We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
PerspectiveFree Access

Lessons learned from regulatory submissions involving endogenous therapeutic analyte bioanalysis

    Chongwoo Yu

    *Author for correspondence: Tel.: +1 301 796 2335;

    E-mail Address: chongwoo.yu@fda.hhs.gov

    Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS), Center for Drug Evaluation & Research (CDER), US FDA), Silver Spring, MD 20993, USA

    ,
    Wenlei Jiang

    Office of Research & Standards (ORS), Office of Generic Drugs (OGD), CDER, US FDA, Silver Spring, MD 20993, USA

    ,
    Murali Matta

    Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS), Center for Drug Evaluation & Research (CDER), US FDA), Silver Spring, MD 20993, USA

    ,
    Rong Wang

    Office of Bioequivalence (OB), OGD, CDER, US FDA, Silver Spring, MD 20993, USA

    ,
    Sam Haidar

    Office of Study Integrity & Surveillance (OSIS), OTS, CDER, US FDA, Silver Spring, MD 20993, USA

    &
    Hyeonglim Seo

    Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS), Center for Drug Evaluation & Research (CDER), US FDA), Silver Spring, MD 20993, USA

    Department of Chemistry & Biochemistry, University of California San Diego, La Jolla, CA 92093, USA

    Published Online:

    Endogenous therapeutic analytes include hormones, neurotransmitters, vitamins, fatty acids and inorganic elements that are naturally present in the body because either the body produces them or they are present in the normal diet. The accurate measurement of endogenous therapeutic analytes poses a challenge when the administered exogenous therapeutic analyte and its endogenous counterpart cannot be distinguished. In this article, real case examples with endogenous therapeutic analyte bioanalysis during drug development in support of regulatory submissions are collected and presented. The article highlights common challenges encountered and lessons learned related to bioanalysis of endogenous therapeutic analytes and provides practical tips and strategies to consider from a regulatory perspective.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. US Food and Drug Administration. Draft guidance for industry: bioequivalence studies with pharmacokinetic endpoints for drugs submitted under an ANDA (2021). www.fda.gov/media/87219/download
    • 2. Hirsch IB, Juneja R, Beals JM, Antalis CJ, Wright EE. The evolution of insulin and how it informs therapy and treatment choices. Endocr. Rev. 41(5), 733–755 (2020).
    • 3. Oscarsson J, Hurt-Camejo E. Omega-3 fatty acids eicosapentaenoic acid and docosahexaenoic acid and their mechanisms of action on apolipoprotein B-containing lipoproteins in humans: a review. Lipids Health Dis 16, 149 (2017).
    • 4. US Food and Drug Administration. Guidance for Industry: Bioanalytical Method Validation (2018). www.fda.gov/media/70858/download •• Provides regulatory recommendation on bioanalysis, including that of endogenous analytes.
    • 5. Yu C, Bashaw ED. Regulatory perspective of biomarker bioanalysis during drug development. Bioanalysis 11(7), 607–610 (2019). •• Regulatory perspective on endogenous biomarker bioanalysis during drug development and relevant case examples.
    • 6. BioPharma Services, Inc. Bioanalysis considerations for endogenous substance drug products. www.biopharmaservices.com/blog/bioanalysis-considerations-for-endogenous-substance-drug-products/
    • 7. Dissanayake S. Assessing the bioequivalence of analogues of endogenous substances (‘endogenous drugs’): considerations to optimize study design. Br. J. Clin. Pharmacol. 69(3), 238–244 (2010). • Shares some strategies to overcome inherent biases in bioequivalence study of endogenous drugs, including approaches employed in bioanalysis.
    • 8. International Conference on Harmonisation. ICH Guidance for Industry: M10 Bioanalytical Method Validation and Study Sample Analysis (2022). https://www.fda.gov/media/162903/download •• Provides internally harmonized guideline in bioanalysis, including that of endogenous analytes.
    • 9. US Food and Drug Administration. NDA 203098 clinical pharmacology review (2012). www.accessdata.fda.gov/drugsatfda_docs/nda/2013/203098Orig1s000ClinpharmR.pdf
    • 10. US Food and Drug Administration. NDA 203098 other reviews (2012). www.accessdata.fda.gov/drugsatfda_docs/nda/2013/203098Orig1s000OtherR.pdf
    • 11. US Food and Drug Administration. NDA 205488 other reviews (2014). www.accessdata.fda.gov/drugsatfda_docs/nda/2014/205488Orig1s000OtherR.pdf
    • 12. Yin A, Htun M, Swerdloff RS et al. Reexamination of pharmacokinetics of oral testosterone undecanoate in hypogonadal men with a new self-emulsifying formulation. J. Androl. 33(2), 190–201 (2012).
