We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Mini Focus Issue: Tiered approach to bioanalysis - Research Article

An industry perspective on tiered approach to the investigation of metabolites in drug development

    Su-Er W Huskey

    Department of Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, One Health Plaza, East Hanover, NJ 07936, USA

    ,
    Wenkui Li

    Department of Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, One Health Plaza, East Hanover, NJ 07936, USA

    ,
    James B Mangold

    Department of Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, One Health Plaza, East Hanover, NJ 07936, USA

    &
    Jimmy Flarakos

    * Author for correspondence

    Department of Drug Metabolism & Pharmacokinetics, Novartis Institutes for Biomedical Research, One Health Plaza, East Hanover, NJ 07936, USA.

    Published Online:https://doi.org/10.4155/bio.14.25

    Background: A tiered approach to drug metabolite measurement and identification is often used industry wide to fulfill regulatory requirements specified in recent US FDA and European Medicines Agency guidance. Although this strategy is structured in its intent it can be customized to address unique challenges which may arise during early and late drug development activities. These unconventional methods can be applied at any stage to facilitate metabolite characterization. Results: Two case studies are described NVS 1 and 2. NVS 1: plasma concentrations, measured using a radiolabeled MS-response factor exploratory method, were comparable to those from a validated bioanalytical method. The NVS 2 example showed how in vitro analysis helped to characterize an unexpectedly abundant circulating plasma metabolite M3. Conclusion: A tiered approach incorporating many aspects of conventional and flexible analytical methodologies can be pulled together to address regulatory questions surrounding drug metabolite characterization.

    References

    • Isoherranen N, Hachad H, Yeung CK, Levy RH. Qualitative analysis of the role of metabolites in inhibitory drug-drug interactions: literature evaluation based on the metabolism and transport drug interaction database. Chem. Res. Toxicol.22(2),294–298 (2009).
    • Hartyánszky I, Kalász H, Adeghate E et al. Active metabolites resulting from decarboxylation, reduction and ester hydrolysis of parent drugs. Curr. Drug Metab.13(6),835–862 (2012).
    • Hutzler J, Obach RS, Dalvie DK, Zientek MA. Strategies for a comprehensive understanding of metabolism by aldehyde oxidase. Expert Opin. Drug Metab. Toxicol.9(2),153–168 (2013).
    • Ito K, Iwatsubo T, Kanamitsu S, Ueda K, Suzuki H, Sugiyama Y. Prediction of pharmacokinetic alterations caused by drug-drug interactions: metabolic interaction in the liver. Pharmacol. Rev.50(3),387–412 (1998).
    • Minagawa T, Nakano K, Furuta S et al. Perspectives on non-clinical safety evaluation of drug metabolites through the JSOT workshop. J. Toxicol. Sci.37(4),667–673 (2012).
    • Obach RS. Pharmacologically active drug metabolites: impact on drug discovery and pharmacotherapy. Pharmacol. Rev.65(2),578–640 (2013).
    • Yu H, Tweedie D. A perspective on the contribution of metabolites to drug drug interaction potential: the need to consider both circulating levels and inhibition potency. Drug Metab. Dispos.41(3),536–540 (2013).
    • Timmerman P, Anders Kall M, Gordon B et al. Best practices in a tiered approach to metabolite quantification: views and recommendations of the European Bioanalysis Forum. Bioanalysis.2(7),1185–1194 (2010).
    • GaoH, Jacobs A, White RE, Booth BP, Obach RS. Meeting report: Metabolites in Safety Testing (MIST) Symposium-Safety Assessment of Human Metabolites: what’s really necessary to ascertain exposure coverage in safety tests? AAPS J.15(4),970–973 (2013).
    • 10  Li W, Zhang J, Tse FL. Strategies in quantitative LC–MS/MS analysis of unstable small molecules in biological matrices. Biomed. Chromatogr.25(1–2),258–277 (2011).
    • 11  D’Esposito F, Tattam BN, Ramzan I, Murray M. A liquid chromatography/electrospray ionization mass spectrometry (LC–MS/MS) assay for the determination of irinotecan (CPT-11) and its two major metabolites in human liver microsomal incubations and human plasma samples. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.875(2),522–530 (2008).
    • 12  Eriksson T, Björkman S. Handling of blood samples for determination of thalidomide. Clin. Chem.43(6 Pt 1),1094–1096 (1997).
    • 13  Sadilkova K, Gospe Jr SM, Hahn SH. Simultaneous determination of alpha-aminoadipic semialdehyde, piperideine-6-carboxylate and pipecolic acid by LC–MS/MS for pyridoxine-dependent seizures and folinic acid-responsive seizures. J. Neurosci. Methods184(1),136–141 (2009).
    • 14  Sadagopan NP, Li W, Cook JA et al. Investigation of EDTA anticoagulant in plasma to improve the throughput of liquid chromatography–tandem mass spectrometric assays. Rapid Commun. Mass Spectrom.17(10),1065–1070 (2003).
    • 15  Yue B, Pattison E, Roberts WL et al. Choline in whole blood and plasma: sample preparation and stability. Clin. Chem.54(3),590–593 (2008).
    • 16  Chen J, Hsieh Y. Stabilizing drug molecules in biological samples. Ther. Drug Monit.27(5),617–624 (2005).
    • 17  Baranda AB, Alonso RM, Jiménez RM, Weinmann W. Instability of calcium channel antagonists during sample preparation for LC–MS-MS analysis of serum samples. Forensic Sci. Int.156(1),23–34 (2006).
    • 18  Hendriks G, Uges DRA, Franke JP. pH adjustment of human blood plasma prior to bioanalytical sample preparation. J. Pharm. Biomed. Anal.47(1),126–133 (2008).
    • 19  Meng M, Reuschel S, Bennett P. Identifying trends and developing solutions for incurred sample reanalysis failure investigations in a bioanalytical CRO. Bioanalysis3(4),449–465 (2011).
    • 20  Jemal M, Ouyang Z, Xia YQ. Systematic LC–MS/MS bioanalytical method development that incorporates plasma phospholipids risk avoidance, usage of incurred sample and well thought-out chromatography. Biomed. Chromatogr.24(1),2–19 (2010).
    • 21  Dell D. Labile metabolites. Chromatographia Supplement59(2),S139–S148 (2004).
    • 22  Krüger R, Vogeser M, Burghardt S, Vogelsberger R, Lackner KJ. Impact of glucuronide interferences on therapeutic drug monitoring of posaconazole by tandem mass spectrometry. Clin. Chem. Lab. Med.48(12),1723–1731 (2010).
    • 23  Li W, Fu Y, Flarakos J, Zhang D. LC–MS bioanalysis of ester prodrugs and other esterase labile molecules. In: Handbook of LC-MS Bioanalysis: Best Practice, Experimental Protocols and Regulations. Li W, Zhang J, Tse F (Eds). Wiley & Sons, Inc., Hoboken, NJ, USA, 431–445 (2013).
    • 24  Vainchtein LD, Rosing H, Mirejovsky D, Lenaz L, Schellens JH, Beijnen JH. Stability experiments in human urine with EO9 (apaziquone): a novel anticancer agent for the intravesical treatment of bladder cancer. J. Pharm. Biomed. Anal.43(1),285–292 (2007).
    • 25  Kamei T, Uchimura T, Nishimiya K, Kawanishi T. Method development and validation of the simultaneous determination of a novel topoisomerase 1 inhibitor, the prodrug, and the active metabolite in human plasma using column-switching LC–MS/MS, and its application in a clinical trial. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.879(30),3415–3422 (2011).
    • 26  Chang Q, Chow MS, Zuo Z. Studies on the influence of esterase inhibitor to the pharmacokinetic profiles of oseltamivir and oseltamivir carboxylate in rats using an improved LC–MS/MS method. Biomed. Chromatogr.23(8),852–857 (2009).
    • 27  Zeng J, Onthank D, Crane P et al. Simultaneous determination of a selective adenosine 2A agonist, BMS-068645, and its acid metabolite in human plasma by liquid chromatography–tandem mass spectrometry – evaluation of the esterase inhibitor, diisopropyl fluorophosphate, in the stabilization of a labile ester-containing drug. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.852(1–2),77–84 (2007).
    • 28  Tang C, Sojinu OS. Simultaneous determination of caffeic acid phenethyl ester and its metabolite caffeic acid in dog plasma using liquid chromatography–tandem mass spectrometry. Talanta94,232–239 (2012).
    • 29  Wang X, Bowman PD, Kerwin SM, Stavchansky S. Stability of caffeic acid phenethyl ester and its fluorinated derivative in rat plasma. Biomed. Chromatogr.21(4),343–350 (2007).
    • 30  Salem II, Alkhatib M, Najib N. LC–MS/MS determination of betamethasone and its phosphate and acetate esters in human plasma after sample stabilization. J. Pharm. Biomed. Anal.56(5),983–991 (2011).
    • 31  Li W, Tse FL. Dried blood spot sampling in combination with LC–MS/MS for quantitative analysis of small molecules. Biomed. Chromatogr.24(1),49–65 (2010).
    • 32  Heinig K, Wirz T, Bucheli F, Gajate-Perez A. Determination of oseltamivir (Tamiflu®) and oseltamivir carboxylate in dried blood spots using offline or online extraction. Bioanalysis3(4),421–372 (2011).
    • 33  D’Arienzo CJ, Ji QC, Discenza L et al. DBS sampling can be used to stabilize prodrugs in drug discovery rodent studies without the addition of esterase inhibitors. Bioanalysis2(8),1415–1422 (2010).
    • 34  Eikel D, Henion JD. Liquid extraction surface analysis (LESA) of food surfaces employing chip-based nano-electrospray mass spectrometry. Rapid Commun. Mass Spectrom.25(16),2345–2354 (2011).
    • 35  Eikel D, Vavrek M, Smith S et al. Liquid extraction surface analysis mass spectrometry (LESA-MS) as a novel profiling tool for drug distribution and metabolism analysis: the terfenadine example. Rapid Commun. Mass Spectrom.25(23),3587–3596 (2011).
    • 36  Schadt S, Kallbach S, Almeida R, Sandel J. Investigation of figopitant and its metabolites in rat tissue by combining whole-body autoradiography with liquid extraction surface analysis mass spectrometry. .Drug Metab. Dispos.40(3),419–425 (2012).
    • 37  Kertesz V, Van Berkel GJ. Automated liquid microjunction surface sampling-HPLC–MS/MS analysis of drugs and metabolites in whole-body thin tissue sections. Bioanalysis5(7),819–826 (2013).
    • 38  Wisztorski M, Fatou B, Franck J et al. Microproteomics by liquid extraction surface analysis: application to FFPE tissue to study the fimbria region of tubo-ovarian cancer. Proteomics Clin. Appl.7(3–4),234–240 (2013).
    • 39  HamiltonR, Garnett W, Kline B. Determination of mean valproic acid serum level by assay of a single pooled sample. Clin. Pharmacol. Ther.29(3),408–413 (1981).
    • 40  Dalvie DK, Zhang C, Chen W, Smolarek T, Obach RS, Loi C-M. Cross species comparison of the metabolism and excretion of zoniporide: contribution of aldehyde oxidase to interspecies differences. Drug Metab. Dispos.38(4),641–654 (2010).
    • 41  Diamond S, Boer J, Maduskuie TP, Falahatpisheh N, Li Y, Yeleswaram S. Species-specific metabolism of SGX523 by aldehyde oxidase and the toxicological implications. Drug Metab. Dispos.38(8),1277–1285 (2010).
    • 101  US FDA. Guidance for Industry: Safety Testing of Drug Metabolites (2008). www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm079266.pdf
    • 102  International Conference of Harmonization M3(R2) Guideline: Guidance on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals (2010). www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm073246.pdf
    • 103  International Conference of Harmonization M3(R2) Guideline: Guidance on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals Questions & Answers (R2) (2012). www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM292340.pdf