Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stephen Fowler is active.

Publication


Featured researches published by Stephen Fowler.


Aaps Journal | 2008

In Vitro Evaluation of Reversible and Irreversible Cytochrome P450 Inhibition: Current Status on Methodologies and their Utility for Predicting Drug–Drug Interactions

Stephen Fowler; Hongjian Zhang

It is widely accepted that today’s practice of polypharmacy inevitably increases the incidence of drug–drug interactions (DDIs). Serious DDI is a major liability for any new chemical entity (NCE) entering the pharmaceutical market. As such, pharmaceutical companies employ various strategies to avoid problematic compounds for clinical development. A key cause for DDIs is the inhibition of cytochrome P450 enzymes (CYPs) that are responsible for metabolic clearance of many drugs. Screening for inhibition potency of CYPs by NCEs has therefore become a routine practice during the drug discovery stage. However, in order to make proper use of DDI data, an understanding of the strengths and weaknesses of the various experimental systems in current use is required. An illustrated review of experimental practices is presented with discussion of likely future developments. The combination of high quality in vitro data generation and the application of in vivo CYP inhibition modelling approaches should allow more informed decisions to be made in the search for drug molecules with acceptable DDI characteristics.


Journal of Pharmacology and Experimental Therapeutics | 2015

A UGT2B10 splicing polymorphism common in African populations may greatly increase drug exposure.

Stephen Fowler; Heidemarie Kletzl; Moshe Finel; Nenad Manevski; Paul Schmid; Dietrich Tuerck; Roger David Norcross; Marius C. Hoener; Olivia Spleiss; Victor A. Iglesias

RO5263397 [(S)-4-(3-fluoro-2-methyl-phenyl)-4,5-dihydro-oxazol-2-ylamine], a new compound that showed promising results in animal models of schizophrenia, is mainly metabolized in humans by N-glucuronidation. Enzyme studies, using the (then) available commercial uridine 5′-diphosphate-glucuronosyltransferases (UGTs), suggested that UGT1A4 is responsible for its conjugation. In the first clinical trial, in which RO5263397 was administered orally to healthy human volunteers, a 136-fold above-average systemic exposure to the parent compound was found in one of the participants. Further administration in this trial identified two more such poor metabolizers, all three of African origin. Additional in vitro studies with recombinant UGTs showed that the contribution of UGT2B10 to RO5263397 glucuronidation is much higher than UGT1A4 at clinically relevant concentrations. DNA sequencing in all of these poor metabolizers identified a previously uncharacterized splice site mutation that prevents assembly of full-length UGT2B10 mRNA and thus functional UGT2B10 protein expression. Further DNA database analyses revealed the UGT2B10 splice site mutation to be highly frequent in individuals of African origin (45%), moderately frequent in Asians (8%) and almost unrepresented in Caucasians (<1%). A prospective study using hepatocytes from 20 individual African donors demonstrated a >100-fold lower intrinsic clearance of RO5263397 in cells homozygous for the splice site variant allele. Our results highlight the need to include UGT2B10 when screening the human UGTs for the enzymes involved in the glucuronidation of a new compound, particularly when there is a possibility of N-glucuronidation. Moreover, this study demonstrates the importance of considering different ethnicities during drug development.


Clinical Therapeutics | 2009

A single-center, open-label, one-sequence study of dalcetrapib coadministered with ketoconazole, and an in vitro study of the S-methyl metabolite of dalcetrapib

Michael Derks; Stephen Fowler; Olaf Kuhlmann

BACKGROUND Dalcetrapib (RO4607381/JTT-705) is currently under clinical investigation for the prevention of cardiovascular events. It inhibits the activity of cholesteryl ester transfer protein and has been reported to increase levels of high-density lipoprotein cholesterol. OBJECTIVE Because dalcetrapib is likely to be coadministered with agents that inhibit the cytochrome P450 (CYP) 3A4 isozyme, this study aimed to determine the effect of ketoconazole, a strong CYP3A4 inhibitor, on the pharmacokinetics of dalcetrapib. METHODS An open-label, 1-sequence study was conducted in 2 cohorts of healthy, nonsmoking male volunteers aged 18 through 65 years, with a body mass index of 18 to 32 kg/m(2). The first cohort received dalcetrapib 600 mg on days 1 and 7 and ketoconazole 400 mg on days 2 through 7, and, based on the results of a planned interim analysis, the second cohort received dalcetrapib 900 mg alone on days 1 and 7 and ketoconazole on days 2 through 7. Pharmacokinetic and safety parameters were assessed at specific times throughout the study. To confirm CYP involvement in the metabolism of the inactive metabolite dalcetrapib-S-methyl, in vitro studies were performed using human liver microsomes and recombinantly expressed CYP isoforms. RESULTS Of the 26 participants, 96% were white, with a mean age of 38.1 years and a mean weight of 78.6 kg. In the in vivo portion of the study, coadministration of ketoconazole with dalcetrapib 600 mg had no significant effect on any pharmacokinetic parameter of dalcetrapib. Coadministration of ketoconazole with dalcetrapib 900 mg was associated with significant decreases in the dalcetrapib C(max) (-23%; P = 0.002) and AUC(0-infinity) (-18%; P = 0.001) and a significant increase in oral clearance (22%; P = 0.001). Significant increases in the C(max) (P = 0.001) and AUC(0-infinity) (P < 0.001) of dalcetrapib-S-methyl were observed with coadministration of ketoconazole. The combination was generally well tolerated, with 32 of 35 adverse events (91.4%) being mild in intensity. The most frequent adverse events were headache (6/26 [23.1%] in the ketoconazole group; 4/18 [22.2%] in the group receiving dalcetrapib 900 mg plus ketoconazole) and diarrhea (4/26 [15.4%] in the ketoconazole group; 2/18 [11.1%] in the group receiving dalcetrapib 900 mg plus ketoconazole). The in vitro studies confirmed the involvement of CYP3A in the metabolism of dalcetrapib-S-methyl. CONCLUSIONS In this clinical study in healthy male volunteers, coadministration of dalcetrapib 600 mg with the CYP3A4 inhibitor ketoconazole was not associated with any significant changes in the pharmacokinetic parameters of the parent compound. Coadministration of dalcetrapib 900 mg with ketoconazole was associated with significant decreases in the dalcetrapib C(max) and AUC, contrary to the increases that would be expected if dalcetrapib were a substrate for CYP3A4. The combination of dalcetrapib and ketoconazole was generally well tolerated.


Current Medical Research and Opinion | 2009

In vitro and in vivo assessment of the effect of dalcetrapib on a panel of CYP substrates

Michael Derks; Stephen Fowler; Olaf Kuhlmann

ABSTRACT Objective: The primary objective of this study was to investigate the drug–drug interaction potential of dalcetrapib on drugs metabolized via major cytochrome P450 (CYP) isoforms using both in vitro and clinical approaches. A secondary objective was to investigate the safety and tolerability of dalcetrapib alone or co-administered either with a combination of five probe drugs or with rosiglitazone. Research design and methods: Human liver microsomes and a panel of substrates for CYP enzymes were used to determine IC50 for inhibition of CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4. In addition, two drug–drug interaction studies were conducted in healthy males: dalcetrapib 900 mg plus the Cooperstown 5 + 1 drug cocktail, which includes substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4, and dalcetrapib 900 mg plus rosiglitazone, a substrate for CYP2C8. Pharmacokinetic and safety parameters were assessed. Results: In vitro, dalcetrapib was inhibitory to all CYP enzymes tested. IC50 values ranged from 1.5 ± 0.1 μM for CYP2C8 to 82 ± 4 μM for CYP2D6. Co-administration of dalcetrapib plus drug cocktail showed no clinically relevant effect of 900 mg dalcetrapib on activity of CYP1A2, CYP2C19, CYP2D6, CYP2C9, or CYP3A4 following repeated administration. Co-administration of dalcetrapib plus rosiglitazone showed no clinically relevant effect of dalcetrapib 900 mg on activity of CYP2C8. Dalcetrapib was generally well tolerated. Conclusions: Although in vitro studies indicated that dalcetrapib inhibits CYP activity, two clinical studies showed no clinically relevant effect on any of the major CYP isoforms at a 900 mg dose, which is higher than the 600 mg dose being explored in phase III studies. Dalcetrapib was generally well tolerated in these studies. However, these studies were limited to a small number of healthy males; additional, larger studies are necessary to study its safety.


Antimicrobial Agents and Chemotherapy | 2009

Nonclinical Pharmacokinetics of Oseltamivir and Oseltamivir Carboxylate in the Central Nervous System

Gerhard Hoffmann; Christoph Funk; Stephen Fowler; Michael B. Otteneder; Alexander Breidenbach; Craig R. Rayner; Tom Chu; Eric Prinssen

ABSTRACT Oseltamivir, a potent and selective inhibitor of influenza A and B virus neuraminidases, is a prodrug that is systemically converted into the active metabolite oseltamivir carboxylate. In light of reported neuropsychiatric events in influenza patients, including some taking oseltamivir, and as part of a full assessment to determine whether oseltamivir could contribute to, or exacerbate, such events, we undertook a series of nonclinical studies. In particular, we investigated (i) the distribution of oseltamivir and oseltamivir carboxylate in the central nervous system of rats after single intravenous doses of oseltamivir and oseltamivir carboxylate and oral doses of oseltamivir, (ii) the active transport of oseltamivir and oseltamivir carboxylate in vitro by transporters located in the blood-brain barrier, and (iii) the extent of local conversion of oseltamivir to oseltamivir carboxylate in brain fractions. In all experiments, results showed that the extent of partitioning of oseltamivir and especially oseltamivir carboxylate to the central nervous system was low. Brain-to-plasma exposure ratios were approximately 0.2 for oseltamivir and 0.01 for oseltamivir carboxylate. Apart from oseltamivir being a good substrate for the P-glycoprotein transporter, no other active transport processes were observed. The conversion of the prodrug to the active metabolite was slow and limited in human and rat brain S9 fractions. Overall, these studies indicate that the potential for oseltamivir and oseltamivir carboxylate to reach the central nervous system in high quantities is low and, together with other analyses and studies, that their involvement in neuropsychiatric events in influenza patients is unlikely.


Aaps Journal | 2017

Metabolic Profiling of Human Long-Term Liver Models and Hepatic Clearance Predictions from In Vitro Data Using Nonlinear Mixed-Effects Modeling

Nicole A. Kratochwil; Christophe Meille; Stephen Fowler; Florian Klammers; Aynur Ekiciler; Birgit Molitor; Sandrine Simon; Isabelle Walter; Claudia McGinnis; Johanna Walther; Brian Leonard; Miriam Triyatni; Hassan Javanbakht; Christoph Funk; Franz Schuler; Thierry Lavé; Neil Parrott

ABSTRACTEarly prediction of human clearance is often challenging, in particular for the growing number of low-clearance compounds. Long-term in vitro models have been developed which enable sophisticated hepatic drug disposition studies and improved clearance predictions. Here, the cell line HepG2, iPSC-derived hepatocytes (iCell®), the hepatic stem cell line HepaRG™, and human hepatocyte co-cultures (HμREL™ and HepatoPac®) were compared to primary hepatocyte suspension cultures with respect to their key metabolic activities. Similar metabolic activities were found for the long-term models HepaRG™, HμREL™, and HepatoPac® and the short-term suspension cultures when averaged across all 11 enzyme markers, although differences were seen in the activities of CYP2D6 and non-CYP enzymes. For iCell® and HepG2, the metabolic activity was more than tenfold lower. The micropatterned HepatoPac® model was further evaluated with respect to clearance prediction. To assess the in vitro parameters, pharmacokinetic modeling was applied. The determination of intrinsic clearance by nonlinear mixed-effects modeling in a long-term model significantly increased the confidence in the parameter estimation and extended the sensitive range towards 3% of liver blood flow, i.e., >10-fold lower as compared to suspension cultures. For in vitro to in vivo extrapolation, the well-stirred model was used. The micropatterned model gave rise to clearance prediction in man within a twofold error for the majority of low-clearance compounds. Further research is needed to understand whether transporter activity and drug metabolism by non-CYP enzymes, such as UGTs, SULTs, AO, and FMO, is comparable to the in vivo situation in these long-term culture models.


Archives of Toxicology | 2017

Humanizing the zebrafish liver shifts drug metabolic profiles and improves pharmacokinetics of CYP3A4 substrates

Kar Lai Poon; Ashley Shu Mei Ng; Wei Huang Goh; Claudia McGinnis; Stephen Fowler; Tom J. Carney; Haishan Wang; Phillip W. Ingham

Abstract Understanding and predicting whether new drug candidates will be safe in the clinic is a critical hurdle in pharmaceutical development, that relies in part on absorption, distribution, metabolism, excretion and toxicology studies in vivo. Zebrafish is a relatively new model system for drug metabolism and toxicity studies, offering whole organism screening coupled with small size and potential for high-throughput screening. Through toxicity and absorption analyses of a number of drugs, we find that zebrafish is generally predictive of drug toxicity, although assay outcomes are influenced by drug lipophilicity which alters drug uptake. In addition, liver microsome assays reveal specific differences in metabolism of compounds between human and zebrafish livers, likely resulting from the divergence of the cytochrome P450 superfamily between species. To reflect human metabolism more accurately, we generated a transgenic “humanized” zebrafish line that expresses the major human phase I detoxifying enzyme, CYP3A4, in the liver. Here, we show that this humanized line shows an elevated metabolism of CYP3A4-specific substrates compared to wild-type zebrafish. The generation of this first described humanized zebrafish liver suggests such approaches can enhance the accuracy of the zebrafish model for toxicity prediction.


Molecular Pharmaceutics | 2013

In vitro to in vivo extrapolation and physiologically based modeling of cytochrome P450 mediated metabolism in beagle dog gut wall and liver.

