Elsevier

Biologicals

Volume 43, Issue 6, November 2015, Pages 457-473
Biologicals

Subvisible (2–100 μm) particle analysis during biotherapeutic drug product development: Part 2, experience with the application of subvisible particle analysis

https://doi.org/10.1016/j.biologicals.2015.07.011Get rights and content

Abstract

Measurement and characterization of subvisible particles (including proteinaceous and non-proteinaceous particulate matter) is an important aspect of the pharmaceutical development process for biotherapeutics. Health authorities have increased expectations for subvisible particle data beyond criteria specified in the pharmacopeia and covering a wider size range. In addition, subvisible particle data is being requested for samples exposed to various stress conditions and to support process/product changes. Consequently, subvisible particle analysis has expanded beyond routine testing of finished dosage forms using traditional compendial methods. Over the past decade, advances have been made in the detection and understanding of subvisible particle formation. This article presents industry case studies to illustrate the implementation of strategies for subvisible particle analysis as a characterization tool to assess the nature of the particulate matter and applications in drug product development, stability studies and post-marketing changes.

Introduction

The compendial method for subvisible particle testing, based on USP <788> and <787>, uses light obscuration for monitoring particles having an equivalent circular diameter (ECD) of ≥10 μm and ≥25 μm [1], [2]. As discussed in a previous manuscript [3], health authorities often request testing beyond the limitations of the compendial method and investigation of particles smaller than 10 μm for more complex biotherapeutic parenteral drug products, which may have the propensity to form proteinaceous particulate matter. Consequently, the application of particle analysis across the entire subvisible particle range has become a key consideration during the development of a biotherapeutic drug product [3], [4], [5], [6], [7], [8], [9], [10], [11], [12], [13], [14], [15], [16], [17]. In this article, we provide examples demonstrating how a variety of subvisible particle analysis techniques can be applied in practice during clinical and commercial development. The term “subvisible” applies to particulate matter of the size range defined as 2–100 μm, which may be proteinaceous or non-proteinaceous.

Subvisible particle analysis requires significant long-term data under actual conditions of drug product storage, with material made at different times and from different facilities, to understand the drug product and method variability. The key to understanding such variability in the particles and amounts seen is to have sufficient data to understand product trends in subvisible particulate matter and the ability to determine if the particulate matter is protein-based or originates from other intrinsic or extrinsic species that are also counted in quantitative subvisible particle assessments [18], [19], [20]. Collection of information on particle morphology and composition may be additional components of particle analysis and trending during development.

The case studies presented are a compilation of the selected experiences of the companies that participated in the preparation of this manuscript, and illustrate ways in which this strategy can be applied. The case studies are organized into two sections: (1) Particle Characterization and (2) Applications of Particle Characterization in Drug Product Development, Stability Studies and Post-Marketing Changes.

While the information presented is illustrative of the types of approaches being taken to evaluate subvisible particles also below 10 μm, the examples are not intended to prescribe strategies or the application of any specific techniques considered necessary to meet requirements in relation to regulatory applications for product development or licensure.

Section snippets

Overall conclusions

In recent years, regulatory expectations for product characterization of more complex biotherapeutic parenteral drug products, which may have the propensity to form proteinaceous particulate matter through self-association, have expanded to extend subvisible particle testing beyond the compendial limits into the 2–10 μm range and to include orthogonal methods for characterization. This regulatory expectation includes collection of data on supporting development studies, drug product clinical

Acknowledgments

The authors thank the following individuals for generated data and/or providing valuable insight into the preparation of this manuscript: Dean C. Ripple, NIST; John Ferbas, Nic Angell, Yijia Jiang, Cynthia Li, Keith Kelley, Nancy Jiao, Amgen; Aniello Regine, Horst Bierau, Merck Serono; Yoen Joo Kim, Roja Narwal, Min Zhu, MedImmune; Andrew Werner, John Dougherty, William Weiss, Eli Lilly & Company; Vincent Laperle, Justin Wright, Jean-Bernard Hamel, Becton Dickinson; Michael Adler, Angela

References (47)

  • K. Wuchner et al.

    Development of a microflow imaging assay to characterize protein particulates during storage of a high concentration IgG1 monoclonal antibody Solution

    J Pharm Sci

    (2010)
  • S. Zölls et al.

    How subvisible particles become invisible-relevance of the refractive index for protein particle analysis

    J Pharm Sci

    (2013)
  • D.K. Sharma et al.

    Quantification and characterization of subvisible proteinaceous particles in opalescent mAb formulations using micro-flow imaging

    J Pharm Sci

    (2010)
  • H. Mach et al.

    The use of flow cytometry for the detection of subvisible particles in therapeutic protein formulations

    J Pharm Sci

    (2011)
  • D.B. Ludwig et al.

    Flow cytometry: a promising technique for the study of silicone oil-induced particulate formation in protein formulations

    Anal Biochem

    (2011)
  • R. Thirumangalathu et al.

    Silicone oil and agitation induced aggregation of a monoclonal antibody in aqueous solution

    J Pharm Sci

    (2009)
  • A.K. Tyagi et al.

    IgG particle formulation during filling pump operation: a case study of heterogeneous nucleation on stainless steel nanoparticles

    J Pharm Sci

    (2009)
  • E.Y. Chi et al.

    Heterogeneous nucleation-controlled particulate formation of recombinant human platelet-activating factor acetylhydrolase in pharmaceutical formulation

    J Pharm Sci

    (2005)
  • C. Kalonia et al.

    Radar chart array analysis to visualize effects of formulation variables on IgG1 particle formation as measured by multiple analytical techniques

    J Pharm Sci

    (2013)
  • H.C. Mahler et al.

    Protein aggregation: pathways, induction factors and analysis

    J Pharm Sci

    (2009)
  • L.S. Jones et al.

    Silicone oil induced aggregation of proteins

    J Pharm Sci

    (2005)
  • S. Kiese et al.

    Shaken, not stirred: mechanical stress testing of an IgG1 antibody

    J Pharm Sci

    (2008)
  • K. Auge et al.

    Demonstrating the stability of albinterferon alfa-2b in the presence of silicone oil

    J Pharm Sci

    (2011)
  • Cited by (59)

    View all citing articles on Scopus
    1

    Current address: MacroGenics, Inc., Rockville, MD, USA.

    2

    Current address: Amgen, West Greenwich, RI, USA.

    3

    Current address: AN Biologics Consulting LLC, PA, USA.

    View full text