Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Chronic Obstr Pulm Dis ; 11(3): 282-292, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809792

ABSTRACT

Background: Alpha-1 antitrypsin deficiency (AATD) is characterized by low alpha-1 antitrypsin (AAT) levels, predisposing individuals to lung disease. The standard of care, plasma-derived AAT (pdAAT), is delivered as weekly infusions to maintain serum AAT concentrations ≥11µM (≈50% of those in healthy individuals). INBRX-101, a recombinant human AAT-Fc fusion protein, was designed to have a longer half-life and achieve higher AAT levels than pdAAT. Methods: In this phase 1 dose-escalation study (N=31), adults with AATD received 1 dose (part 1) or 3 doses (part 2) of 10 (part 1), 40, 80, or 120mg/kg INBRX-101 every 3 weeks (Q3W) via intravenous infusion. The primary endpoint was safety and tolerability. Secondary endpoints were pharmacokinetics (PK), pharmacodynamics (PD), and immunogenicity of INBRX-101. Results: INBRX-101 was well tolerated. Most treatment-emergent adverse events were grade ≤2. In part 2 (n=18; each dose, n=6), dose-related increases in serum functional AAT (fAAT) were observed; mean fAAT levels remained above the 21 µM target for up to 4 weeks after the final dose in the 120-mg/kg cohort. Antidrug antibodies had no meaningful impact on PK or PD. INBRX-101 was detected in pulmonary epithelial lining fluid (PELF) from all patients assessed (n=11), and PELF fAAT increased after dosing. PK/PD modeling projected steady-state serum fAAT ≥21µM at 120 mg/kg Q3W (average concentration ≈43µM; trough concentration ≈28µM) and Q4W (≈34µM; ≈21µM). Conclusion: The favorable safety profile and ability to maintain serum fAAT levels >21µM with extended-interval dosing, support a phase 2 trial evaluating Q3W and Q4W dosing of INBRX-101.

2.
Clin Cancer Res ; 29(16): 2988-3003, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37265425

ABSTRACT

PURPOSE: Patients with unresectable/metastatic chondrosarcoma have poor prognoses; conventional chondrosarcoma is associated with a median progression-free survival (PFS) of <4 months after first-line chemotherapy. No standard targeted therapies are available. We present the preclinical characterization of INBRX-109, a third-generation death receptor 5 (DR5) agonist, and clinical findings from a phase I trial of INBRX-109 in unresectable/metastatic chondrosarcoma (NCT03715933). PATIENTS AND METHODS: INBRX-109 was first characterized preclinically as a DR5 agonist, with binding specificity and hepatotoxicity evaluated in vitro and antitumor activity evaluated both in vitro and in vivo. INBRX-109 (3 mg/kg every 3 weeks) was then evaluated in a phase I study of solid tumors, which included a cohort with any subtype of chondrosarcoma and a cohort with IDH1/IDH2-mutant conventional chondrosarcoma. The primary endpoint was safety. Efficacy was an exploratory endpoint, with measures including objective response, disease control rate, and PFS. RESULTS: In preclinical studies, INBRX-109 led to antitumor activity in vitro and in patient-derived xenograft models, with minimal hepatotoxicity. In the phase I study, INBRX-109 was well tolerated and demonstrated antitumor activity in unresectable/metastatic chondrosarcoma. INBRX-109 led to a disease control rate of 87.1% [27/31; durable clinical benefit, 40.7% (11/27)], including two partial responses, and median PFS of 7.6 months. Most treatment-related adverse events, including liver-related events, were low grade (grade ≥3 events in chondrosarcoma cohorts, 5.7%). CONCLUSIONS: INBRX-109 demonstrated encouraging antitumor activity with a favorable safety profile in patients with unresectable/metastatic chondrosarcoma. A randomized, placebo-controlled, phase II trial (ChonDRAgon, NCT04950075) will further evaluate INBRX-109 in conventional chondrosarcoma.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Receptors, TNF-Related Apoptosis-Inducing Ligand , Humans , Antibodies, Monoclonal/therapeutic use , Bone Neoplasms/drug therapy , Chemical and Drug Induced Liver Injury , Chondrosarcoma/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
3.
Int J Hematol ; 112(2): 223-233, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32564243

ABSTRACT

Novel therapies are needed for children with relapsed/refractory (R/R) B-cell precursor acute lymphoblastic leukemia (ALL). Blinatumomab is a bispecific T-cell engager immunotherapy that simultaneously binds to CD3-positive cytotoxic T cells and CD19-positive B cells and redirects the patient's T cells to lyse malignant and normal B cells. We conducted an open-label phase 1b study to determine the safety, pharmacokinetics, efficacy, and recommended dose of blinatumomab in Japanese children with R/R B-cell precursor ALL. Patients received induction blinatumomab for 4 weeks (5 µg/m2/day week 1; 15 µg/m2/day weeks 2-4), followed by a 2-week treatment-free interval (6-week cycle). In subsequent cycles, patients received blinatumomab 15 µg/m2/day. The primary end point was the incidence of dose-limiting toxicities. Nine patients received blinatumomab. Since no dose-limiting toxicities were reported, the maximum tolerated dose was 5 µg/m2/day for week 1, followed by 15 µg/m2/day weeks 2-4 (5-15 µg/m2/day, the global recommended dose of blinatumomab). All patients had ≥ 1 grade ≥ 3 adverse events; 89% had grade ≥ 3 treatment-related adverse events. M1 remission rate within the first two cycles of treatment was 56%; one patient had a minimal residual disease response. Consistent with global studies, blinatumomab appeared to be safe with preliminary evidence of efficacy in Japanese children with R/R B-cell precursor ALL.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antineoplastic Agents/administration & dosage , B-Lymphocytes , Immunotherapy , T-Lymphocytes , Adolescent , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/metabolism , Antineoplastic Agents/adverse effects , Antineoplastic Agents/metabolism , Asian People , B-Lymphocytes/metabolism , Child , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Remission Induction , Safety , T-Lymphocytes/metabolism , Treatment Outcome
4.
Cancer Sci ; 111(4): 1314-1323, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31971321

ABSTRACT

Adult patients with relapsed/refractory (R/R) B-precursor acute lymphoblastic leukemia (ALL) have a poor prognosis. Blinatumomab is a bispecific T-cell engager (BiTE) immuno-oncology therapy with dual specificity for CD19 and CD3 that redirects patients' CD3-positive cytotoxic T cells to lyse malignant and normal B cells. We conducted an open-label, phase 1b/2 study to determine the safety, pharmacokinetics, efficacy and recommended dose of blinatumomab in Japanese adults with R/R B-precursor ALL. Patients received 9 µg/day blinatumomab during week 1 and 28 µg/day during weeks 2-4, with a 2-week treatment-free interval (6-week cycle); patients received 28 µg/day blinatumomab in subsequent cycles. Primary endpoints were the incidence of dose-limiting toxicities (DLT) in phase 1b and complete remission (CR)/CR with partial hematologic recovery (CRh) within the first two cycles in phase 2. A total of 26 patients enrolled and 25 (96%) reported grade ≥3 adverse events (mostly cytopenias). There were no DLT. CR/CRh within two cycles was achieved by 4 of 5 patients (80%) in phase 1b and 8 of 21 patients (38%) in phase 2. Among patients with evaluable minimal residual disease, 4 (100%) in phase 1b and 3 (38%) in phase 2 had a complete MRD response. Median RFS for 8 patients who achieved CR/CRh in phase 2 was 5 (95% CI: 3.5-6.4) months; median OS was not estimable. There were no significant associations between maximum cytokine levels or percentage of specific cell types during cycle 1 and response. Consistent with global studies, blinatumomab appeared to be safe and efficacious in Japanese adults with R/R ALL.


