Skip to main content
Log in

Factors Governing the Precision of Subvisible Particle Measurement Methods – A Case Study with a Low-Concentration Therapeutic Protein Product in a Prefilled Syringe

  • Research Paper
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose

The current study was performed to assess the precision of the principal subvisible particle measurement methods available today. Special attention was given to identifying the sources of error and the factors governing analytical performance.

Methods

The performance of individual techniques was evaluated using a commercial biologic drug product in a prefilled syringe container. In control experiments, latex spheres were used as standards and instrument calibration suspensions.

Results

The results reported in this manuscript clearly demonstrated that the particle measurement techniques operating in the submicrometer range have much lower precision than the micrometer size-range methods. It was established that the main factor governing the relatively poor precision of submicrometer methods in general and inherently, is their low sampling volume and the corresponding large extrapolation factors for calculating final results.

Conclusions

The variety of new methods for submicrometer particle analysis may in the future support product characterization; however, the performance of the existing methods does not yet allow for their use in routine practice and quality control.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Abbreviations

CC:

Coulter counter

CV:

Coefficient of variance

ECD:

Equivalent circular diameter

MFI:

Micro flow imaging

LO:

Light obscuration

NTA:

Nanoparticle tracking analysis

PFS:

Pre-filled syringe

r.h.:

Relative humidity

RMM:

Resonant mass measurement

RMM (+):

Positively buoyant particles detected by RMM

RMM (−):

Negatively buoyant particles detected by RMM

References

  1. Carpenter JF, Randolph TW, Jiskoot W, Crommelin DJA, Middaugh CR, Winter G, et al. Overlooking subvisible particles in therapeutic protein products: gaps that may compromise product quality. J Pharm Sci. 2009;98(4):1201–5.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  2. Singh SK, Afonina N, Awwad M, Bechtold-Peters K, Blue JT, Chou D, et al. An industry perspective on the monitoring of subvisible particles as a quality attribute for protein therapeutics. J Pharm Sci. 2010;99(8):3302–21.

    Article  CAS  PubMed  Google Scholar 

  3. Rosenberg AS, Verthelyi D, Cherney BW. Managing uncertainty: a perspective on risk pertaining to product quality attributes as they bear on immunogenicity of therapeutic proteins. J Pharm Sci. 2012;101(10):3560–7.

    Article  CAS  PubMed  Google Scholar 

  4. Kiese S, Papppenberger A, Friess W, Mahler H-C. Shaken, not stirred: mechanical stress testing of an IgG1 antibody. J Pharm Sci. 2008;97(10):4347–66.

    Article  CAS  PubMed  Google Scholar 

  5. Mahler H-C, Friess W, Grauschopf U, Kiese S. Protein aggregation: pathways, induction factors and analysis. J Pharm Sci. 2009;98(9):2909–34.

    Article  CAS  PubMed  Google Scholar 

  6. Barnard JG, Singh S, Randolph TW, Carpenter JF. Subvisible particle counting provides a sensitive method of detecting and quantifying aggregation of monoclonal antibody caused by freeze-thawing: insights into the roles of particles in the protein aggregation pathway. J Pharm Sci. 2011;100(2):492–503.

    Article  CAS  PubMed  Google Scholar 

  7. Jiskoot W, Randolph TW, Volkin DB, Middaugh CR, Schoneich C, Winter G, et al. Protein instability and immunogenicity: roadblocks to clinical application of injectable protein delivery systems for sustained release. J Pharm Sci. 2012;101(3):946–54.

    Article  CAS  PubMed  Google Scholar 

  8. Barnard JG, Babcock K, Carpenter JF. Characterization and quantitation of aggregates and particles in interferon- products: potential links between product quality attributes and immunogenicity. J Pharm Sci. 2013;102(3):915–28.

    Article  CAS  PubMed  Google Scholar 

  9. Philo JS. Is any measurement method optimal for all aggregate sizes and types? AAPS J. 2006;8(3):E564–71.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  10. Carpenter JF, Randolph TW, Jiskoot W, Crommelin DJA, Middaugh CR, Winter G. Potential inaccurate quantitation and sizing of protein aggregates by size exclusion chromatography: essential need to use orthogonal methods to assure the quality of therapeutic protein products. J Pharm Sci. 2010;99(5):2200–8.