    • 13. US Food and Drug Administration. NDA 213953 Multi-disciplinary Review and Evaluation (2021). www.accessdata.fda.gov/drugsatfda_docs/nda/2022/213953Orig1s000MultidisciplineR.pdf
    • 14. Lachance S, Dhingra O, Bernstein J et al. Importance of measuring testosterone in enzyme-inhibited plasma for oral testosterone undecanoate androgen replacement therapy clinical trials. Future Sci. OA 1(4), FSO55 (2015).
    • 15. US Food and Drug Administration. FDA Briefing Package for Advisory Committee Meeting for NDA 206089 (2014). https://wayback.archive-it.org/7993/20170113050245/http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/ReproductiveHealthDrugsAdvisoryCommittee/UCM414593.pdf
    • 16. US Food and Drug Administration. Product Specific Guidance on Calcitriol Capsules, Recommended July 2008 (2010). www.accessdata.fda.gov/drugsatfda_docs/psg/Calcitriol_cap%20_18044_RC7-08.pdf
    • 17. Jenkinson C, Taylor AE, Hassan-Smith ZK et al. High throughput LC–MS/MS method for the simultaneous analysis of multiple vitamin D analytes in serum. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1014, 56–63 (2016).
    • 18. Thakare R, Chhonker YS, Gautam N, Alamoudi JA, Alnouti Y. Quantitative analysis of endogenous compounds. J. Pharm. Biomed. Anal. 128, 426–437 (2016).
    • 19. US FDA. Draft Product Specific Guidance on Conjugated Estrogens (2014). https://www.accessdata.fda.gov/drugsatfda_docs/psg/Conjugated_estrogens_004782_RC12-14.pdf
    • 20. Patrick A, Maxim N, Cabooter D. Strategies for the quantification of endogenously present small molecules in biological samples. LCGC Asia Pacific 22, 354–363 (2019).
    • 21. Wakamatsu A, Ochiai S, Suzuki E et al. Proposed selection strategy of surrogate matrix to quantify endogenous substances by Japan Bioanalysis Forum DG2015-15. Bioanalysis 10(17), 1349–1360 (2018).
    • 22. Jenkinson C, Taylor AE, Hassan-Smith ZK et al. High throughput LC–MS/MS method for the simultaneous analysis of multiple vitamin D analytes in serum. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1014, 56–63 (2016).
    • 23. Jones BR, Schultz GA, Eckstein JA, Ackermann BL. Surrogate matrix and surrogate analyte approaches for definitive quantitation of endogenous biomolecules. Bioanalysis 4(19), 2343–2356 (2012). • Discusses the things to consider in using surrogate matrix and surrogate analyte approaches for definitive quantitation of endogenous biomolecules, with case examples.
    • 24. Agrawal K, Voggu RR, Pisek D et al. A validated surrogate analyte LC–MS/MS assay for quantification of endogenous cortisol in human whole blood. J. Pharm. Biomed. Anal. 198, 114028 (2021).
    • 25. Liu L, Cui Z, Deng Y, Dean B, Hop CECA, Liang X. Surrogate analyte approach for quantitation of endogenous NAD(+) in human acidified blood samples using liquid chromatography coupled with electrospray ionization tandem mass spectrometry. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1011, 69–76 (2016).
    • 26. Florian J, Gershuny V, Sun Q et al. Considerations for use of pharmacodynamic biomarkers to support biosimilar development – (III) a randomized trial with interferon beta-1a products. Clin. Pharmacol. Ther. 113(2), 339–348 (2023).
    • 27. Khamis MM, Adamko DJ, El-Aneed A. Strategies and challenges in method development and validation for the absolute quantification of endogenous biomarker metabolites using liquid chromatography-tandem mass spectrometry. Mass Spectrom. Rev. 40(1), 31–52 (2021).
    • 28. Huang S, Chen G, Ye N et al. Solid-phase microextraction: an appealing alternative for the determination of endogenous substances – a review. Anal. Chim. Acta 1077, 67–86 (2019).
    • 29. Yuan J, Li A, Chen T, Du J, Ma A, Pan J. Micelle-dominated distribution strategy for non-matrix matched calibration without an internal standard: ‘extract-and-shoot’ approach for analyzing hydrophilic targets in blood and cell samples. Anal. Chim. Acta 1102, 24–35 (2020).
    • 30. Song X, Huang X. Recent developments in microextraction techniques for detection and speciation of heavy metals. Adv. Sample Prep. 2, 100019 (2022).
    • 31. Yu C, Bashaw E. Regulatory perspective of bioanalysis from a clinical pharmacology reviewer standpoint: do you see what I see? Bioanalysis 6(4), 505–510 (2014). •• Shares a regulatory perspective on bioanalysis from a clinical pharmacology reviewer standpoint, with relevant case examples.
    • 32. Klont F, Hopfgartner G. Mass spectrometry based approaches and strategies in bioanalysis for qualitative and quantitative analysis of pharmaceutically relevant molecules. Drug Discov. Today Technol. 40, 64–68 (2021).