Subject(s)
Antibodies, Bispecific/administration & dosage , Lymphoma, B-Cell/drug therapy , Neoplasm, Residual/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adult , Aged , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/blood , Antibodies, Bispecific/pharmacokinetics , Antigens, CD19/genetics , Antigens, CD19/immunology , B-Lymphocytes/pathology , CD3 Complex/genetics , CD3 Complex/immunology , Dexamethasone/administration & dosage , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Interferon-gamma/blood , Kaplan-Meier Estimate , Lymphoma, B-Cell/blood , Lymphoma, B-Cell/pathology , Male , Middle Aged , Neoplasm, Residual/blood , Neoplasm, Residual/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Remission Induction , T-Lymphocytes/immunology
5.
J Clin Oncol ; 38(8): 775-783, 2020 03 10.
Article in English | MEDLINE | ID: mdl-31895611

ABSTRACT

PURPOSE: The anti-B-cell maturation antigen BiTE molecule AMG 420 was assessed in patients with relapsed/refractory multiple myeloma. PATIENTS AND METHODS: In this first-in-human study, up to 10 cycles of AMG 420 were given (4-week infusions/6-week cycles). Patients had progression after ≥ 2 lines of prior therapy and no extramedullary disease. Minimal residual disease (MRD) response was defined as < 1 tumor cell/104 bone marrow cells by flow cytometry. RESULTS: Forty-two patients received AMG 420 at 0.2-800 µg/d. Median age was 65 years, and median disease duration was 5.2 years. Median exposure was 1 cycle (range, 1-10 cycles) and 7 cycles (range, 1-10 cycles) for responders. Patients discontinued for disease progression (n = 25), adverse events (AEs; n = 7), death (n = 4), completion of 10 cycles (n = 3), and consent withdrawal (n = 1). Two patients remain on treatment. There were 2 nontreatment-related deaths from AEs, influenza/aspergillosis and adenovirus-related hepatitis. Serious AEs (n = 20; 48%) included infections (n = 14) and polyneuropathy (n = 2); treatment-related serious AEs included 2 grade 3 polyneuropathies and 1 grade 3 edema. There were no grade ≥ 3 CNS toxicities or anti-AMG 420 antibodies. In this study, 800 µg/d was considered to not be tolerable because of 1 instance each of grade 3 cytokine release syndrome and grade 3 polyneuropathy, both of which resolved. The overall response rate was 31% (n = 13 of 42). At the maximum tolerated dose (MTD) of 400 µg/d, the response rate was 70% (n = 7 of 10). Of these, five patients experienced MRD-negative complete responses, and 1 had a partial response, and 1 had a very good partial response; all 7 patients responded during the first cycle, and some responses lasted > 1 year. CONCLUSION: In this study of AMG 420 in patients with relapsed/refractory multiple myeloma, the response rate was 70%, including 50% MRD-negative complete responses, at 400 µg/d, the MTD for this study.


Subject(s)
Antibodies, Bispecific/administration & dosage , B-Cell Maturation Antigen/antagonists & inhibitors , Multiple Myeloma/therapy , Adult , Aged , Antibodies, Bispecific/immunology , B-Cell Maturation Antigen/immunology , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Multiple Myeloma/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
6.
Pharmacogenomics ; 17(6): 615-31, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27045656

ABSTRACT

Genetic variants of drug metabolism enzymes and transporters can result in high pharmacokinetic and pharmacodynamic variability, unwanted characteristics of efficacious and safe drugs. Ideally, the contributions of these enzymes and transporters to drug disposition can be predicted from in vitro experiments and in silico modeling in discovery or early development, and then be utilized during clinical development. Recently, regulatory agencies have provided guidance on the preclinical investigation of pharmacogenetics, for application to clinical drug development. This white paper summarizes the results of an industry survey conducted by the Industry Pharmacogenomics Working Group on current practice and challenges with using in vitro systems and in silico models to understand pharmacogenetic causes of variability in drug disposition.


Subject(s)
Genetic Variation/genetics , Inactivation, Metabolic/genetics , Membrane Transport Proteins/genetics , Drug Discovery/methods , Humans , Pharmacogenetics/methods
7.
Pharm Res ; 31(7): 1644-55, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24562808

