exposure. Concentration-time data for TAM and 6 metabolites come from a prospective, multicenter, 3-year follow-up study of adjuvant TAM (20 mg/day) in patients with breast cancer, with plasma samples drawn every 6 months, and genotypes for 63 genetic polymorphisms (PHACS study, NCT01127295). Concentration data for TAM and 6 metabolites from 928 patients (n = 27,433 concentrations) were analyzed simultaneously with a 7-compartment PopPK model. CYP2D6 phenotype (poor metabolizer (PM), intermediate metabolizer (IM), normal metabolizer (NM), and ultra-rapid metabolizer (UM)), CYP3A4*22, CYP2C19*2, and CYP2B6*6 genotypes, concomitant CYP2D6 inhibitors, age, and body weight had a significant impact on TAM metabolism. Formation of ENDO from N-desmethyltamoxifen was decreased by 84% (relative standard error (RSE) = 14%) in PM patients and by 47% (RSE = 9%) in IM patients and increased in UM patients by 27% (RSE = 12%) compared with NM patients. Dose-adjustment simulations support an increase from 20 mg/day to 40 and 80 mg/day in IM patients and PM patients, respectively, to reach ENDO levels similar to those in NM patients. However, when considering Antiestrogenic Activity Score (AAS), a dose increase to 60 mg/day in PM patients seems sufficient. This PopPK model can be used as a tool to predict ENDO levels or AAS according to the patient's CYP2D6 phenotype for TAM dose adaptation.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cytochrome P-450 CYP2D6/genetics , Polymorphism, Genetic , Tamoxifen/pharmacokinetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/pharmacokinetics , Female , Humans , Longitudinal Studies , Middle Aged , Models, Biological , Pharmacogenomic Variants , Tamoxifen/administration & dosage , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism
7.
Cancers (Basel) ; 11(3)2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30832225

ABSTRACT

GA101/obinutuzumab is a novel type II anti-CD20 monoclonal antibody (mAb), which is more effective than rituximab (RTX) in preclinical and clinical studies when used in combination with chemotherapy. Ca2+ signaling was shown to play a role in RTX-induced cell death. This report concerns the effect of GA101 on Ca2+ signaling and its involvement in the direct cell death induced by GA101. We reveal that GA101 triggered an intracellular Ca2+ increase by mobilizing intracellular Ca2+ stores and activating Orai1-dependent Ca2+ influx in non-Hodgkin lymphoma cell lines and primary B-Cell Chronic Lymphocytic Leukemia (B-CLL) cells. According to the cell type, Ca2+ was mobilized from two distinct intracellular compartments. In Raji, BL2, and B-CLL cells, GA101 induced a Ca2+ release from lysosomes, leading to the subsequent lysosomal membrane permeabilization and cell death. Inhibition of this calcium signaling reduced GA101-induced cell death in these cells. In SU-DHL-4 cells, GA101 mobilized Ca2+ from the endoplasmic reticulum (ER). Inhibition of ER replenishment, by blocking Orai1-dependent Ca2+ influx, led to an ER stress and unfolded protein response (UPR) which sensitized these cells to GA101-induced cell death. These results revealed the central role of Ca2+ signaling in GA101's action mechanism, which may contribute to designing new rational drug combinations improving its clinical efficacy.

