Skip to main content

Regulatory Aspects of Freeze-Drying

  • Protocol
  • First Online:
Lyophilization of Pharmaceuticals and Biologicals

Part of the book series: Methods in Pharmacology and Toxicology ((MIPT))

Abstract

Freeze-drying or lyophilization has become an important formulation and stabilization strategy in the pharmaceutical industry for drugs, vaccines, antibodies, and other biological materials, for improving their long-term stability. For biological products whose storage in aqueous solution results in instability, freeze-drying prolongs the shelf-life by inhibiting chemical, physical, and microbiological pathways that occur in the presence of moisture. Due to the complex multi-step processes involved in lyophilization, it is crucial to ensure a robust and efficient cycle, which in turn ensures that high-quality products are consistently manufactured that meet the labelled therapeutic claim over the shelf life of the product. Most lyophilized small molecule drug products are usually very simple formulations containing the active ingredient, water, and in some cases, buffer. Even with these simple formulations, a number of post-approval manufacturing process changes occur occasionally due to their failure to consistently meet desirable target qualities such as reconstitution time, low moisture content, and stability. In most cases, the root causes for such failure are identified as poor lyophilization cycles or a lack of operator training.

An overall understanding of the lyophilization process, as well as use of optimum lyophilization process parameters, would provide the Agency with a greater level of knowledge and help with the approval process. This will also facilitate a greater understanding between the FDA and industrial sponsors on the regulatory aspects of lyophilized drug products.

This chapter provides an overview of the application of risk analysis to defining and measuring the critical parameters of lyophilization, for which a specific level of scrutiny is required, to ensure a robust and controlled process. Particular focus will be on the key steps of risk management through ICH Q9 applied to the freeze-drying process, with specific examples, as applied to the lyophilization of a model monoclonal antibody. The application of various process analytical technology (PAT) tools for monitoring and controlling the various processes, with specific examples, will also be discussed.

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

Access this chapter

Subscribe and save

Springer+ Basic
$34.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 149.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 199.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  1. Kasper JC, Friess W (2011) The freezing step in lyophilization: physico-chemical fundamentals, freezing methods and consequences on process performance and quality attributes of biopharmaceuticals. Eur J Pharm Biopharm 78:248–263

    Article  CAS  Google Scholar 

  2. Vasconcelos T, Sarmento B, Costa P (2007) Solid dispersions as strategy to improve oral bioavailability of poor water souluble drugs. Drug Discov Today 12:1068–1075

    Article  CAS  Google Scholar 

  3. Reichert JM (2016) Antibodies to watch in 2016. MAbs 8(2):197–204. https://doi.org/10.1080/19420862.2015.1125583

    Article  Google Scholar 

  4. Pikal MJ, Rambhatla S, Ramot R (2002) The impact of the freezing stage in lyophilization: effects of the ice nucleation temperature on process design and product quality. Am Pharm Rev 5:48–53

    Google Scholar 

  5. Tang X, Pikal M (2004) Design of freeze-drying processes for pharmaceuticals: practical advice. Pharm Res 21:191–200

    Article  CAS  Google Scholar 

  6. Chang BS, Kendrick BS, Carpenter JS (1996) Surface-induced denaturation of proteins during freezing and its inhibition by surfactants. J Pharm Sci 85(12):1325–1330

    Article  CAS  Google Scholar 

  7. Bedu-Addo FK (2004) Understanding lyophilization formulation development. Pharmaceutical Technology – Lyophilization pp 10–18

    Google Scholar 

  8. Food and Drug Administration (2009) Q8 (R2) pharmaceutical development. https://www.fda.gov/downloads/drugs/guidances/ucm073507.pdf. Accessed on 23 May 2017

  9. Jameel F, Khan MA (2009) Quality-by-design as applied to the development and manufacturing of a lyophilized protein product. Am Pharm Rev (November/December): 20–24

    Google Scholar 

  10. Guidance for industry: PAT — a framework for innovative pharmaceutical development, manufacturing and quality assurance (2004) https://www.fda.gov/downloads/drugs/guidances/ucm070305.pdf. Accessed on 23 May 2017

  11. Woodcock J (2005) Pharmaceutical quality in the 21st century – an integrated systems approach. In: AAPS Workshop on Pharmaceutical Quality Assessment – A Science and Risk-Based CMC Approach in the 21st Century

    Google Scholar 

  12. Guidance for industry; Q9 quality risk management (2006) https://www.fda.gov/downloads/Drugs/Guidances/ucm073511.pdf. Accessed on 23 May 2017

  13. Guidance for industry: Q10 pharmaceutical quality systems (2009) https://www.fda.gov/downloads/Drugs/.../Guidances/ucm073517.pdf. Accessed 23 May 2017

  14. Nasr MM (2011) Implementation of Quality by Design (QbD) – current perspectives on opportunities and challenges topic introduction and ICH update. Presented at Advisory Committee for Pharmaceutical Science and Clinical Pharmacology, US Food and drug Administration, 27 July, 2011

    Google Scholar 

  15. Rathore SA, Winkle H (2009) Quality by Design for biopharmaceuticals. Nat Biotechnol 27(1):26–34

    Article  CAS  Google Scholar 

  16. Kenett RS, Kenett DA (2008) Quality by Design applications in biosimilar pharmaceutical products. Accred Qual Assur 13:681–690