ABSTRACT

PURPOSE: The intracardiac synthesis of anthracycline alcohol metabolites (e.g., daunorubicinol) contributes to the pathogenesis of anthracycline-related cardiotoxicity. Cancer patients with Down syndrome (DS) are at increased risk for anthracycline-related cardiotoxicity. We profiled the expression of anthracycline metabolizing enzymes in hearts from donors with- and without- DS. METHODS: Cardiac expression of CBR1, CBR3, AKR1A1, AKR1C3 and AKR7A2 was examined by quantitative real time PCR, quantitative immunoblotting, and enzyme activity assays using daunorubicin. The CBR1 polymorphism rs9024 was investigated by allelic discrimination with fluorescent probes. The contribution of CBRs/AKRs proteins to daunorubicin reductase activity was examined by multiple linear regression. RESULTS: CBR1 was the most abundant transcript (average relative expression; DS: 81%, non-DS: 58%), and AKR7A2 was the most abundant protein (average relative expression; DS: 38%, non-DS: 35%). Positive associations between cardiac CBR1 protein levels and daunorubicin reductase activity were found for samples from donors with- and without- DS. Regression analysis suggests that sex, CBR1, AKR1A1, and AKR7A2 protein levels were significant contributors to cardiac daunorubicin reductase activity. CBR1 rs9024 genotype status impacts on cardiac CBR1 expression in non-DS hearts. CONCLUSIONS: CBR1, AKR1A1, and AKR7A2 protein levels point to be important determinants for predicting the synthesis of cardiotoxic daunorubicinol in heart.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Alcohol Oxidoreductases/metabolism , Aldehyde Reductase/metabolism , Anthracyclines/metabolism , Down Syndrome/enzymology , Heart/drug effects , Hydroxyprostaglandin Dehydrogenases/metabolism , Myocardium/enzymology , 3-Hydroxysteroid Dehydrogenases/analysis , 3-Hydroxysteroid Dehydrogenases/genetics , Alcohol Oxidoreductases/analysis , Alcohol Oxidoreductases/genetics , Aldehyde Reductase/analysis , Aldehyde Reductase/genetics , Aldo-Keto Reductase Family 1 Member C3 , Anthracyclines/adverse effects , Cardiotoxins/adverse effects , Cardiotoxins/metabolism , Daunorubicin/adverse effects , Daunorubicin/analogs & derivatives , Daunorubicin/metabolism , Down Syndrome/complications , Down Syndrome/drug therapy , Down Syndrome/genetics , Female , Gene Expression , Genotype , Humans , Hydroxyprostaglandin Dehydrogenases/analysis , Hydroxyprostaglandin Dehydrogenases/genetics , Male , Myocardium/metabolism , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics
8.
PLoS One ; 7(11): e48622, 2012.
Article in English | MEDLINE | ID: mdl-23133646

ABSTRACT

MicroRNAs (miRNAs) are small RNAs responsible for the post-transcriptional regulation of a variety of human genes. To date, their involvement in the regulation of CBR1 is unknown. This study reports for the first time the identification of microRNA-574-5p (hsa-miR-574-5p) and microRNA-921 (hsa-miR-921) as two miRNAs capable of interacting with the 3'-untranslated region (3'-UTR) of the CBR1 gene and downregulating CBR1 expression. Furthermore, we demonstrate that a common single-nucleotide polymorphism (SNP) in the CBR1 3'-UTR (rs9024, CBR1 1096G>A) differentially impacts the regulation of CBR1 by hsa-miR-574-5p and hsa-miR-921 dependent on genotype. First, four candidate miRNAs were selected based on bioinformatic analyses, and were tested in Chinese hamster ovary (CHO) cells transfected with CBR1 3'-UTR constructs harboring either the G or A allele for rs9024. We found that hsa-miR-574-5p and hsa-miR-921 significantly decreased luciferase activity in CHO cells transfected with the CBR1 3'-UTR construct carrying the major rs9024 G allele by 35% and 46%, respectively. The influence of these miRNAs was different in cells transfected with a CBR1 3'-UTR construct containing the minor rs9024 A allele in that only hsa-miR-574-5p had a demonstrable effect (i.e., 52% decrease in lucifersase activity). To further determine the functional effects of miRNA-mediated regulation of polymorphic CBR1, we assessed CBR1 protein expression and CBR1 enzymatic activity for the prototypical substrate menadione in human lymphoblastoid cell lines with distinct rs9024 genotypes. We found that hsa-miR-574-5p and hsa-miR-921 significantly decreased CBR1 protein (48% and 40%, respectively) and CBR1 menadione activity (54% and 18%, respectively) in lymphoblastoid cells homozygous for the major rs9024 G allele. In contrast, only hsa-miR-574-5p decreased CBR1 protein and CBR1 activity in cells homozygous for the minor rs9024 A allele, and did so by 49% and 56%, respectively. These results suggest that regulation of human CBR1 expression by hsa-miR-574-5p and hsa-miR-921 depends upon rs9024 genotype status.


Subject(s)
Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/physiology , Gene Expression Regulation , MicroRNAs/metabolism , 3' Untranslated Regions , Alleles , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cytosol/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Variation , Genotype , Humans , Kinetics , MicroRNAs/biosynthesis , Models, Genetic , Polymorphism, Genetic , Thermodynamics , Vitamin K 3/pharmacology
9.
Toxicol Lett ; 211(3): 266-73, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22531821

ABSTRACT

Carbonyl reductase 1 (CBR1) reduces various xenobiotic carbonyl substrates to corresponding alcohol metabolites. Here we demonstrated that benzo[a]pyrene (B[a]P), a potent pro-carcinogen and predominant polycyclic aromatic hydrocarbon (PAH) compound in cigarette smoke and air pollutants, upregulates CBR1 gene expression in vitro and in vivo, and that a proximal xenobiotic response element (XRE) motif (₋122XRE) mediates the induction effect of B[a]P. First, we observed 46% and 50% increases in CBR1 mRNA and CBR1 protein levels, respectively, in human lung tissue samples from smokers compared to never-smokers. Second, we detected 3.0-fold (p<0.0001) induction of CBR1 mRNA and 1.5-fold (p<0.01) induction of CBR1 protein levels in cells of the human lung cancer cell line A549 incubated with 2.5 µM B[a]P for 24h. Third, results from experiments with CBR1 promoter constructs indicated that a proximal XRE motif ₋122XRE) mediates induction of reporter activity in response to B[a]P. Furthermore, we detected enhanced nuclear translocation of aryl hydrocarbon receptor (AhR) following B[a]P exposure in A549 cells. Finally, we demonstrated increased binding of specific protein complexes to ₋122XRE in nuclear extracts from B[a]P-treated cells and the presence of the AhR/Arnt complex in the specific nuclear protein ₋122XRE complexes.


Subject(s)
Alcohol Oxidoreductases/biosynthesis , Benzo(a)pyrene/toxicity , Environmental Pollutants/toxicity , Lung Neoplasms/enzymology , Lung/enzymology , Nicotiana/adverse effects , Alcohol Oxidoreductases/genetics , Blotting, Western , Cell Line, Tumor , Cell Nucleus/metabolism , Cytosol/metabolism , Electrophoretic Mobility Shift Assay , Enzyme Induction/drug effects , Humans , Luciferases/genetics , Lung/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/physiology , Smoke/adverse effects , Smoke/analysis , Smoking/metabolism , Nicotiana/chemistry , Transfection
10.
BMC Genomics ; 13: 44, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22276777

ABSTRACT

BACKGROUND: Single nucleotide polymorphisms (SNPs) can lead to the susceptibility and onset of diseases through their effects on gene expression at the posttranscriptional level. Recent findings indicate that SNPs could create, destroy, or modify the efficiency of miRNA binding to the 3'UTR of a gene, resulting in gene dysregulation. With the rapidly growing number of published disease-associated SNPs (dSNPs), there is a strong need for resources specifically recording dSNPs on the 3'UTRs and their nucleotide distance from miRNA target sites. We present here miRdSNP, a database incorporating three important areas of dSNPs, miRNA target sites, and diseases. DESCRIPTION: miRdSNP provides a unique database of dSNPs on the 3'UTRs of human genes manually curated from PubMed. The current release includes 786 dSNP-disease associations for 630 unique dSNPs and 204 disease types. miRdSNP annotates genes with experimentally confirmed targeting by miRNAs and indexes miRNA target sites predicted by TargetScan and PicTar as well as potential miRNA target sites newly generated by dSNPs. A robust web interface and search tools are provided for studying the proximity of miRNA binding sites to dSNPs in relation to human diseases. Searches can be dynamically filtered by gene name, miRBase ID, target prediction algorithm, disease, and any nucleotide distance between dSNPs and miRNA target sites. Results can be viewed at the sequence level showing the annotated locations for miRNA target sites and dSNPs on the entire 3'UTR sequences. The integration of dSNPs with the UCSC Genome browser is also supported. CONCLUSION: miRdSNP provides a comprehensive data source of dSNPs and robust tools for exploring their distance from miRNA target sites on the 3'UTRs of human genes. miRdSNP enables researchers to further explore the molecular mechanism of gene dysregulation for dSNPs at posttranscriptional level. miRdSNP is freely available on the web at http://mirdsnp.ccr.buffalo.edu.


