Skip to main content

Advertisement

Log in

An improved workflow for the development of MS-compatible liquid chromatography assay purity and purification methods by using automated LC Screening instrumentation and in silico modeling

  • Research Paper
  • Published:
Analytical and Bioanalytical Chemistry Aims and scope Submit manuscript

Abstract

The development of liquid chromatography UV and mass spectrometry (LC-UV–MS) assays in pharmaceutical analysis is pivotal to improve quality control by providing critical information about drug purity, stability, and presence and identity of byproducts and impurities. Analytical method development of these assays is time-consuming, which often causes it to become a bottle neck in drug development and poses a challenge for process chemists to quickly improve the chemistry. In this study, a systematic and efficient workflow was designed to develop purity assay and purification methods for a wide range of compounds including peptides, proteins, and small molecules with MS-compatible mobile phases (MP) by using automated LC screening instrumentation and in silico modeling tools. Initial LC MPs and chromatography column screening experiments enabled quick identification of conditions which provided the best resolution in the vicinity of the target compounds, which is further optimized using computer-assisted modeling (LC Simulator from ACD/Labs). The experimental retention times were in good agreement with the predicted retention times from LC Simulator (ΔtR < 7%). This workflow presents a practical workflow to significantly expedite the time needed to develop optimized LC-UV–MS methods, allowing for a facile, automatic method optimization and reducing the amount of manual work involved in developing new methods during drug development.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Scheme 1
Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Ganorkar SB, Shirkhedkar AA. Design of experiments in liquid chromatography (HPLC) analysis of pharmaceuticals: analytics, applications, implications and future prospects. Rev Anal Chem. 2017;36(3):20160025.

  2. Mennen SM, Alhambra C, Allen CL, Barberis M, Berritt S, Brandt TA, et al. The evolution of high-throughput experimentation in pharmaceutical development and perspectives on the future. Org Process Res Dev. 2019;23(6):1213–42.

    Article  CAS  Google Scholar 

  3. Tome T, Žigart N, Časar Z, Obreza A. Development and optimization of liquid chromatography analytical methods by using AQbD principles: overview and recent advances. Org Process Res Dev. 2019;23(9):1784–802.

    Article  CAS  Google Scholar 

  4. Liu Y, Zhang C, Chen J, Fernandez J, Vellala P, Kulkarni TA, et al. A fully integrated online platform for real time monitoring of multiple product quality attributes in biopharmaceutical processes for monoclonal antibody therapeutics. J Pharm Sci. 2022;111(2):358–67.

    Article  CAS  PubMed  Google Scholar 

  5. Wang Y-L, Liang Y-Z, Chen B-M, He Y-K, Li B-Y, Hu Q-N. LC-DAD-APCI-MS-based screening and analysis of the absorption and metabolite components in plasma from a rabbit administered an oral solution of danggui. Anal Bioanal Chem. 2005;383:247–54.

    Article  CAS  PubMed  Google Scholar 

  6. Schmitz OJ. Klaus Wanner and Georg Höfner (Eds.): Mass spectrometry in medicinal chemistry. Applications in drug discovery. Anal Bioanal Chem. 2008;391(6):2101–2.

  7. Xu C, Khanal S, Pierson NA, Quiroz J, Kochert B, Yang X, et al. Development, validation, and implementation of a robust and quality control-friendly focused peptide mapping method for monitoring oxidation of co-formulated monoclonal antibodies. Anal Bioanal Chem. 2022;414(29–30):8317–30. 

  8. Barrientos RC, Losacco GL, Azizi M, Wang H, Nguyen AN, Shchurik V, et al. Automated hydrophobic interaction chromatography screening combined with in silico optimization as a framework for nondenaturing analysis and purification of biopharmaceuticals. Anal Chem. 2022;94(49):17131–41.

  9. Regalado EL, Haidar Ahmad IA, Bennett R, D’Atri V, Makarov AA, Humphrey GR, et al. The emergence of universal chromatographic methods in the research and development of new drug substances. Acc Chem Res. 2019;52(7):1990–2002.

    Article  CAS  PubMed  Google Scholar 

  10. Haidar Ahmad IA, Kiffer A, Barrientos RC, Losacco GL, Singh A, Shchurik V, et al. In silico method development of achiral and chiral tandem column reversed-phase liquid chromatography for multicomponent pharmaceutical mixtures. Anal Chem. 2022;94(9):4065–71.

    Article  CAS  PubMed  Google Scholar 

  11. Losacco GL, Hicks MB, DaSilva JO, Wang H, Potapenko M, Tsay F-R, et al. Automated ion exchange chromatography screening combined with in silico multifactorial simulation for efficient method development and purification of biopharmaceutical targets. Anal Bioanal Chem. 2022;414(12):3581–91.

    Article  CAS  PubMed  Google Scholar 

  12. Losacco GL, Wang H, Haidar Ahmad IA, DaSilva J, Makarov AA, Mangion I, et al. Enantioselective UHPLC screening combined with in silico modeling for streamlined development of ultrafast enantiopurity assays. Anal Chem. 2021;94(3):1804–12.

    Article  PubMed  Google Scholar 

  13. Haidar Ahmad IA. Automated column screening and computer-assisted modeling for analysis of complex drug samples in pharmaceutical laboratories. Chromatographia. 2022;85(10–11):977–84.

    Article  CAS  Google Scholar 

  14. Zou L, Bennett R, Haidar Ahmad IA, Jocher BM, Zhang L, Bu X, et al. Generic ion chromatography–conductivity detection method for analysis of palladium scavengers in new drug substances. Org Process Res Dev. 2019;23(5):1060–8.

    Article  CAS  Google Scholar 

  15. Ahmad IAH, Bennett R, Makey D, Shchurik V, Lhotka H, Mann BF, et al. In silico method development for the reversed-phase liquid chromatography separation of proteins using chaotropic mobile phase modifiers. J Chromatogr B. 2021;1173: 122587.

    Article  Google Scholar 

  16. Haidar Ahmad IA, Shchurik V, Nowak T, Mann BF, Regalado EL. Introducing multifactorial peak crossover in analytical and preparative chromatography via computer-assisted modeling. Anal Chem. 2020;92(19):13443–51.

    Article  CAS  PubMed  Google Scholar 

  17. Guillarme D, Rouvière F, Heinisch S. Theoretical and practical comparison of RPLC and RPLC× RPLC: how to consider dilution effects and sensitivity in addition to separation power? Anal Bioanal Chem. 2023;415(13):2357–69.

    Article  CAS  PubMed  Google Scholar 

  18. Kalariya PD, Namdev D, Srinivas R, Gananadhamu S. Application of experimental design and response surface technique for selecting the optimum RP-HPLC conditions for the determination of moxifloxacin HCl and ketorolac tromethamine in eye drops. J Saudi Chem Soc. 2017;21:S373–82.

    Article  CAS  Google Scholar 

  19. Haidar Ahmad IA. Automated column screening and computer-assisted modeling for analysis of complex drug samples in pharmaceutical laboratories. Chromatographia. 2022;85(10–11):977–84. 

  20. Van Heukelem L, Thomas CS. Computer-assisted high-performance liquid chromatography method development with applications to the isolation and analysis of phytoplankton pigments. J Chromatogr A. 2001;910(1):31–49.

    Article  PubMed  Google Scholar 

  21. Obradović D, Radan M, Đikić T, Nikolić MP, Oljačić S, Nikolić K. The evaluation of drug-plasma protein binding interaction on immobilized human serum albumin stationary phase, aided by different computational approaches. J Pharm Biomed Anal. 2022;211: 114593.

    Article  PubMed  Google Scholar 

  22. Liapikos T, Zisi C, Kodra D, Kademoglou K, Diamantidou D, Begou O, et al. Quantitative structure retention relationship (QSRR) modelling for analytes’ retention prediction in LC-HRMS by applying different machine Learning algorithms and evaluating their performance. J Chromatogr B. 2022;1191: 123132.

