Archives

  • 2025-11
  • 2025-10
  • 2025-09
  • 2025-04
  • 2025-03
  • 2025-02
  • 2025-01
  • 2024-12
  • 2024-11
  • 2024-10
  • 2024-09
  • 2024-08
  • 2024-07
  • 2024-06
  • 2024-05
  • 2024-04
  • 2024-03
  • 2024-02
  • 2024-01
  • 2023-12
  • 2023-11
  • 2023-10
  • 2023-09
  • 2023-08
  • 2023-07
  • 2023-06
  • 2023-05
  • 2023-04
  • 2023-03
  • 2023-02
  • 2023-01
  • 2022-12
  • 2022-11
  • 2022-10
  • 2022-09
  • 2022-08
  • 2022-07
  • 2022-06
  • 2022-05
  • 2022-04
  • 2022-03
  • 2022-02
  • 2022-01
  • 2021-12
  • 2021-11
  • 2021-10
  • 2021-09
  • 2021-08
  • 2021-07
  • 2021-06
  • 2021-05
  • 2021-04
  • 2021-03
  • 2021-02
  • 2021-01
  • 2020-12
  • 2020-11
  • 2020-10
  • 2020-09
  • 2020-08
  • 2020-07
  • 2020-06
  • 2020-05
  • 2020-04
  • 2020-03
  • 2020-02
  • 2020-01
  • 2019-12
  • 2019-11
  • 2019-10
  • 2019-09
  • 2019-08
  • 2019-07
  • 2019-06
  • 2019-05
  • 2019-04
  • 2018-11
  • 2018-10
  • 2018-07
  • (S)-Mephenytoin: A Gold-Standard CYP2C19 Substrate for In...

    2025-10-04

    (S)-Mephenytoin: A Gold-Standard CYP2C19 Substrate for Intestinal Organoid Metabolism Studies

    Principle Overview: Harnessing (S)-Mephenytoin in Cytochrome P450 Metabolism Research

    (S)-Mephenytoin, chemically known as (5S)-5-ethyl-3-methyl-5-phenyl-2,4-imidazolidinedione, has emerged as the gold-standard substrate for evaluating mephenytoin 4-hydroxylase activity, specifically the cytochrome P450 isoform CYP2C19. Its unique metabolic pathway—undergoing N-demethylation and 4-hydroxylation—makes it an indispensable probe in oxidative drug metabolism, particularly for anticonvulsive drug metabolism and broader pharmacokinetic studies. The capacity to quantify its conversion by CYP2C19 underpins not only robust in vitro CYP enzyme assays but also provides a direct window into CYP2C19 genetic polymorphism and inter-individual metabolic variability.

    Traditional models, such as animal systems and Caco-2 cells, fall short due to species differences or suboptimal enzyme expression. Recent advances in human induced pluripotent stem cell (hiPSC)-derived intestinal organoids, as detailed in the seminal 2025 study by Saito et al., have propelled CYP2C19 substrate research into a new era. These organoid models not only recapitulate the cellular diversity and transporter/enzyme activity of native human intestine but also support long-term propagation and functional maturation, enabling more predictive human-relevant drug metabolism assessments.

    As a highly pure (98%), well-characterized compound with favorable solubility (up to 25 mg/ml in DMSO or DMF), (S)-Mephenytoin is ideal for both routine and advanced pharmacokinetic profiling. Its kinetic parameters—Km of 1.25 mM and Vmax values of 0.8–1.25 nmol/min/nmol P450—allow for sensitive detection of CYP2C19 activity and facilitate cross-study comparability.

    Step-by-Step Workflow: Integrating (S)-Mephenytoin into Intestinal Organoid CYP2C19 Assays

    1. Preparation and Storage

    • Dissolve (S)-Mephenytoin in DMSO (recommended) at up to 25 mg/ml. For best results, prepare fresh aliquots for each experiment; long-term storage of solutions is not recommended due to potential degradation.
    • Store crystalline (S)-Mephenytoin at -20°C in tightly sealed containers. Shipments should arrive on blue ice to preserve integrity.

    2. Culturing hiPSC-Derived Intestinal Organoids

    • Follow the direct 3D cluster culture protocol described by Saito et al. (2025) to generate expandable hiPSC-derived intestinal organoids (iPSC-IOs).
    • Differentiate organoids into mature intestinal epithelial cells (IECs) by seeding onto a 2D monolayer. Confirm the presence of enterocyte markers and CYP2C19 activity prior to drug metabolism assays.

    3. CYP2C19 Enzyme Assay Setup

    • Prepare IEC monolayers in appropriate media, ensuring confluency and viability.
    • Add (S)-Mephenytoin at a range of concentrations (e.g., 0.5–2 mM) to capture kinetic parameters. Include cytochrome b5 if assessing maximal metabolic rates.
    • Incubate for defined timepoints (commonly 15–60 minutes) at 37°C, sampling supernatant at intervals for quantification of 4-hydroxy-mephenytoin product.

    4. Quantification and Data Analysis

    • Analyze samples via LC-MS/MS or HPLC, using internal standards for accurate quantification of (S)-Mephenytoin and its metabolites.
    • Calculate enzyme kinetics (Km, Vmax) and compare to established benchmarks (Km ~1.25 mM; Vmax 0.8–1.25 nmol/min/nmol P450).

    Advanced Applications and Comparative Advantages: Beyond Conventional Models

    The use of (S)-Mephenytoin in hiPSC-derived intestinal organoids represents a major leap for in vitro CYP2C19 substrate studies. Unlike Caco-2 cells—which underexpress key cytochrome P450 enzymes—or animal models subject to interspecies metabolic discrepancies, intestinal organoids recapitulate native human enzyme expression and transporter activity, as shown in the referenced 2025 study.

    Key advantages include:

    • Precision in CYP2C19 Polymorphism Research: By leveraging patient-specific hiPSC lines, researchers can model genetic variants in CYP2C19 and directly assess their impact on (S)-Mephenytoin metabolism, supporting precision pharmacogenomics.
    • High Predictive Value for Human Pharmacokinetics: The kinetic parameters obtained using intestinal organoids closely mirror in vivo metabolism, enhancing the translational relevance of preclinical drug metabolism enzyme substrate studies.
    • Scalability and Cryopreservation: Organoids can be expanded and cryopreserved without loss of function, enabling standardized, high-throughput pharmacokinetic screening.

    These strengths are highlighted in the article “Precision CYP2C19 Substrate for Organoid Research”, which complements this workflow by offering detailed protocols and troubleshooting advice for organoid-based CYP2C19 assays. In contrast, “A Precision CYP2C19 Substrate for In Vitro Assays” extends the discussion to advanced enzyme assay formats, including microsomal and recombinant systems, allowing researchers to benchmark their results across diverse platforms. Finally, “(S)-Mephenytoin in Human Intestinal Organoids: Redefining Models” explores translational and assay design implications, reinforcing the comparative advantages of organoid models in drug metabolism studies.

    Troubleshooting & Optimization: Maximizing CYP2C19 Assay Performance

    • Low Metabolite Yield: If poor 4-hydroxy-mephenytoin formation is observed, verify organoid differentiation is complete and that CYP2C19 is expressed at functional levels (via RT-qPCR or immunostaining). Consider supplementing cultures with cytochrome b5, which has been shown to enhance Vmax values (up to 1.25 nmol/min/nmol P450).
    • Compound Solubility Issues: Always use freshly prepared DMSO or DMF solutions and avoid exceeding the recommended 25 mg/ml concentration. Ensure DMSO content in the final assay is ≤1% to prevent cytotoxicity or enzyme inhibition.
    • Batch Variability: Standardize organoid passage number and differentiation duration. Use pooled organoid batches or replicate patient-derived lines to minimize biological variability and capture the influence of CYP2C19 genetic polymorphism.
    • Assay Sensitivity: Employ highly sensitive detection methods (LC-MS/MS preferred) and include calibration curves to ensure reliable quantitation at low substrate concentrations.
    • Compound Stability: Store (S)-Mephenytoin powder at -20°C, protect from light, and avoid repeated freeze-thaw cycles. Prepare fresh working solutions immediately prior to use to maintain integrity.

    For more hands-on troubleshooting and protocol refinement, readers are encouraged to consult the organoid-specific troubleshooting guide, which details common pitfalls and solutions for maximizing metabolic readouts in these advanced systems.

    Future Outlook: (S)-Mephenytoin and the Next Generation of Translational Drug Metabolism Models

    The integration of (S)-Mephenytoin as a CYP2C19 substrate in hiPSC-derived intestinal organoids is redefining the landscape of human drug metabolism research. As organoid technology matures, we anticipate a surge in high-throughput, patient-specific pharmacokinetic studies, enabling rapid screening of drug candidates and elucidation of genotype-phenotype relationships in oxidative drug metabolism. The continued evolution of single-cell analytics, CRISPR-based gene editing, and multi-omics readouts will further empower researchers to dissect the nuances of CYP2C19 activity—transforming how we predict and personalize anticonvulsive drug metabolism.

    Moreover, as highlighted by “Redefining Human Drug Metabolism”, the translational impact of these models extends beyond academic research, informing regulatory strategies, guiding clinical trial design, and supporting the development of safer, more effective therapeutics.

    In conclusion, (S)-Mephenytoin remains an essential tool for scientists seeking reproducible, human-relevant data in CYP2C19 substrate studies, especially within advanced intestinal organoid systems. By combining robust workflows, rigorous troubleshooting, and next-generation models, researchers can unlock new frontiers in cytochrome P450 metabolism and pharmacokinetic science.