Subject(s)
Databases, Genetic , Disease/genetics , MicroRNAs/metabolism , Polymorphism, Single Nucleotide , 3' Untranslated Regions , Algorithms , Humans , Internet , Software
11.
Biochem Pharmacol ; 83(1): 139-48, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22001310

ABSTRACT

Carbonyl reductase activity catalyzes the two electron reduction of several endogenous and exogenous carbonyl substrates. Recent data indicate that the expression of human carbonyl reductase 3 (CBR3) is regulated by the master redox switch Nrf2. Nrf2 binds to conserved antioxidant response elements (AREs) in the promoters of target genes. The presence of functional AREs in the CBR3 promoter has not yet been reported. In this study, experiments with reporter constructs showed that the prototypical Nrf2 activator tert-butyl hydroquinone (t-BHQ) induces CBR3 promoter activity in cultures of HepG2 (2.7-fold; p<0.05) and MCF-7 cells (22-fold; p<0.01). Computational searches identified a conserved ARE in the distal CBR3 promoter region ((-2698)ARE). Deletion of this ARE from a 4212-bp CBR3 promoter construct impacted basal promoter activity and induction of promoter activity in response to treatment with t-BHQ. Deletion of (-2698)ARE also impacted the induction of CBR3 promoter activity in cells overexpressing Nrf2. Electrophoretic mobility shift assays (EMSA) demonstrated increased binding of specific protein complexes to (-2698)ARE in nuclear extracts from t-BHQ treated cells. The presence of Nrf2 in the specific nuclear protein-(-2698)ARE complexes was evidenced in EMSA experiments with anti-Nrf2 antibodies. These data suggest that the distal (-2698)ARE mediates the induction of human CBR3 in response to prototypical activators of Nrf2.


Subject(s)
Alcohol Oxidoreductases/genetics , Antioxidants/physiology , Conserved Sequence/genetics , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Promoter Regions, Genetic/genetics , Response Elements/genetics , Enzyme Induction/genetics , Hep G2 Cells , Humans , Oxidation-Reduction
12.
Drug Metab Dispos ; 38(12): 2096-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20729274

ABSTRACT

Cancer patients with Down syndrome (DS) are susceptible to developing anthracycline-related cardiotoxicity. The pathogenesis of anthracycline-related cardiotoxicity has been linked to the intracardiac synthesis of alcohol metabolites by carbonyl reductase 1 (CBR1). CBR1 is located in the DS critical region (21q22.12). The expression of CBR1 in hearts from individuals with DS has not been characterized. This study documented CBR1 expression in hearts from donors with DS (n = 4) and donors without DS (n = 15). The DS samples showed 1.8-fold higher CBR1 mRNA levels compared to the non-DS samples (levels in DS samples were 3.3-relative fold, and those in non-DS were 1.8-relative fold; p = 0.012). CBR1 protein levels were 1.9-fold higher in DS samples than in non-DS samples (13.5 ± 7.7 versus 7.2 ± 3.9 nmol/g cytosolic protein, respectively; p = 0.029). CBR1 activity for daunorubicin was 1.7-fold higher in DS samples than in non-DS samples (3.8 ± 0.1 versus 2.3 ± 0.2 nmol daunol/min · mg, respectively; p = 0.050). CBR1 1096G>A (rs9024) affects CBR1 activity, and one heart trisomic for the variant A allele (A/A/A) exhibited low enzymatic activity. These findings suggest that increased CBR1 expression in the hearts of individuals with DS may contribute to the risk of anthracycline-related cardiotoxicity.