8.
Clin Pharmacol Ther ; 106(3): 585-595, 2019 09.
Article in English | MEDLINE | ID: mdl-30786012

ABSTRACT

In addition to the effect of cytochrome P450 (CYP) 2D6 genetic polymorphisms, the metabolism of tamoxifen may be impacted by other factors with possible consequences on therapeutic outcome (efficacy and toxicity). This analysis focused on the pharmacokinetic (PK)-pharmacogenetic evaluation of tamoxifen in 730 patients with adjuvant breast cancer included in a prospective multicenter study. Plasma concentrations of tamoxifen and six major metabolites, the genotype for 63 single-nucleotide polymorphisms, and comedications were obtained 6 months after treatment initiation. Plasma concentrations of endoxifen were significantly associated with CYP2D6 diplotype (P < 0.0001), CYP3A4*22 genotype (P = 0.0003), and concomitant intake of potent CYP2D6 inhibitors (P < 0.001). Comparison of endoxifen levels showed that the CYP2D6 phenotype classification could be improved by grouping intermediate metabolizer (IM)/IM and IM/poor metabolizer diplotype into IM phenotype for future use in tamoxifen therapy optimization. Finally, the multivariable regression analysis showed that formation of tamoxifen metabolites was independently impacted by CYP2D6 diplotype and CYP3A4*22, CYP2C19*2, and CYP2B6*6 genetic polymorphisms.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/pharmacokinetics , Breast Neoplasms/drug therapy , Cytochrome P-450 Enzyme System/genetics , Tamoxifen/administration & dosage , Tamoxifen/pharmacokinetics , Adult , Aged , Antineoplastic Agents, Hormonal/therapeutic use , Cytochrome P-450 CYP2B6/genetics , Cytochrome P-450 CYP2C19 Inhibitors , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP3A/genetics , Female , Genotype , Humans , Middle Aged , Pharmacogenetics , Polymorphism, Single Nucleotide , Prospective Studies , Tamoxifen/analogs & derivatives , Tamoxifen/blood , Tamoxifen/therapeutic use
9.
Cancer Chemother Pharmacol ; 61(3): 481-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17483951

ABSTRACT

PURPOSE: Carboxylesterase 2 (CES2) is involved in the activation of the anticancer drug irinotecan to its active metabolite SN-38. We previously identified a single nucleotide polymorphism (SNP), with an allele frequency around 10%, as possibly involved in enzyme expression (Clin Pharmacol Ther 76:528-535, 2004), which could explain the large individual variation in SN-38 disposition. METHODS: The 830C>G SNP, located in the 5' untranslated region of the gene, was analysed in various DNA samples extracted from: (1) the National Cancer Institute NCI-60 panel of human tumour cell lines; (2) a collection of 104 samples of normal tissue from colorectal cancer patients; (3) blood samples from a population of 95 normal subjects; (4) a collection of 285 human livers. CES2 genotypes were tentatively related to irinotecan cytotoxicity and CES2 expression in the NCI-60 panel; to response to treatment and event-free survival in colorectal cancer patients; and to CES2 expression and catalytic activity in subsets of the human liver collection. RESULTS: No significant relationship was found in the NCI-60 panel between CES2 830C>G genotype and irinotecan cytotoxicity or CES2 expression. No significant relationship was found between CES2 830C>G genotype and the toxicity and therapeutic efficacy (tumour response, event-free survival) of irinotecan in colorectal cancer patients. There was no significant relationship between CES2 830C>G genotype and CES2 expression and catalytic activity determined in a subset of genotype-selected liver samples. CONCLUSION: The 830C>G SNP of CES2 is unlikely to have significant functional consequences on CES2 expression, activity or function.


Subject(s)
Carboxylesterase/genetics , Polymorphism, Single Nucleotide/genetics , Alleles , Amino Acid Substitution , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA/genetics , Genotype , Humans , Irinotecan , Liver/chemistry , Paraffin Embedding , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
10.
Eur J Cancer ; 43(5): 955-62, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17317154