Aki T. Heikkinen; Stephen Fowler; Lynn Gray; Jia Li; Ying Peng; Preeti Yadava; Aruna Railkar; Neil Parrott

The beagle dog is a widely used in vivo model to guide clinical formulation development and to explore the potential for food effects. However, the results in dogs are often not directly translatable to humans. Consequently, a physiologically based modeling strategy has been proposed, using the dog as a validation step to verify model assumptions before making predictions in humans. One current weakness in this strategy is the lack of validated tools to incorporate gut wall metabolism into the dog model. In this study, in vitro to in vivo extrapolation factors for CYP2B11 and CYP3A12 mediated metabolism were established based on tissue enzyme abundance data reported earlier. Thereafter, physiologically based modeling of intestinal absorption in beagle dog was conducted in GastroPlus using V(max) and K(m) determined in recombinant enzymes as inputs for metabolic turnover. The predicted fraction of absorbed dose escaping the gut wall metabolism (F(g)) of all five reference compounds studied (domperidone, felodipine, nitrendipine, quinidine, and sildenafil) were within a two-fold range of the value estimated from in vivo data at single dose levels. However, further in vivo studies and analysis of the dose-dependent pharmacokinetics of felodipine and nitrendipine showed that more work is required for robust forecasting of nonlinearities. In conclusion, this study demonstrates an approach for prediction of the gut wall extraction of CYP substrates in the beagle dog, thus enhancing the value of dog studies as a component in a strategy for the prediction of human pharmacokinetics.


Xenobiotica | 2015

In vitro profiling of the metabolism and drug–drug interaction of tofogliflozin, a potent and highly specific sodium-glucose co-transporter 2 inhibitor, using human liver microsomes, human hepatocytes, and recombinant human CYP

Mizuki Yamane; Kosuke Kawashima; Koji Yamaguchi; Shunsuke Nagao; Mika Sato; Masayuki Suzuki; Kiyofumi Honda; Hitoshi Hagita; Olaf Kuhlmann; Agnès Poirier; Stephen Fowler; Christoph Funk; Sandrine Simon; Yoshinori Aso; Sachiya Ikeda; Masaki Ishigai

Abstract 1. The metabolism and drug–drug interaction (DDI) risk of tofogliflozin, a potent and highly specific sodium-glucose co-transporter 2 inhibitor, were evaluated by in vitro studies using human liver microsomes, human hepatocytes, and recombinant human CYPs. 2. The main metabolite of tofogliflozin was the carboxylated derivative (M1) in human hepatocytes, which was the same as in vivo. The metabolic pathway of tofogliflozin to M1 was considered to be as follows: first, tofogliflozin was catalyzed to the primary hydroxylated derivative (M4) by CYP2C18, CYP4A11 and CYP4F3B, then M4 was oxidized to M1. 3. Tofogliflozin had no induction potential on CYP1A2 and CYP3A4. Neither tofogliflozin nor M1 had inhibition potential on CYPs, with the exception of a weak CYP2C19 inhibition by M1. 4. Not only are multiple metabolic enzymes involved in the tofogliflozin metabolism, but the drug is also excreted into urine after oral administration, indicating that tofogliflozin is eliminated through multiple pathways. Thus, the exposure of tofogliflozin would not be significantly altered by DDI caused by any co-administered drugs. Also, tofogliflozin seems not to cause significant DDI of co-administered drugs because tofogliflozin has no CYP induction or inhibition potency, and the main metabolite M1 has no clinically relevant CYP inhibition potency.


Xenobiotica | 2018

In vitro metabolism of alectinib, a novel potent ALK inhibitor, in human: contribution of CYP3A enzymes

Toshito Nakagawa; Stephen Fowler; Kenji Takanashi; Kuresh Youdim; Tsuyoshi Yamauchi; Kosuke Kawashima; Mika Sato-Nakai; Li Yu; Masaki Ishigai

Abstract 1. The in vitro metabolism of alectinib, a potent and highly selective oral anaplastic lymphoma kinase inhibitor, was investigated. 2. The main metabolite (M4) in primary human hepatocytes was identified, which is produced by deethylation at the morpholine ring. Three minor metabolites (M6, M1a, and M1b) were also identified, and a minor peak of hydroxylated alectinib (M5) was detected as a possible precursor of M4, M1a, and M1b. 3. M4, an important active major metabolite, was produced and further metabolized to M6 by CYP3A, indicating that CYP3A enzymes were the principal contributors to this route. M5 is possibly produced by CYP3A and other isoforms as the primary step in metabolism, followed by oxidation to M4 mainly by CYP3A. Alternatively, M5 could be oxidized to M1a and M1b via an NAD-dependent process. None of the non-CYP3A-mediated metabolism appeared to be major. 4. In conclusion, this study suggests that involvement of multiple enzymes in the metabolism of alectinib reduces its potential for drug–drug interactions.

Collaboration


Dive into the Stephen Fowler's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masaki Ishigai

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge