Network


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

Hotspot


Dive into the research topics where Christopher R. Gibson is active.

Publication


Featured researches published by Christopher R. Gibson.


Clinical Pharmacology & Therapeutics | 2015

Physiologically based pharmacokinetic modeling in drug discovery and development: A pharmaceutical industry perspective

Hannah M. Jones; Yuan Chen; Christopher R. Gibson; Tycho Heimbach; Neil Parrott; Sheila Annie Peters; Jan Snoeys; Vijay Upreti; Ming Zheng; Stephen Hall

The application of physiologically based pharmacokinetic (PBPK) modeling has developed rapidly within the pharmaceutical industry and is becoming an integral part of drug discovery and development. In this study, we provide a cross pharmaceutical industry position on “how PBPK modeling can be applied in industry” focusing on the strategies for application of PBPK at different stages, an associated perspective on the confidence and challenges, as well as guidance on interacting with regulatory agencies and internal best practices.


Aaps Journal | 2013

Drug-drug interaction studies: regulatory guidance and an industry perspective.

Thomayant Prueksaritanont; Xiaoyan Chu; Christopher R. Gibson; Donghui Cui; Ka Lai Yee; Jeanine Ballard; Tamara Cabalu; Jerome H. Hochman

Recently, the US Food and Drug Administration and European Medicines Agency have issued new guidance for industry on drug interaction studies, which outline comprehensive recommendations on a broad range of in vitro and in vivo studies to evaluate drug–drug interaction (DDI) potential. This paper aims to provide an overview of these new recommendations and an in-depth scientifically based perspective on issues surrounding some of the recommended approaches in emerging areas, particularly, transporters and complex DDIs. We present a number of theoretical considerations and several case examples to demonstrate complexities in applying (1) the proposed transporter decision trees and associated criteria for studying a broad spectrum of transporters to derive actionable information and (2) the recommended model-based approaches at an early stage of drug development to prospectively predict DDIs involving time-dependent inhibition and mixed inhibition/induction of drug metabolizing enzymes. We hope to convey the need for conducting DDI studies on a case-by-case basis using a holistic scientifically based interrogative approach and to communicate the need for additional research to fill in knowledge gaps in these areas where the science is rapidly evolving to better ensure the safety and efficacy of new therapeutic agents.


Bioorganic & Medicinal Chemistry Letters | 2009

Discovery of GlyT1 inhibitors with improved pharmacokinetic properties

Scott E. Wolkenberg; Zhijian Zhao; David D. Wisnoski; William Leister; Julie A. O’Brien; Wei Lemaire; David L. Williams; Marlene A. Jacobson; Cyrille Sur; Gene G. Kinney; Doug J. Pettibone; Philip R. Tiller; Sheri Smith; Christopher R. Gibson; Bennett Ma; Stacey L. Polsky-Fisher; Craig W. Lindsley; George D. Hartman

Glycine transporter 1 (GlyT1) represents a novel target for the treatment of schizophrenia via the potentiation of glutamatergic NMDA receptors. The discovery of 4,4-disubstituted piperidine inhibitors of GlyT1 which exhibit improved pharmacokinetic properties, including oral bioavailability, is discussed.


Drug Metabolism and Disposition | 2006

Effect of cytochromes P450 chemical inhibitors and monoclonal antibodies on human liver microsomal esterase activity.

Stacey L. Polsky-Fisher; Hong Cao; Ping Lu; Christopher R. Gibson

Selective and nonselective cytochromes P450 (P450) chemical inhibitors and monoclonal antibodies (mAbs) are routinely used to determine the contribution of P450 enzymes involved in the biotransformation of a drug. A fluorometric assay has been established using fluorescein diacetate as a model substrate to determine the effect of some commonly used P450 inhibitors and mAbs on human liver microsomal esterase activity. Of those inhibitors studied, only α-naphthoflavone, clotrimazole, ketoconazole, miconazole, nicardipine, and verapamil significantly inhibited human liver microsomal esterase activity, with apparent IC50 values of 18.0, 20.5, 6.5, 15.0, 19.4, and 5.4 μM, respectively. All of these showed ≥20% inhibition of human liver microsomal esterase activity at concentrations typically used for P450 reaction phenotyping studies, with clotrimazole, miconazole, nicardipine, and verapamil showing >60% inhibition. Unlike the chemical inhibitors, no inhibition of human liver microsomal esterase activity was observed in the presence of mAb to CYP1A2, 2C8, 2C9, 2C19, 2D6, and 3A4. These results suggest that P450 chemical inhibitors are capable of inhibiting human liver microsomal esterase activity and should not be used to assess the role of P450 enzymes in the biotransformation of esters. The lack of inhibition of human liver microsomal esterase activity by P450-specific monoclonal antibodies suggests that they may be used to assess the role of P450 enzymes in the biotransformation of esters. Additional experiments to assess the contribution of oxidative enzymes in the metabolism of esters may include incubations in the presence and absence of β-nicotinamide adenine dinucleotide 2′-phosphate reduced.


ACS Chemical Neuroscience | 2012

Characterization of non-nitrocatechol pan and isoform specific catechol-O-methyltransferase inhibitors and substrates.

Ronald G. Robinson; Sean M. Smith; Scott E. Wolkenberg; Monika Kandebo; Lihang Yao; Christopher R. Gibson; Scott T. Harrison; Stacey L. Polsky-Fisher; James C. Barrow; Peter J. Manley; James Mulhearn; Kausik K. Nanda; Jeffrey W. Schubert; B. Wesley Trotter; Zhijian Zhao; John M. Sanders; Robert F. Smith; Debra McLoughlin; Sujata Sharma; Dawn L. Hall; Tiffany L. Walker; Jennifer L. Kershner; Neetesh Bhandari; Pete H. Hutson; Nancy Sachs

Reduced dopamine neurotransmission in the prefrontal cortex has been implicated as causal for the negative symptoms and cognitive deficit associated with schizophrenia; thus, a compound which selectively enhances dopamine neurotransmission in the prefrontal cortex may have therapeutic potential. Inhibition of catechol-O-methyltransferase (COMT, EC 2.1.1.6) offers a unique advantage, since this enzyme is the primary mechanism for the elimination of dopamine in cortical areas. Since membrane bound COMT (MB-COMT) is the predominant isoform in human brain, a high throughput screen (HTS) to identify novel MB-COMT specific inhibitors was completed. Subsequent optimization led to the identification of novel, non-nitrocatechol COMT inhibitors, some of which interact specifically with MB-COMT. Compounds were characterized for in vitro efficacy versus human and rat MB and soluble (S)-COMT. Select compounds were administered to male Wistar rats, and ex vivo COMT activity, compound levels in plasma and cerebrospinal fluid (CSF), and CSF dopamine metabolite levels were determined as measures of preclinical efficacy. Finally, novel non-nitrocatechol COMT inhibitors displayed less potent uncoupling of the mitochondrial membrane potential (MMP) compared to tolcapone as well as nonhepatotoxic entacapone, thus mitigating the risk of hepatotoxicity.


Future Medicinal Chemistry | 2015

Optimization of human dose prediction by using quantitative and translational pharmacology in drug discovery

Tjerk Bueters; Christopher R. Gibson; Sandra A. G. Visser

In this perspective article, we explain how quantitative and translational pharmacology, when well-implemented, is believed to lead to improved clinical candidates and drug targets that are differentiated from current treatment options. Quantitative and translational pharmacology aims to build and continuously improve the quantitative relationship between drug exposure, target engagement, efficacy, safety and its interspecies relationship at every phase of drug discovery. Drug hunters should consider and apply these concepts to develop compounds with a higher probability of interrogating the clinical biological hypothesis. We offer different approaches to set an initial effective concentration or pharmacokinetic-pharmacodynamic target in man and to predict human pharmacokinetics that determine together the predicted human dose and dose schedule. All concepts are illustrated with ample literature examples.


The Journal of Clinical Pharmacology | 2014

Prednisone has no effect on the pharmacokinetics of CYP3A4 metabolized drugs – midazolam and odanacatib

Eugene E. Marcantonio; Jeanine Ballard; Christopher R. Gibson; Kelem Kassahun; Jairam Palamanda; Cuyue Tang; Raymond Evers; Chengcheng Liu; Stefan Zajic; Chantal Mahon; Kate Mostoller; David Hreniuk; Anish Mehta; Denise Morris; John A. Wagner; S. Aubrey Stoch

We evaluated the effect of prednisone on midazolam and odanacatib pharmacokinetics. In this open‐label, 2‐period crossover study in healthy male subjects, midazolam 2 mg was administered (Day −1) followed by odanacatib 50 mg (Day 1) during Part 1. In Period 2, prednisone 10 mg once daily (qd) was administered on Days 1–28; odanacatib was co‐administered on Day 14 and midazolam 2 mg was co‐administered on Days 1 and 28. Subjects were administered midazolam 2 mg on Days 42 and 56. Safety and tolerability were assessed throughout the study. A physiologically‐based pharmacokinetic (PBPK) model was also built. There were 15 subjects enrolled; mean age was 31 years. The odanacatib AUC0−∞ GMR (90% CI) [odanacatib + prednisone (Day 14, Period 2)/odanacatib alone (Day 1, Period 1] was 1.06 (0.96, 1.17). AUC0−∞ GMR (90%CI) [midazolam + prednisone (Day 28, Period 2)/midazolam alone (Day −1, Period 1] was 1.08 (0.93,1.26). There were no serious AEs or AEs leading to discontinuation. PBPK modeling showed that prednisone does not cause significant effects on the exposure of sensitive CYP3A4 substrates in vivo at therapeutic doses. Co‐administration of prednisone 10 mg qd had no effect on pharmacokinetics of either odanacatib 10 mg or midazolam 2 mg.


Drug Metabolism and Disposition | 2011

Novel Cytochrome P450-Mediated Ring Opening of the 1,3,4-Oxadiazole in Setileuton, a 5-Lipoxygenase Inhibitor

Cheri Maciolek; Bennett Ma; Karsten Menzel; Sebastien Laliberte; Kevin P. Bateman; Paul Krolikowski; Christopher R. Gibson

Setileuton [4-(4-fluorophenyl)-7-[({5-[(1S)-1-hydroxy-1-(trifluoromethyl)propyl]-1,3,4-oxadiazol-2-yl}amino)methyl]-2H-1-benzopyran-2-one] is a selective inhibitor of the 5-lipoxygenase enzyme, which is under investigation for the treatment of asthma and atherosclerosis. During the development of setileuton, a metabolite (M5) was identified in incubations with rat, dog, and human liver microsomes that represented the addition of 18 Da to the 1,3,4-oxadiazole portion of the molecule. Based on mass spectral data, a ring opened structure was proposed and confirmed through comparison with a synthetic standard. The metabolic ring opening was examined in vitro in rat liver microsomes and was determined to be mediated by cytochrome P450s (P450s). Upon examination of the specific P450s involved using cDNA-expressed rat P450s, it was shown that CYP1A2 likely was the major isoform contributing to the formation of M5. Studies using stable labeled molecular oxygen and water demonstrated that the oxygen was incorporated from molecular oxygen, rather than water, and confirmed that the metabolic formation was oxidative. An alternative, comparatively slow pathway of chemical hydrolysis also was identified and described. Three potential mechanisms for the two-step metabolic ring opening of the 1,3,4-oxadizole are proposed.


Bioorganic & Medicinal Chemistry Letters | 2016

Synthesis and optimization of N-heterocyclic pyridinones as catechol-O-methyltransferase (COMT) inhibitors.

Zhijian Zhao; Scott T. Harrison; Jeffrey W. Schubert; John M. Sanders; Stacey L. Polsky-Fisher; Nanyan Rena Zhang; Debra McLoughlin; Christopher R. Gibson; Ronald G. Robinson; Nancy Sachs; Monika Kandebo; Lihang Yao; Sean M. Smith; Pete H. Hutson; Scott E. Wolkenberg; James C. Barrow

A series of N-heterocyclic pyridinone catechol-O-methyltransferase (COMT) inhibitors were synthesized. Physicochemical properties, including ligand lipophilic efficiency (LLE) and clogP, were used to guide compound design and attempt to improve inhibitor pharmacokinetics. Incorporation of heterocyclic central rings provided improvements in physicochemical parameters but did not significantly reduce in vitro or in vivo clearance. Nevertheless, compound 11 was identified as a potent inhibitor with sufficient in vivo exposure to significantly affect the dopamine metabolites homovanillic acid (HVA) and dihydroxyphenylacetic acid (DOPAC), and indicate central COMT inhibition.


Clinical Pharmacology & Therapeutics | 2018

Application of Physiologically‐Based Pharmacokinetic Modeling to Predict Pharmacokinetics in Healthy Japanese Subjects

Yuki Matsumoto; Tamara Cabalu; Punam Sandhu; Georgy Hartmann; Takashi Iwasa; Hiroyuki Yoshitsugu; Christopher R. Gibson; Naoto Uemura

Pharmacokinetics (PKs) in Japanese healthy subjects were simulated for nine compounds using physiologically based PK (PBPK) models parameterized with physicochemical properties, preclinical absorption, distribution, metabolism, and excretion (ADME) data, and clinical PK data from non‐Japanese subjects. For each dosing regimen, 100 virtual trials were simulated and predicted/observed ratios for peak plasma concentration (Cmax) and area under the curve (AUC) were calculated. As qualification criteria, it was prespecified that >80% of simulated trials should demonstrate ratios to observed data ranging from 0.5–2.0. Across all compounds and dose regimens studied, 93% of simulated Cmax values in Japanese subjects fulfilled the criteria. Similarly, for AUC, 77% of single‐dosing regimens and 100% of multiple‐dosing regimens fulfilled the criteria. In summary, mechanistically incorporating the appropriate ADME properties into PBPK models, followed by qualification using non‐Japanese clinical data, can predict PKs in the Japanese population and lead to efficient trial design and conduct of Japanese phase I studies.

Collaboration


Dive into the Christopher R. Gibson's collaboration.

Researchain Logo
Decentralizing Knowledge