ABSTRACT

A significant association has been established, in clinical studies, between the expression or activity of thymidylate synthase (TYMS) and the efficiency of fluorouracil. TYMS expression is partly under the dependence of gene polymorphisms in the 5' and 3' untranslated regions (UTR), but conflicting results have been obtained about their roles on fluorouracil efficiency. In this study, we wanted to use the National Cancer Institute (NCI) panel of 60 human tumour cell lines to clarify this problem. Three relevant polymorphisms of the TYMS gene were studied: (i) the 5'UTR tandem repeat of 28-bp (2R/3R polymorphism); (ii) the single nucleotide polymorphism (SNP) within the second repeat (3C/3G polymorphism); (iii) the 3'UTR 6-bp deletion (+6/-6 polymorphism). Allele frequencies were close to those expected in a Caucasian population (2R/3C/3G: 53/29/18%; +6/-6: 68/32%), but the proportion of heterozygous genotypes was lower than expected from allele frequencies. The 2R and 3G alleles were significantly associated with the +6 and the -6 alleles, respectively. There was a significant association between the presence of the 3G allele and TYMS mRNA expression and catalytic activity, particularly in p53-mutated cell lines. However, no significant correlation existed between fluorouracil cytotoxicity, as extracted from the NCI databases, and TYMS expression, activity or polymorphisms.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Drug Resistance, Neoplasm/genetics , Fluorouracil/therapeutic use , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Polymorphism, Genetic/genetics , Thymidylate Synthase/genetics , Cell Line, Tumor , Gene Expression , Gene Frequency , Humans , Neoplasms/drug therapy , Neoplasms/genetics
11.
Pharmacogenomics ; 8(12): 1693-703, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18085999

ABSTRACT

INTRODUCTION: Glutathione S-transferase P1 (GSTP1) and excision-repair cross-complementing repair deficiency group 2 protein (ERCC2 or XPD) may modulate the activity of platinum derivatives. The SNPs, Ile105Val for GSTP1 and Lys751Gln for ERCC2, may affect the efficiency of oxaliplatin in patients treated with an oxaliplatin-based regimen for metastatic colorectal carcinoma. PATIENTS & METHODS: A total of 107 patients treated with first-line chemotherapy, 59 with an oxaliplatin-based regimen and 48 with an irinotecan-based regimen, were included retrospectively. GSTP1 and ERCC2 genotypes were identified on DNA samples extracted from paraffin blocks containing either normal tissue (nodes) or tumor tissue. We analyzed treatment response, event-free and overall survival. RESULTS: GSTP1 genotype distribution was Ile/Ile 58%, Ile/Val 35% and Val/Val 7%. ERCC2 genotype distribution was Lys/Lys 49%, Lys/Gln 44%, Gln/Gln 7%. Event-free and overall survivals were not significantly different as a function of the GSTP1 genotype, whatever the treatment received. Event-free survival was significantly different as a function of the ERCC2 genotype only in patients receiving oxaliplatin: patients having at least one variant allele had a shorter median event-free survival (6 months) than those having no variant allele (11.6 months, p = 0.008). This difference was maintained for median overall survival (15.6 vs 25.3 months, p = 0.016). Using univariate analysis, ERCC2 genotype, hemoglobinemia and carbohydrate antigen 19.9 plasma levels were significantly related to overall and event-free survival in patients receiving oxaliplatin. CONCLUSION: The ERCC2 genotype appears as an important predictive factor of the survival of patients treated with oxaliplatin in first-line therapy for metastatic colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Glutathione S-Transferase pi/genetics , Xeroderma Pigmentosum Group D Protein/genetics , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Female , Genotype , Humans , Male , Middle Aged , Treatment Outcome
12.
Oncol Rep ; 17(5): 1213-21, 2007 May.
Article in English | MEDLINE | ID: mdl-17390068

ABSTRACT

The objective was to relate the gene expression profiles of colorectal cancer cells in culture to the in vitro cytotoxicity of cisplatin and oxaliplatin. We studied the gene expression profiles of six human colorectal cancer cell lines, using the Atlas Plastic Human 8K Microarray from Clontech, and related it to the in vitro cytotoxicities of oxaliplatin and cisplatin obtained by inhibition of exponential growth of cells. We calculated the Pearson's coefficients of correlation (r) between gene expression and drug IC50. A functional analysis was performed using the Gene Ontology Consortium database. Results were validated on a series of representative genes by real-time quantitative PCR. Validation of the significance of the coefficients of correlation was also performed using a leave-one-out analysis. We identified 394 genes whose expression was significantly correlated (P<0.05) to oxaliplatin cytotoxicity and 40 with cisplatin cytotoxicity. Three major functions were preferentially involved in oxaliplatin activity: protein synthesis, cell energetics and response to oxidative stress. No significant correlation was observed between oxaliplatin or cisplatin cytotoxicity and the expression of genes involved in DNA repair, cell proliferation or cell adhesion. A strongly significant correlation was found between the microarray and the rt-PCR approaches (r=0.968, P<10(-6)). The leave-one-out analysis showed that the same functions still appeared significantly involved in the activity of both drugs. Based on the functional analysis, we hypothesized that oxaliplatin would specifically form protein adducts during synthesis, thus exposing their thiol groups, which are known to be especially vulnerable to reactive oxygen species.