    Article  CAS  Google Scholar 

  17. Awotwe-Otoo D, Agarabi C, Wu GK et al (2012) Quality by design: impact of formulation variables and their interactions on quality attributes of a lyophilized monoclonal antibody. Int J Pharm 438(1-2):167–175

    Article  CAS  Google Scholar 

  18. Tang X, Nail SL, Pikal MJ (2006) Evaluation of manometric temperature measurement, a process analytical technology tool for freeze-drying: Part I, Product temperature measurement. AAPS PharmSciTech 7(1):E1–E9

    Article  Google Scholar 

  19. Nail SL, Searles JA (2008) Elements of quality by design in development and scale-up of freezed-dried parenterals. Bio Pharm Int 21(1):44–50

    CAS  Google Scholar 

  20. Kasper JC, Winter G, Freiss W (2013) Recent advances and further challenges in lyophilization. Eur J Pharm Biopharm 85:162–169

    Article  CAS  Google Scholar 

  21. U.S. Food and Drug Administration. Guide to inspections of lyophilization of parenterals. http://www.fda.gov/ICECI/Inspections/Inspectionguides/UCM074909.htm. Accessed 3 Nov 2017

  22. Skrabanja ATP, de Meere ALJ, Rien d R, van der Oetelaar PJM (1994) Lyophilization of biotechnology products. PDA. J Pharm Sci Technol 48(6):311–317

    CAS  Google Scholar 

  23. Meister E, Gieseler H (2009) Freeze-dry microscopy of protein/sugar mixtures: drying behavior, interpretation of collapse temperatures and a comparison to corresponding glass transition data. J Pharm Sci 98(9):3072–3087

    Article  CAS  Google Scholar 

  24. Mujat M, Greco K, Galbally-Kinney KL, Hammer DX, Ferguson RD, Iftimia N et al (2012) Optical coherence tomography-based freeze-drying microscopy. Biomed Opt Express 3(1):55–63

    Article  Google Scholar 

  25. Greco K, Mujat M, Galbally-Kinney KL, Hammer DX, Ferguson RD, Iftimia N et al (2013) Accurate prediction of collapse temperature using optical coherence tomography-based freeze-drying microscopy. J Pharm Sci 102(6):1773–1785

    Article  CAS  Google Scholar 

  26. Awotwe-Otoo D, Agarabi C, Read EK et al (2013) Impact of controlled ice nucleation on process performance and quality attributes of a lyophilized monoclonal antibody. Int J Pharm 450(1-2):70–78

    Article  CAS  Google Scholar 

  27. Konstantinidis AK, Luu W, Otten L, Nail SL, Sever RR (2011) Controlled nucleation in freeze-drying: effects on pore size in the dried product layer, mass transfer resistance and primary drying rate. J Pharm Sci 100:3453–3347

    Article  CAS  Google Scholar 

  28. Searles JA, Carpenter T, Randolph TW (2001) The ice nucleation temperature determines the primary drying rate of lyophilization for samples frozen on a temperature controlled shelf. J Pharm Sci 90:860–871

    Article  CAS  Google Scholar 

  29. Tang X, Nail SL, Pikal MJ (2005) Freeze-drying process design by manometric temperature measurement: design of a smart freeze dryer. Pharm Res 22:685–700

    Article  CAS  Google Scholar 

  30. De Beer TRM, Vercruysse P, Burggraeve A, Quinten T, Ouyang J, Zhang X, Vervaet C, Remon JP, Baeyens WR (2009) In-line and real-time process monitoring of a freeze-drying process using Raman and NIR spectroscopy as complementary process analytical technology (PAT) tools. J Pharm Sci 98(9):3430–3446

    Article  Google Scholar 

  31. Patel SM, Pikal MJ (2009) Process analytical technologies (PAT) in freeze-drying of parenteral products. Pharm Dev Technol 14(6):567–587

    Article  CAS  Google Scholar 

  32. Awotwe-Otoo D, Agarabi C, Khan MA (2014) An integrated process analytical technology (PAT) approach to monitoring the effect of supercooling on lyophilization product and process parameters of model monoclonal antibody formulations. J Pharm Sci 103:2042–2052

    Article  CAS  Google Scholar 

  33. Patel SM, Doen T, Pikal MJ (2010) Determination of end-point of primary drying in freeze-drying process control. AAPS PharmSciTech 11(1):73–84

    Article  CAS  Google Scholar 

  34. De Beer TR, Allesø M, Goethals F et al (2007) Implementation of a process analytical technology system in a freeze-drying process using Raman spectroscopy for in-line process monitoring. Anal Chem 79(21):7992–8003

    Article  Google Scholar 

  35. Schneid SC, Gieseler H, Kessler WJ, Luthra SA, Pikal MJ (2011) Tunable diode laser absorption spectroscopy (TDLAS) as a residual moisture monitor for the secondary drying stage of freeze drying. AAPS PharmSciTech 12(1):379–387

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David Awotwe-Otoo .

Editor information

Editors and Affiliations

Additional information

Disclaimer: The views expressed in this chapter are those of the authors and do not necessarily represent the position of the Agency.

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Awotwe-Otoo, D., Khan, M. (2019). Regulatory Aspects of Freeze-Drying. In: Ward, K., Matejtschuk, P. (eds) Lyophilization of Pharmaceuticals and Biologicals. Methods in Pharmacology and Toxicology. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-8928-7_8

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-8928-7_8

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-8927-0

  • Online ISBN: 978-1-4939-8928-7

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics