Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Br J Clin Pharmacol ; 89(10): 3079-3091, 2023 10.
Article in English | MEDLINE | ID: mdl-37264516

ABSTRACT

AIMS: RO7049389 (linvencorvir) is a developmental oral treatment for chronic hepatitis B virus infection. The aim of this work was to conduct mass balance (MB) and absolute bioavailability (BA) analyses in healthy volunteers, alongside in vitro evaluations of the metabolism of RO7049389 and a major circulating active metabolite M5 in human hepatocytes, and physiologically based pharmacokinetic (PBPK) modelling to refine the underlying drug disposition paradigm. METHODS: Participants in the clinical study (MB: Caucasian, male, n = 6; BA: Caucasian and Asian, male and female, n = 16, 8 in each ethnic groups) received oral [14 C] or unlabelled RO7049389 (600/1000 mg) followed by 100 µg intravenous [13 C]RO7049389. Metabolic pathways with fractions metabolized-obtained from the in vitro incubation results of 10 µM [14 C]RO7049389 and 1 µM M5 with (long-term cocultured) human hepatocytes in the absence and presence of the cytochrome P450 3A4 (CYP3A4) inhibitor itraconazole-were used to complement the PBPK models, alongside the clinical MB and BA data. RESULTS: The model performance in predicting the pharmacokinetic profiles of RO7049389 and M5 aligned with clinical observations in Caucasians and was also successfully applied to Asians. Accordingly, the drug disposition pathways for RO7049389 were postulated with newly characterized estimates of the fractions: biliary excretion by P-glycoprotein (~41%), direct glucuronidation via uridine 5'-diphosphoglucuronosyltransferase 1A3 (~11%), hexose conjugation (~6%), oxidation by CYP3A4 (~28%) and other oxidation reactions (~9%). CONCLUSION: These results support the ongoing clinical development program for RO7049389 and highlight the broader value of PBPK and MB analyses in drug development.


Subject(s)
Cytochrome P-450 CYP3A , Hepatitis B, Chronic , Humans , Male , Female , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Models, Biological , Administration, Oral
2.
Drug Discov Today ; 27(6): 1604-1621, 2022 06.
Article in English | MEDLINE | ID: mdl-35304340

ABSTRACT

Many in vitro and in vivo models are used in pharmacological research to evaluate the role of targeted proteins in a disease. Understanding the translational relevance and limitation of these models for analyzing a drug's disposition, pharmacokinetic/pharmacodynamic (PK/PD) profile, mechanism, and efficacy, is essential when selecting the most appropriate model of the disease of interest and predicting clinically efficacious doses of the investigational drug. Selected animal models used in ophthalmology, infectious diseases, oncology, autoimmune diseases, and neuroscience are reviewed here. Each area has specific challenges around translatability and determination of an efficacious dose: new patient-specific dosing methods may help overcome these limitations.


Subject(s)
Drugs, Investigational , Medical Oncology , Animals , Models, Biological
3.
Cancer Immunol Res ; 10(1): 87-107, 2022 01.
Article in English | MEDLINE | ID: mdl-34782346

ABSTRACT

Targeting chromatin binding proteins and modifying enzymes can concomitantly affect tumor cell proliferation and survival, as well as enhance antitumor immunity and augment cancer immunotherapies. By screening a small-molecule library of epigenetics-based therapeutics, BET (bromo- and extra-terminal domain) inhibitors (BETi) were identified as agents that sensitize tumor cells to the antitumor activity of CD8+ T cells. BETi modulated tumor cells to be sensitized to the cytotoxic effects of the proinflammatory cytokine TNF. By preventing the recruitment of BRD4 to p65-bound cis-regulatory elements, BETi suppressed the induction of inflammatory gene expression, including the key NF-κB target genes BIRC2 (cIAP1) and BIRC3 (cIAP2). Disruption of prosurvival NF-κB signaling by BETi led to unrestrained TNF-mediated activation of the extrinsic apoptotic cascade and tumor cell death. Administration of BETi in combination with T-cell bispecific antibodies (TCB) or immune-checkpoint blockade increased bystander killing of tumor cells and enhanced tumor growth inhibition in vivo in a TNF-dependent manner. This novel epigenetic mechanism of immunomodulation may guide future use of BETi as adjuvants for immune-oncology agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
Expert Opin Investig Drugs ; 30(8): 893-901, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34176392