Subject(s)
Cisplatin/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Gene Expression Profiling , HCT116 Cells , HT29 Cells , Humans , Oligonucleotide Array Sequence Analysis , Oxaliplatin
13.
Pharmacogenomics ; 7(6): 843-52, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16981845

ABSTRACT

INTRODUCTION: The National Cancer Institute (NCI)-60 panel consists of 60 human tumor cell lines initially established for screening thousands of molecules for antiproliferative activity. It has been powerful for deciphering the relationships between anticancer drug cytotoxicity and cell molecular characteristics. We tested its potential interest for establishing relationships between the polymorphism of genes involved in drug metabolism and transport or in DNA repair, and drug cytotoxicity extracted from NCI databases. METHODS: Using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) techniques, three frequent single nucleotide polymorphisms (SNPs) were analyzed: Lys751Gln in the Xeroderma pigmentosum complementation group D (XPD, ERCC2) gene, Asp1104His in the Xeroderma pigmentosum complementation group G (XPG, ERCC5) gene and Ile105Val in the glutathione S-transferase P1 (GSTP1) gene. RESULTS: The allelic frequencies of the variants were 33% for ERCC2, 23% for ERCC5 and 39% for GSTP1. The ERCC2 polymorphism appeared to be a strong determinant of the in vitro cytotoxicity of most anticancer agents, with lower half maximal inhibitory concentration (IC50) values in variant homozygous lines than in common homozygous or heterozygous cell lines. Unexpectedly, the cytotoxicity of taxanes appeared markedly dependent upon the ERCC2 genotype, with threefold lower mean IC50 values in variant homozygous cell lines. The ERCC5 genotype appeared to be important only for taxanes, with fourfold higher IC50 values in variant homozygous cell lines. The GSTP1 polymorphism was related to the cytotoxicity of several drug classes, especially topoisomerase inhibitors, antimetabolites and N7 alkylating agents. CONCLUSION: The NCI-60 panel is capable of providing clues and tracks for the establishment of clinically useful relationships between a given genotype and the cytotoxicity of an anticancer agent.


Subject(s)
Antineoplastic Agents/toxicity , Polymorphism, Single Nucleotide , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Base Sequence , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , DNA Primers/genetics , DNA-Binding Proteins/genetics , Drug Screening Assays, Antitumor , Endonucleases/genetics , Female , Glutathione S-Transferase pi/genetics , Humans , Male , National Institutes of Health (U.S.) , Nuclear Proteins/genetics , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Toxicogenetics , Transcription Factors/genetics , United States , Xeroderma Pigmentosum Group D Protein/genetics
14.
Fundam Clin Pharmacol ; 29(3): 219-37, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25817555