Subject(s)
Alcohol Oxidoreductases/genetics , Anthracyclines/metabolism , Down Syndrome/enzymology , Myocardium/enzymology , Tissue Donors , Anthracyclines/toxicity , Genotype , Heart/drug effects , Humans , RNA, Messenger/analysis
13.
Drug Metab Dispos ; 37(2): 400-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19022938

ABSTRACT

Carbonyl reductase 1 (CBR1) reduces the anticancer drug doxorubicin into the cardiotoxic metabolite doxorubicinol. We documented the hepatic expression of CBR1 in samples from white and black donors. Concordance between ethnicity and geographical ancestry was examined with ancestry informative markers. Livers from blacks and whites showed similar CBR1 mRNA levels (CBR1 mRNA(blacks) = 4.8 +/- 4.3 relative -fold versus CBR1 mRNA(whites) = 3.6 +/- 3.6 relative -fold; p = 0.217). CBR1 protein levels did not differ between both groups (CBR1(blacks) = 8.0 +/- 3.4 nmol/g cytosolic protein versus CBR1(whites) = 9.0 +/- 4.6 nmol/g cytosolic protein; p = 0.347). The CBR1 3'-untranslated region polymorphism 1096G>A was detected in DNA samples from whites (p = 0.875; q = 0.125), and livers with homozygous G/G genotypes showed a trend toward higher CBR1 mRNA levels compared with samples with heterozygous G/A genotypes [CBR1 1096G>A((G/G)) = 4.1 +/- 4.1 relative -fold versus CBR1 1096G>A((G/A)) = 3.0 +/- 2.5 relative-fold; p = 0.266]. CBR1 1096G>A genotype status was associated with CBR1 protein levels (p = 0.030) and CBR activity expressed as the rate of synthesis of doxorubicinol (p = 0.028). Our findings warrant further studies to evaluate the impact of CBR1 1096G>A genotype status on the variable pharmacodynamics of anthracycline drugs.


Subject(s)
Black People/genetics , Polymorphism, Genetic , White People/genetics , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Genotype , Humans , Pharmacogenetics , Tissue Donors
14.
Pharm Res ; 25(7): 1730-4, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18449627

ABSTRACT

PURPOSE: Carbonyl reductase 1 (CBR1) reduces the anticancer anthracyclines doxorubicin and daunorubicin into the cardiotoxic metabolites doxorubicinol and daunorubicinol. We evaluated whether the cardioprotectant monoHER inhibits the activity of polymorphic CBR1. METHODS: We performed enzyme kinetic studies with monoHER, CBR1 (CBR1 V88 and CBR1 I88) and anthracycline substrates. We also characterized CBR1 inhibition by the related flavonoids triHER and quercetin. RESULTS: MonoHER inhibited the activity of CBR1 V88 and CBR1 I88 in a concentration-dependent manner. The IC(50) values of monoHER were lower for CBR1 I88 compared to CBR1 V88 for the substrates daunorubicin and doxorubicin (daunorubicin, IC(50)-CBR1 I88 = 164 microM vs. IC(50)-CBR1 V88 = 219 microM; doxorubicin, IC(50)-CBR1 I88 = 37 microM vs. IC(50)-CBR1 V88 = 59 microM; p < 0.001). Similarly, the flavonoids triHER and quercetin exhibited lower IC(50) values for CBR1 I88 compared to CBR1 V88 (p < 0.001). MonoHER acted as a competitive CBR1 inhibitor when using daunorubicin as a substrate Ki = 45 +/- 18 microM. MonoHER acted as an uncompetitive CBR1 inhibitor for the small quinone substrate menadione Ki = 33 +/- 17 microM. CONCLUSIONS: The cardioprotectant monoHER inhibits CBR1 activity. CBR1 V88I genotype status and the type of anthracycline substrate dictate the inhibition of CBR1 activity.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydroxyethylrutoside/analogs & derivatives , Alcohol Oxidoreductases/genetics , Antibiotics, Antineoplastic/pharmacology , Data Interpretation, Statistical , Daunorubicin/pharmacology , Doxorubicin/pharmacology , Genotype , Humans , Hydroxyethylrutoside/pharmacology , Kinetics , Recombinant Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...