Elsevier

Journal of Chromatography B

Volume 1139, 15 February 2020, 121885
Journal of Chromatography B

Development and validation of a mass spectrometric method to determine the identity of rituximab based on its microheterogeneity profile

https://doi.org/10.1016/j.jchromb.2019.121885Get rights and content

Highlights

  • First reported validation design of an identity MS-UPLC method for biotherapeutic analysis.

  • Validation was designed according to ICH Q2 (R1).

  • The proposed validation could be applied to other similar analytical methods for mAbs analysis.

Abstract

Analytical methods have been considered the “eyes” for development, characterization and batch releasing of biotherapeutics over the past 40 years. One of the most powerful analytical platform for biotherapeutic analysis is mass spectrometry coupled to liquid chromatography (LC-MS). Due to its wide flexibility and instrumental configurations, LC-MS can determine different physicochemical attributes of proteins, e.g. molecular mass, primary sequence, and posttranslational modifications. Intact molecular mass analysis of therapeutic proteins is essential to confirm their identity. Analytical methods must be validated to support drug quality information during its approval process. Although there are international guidelines that provide general information on validation of analytical methods, practical examples about the design, selection of validation attributes and acceptance criteria of identity LC-MS methods are scarce. Here, according to the recommendations of Q2R1 ICH guideline, we showcase the validation of an LC-MS-TOF method to identity rituximab by determining its intact and deglycosylated molecular mass profiles. The proposed method specifically identified the m/z profile and deconvoluted mass profile of rituximab from deglycosylated rituximab and from excipient blank (specificity) with a maximum error of 76.63 ppm (accuracy) and a maximum Relative Standard Deviation (RSD) of 0.00315% (precision). Besides, the system suitability test, which was based on the expected mass value of the mass calibrator, confirmed the reliability of the analytical results. In summary, validation showed that the proposed method is suitable for identifying rituximab based on its glycosylated (intact) and deglycosylated mass profile.

Introduction

Batch-to-batch consistency of biotherapeutics is demanded by regulatory agencies during the approval process of any biotherapeutic, given that it is crucial to guarantee product quality and to ensure the expected efficacy and safety [1], [2], [3], [4]. Batch reproducibility is a valuable piece of information of the Quality Module of the Common Technical Document (CTD), the document that follows the ICH guidelines used to submit drug product information to regulatory agencies for approval purposes [5]. Also, Quality Module contains the evidence of the characterization assays that describe the physicochemical and functional properties of the biotherapeutic, which are taken into account to identify critical quality attributes (CQAs) and quality specifications of the drug product [6]. Notwithstanding, some CQAs must be supported by cutting-edge analytical technology due to the complexity of biotherapeutics.

It is well-known that most of the therapeutic recombinant proteins are subjected to posttranslational modifications during biosynthesis and to chemical modifications during purification, formulation and storage [7], [8], [9]; both processes confer the characteristic microheterogeneity profile to each biotherapeutic. Microheterogeneity of monoclonal antibodies (mAbs) is mainly related to a conserved N-glycosylations at Asp 297 on both heavy chains, where three different oligosaccharide structure could be attached: high mannose, complex and hybrid, with or without a core of fucosylation (F) and/or sialic acid (S) [10], [11]. In consequence, glycosylated mAbs exhibit different pairs of glycan structures, namely G0(F), G1(F), G2(F), G2S1(F) and G2S2(F) as described elsewhere [12]. On the other hand, analysis of deglycosylated mAbs can easily detect isoforms related to one or two additional C-Lys residues at C-terminal of heavy chains, or spontaneous glutamic acid cyclization (pyroglutamic acid) of Gln residues at N-terminal of light or heavy chain (pQ isoform) [13]. Oxidation, deamination, isomerization and proteolytic cleavage of some amino acid residues could be also detected [14].

The combination of posttranslational and chemical modifications gives the characteristic molecular mass profile of biotherapeutics, which regulatory agencies strongly recommend to characterize considering its high impact on safety (immunogenicity) and efficacy (pharmacodynamics and pharmacokinetics) [15], [16]. In this sense, it is well known that mAbs with a higher content of fucosylated isoforms exhibit less Antibody-dependent Cell-mediated Cytotoxicity (ADCC), which might compromise efficacy [17]. In contrast, extra C-terminal lysine residues induce basic variants and increase isoelectric point values, but it does not alter in vivo activity given that extra C-terminal lysines are removed by endogenous carboxypeptidases [18], [19], [20].