    Article  CAS  Google Scholar 

  23. Samuelsson J, Eiriksson FF, Åsberg D, Thorsteinsdóttir M, Fornstedt T. Determining gradient conditions for peptide purification in RPLC with machine-learning-based retention time predictions. J Chromatogr A. 2019;1598:92–100.

    Article  CAS  PubMed  Google Scholar 

  24. Jovanović M, Rakić T, Jančić-Stojanović B, Ivanović D, Medenica M. Retention mechanism assessment and method development for the analysis of iohexol and its related compounds in hydrophilic interaction liquid chromatography. Anal Bioanal Chem. 2014;406:4217–32.

    Article  PubMed  Google Scholar 

  25. Pappa-Louisi A, Agrafiotou P, Papachristos K. Retention modeling under organic modifier gradient conditions in ion-pair reversed-phase chromatography. Application to the separation of a set of underivatized amino acids. Anal Bioanal Chem. 2010;397:2151–9.

    Article  CAS  PubMed  Google Scholar 

  26. Baeza-Baeza J, García-Alvarez-Coque M. Extension of the linear solvent strength retention model including a parameter that describes the elution strength changes in liquid chromatography. J Chromatogr A. 2020;1615: 460757.

    Article  CAS  PubMed  Google Scholar 

  27. Soriano-Meseguer S, Fuguet E, Abraham MH, Port A, Rosés M. Linear free energy relationship models for the retention of partially ionized acid-base compounds in reversed-phase liquid chromatography. J Chromatogr A. 2021;1635: 461720.

    Article  CAS  PubMed  Google Scholar 

  28. Haddad PR, Taraji M, Szücs R. Prediction of analyte retention time in liquid chromatography. Anal Chem. 2020;93(1):228–56.

    Article  PubMed  Google Scholar 

  29. Besenhard MO, Tsatse A, Mazzei L, Sorensen E. Recent advances in modelling and control of liquid chromatography. Curr Opin Chem Eng. 2021;32: 100685.

    Article  Google Scholar 

  30. Bennett R, Haidar Ahmad IA, DaSilva J, Figus M, Hullen K, Tsay F-R, et al. Mapping the separation landscape of pharmaceuticals: rapid and efficient scale-up of preparative purifications enabled by computer-assisted chromatographic method development. Org Process Res Dev. 2019;23(12):2678–84.

    Article  CAS  Google Scholar 

  31. Szucs R, Brown R, Brunelli C, Heaton JC, Hradski J. Structure driven prediction of chromatographic retention times: applications to pharmaceutical analysis. Int J Mol Sci. 2021;22(8):3848.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Dolan J, Snyder L, Djordjevic N, Hill D, Saunders D, Van Heukelem L, et al. Simultaneous variation of temperature and gradient steepness for reversed-phase high-performance liquid chromatography method development: I. Application to 14 different samples using computer simulation. J Chromatogr A. 1998;803(1–2):1–31.

    Article  CAS  PubMed  Google Scholar 

  33. Melander W, Campbell DE, Horváth C. Enthalpy—entropy compensation in reversed-phase chromatography. J Chromatogr A. 1978;158:215–25.

    Article  CAS  Google Scholar 

  34. Philipsen H, Claessens H, Lind H, Klumperman B, German A. Study on the retention behaviour of low-molar-mass polystyrenes and polyesters in reversed-phase liquid chromatography by evaluation of thermodynamic parameters. J Chromatogr A. 1997;790(1–2):101–16.

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jeremy Manheim.

Ethics declarations

Conflict of interest

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Jeremy Manheim and Andrew N. Singh are the first authors.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Manheim, J., Singh, A.N., Aggarwal, P. et al. An improved workflow for the development of MS-compatible liquid chromatography assay purity and purification methods by using automated LC Screening instrumentation and in silico modeling. Anal Bioanal Chem 416, 1269–1279 (2024). https://doi.org/10.1007/s00216-023-05118-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00216-023-05118-3

Keywords

Navigation