ABSTRACT

BACKGROUND: Balovaptan is a potent, selective vasopressin 1a receptor antagonist. The early-phase pharmacokinetics (PK) of balovaptan are reported. RESEARCH DESIGN AND METHODS: Two Phase 1 studies (overall N = 93) assessed single- and multiple-dose balovaptan PK in healthy adults. One (N = 16) assessed absolute oral bioavailability (10 mg or 50 mg) vs a [13C]-balovaptan microdose. The other (N = 77) explored single- (0.5-76 mg) and multiple-dose (14 days; 12-52 mg/day) - randomized 6:2 balovaptan:placebo per dose - PK, dose proportionality, and the effect of food on single-dose (32 mg) Cmax and AUCinf. RESULTS: Absolute balovaptan bioavailability was high (103-116%). Steady-state (Day 14) balovaptan PK was approximately dose proportional with a half-life of 45-47 hours, but single-dose Cmax increased more than dose proportionally and half-life was inversely dose-proportional - a discordance partially attributable to a dose-and-time-dependent volume of distribution. Accumulation (Day 1-Day 14) was inversely dose-proportional (~3.5 [12 mg] to ~1.8 [52 mg]). There was no relevant effect of a high-fat meal on single-dose balovaptan exposure. There were no safety signals: 2/93 subjects discontinued for adverse events. CONCLUSIONS: Balovaptan was well tolerated at single (≤76 mg) and multiple (≤52 mg/day) doses, with a PK profile supportive of once-daily administration without food restrictions. TRIAL REGISTRATION: ClinicalTrials.gov NCT03764449; NCT01418963.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/administration & dosage , Benzodiazepines/administration & dosage , Food-Drug Interactions , Pyridines/administration & dosage , Triazoles/administration & dosage , Administration, Oral , Adolescent , Adult , Antidiuretic Hormone Receptor Antagonists/adverse effects , Antidiuretic Hormone Receptor Antagonists/pharmacokinetics , Area Under Curve , Benzodiazepines/adverse effects , Benzodiazepines/pharmacokinetics , Biological Availability , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Male , Middle Aged , Pyridines/adverse effects , Pyridines/pharmacokinetics , Time Factors , Tissue Distribution , Triazoles/adverse effects , Triazoles/pharmacokinetics , Young Adult
5.
Clin Pharmacol Ther ; 109(4): 1081-1091, 2021 04.
Article in English | MEDLINE | ID: mdl-33523474

ABSTRACT

The pharmacokinetics (PK) of RO7049389, a new hepatitis B virus (HBV) core protein allosteric modulator of class I, and of its active metabolite M5 were studied in fasted and fed conditions after single and multiple once-a-day and twice-a-day doses in healthy subjects and patients with HBV. The nonlinearity of the pharmacokinetics, the large variability, the small sample size per dose arms, the higher plasma exposure in Asians, and the heterogeneity in patient baseline characteristics seen in phase I studies made the ethnic sensitivity assessment and the selection of the recommended phase II dose difficult. A population PK model, simultaneously modeling RO7049389 and M5, was developed to characterize the complex PK, quantify ethnicity (i.e., Asian vs. non-Asian) and gender effects on the PK of RO7049389 and M5, and infer the quantity of RO7049389 in liver relative to plasma. Exposures in the liver are of particular importance for dose selection since the liver is the site of action of the compound. The model described and reproduced the population PK profiles as well as the between-subject variability of RO7049389 and its metabolite. It could show that the PK is similar between healthy subjects and in HBV patients, once the ethnicity and gender effects are accounted for. The model predicts that, despite a large difference in the plasma exposure of RO7049389 between Asians and non-Asians, the exposure in the liver is comparable, allowing the use of the same dose to treat Asian and non-Asian patients. This model provides a valuable basis to develop this new anti-HBV drug and to define optimal dosing.