Several orthogonal analytical methods based on cutting-edge technologies have been rapidly adopted by pharmaceutical industries for characterization and batch releasing of biotherapeutics. Among them, LC-MS has been evolving to become a confident high-throughput technology routinely employed for characterization and quality control of therapeutic proteins [21], [22], [23]. For example, intact mass analysis of monoclonal antibodies by LC-MS allows determining the microheterogeneity profile based on their posttranslational and chemical modifications [24], [25], [26], [27]. Notwithstanding, despite MS determines an absolute characteristic of the analyte (Molecular Mass), validation of LC-MS methods is mandatory when used for pharmaceutical purposes. Nowadays, regulatory agencies have published guidelines on validation of LC-MS methods for determining the identity of therapeutic proteins, but a practical validation strategy has not been published yet. Here, we describe the development and validation of an LC-MS for determining the identity of a monoclonal antibody based on its intact (glycosylated) mass according to Q2R1 ICH guideline using rituximab as model.

Section snippets

Chemicals and reagents

Solvents and the pH modifier used to prepare chromatographic solutions were MS grade: formic acid (FA) and acetonitrile (AcN) were purchased from Thermo Fisher Scientific (MA, USA), and water from Honeywell® (NJ, USA). The spectrometric mass corrector [Glu1]-fibrinopeptide B (Glufib) was obtained from Waters® (MA, USA). Deglycosylated rituximab samples were prepared using sequencing grade PNGase F, which was acquired from New England Biolabs (MA, USA). Deglycosylation procedure also involved

Method development

The first step to develop an identity MS method comprises (i) tuning the mass spectrometer acquisition parameters, (ii) the ion source parameters and (iii) determining the concentration of the analyte samples to obtain suitable m/z signals, whose signal/noise ratio should be higher than 10 [31], [32]. This value is highly recommended to reduce the variability of method accuracy and precision. In this sense, the optimal m/z signal of rituximab samples was obtained at 2 mg/mL employing the MS

Conclusion

International guidelines, such as ICH Q2 (R1), provide valuable suggestions to perform the validation of pharmaceutical analytical methods. Notwithstanding, the design should be adapted depending on the know-how of the analytical method developer and the intended purpose of the analytical method. Here, we describe the design and performance of an MS-UPLC method for determining the identity of rituximab based on its microheterogeneity (glycosylated and deglycosylated variants). The obtained

Declaration of Competing Interest

Authors declare no conflict of interest.

Acknowledgements

This work was carried out with equipment of “Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) para Farmoquímicos y Biotecnológicos”, LANSEIDI-FarBiotec-CONACyT, which is part of Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI)-IPN”.

References (37)

  • A.S. Rathore et al.

    Process development in the QbD paradigm: Role of process integration in process optimization for production of biotherapeutics

    Biotechnol. Prog.

    (2016)
  • International Council for Harmonisation (ICH)

    ICH guideline Q11 on development and manufacture of drug substances (chemical entities and biotechnological/ biological entities)

    (2012)
  • International Council for Harmonisation (ICH)

    ICH guideline M4 on the organisation of the common technical document for the registration of pharmaceuticals for human use

    (2016)
  • International Council for Harmonisation (ICH)

    ICH guideline Q6B on specifications: test procedures and acceptance criteria for biotechnological/biological products

    (1999)
  • S.K. Gupta et al.

    Glycosylation control technologies for recombinant therapeutic proteins

    Appl. Microbiol. Biotechnol.

    (2018)
  • K. Sankar

    Prediction of methionine oxidation risk in monoclonal antibodies using a machine learning method

    MAbs

    (2018)
  • B. Sissolak

    Impact of mammalian cell culture conditions on monoclonal antibody charge heterogeneity: an accessory monitoring tool for process development

    J. Ind. Microbiol. Biotechnol.

    (2019)
  • M. Bas

    Fc sialylation prolongs serum half-life of therapeutic antibodies

    J. Immunol.

    (2019)
  • Cited by (9)

    View all citing articles on Scopus
    1

    Both authors contributed equally to this work.

    View full text