    Article  CAS  PubMed  Google Scholar 

  11. Demeule B, Palais C, Machaidze G, Gurny R, Arvinte T. New methods allowing the detection of protein aggregates A case study on trastuzumab. MAbs. 2009;1(2):142–50.

    Article  PubMed Central  PubMed  Google Scholar 

  12. Narhi LO, Jiang Y, Cao S, Benedek K, Shnek D. A critical review of analytical methods for subvisible and visible particles. Curr Pharm Biotechnol. 2009;10(4):373–81.

    Article  CAS  PubMed  Google Scholar 

  13. Zölls S, Tantipolphan R, Wiggenhorn M, Winter G, Jiskoot W, Friess W, et al. Particles in therapeutic protein formulations, Part 1: overview of analytical methods. J Pharm Sci. 2012;101(3):914–35.

    Article  PubMed  Google Scholar 

  14. Filipe V, Hawe A, Carpenter JF, Jiskoot W. Analytical approaches to assess the degradation of therapeutic proteins. Trac-Trends Anal Chem. 2013;49:118–25.

    Article  CAS  Google Scholar 

  15. Hamrang Z, Rattray NJW, Pluen A. Proteins behaving badly: emerging technologies in profiling biopharmaceutical aggregation. Trends Biotechnol. 2013;31(8):448–58.

    Article  CAS  PubMed  Google Scholar 

  16. Cao S, Jiang Y, Narhi L. A light-obscuration method specific for quantifying subvisible particles in protein therapeutics. Pharmacopeial Forum. 2010;36(3):10.

    Google Scholar 

  17. Hawe A, Schaubhut F, Geidobler R, Wiggenhorn M, Friess W, Rast M, et al. Pharmaceutical feasibility of sub-visible particle analysis in parenterals with reduced volume light obscuration methods. Eur J Pharm Biopharm. 2013;85(3):1084–7.

    Article  CAS  PubMed  Google Scholar 

  18. Werk T, Volkin DB, Mahler HC. Effect of solution properties on the counting and sizing of subvisible particle standards as measured by light obscuration and digital imaging methods. Eur J Pharm Sci. 2014;53:95–108.

    Article  CAS  PubMed  Google Scholar 

  19. Filipe V, Hawe A, Carpenter JF, Jiskoot W. Analytical approaches to assess the degradation of therapeutic proteins. Trac-Trends Anal Chem. 2013;49:118–25.

    Article  CAS  Google Scholar 

  20. Zhao H, Diez M, Koulov A, Bozova M, Bluemel M, Forrer K. Characterization of aggregates and particles using emerging techniques. In: Mahler H-C, Jiskoot W, editors. Analysis of aggregates and particles in protein pharmaceuticals. New York: Wiley; 2012. p. 133–67.

    Chapter  Google Scholar 

  21. Ripple DC, Dimitrova MN. Protein particles: what we know and what we do not know. J Pharm Sci. 2012;101(10):3568–79.

    Article  CAS  PubMed  Google Scholar 

Download references

ACKNOWLEDGEMENTS AND DISCLOSURES

The authors report no conflict of interest and would like to thank Rita Gruebel for technical support and Felix Heise for helpful discussions.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Atanas V. Koulov.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Fig. S1

Boxplot of CV%, each estimated from 3 random values based on a Poisson distribution with expected number of particles λ and originating number of particles n1. (GIF 71 kb)

High Resolution Image (TIFF 107 kb)

Fig. S2

Pool variability in HIAC and CC. Variability of six independent and identically prepared pools of 10 units of PFS measured in HIAC (a) and CC (b). The large pool size originates low variability in the measurements. (GIF 17 kb)

High Resolution Image (TIFF 25 kb)

Fig. S3

Collage of randomly selected MFI image and larger than 5 μm of the protein product used in this study. Mainly silicon oil-like particles can be observed. (DOCX 407 kb)

Table S1

(DOCX 52 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ríos Quiroz, A., Lamerz, J., Da Cunha, T. et al. Factors Governing the Precision of Subvisible Particle Measurement Methods – A Case Study with a Low-Concentration Therapeutic Protein Product in a Prefilled Syringe. Pharm Res 33, 450–461 (2016). https://doi.org/10.1007/s11095-015-1801-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11095-015-1801-4

KEY WORDS

Navigation