ABSTRACT

Irinotecan is a major drug in the treatment of advanced colorectal cancer. Its active form is the SN38 metabolite, which is cleared by the biliary route after glucuronidation by uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). UGT1A1 activity exhibits a wide intersubject variability, in part related to UGT1A1 gene polymorphisms. The present review on the impact of the deficient UGT1A1*28 variant on irinotecan efficacy and toxicity was produced by a French joint workgroup comprising the Group of Clinical Onco-pharmacology (GPCO-Unicancer) and the National Pharmacogenetics Network (RNPGx). It clearly emerges that for irinotecan doses at least equal to 180 mg/m(2) , patients homozygous for the UGT1A1*28 allele are at increased risk of developing hematological and/or digestive toxicities. Irinotecan dose reduction is thus recommended in homozygous *28/*28 patients. In addition, this personalized medicine strategy aims to secure high-dose irinotecan administration (≥240 mg/m(2) ) that have proven to be safe in homozygous *1/*1 patients only. The clinical relevance of this test is discussed in terms of treatment efficacy improvement, as increasing the irinotecan dose appears to be safe in patients not bearing a deficient allele. Best execution practices, cost-effectiveness, and result interpretation are discussed with the aim of facilitating the implementation of this analysis in clinical practice. The existence of networks of laboratories performing this test in routine hospital treatment, as in France, offers the prospect of widespread screening, thus guaranteeing equal access to safe treatment and optimized therapy for patients receiving irinotecan-based therapy in advanced colorectal cancer.

15.
Lipids ; 37(8): 767-72, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12371747

ABSTRACT

In humans, diets rich in fish oil (containing n-3 FA) decrease the incidence of coronary artery diseases. This is thought to be caused by the induction in liver and skeletal muscle of genes involved in lipid oxidation, and to the repression in liver and adipose tissue of genes responsible for lipogenesis. n-3 FA are known to reduce the synthesis of FA and TG in the liver, resulting in a decrease of plasma concentrations of TG-rich lipoproteins. On the other hand, little is known of a possible effect of n-3 FA on HDL metabolism. To investigate this question, female C57Bl/6J mice were fed an n-3 FA-enriched diet for 16 wk. As expected from previous studies, we found that total cholesterol, TG, and phospholipids were reduced in the plasma of treated mice. We also found that HDL-cholesterol decreased after this treatment and that the in vivo fractional catabolic rate of HDL-cholesteryl ester was significantly higher in treated mice than in control mice fed a standard diet. Consistent with these results, treated mice exhibited increased uptake of HDL-cholesteryl ester in the liver. Moreover, quantitative reverse transcriptase-PCR analysis showed a two- to threefold increase in scavenger receptor B-1 gene expression. Taken together, these results suggest that an n-3 FA-enriched diet stimulates one step in the reverse cholesterol transport in mice, probably by increasing the amount of the scavenger receptor class B-1. These effects of n-3 FA on HDL metabolism may contribute to their beneficial effects on the vasculature.


Subject(s)
Cholesterol, HDL/metabolism , Fatty Acids, Omega-3/pharmacology , Liver/metabolism , Membrane Proteins , Receptors, Immunologic , Receptors, Lipoprotein , Animals , Blotting, Northern , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cholesterol/blood , Cholesterol Esters/metabolism , Fatty Acids, Omega-3/administration & dosage , Female , Gene Expression/drug effects , Lipoproteins, HDL/metabolism , Liver/drug effects , Mice , Mice, Inbred C57BL , Phospholipids/blood , RNA, Messenger/biosynthesis , Receptors, Scavenger , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class B , Time Factors , Triglycerides/blood , Tritium
16.
Eur J Cancer ; 50(15): 2532-43, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25103456

ABSTRACT

There are an increasing number of studies devoted to the identification of associations between anticancer drug efficacy and toxicity and common polymorphisms present in the patients' genome. However, many articles presenting the results of such studies do not bring the simple and necessary background information allowing the evaluation of the relevance of the study, its significance and its potential importance for patients' treatment. This position paper first addresses clinical oncologists with the aim of giving them the basic knowledge on pharmacogenetics and on the potential use of gene polymorphisms as predictive biomarkers in routine and clinical research. A secondary objective is to give molecular biologists some recommendations on how to conceive protocols and how to publish their results when they develop pharmacogenetic studies appended to clinical trials or with autonomous goals.


Subject(s)
Clinical Trials as Topic/methods , Neoplasms/drug therapy , Neoplasms/genetics , Polymorphism, Genetic , Clinical Trials as Topic/statistics & numerical data , Gene Frequency , Genetic Predisposition to Disease/genetics , Genotype , Humans , Pharmacogenetics/methods , Pharmacogenetics/statistics & numerical data , Treatment Outcome
17.
Med Oncol ; 31(5): 942, 2014 May.
Article in English | MEDLINE | ID: mdl-24687779

ABSTRACT

Colorectal cancer (CRC) is a complex and multifactorial disease, in which genetic and environmental factors both seem to play a part. Many epidemiological studies have explored the association between genetic polymorphisms of X-ray repair cross-complementing group 3 (XRCC3) (Thr241Met) and Xeroderma pigmentosum group D (XPD) lysine to glutamine at codon 751 (Lys751Gln) and risk of CRC in various populations; however, the results are controversial. We conducted this case-control study in a West Algerian population to assess the potential role of this genetic polymorphism on the risk of CRC in this population. Genomic DNA was extracted from blood samples collected from 129 sporadic CRC patients and 148 normal controls. The polymorphisms were determined by pyrosequencing technique. The distribution of XRCC3 Thr241Met and XPD Lys751Gln genotypes among controls did not differ significantly from those predicted by the Hardy-Weinberg distribution (p > 0.05). There were no significant differences in the genotypes distribution and allele frequencies between CRC patients and controls. A significant association was found between the combined heterozygous of XRCC3 and homozygous variant of XPD gene and CRC. This is the first study on DNA repair genetic polymorphisms in West Algerian population, and it suggests that the XRCC3 Thr241Met and XPD Lys751Gln polymorphisms may not be associated with the CRC risk in this population.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Polymorphism, Genetic/genetics , Adult , Aged , Algeria/epidemiology , Case-Control Studies , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Neoplasm Staging , Polymerase Chain Reaction , Prognosis , Risk Factors
18.
Dalton Trans ; 43(2): 439-50, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24061542

ABSTRACT

A novel bimodal fluorescent/radiolabelled probe based on a pyridyltriazole scaffold (known as pyta) is reported here. The final dual imaging agent combines carboxylate functionalization, for biomolecule conjugation, with two distinct metal chelating sites: a pyta-based tricarbonylrhenium moiety as a fluorescent probe and a (99m)Tc(CO3)(+) core through the tridentate chelating iminodiacetic acid (IDA) clamp as a SPECT reporter. The heterodinuclear (99m)Tc/Re complex , as well as its non-radioactive dirhenium analog , was prepared in six steps. The (99m)Tc/Re agent is water-soluble and stable against histidine challenge. Its structural characterization was achieved by HPLC comparison with the non-radioactive complex . Upon excitation in the MLCT band at 321 nm, the compound exhibits a bright green luminescence centered at 496 nm, with a quantum yield of 0.86% in Tris buffer, pH 7.4. Additionally, the influence of this compound on cell viability was tested on malignant cell lines (A549, HT29 and MCF-7 human lung, colon and breast carcinomas, respectively). Cell viability after 72 h incubation at 37 °C with 300 µmol of complex was >60% for all cell lines. Finally, cellular uptake studies of compound were performed by fluorescent microscopy, showing that the complex was clearly detected at the cellular level in A549 cells and to a lesser extent in HT29 cells. Taking into consideration the luminescent properties, the good radiochemical purity and the promising biological data (in vitro stability, non-toxicity, and cell tracking in two cell lines), the functionalized (99m)Tc/Re dinuclear compound can be considered a potential pre- and intraoperative diagnostic probe.

19.
BMC Res Notes ; 7: 260, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24758527

ABSTRACT

BACKGROUND: Over 50% of colorectal cancer (CRC) patients develop metastases. The aim of this study was to evaluate efficacy and tolerance of first-line FOLFIRI® + bevacizumab (B) treatment for metastatic CRC, and to assess genetic polymorphisms as potential markers. METHODS: Adult patients with histologically-proven, non-resectable metastatic CRC and ECOG ≤ 2 were included. 14-day cycles consisted of bevacizumab (5 mg/kg), irinotecan (180 mg/m2), bolus FU (400 mg/m2) and leucovorin (400 mg/m2), followed by 46-hour FU infusions (2400 mg/m2). Primary endpoint was response rate according to RECIST criteria. Secondary endpoints were overall (OS) and progression-free (PFS) survivals, response duration, and toxicity. Associations between clinical data, UGT1A1, thymidylate synthase, VEGFA polymorphisms and PFS, OS and toxicity were analyzed. RESULTS: Sixty-two patients were enrolled (median age 68y). 59/62 patients were eligible and evaluable for response at 6 months: 28 showed partial response (47.5%; 95% CI; 34.3-60.9), 20 stable disease (33.9%) and 11 progression (18.6%). Grade 3/4 toxicities were as follows: neutropenia 16.1%; diarrhea 11.3%; nausea-vomiting 1.6%. Median response duration was 9.5 months (range 2.7-20); median PFS 10.3 months (range 8.8-11.7); and median OS 25.7 months (range 20.2-29.7). 11/59 initially unresectable patients were resectable after treatment. VEGFA polymorphism (rs25648) was associated with better OS (HR: 3.61; 95% CI: 1.57-8.30). CONCLUSIONS: FOLFIRI® + bevacizumab is active with good response rate, long median OS, and a good safety profile. A VEGFA polymorphism might have a prognostic value in this malignancy. TRIAL REGISTRATION: Clinicaltrials.gov: NCT00467142 (registration date: April 25, 2007).


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Polymorphism, Genetic , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Camptothecin/adverse effects , Camptothecin/therapeutic use , Demography , Disease-Free Survival , Female , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Genotyping Techniques , Humans , Leucovorin/adverse effects , Leucovorin/therapeutic use , Male , Multivariate Analysis , Treatment Outcome
20.
Bull Cancer ; 101(6): 533-53, 2014 Jun.
Article in French | MEDLINE | ID: mdl-24977443

ABSTRACT

Irinotecan is a cytotoxic agent administered by IV infusion in the treatment of advanced colorectal cancer. Its anticancer activity results from its bioactivation into SN-38 metabolite, which is cleared through glucuronidation by the hepatic enzyme uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). In the general population, there is wide inter-subject variability in UGT1A1 enzyme activity related to UGT1A1 gene polymorphisms. The French joint workgroup coming from the National Pharmacogenetic Network (RNPGx) and the Group of Clinical Oncologic Pharmacology (GPCO) herein presents an updated review dealing with efficacy and toxicity clinical studies related to UGT1A1 genetic variants. From a critical analysis of this review it clearly emerges that, for doses higher than 180 mg/m(2), hematologic and digestive irinotecan-induced toxicities could be prevented in daily clinical practice by generalizing the use of a simple pharmacogenetic test before starting treatment. The clinical relevance of this test is also discussed in terms of treatment efficacy improvement, with the possibility of increasing the irinotecan dose in patients not bearing the deleterious allele. This test involves using a blood sample to analyze the promoter region of the UGT1A1 gene (UGT1A1*28 allele). Best execution practices, laboratory costs, as well as results interpretation are described with the aim of facilitating the implementation of this analysis in clinical routine. The existence of a French laboratories network performing this test in clinical routine makes it possible to generalize UGT1A1 deficiency screening, so as to guarantee equal access to safe treatment and optimized irinorecan-based therapy for the many patients receiving irinotecan-based therapy in advanced colorectal cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/analogs & derivatives , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Glucuronosyltransferase/genetics , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asian People , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/pharmacokinetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/ethnology , Colorectal Neoplasms/genetics , France , Genotype , Gilbert Disease/genetics , Glucuronosyltransferase/metabolism , Humans , Irinotecan , Pharmacovigilance , Phenotype , Polymorphism, Genetic , Treatment Outcome , United States , White People
SELECTION OF CITATIONS
SEARCH DETAIL