Subject(s)
Antiviral Agents/pharmacokinetics , Hepatitis B/drug therapy , Adult , Biological Transport , Dose-Response Relationship, Drug , Fasting , Female , Healthy Volunteers , Hepatitis B/ethnology , Humans , Liver , Male , Models, Biological , Racial Groups , Sex Factors
6.
Neuro Oncol ; 22(6): 819-829, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32383735

ABSTRACT

BACKGROUND: Studies evaluating the CNS penetration of a novel tyrosine kinase inhibitor, entrectinib, proved challenging, particularly due to discrepancies across earlier experiments regarding P-glycoprotein (P-gp) interaction and brain distribution. To address this question, we used a novel "apical efflux ratio" (AP-ER) model to assess P-gp interaction with entrectinib, crizotinib, and larotrectinib, and compared their brain-penetration properties. METHODS: AP-ER was designed to calculate P-gp interaction with the 3 drugs in vitro using P-gp-overexpressing cells. Brain penetration was studied in rat plasma, brain, and cerebrospinal fluid (CSF) samples after intravenous drug infusion. Unbound brain concentrations were estimated through kinetic lipid membrane binding assays and ex vivo experiments, while the antitumor activity of entrectinib was evaluated in a clinically relevant setting using an intracranial tumor mouse model. RESULTS: Entrectinib showed lower AP-ER (1.1-1.15) than crizotinib and larotrectinib (≥2.8). Despite not reaching steady-state brain exposures in rats after 6 hours, entrectinib presented a more favorable CSF-to-unbound concentration in plasma (CSF/Cu,p) ratio (>0.2) than crizotinib and larotrectinib at steady state (both: CSF/Cu,p ~0.03). In vivo experiments validated the AP-ER approach. Entrectinib treatment resulted in strong tumor inhibition and full survival benefit in the intracranial tumor model at clinically relevant systemic exposures. CONCLUSIONS: Entrectinib, unlike crizotinib and larotrectinib, is a weak P-gp substrate that can sustain CNS exposure based on our novel in vitro and in vivo experiments. This is consistent with the observed preclinical and clinical efficacy of entrectinib in neurotrophic tropomyosin receptor kinase (NTRK) and ROS1 fusion-positive CNS tumors and secondary CNS metastases.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Protein-Tyrosine Kinases , ATP Binding Cassette Transporter, Subfamily B , Animals , Benzamides , Cell Differentiation , Indazoles , Mice , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins , Rats
7.
Birth Defects Res B Dev Reprod Toxicol ; 107(3): 148-56, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27221585

ABSTRACT

Bitopertin is a glycine type 1 (GlyT1) inhibitor intended for the treatment of psychiatric disorders. The principle adverse effect in the regulatory reproductive toxicity studies was peri-natal pup death when rat dams were treated during parturition at a dose resulting in five-times the human therapeutic exposure (AUC). Cessation of dosing two days before parturition prevented the pup deaths. Investigatory experiments and pharmacokinetic modelling suggested that the neonatal mortality was related to transplacental passage of bitopertin leading to high systemic levels in the newborn pups. Brain levels of bitopertin in the rat fetus and neonate were two-fold higher than in the mother. As illustrated by knock-out mice models, GlyT1 function is essential for neonatal pup survival in rodents, but is not necessary for normal prenatal morphological development. The glycine transport systems are immature at birth in the rat, but are functionally well-developed in the human newborn. While the relevance to humans of the neonatal mortality seen in rats following late gestational exposure is unknown, bitopertin would not be recommended for use during late pregnancy unless the anticipated benefit for the mother outweighs the potential risk to the newborn.


Subject(s)
Glycine Plasma Membrane Transport Proteins/genetics , Maternal Exposure/adverse effects , Piperazines/toxicity , Sulfones/toxicity , Animals , Databases, Factual , Disease Models, Animal , Dose-Response Relationship, Drug , Embryonic Development/drug effects , Female , Fetal Development/drug effects , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Glycine Plasma Membrane Transport Proteins/metabolism , Humans , Piperazines/pharmacokinetics , Pregnancy , Sulfones/pharmacokinetics
8.
J Pharmacol Exp Ther ; 353(1): 213-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25665805

ABSTRACT

Major depressive disorder (MDD) is a serious public health burden and a leading cause of disability. Its pharmacotherapy is currently limited to modulators of monoamine neurotransmitters and second-generation antipsychotics. Recently, glutamatergic approaches for the treatment of MDD have increasingly received attention, and preclinical research suggests that metabotropic glutamate receptor 5 (mGlu5) inhibitors have antidepressant-like properties. Basimglurant (2-chloro-4-[1-(4-fluoro-phenyl)-2,5-dimethyl-1H-imidazol-4-ylethynyl]-pyridine) is a novel mGlu5 negative allosteric modulator currently in phase 2 clinical development for MDD and fragile X syndrome. Here, the comprehensive preclinical pharmacological profile of basimglurant is presented with a focus on its therapeutic potential for MDD and drug-like properties. Basimglurant is a potent, selective, and safe mGlu5 inhibitor with good oral bioavailability and long half-life supportive of once-daily administration, good brain penetration, and high in vivo potency. It has antidepressant properties that are corroborated by its functional magnetic imaging profile as well as anxiolytic-like and antinociceptive features. In electroencephalography recordings, basimglurant shows wake-promoting effects followed by increased delta power during subsequent non-rapid eye movement sleep. In microdialysis studies, basimglurant had no effect on monoamine transmitter levels in the frontal cortex or nucleus accumbens except for a moderate increase of accumbal dopamine, which is in line with its lack of pharmacological activity on monoamine reuptake transporters. These data taken together, basimglurant has favorable drug-like properties, a differentiated molecular mechanism of action, and antidepressant-like features that suggest the possibility of also addressing important comorbidities of MDD including anxiety and pain as well as daytime sleepiness and apathy or lethargy.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Depression/drug therapy , Imidazoles/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Allosteric Regulation , Animals , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/therapeutic use , Biogenic Monoamines/metabolism , Brain/metabolism , Cells, Cultured , Cricetulus , Depression/metabolism , Depression/psychology , Drug Inverse Agonism , Electroencephalography , Female , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Macaca fascicularis , Male , Mice , Pain/drug therapy , Pain/physiopathology , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Radioligand Assay , Rats, Sprague-Dawley , Rats, Wistar , Receptor, Metabotropic Glutamate 5/metabolism , Urinary Bladder, Overactive/drug therapy , Urinary Bladder, Overactive/physiopathology
9.
Clin Ther ; 34(2): 420-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22244809

ABSTRACT

BACKGROUND: Aleglitazar is a dual peroxisome proliferator-activated receptor (PPAR)-α/γ agonist with a balanced activity (similar half-maximal effective concentrations) toward PPAR-α and -γ that is in clinical development for the treatment of patients who have experienced an acute coronary syndrome and have type 2 diabetes mellitus. OBJECTIVE: This study aimed to characterize the metabolic profile and the routes and rates of elimination of aleglitazar and its major metabolites in humans. METHODS: In this Phase I, nonrandomized, open-label, single-center, single-dose study, 6 healthy male subjects each received a single oral dose of 300 µg [(14)C]-labeled aleglitazar. Total urine and feces were collected for up to 15 days. Venous blood samples were collected to determine the plasma concentrations of aleglitazar and its metabolites and for radioactivity counting. RESULTS: The median age (range) and mean (SD) body mass index of subjects were 48 (41-60) years and 24.8 (3.0) kg/m(2), respectively. Recovery of total radioactivity, as a percentage of the dose administered, was high (93 [3]%). Aleglitazar was predominantly eliminated in feces (mean, 66% [range, 55%-74%]), with only 28% (range, 22%-36%) of the radioactivity recovered in urine. Only a mean (SD) of 1.8 (0.8)% of aleglitazar was eliminated unchanged as parent compound in feces and only 0.3 (0.4)% was eliminated in urine. Almost all excreted drug-related material could be attributed to its 2 main metabolites, M1 (21%) and M6 (38%). Treatment with aleglitazar was well tolerated, and no serious adverse events were reported. CONCLUSIONS: In healthy volunteers, aleglitazar was excreted mainly in the form of inactive metabolites, mostly M1 and M6, with only a small proportion eliminated unchanged.


Subject(s)
Carbon Radioisotopes , Oxazoles/pharmacokinetics , PPAR alpha/agonists , PPAR gamma/agonists , Thiophenes/pharmacokinetics , Adult , Aged , Humans , Male , Middle Aged , Oxazoles/adverse effects , PPAR alpha/metabolism , PPAR gamma/metabolism , Thiophenes/adverse effects
10.
Xenobiotica ; 40(12): 840-52, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20868265

ABSTRACT

The pharmacokinetics and excretion of carmegliptin, a novel dipeptidyl peptidase IV inhibitor, were examined in rats, dogs, and cynomolgus monkeys. Carmegliptin exhibited a moderate clearance, extensive tissue distribution, and a variable oral bioavailability of 28-174%. Due to saturation of intestinal active secretion, the area under the plasma concentration-time curve (AUC) in dogs and monkeys increased in a more than dose-proportional manner over an oral dose range of 2.5-10 mg/kg. Following oral administration of [(14)C]carmegliptin at 3 mg/kg, > 94% of the radioactive dose was recovered in 72-h post-dose from Wistar rats and Beagle dogs. Virtually, the entire administered radioactive dose was excreted unchanged in urine, intestinal lumen, and bile. Approximately 36%, 29%, and 19% of the dose were excreted by respective routes. Consistently, in vitro, carmegliptin was highly resistant to hepatic metabolism in all species tested. Based on in vitro studies, carmegliptin is a good substrate for Mdr1/MDR1. Breast cancer resistance protein (Bcrp) is not expected to be involved in the transport of carmegliptin since in vitro carmegliptin was not significantly transported by this transporter. The very high extravascular distribution of carmegliptin in the intestinal tissues, as demonstrated in Wistar rats and Beagle dogs, could play a significant role in its therapeutic effect.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Quinolizines/metabolism , Quinolizines/pharmacokinetics , Absorption , Animals , Autoradiography , Biological Availability , Biotransformation , Blood Proteins/metabolism , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dogs , Dose-Response Relationship, Drug , Feces/chemistry , Haplorhini , Injections, Intravenous , Membrane Transport Proteins/metabolism , Quinolizines/administration & dosage , Quinolizines/chemistry , Rats , Tissue Distribution
11.
Drug Metab Lett ; 4(2): 77-87, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20446913

ABSTRACT

Imiloxan is an alpha2 adrenoceptor antagonist and was developed for depression in the 1980's. In Phase 1 clinical trials imiloxan dosing led to hypersensitivity reactions; the molecule's development was discontinued. The present study revisits the in vitro metabolism of imiloxan using modern analytical methods. Human and rat liver microsomes convert imiloxan into a variety of metabolites many of which are unstable and or reactive. Imiloxan also yields high protein covalent binding in microsomal assays. Imiloxan is a useful test molecule for defining the relationship between liver covalent binding and idiosyncratic toxicity.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/analysis , Adrenergic alpha-2 Receptor Antagonists/metabolism , Imidazoles/analysis , Imidazoles/metabolism , Animals , Biotransformation , Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/metabolism , Humans , In Vitro Techniques , Liver/metabolism , Microsomes, Liver/metabolism , Protein Binding , Rats
13.
Toxicol Lett ; 190(3): 310-6, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19695316

ABSTRACT

In order to support the toxicological risk assessment for the ethyl methanesulfonate (EMS) exposure of patients ingesting contaminated Viracept tablets (Müller and Singer, 2009), there was a need to correlate the effects observed in in vivo genotoxicity studies with mice to EMS exposure and to estimate human exposure to EMS at the level of contamination of Viracept tablets. The species differences in volume of distribution of EMS, a key factor for determination of its C(max), were small in the species investigated (mouse, rat, monkey), the species differences in clearance, the key factor involved in AUC assessment, were large (Lavé et al., 2009). Because of this uncertainty in extrapolation of clearance across species we used a conservative approach for human exposure predictions in terms of AUC where clearance was assumed to solely reflect the chemical stability of EMS neglecting additional clearance pathways such as metabolism and exhalation. This approach was compared to the estimates obtained from allometric scaling based on rat clearance, the species leading to the lowest clearance predicted in man. We found that both approaches led to nearly identical predictions of the human AUC. Thus, we predict a human AUC of 13 microM h for patients ingesting the most contaminated Viracept tablets, corresponding to a maximal daily intake of 0.055 mg/kg of EMS. The C(max) of EMS in these patients is predicted to be 0.85 microM. In order to provide a basis for toxicological risk assessment, these maximal human AUC and C(max) values are to be compared to the AUC and C(max) values in mice at the EMS dose of 25mg/kg which was found to be the threshold dose for induction of mutagenic effects, i.e. the dose at which no mutagenic effects were observed (Gocke et al., 2009-a). We calculate AUC and C(max) in mice at the threshold dose to be 350 microM h and 315 microM, respectively. Thus we conclude that a large safety factor can be deduced, whatever the basis of comparison, as is discussed in detail by Müller et al. (2009).


Subject(s)
Alkylating Agents/pharmacokinetics , Drug Contamination , Ethyl Methanesulfonate/pharmacokinetics , Mutagens/pharmacokinetics , Animals , Area Under Curve , Computer Simulation , Dose-Response Relationship, Drug , HIV Protease Inhibitors/chemistry , Hemoglobins/chemistry , Hemoglobins/drug effects , Hemoglobins/metabolism , Humans , In Vitro Techniques , Macaca fascicularis , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Biological , Nelfinavir/chemistry , Rats , Risk Assessment , Species Specificity , Valine/analogs & derivatives , Valine/chemistry , Valine/metabolism
14.
Chem Res Toxicol ; 15(4): 551-61, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11952342

ABSTRACT

Liquid chromatography with electrospray ionization tandem mass spectrometry (LC/ESI-MS/MS) was used to measure DNA adducts of the carcinogen 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx) with a microbore C-18 reversed-phase column. Quantification of the isomeric adducts N-(deoxyguanosin-8-yl)-2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (dG-C8-MeIQx) and 5-(deoxyguanosin-N(2)-yl)-2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (dG-N(2)-MeIQx) was achieved using synthetic, isotopically labeled internal standards. The reaction of the N-acetoxy ester of 2-(hydroxyamino)-3,8-dimethylimidazo[4,5-f]quinoxaline (HONH-MeIQx) with calf thymus DNA (ct DNA) resulted in formation of these adducts in a ratio of 5:1 (dG-C8-MeIQx:dG-N(2)-MeIQx). The detection limit by LC/ESI-MS/MS in the selected reaction monitoring (SRM) mode ([MH(+) --> MH - 116](+)) (loss of deoxyribose) approached 500 fg (1 fmol) of adduct standard, and 1 adduct per 10(8) DNA bases using 100 microg of DNA following solid-phase extraction. The SRM analysis of rat liver DNA 24 h after an oral dose of MeIQx (10 and 0.5 mg/kg) revealed the presence of isomeric dG-MeIQx adducts at levels of 3.07 +/- 0.84 and 0.45 +/- 0.27 adducts per 10(7) bases, respectively. LC/ESI-MS/MS product ion spectra were acquired on both adducts from the elevated dose of MeIQx for unambiguous adduct identification. The contribution of dG-N(2)-MeIQx to the total adducts in vivo was significantly more important than that observed in vitro. dG-C8-MeIQx was the principal adduct formed at the 10 mg/kg dose, (dG-C8-MeIQx:dG-N(2)-MeIQx (3:2)); however, dG-N(2)-MeIQx was the major lesion detected at the 0.5 mg/kg dose (dG-C8-MeIQx:dG-N(2)-MeIQx 1:10). The striking differences between the relative amounts of dG-C8-MeIQx and dG-N(2)-MeIQx formed in vivo as a function of dose suggest that reactive esters of HONH-MeIQx other than N-acetoxy-MeIQx may be formed in vivo and react preferentially with the N(2) atom of guanine, or that dG-C8-MeIQx is removed at a significantly more rapid rate than dG-N(2)-MeIQx. The dG-N(2)-MeIQx adduct, previously thought to be a minor adduct, is likely to be an important contributor to the genotoxic damage of MeIQx.


Subject(s)
Carcinogens/chemistry , DNA Adducts/analysis , Liver/chemistry , Quinoxalines/chemistry , Animals , Carcinogens/metabolism , Carcinogens/toxicity , Chromatography, High Pressure Liquid , DNA/chemistry , DNA/drug effects , DNA/metabolism , DNA Adducts/drug effects , DNA Adducts/metabolism , Liver/drug effects , Liver/metabolism , Male , Quinoxalines/metabolism , Quinoxalines/toxicity , Rats , Rats, Inbred F344 , Spectrometry, Mass, Electrospray Ionization
15.
Carcinogenesis ; 23(1): 115-22, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11756232

ABSTRACT

Metabolism of the carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) has been compared in human and rat hepatocytes. The identities of seven metabolites were confirmed by UV and mass spectroscopy and by co-elution with reference standards using HPLC. In human hepatocytes, the major biotransformation pathway of PhIP was cytochrome P4501A2 (CYP1A2)-mediated N-oxidation to form the genotoxic metabolite 2-(hydroxyamino)-1-methyl-6-phenylimidazo[4,5-b]pyridine (HONH-PhIP), which underwent glucuronidation at the N(2) and N3 positions of PhIP to form stable conjugates. These products combined accounted for as much as 60% of the added PhIP. Direct glucuronidation of PhIP at the N(2) and N3 positions also occurred, accounting for up to 20% of the amount added. Glucuronide and sulfate conjugates of 2-amino-4'-hydroxy-1-methyl-6-phenylimidazo[4,5-b]pyridine (4'-HO-PhIP) were also detected, comprising 5 and 12% of the products, respectively. The CYP1A2 inhibitor furafylline diminished the formation of both HONH-PhIP glucuronide conjugates in a concentration-dependent manner, however, levels of 4'-HO-PhIP were unchanged, indicating that CYP1A2 does not significantly contribute to 4'-hydroxylation of PhIP. Hepatocytes of male rats, both untreated and pretreated with the CYP1A2 inducer 3-methylcholanthrene (3-MC) transformed PhIP into 4'-HO-PhIP as the prominent product. Unconjugated and conjugated 4'-HO-PhIP metabolites combined accounted for 18 and 46% of the PhIP products in untreated and in 3-MC-pretreated rat hepatocytes, respectively. The isomeric glucuronide conjugates of HONH-PhIP combined accounted for 11 and 26% of the PhIP, respectively, in untreated and 3-MC-pretreated hepatocytes. The regioselectivity of glucuronidation of PhIP was different in human and rat hepatocytes. Human liver UDP-glucuronosyltransferases favored conjugation to the N(2) positions of PhIP and HONH-PhIP, while the N3 atom was the preferred site of conjugation for the rat enzymes. Thus, important differences exist between human and rat enzymes in catalytic activity and regioselectivity of PhIP metabolism. Some human hepatocyte populations are more active at transforming PhIP to a genotoxic species than rat hepatocytes pretreated with the potent CYP1A2 inducer 3-MC.


Subject(s)
Hepatocytes/metabolism , Imidazoles/metabolism , Theophylline/analogs & derivatives , Animals , Biotransformation/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A2/metabolism , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Humans , Imidazoles/administration & dosage , Imidazoles/chemistry , Isoenzymes/metabolism , Magnetic Resonance Spectroscopy , Rats , Species Specificity , Spectrometry, Mass, Electrospray Ionization , Theophylline/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...