Skip to main content
Log in

Best Practices for Drug Substance Stress and Stability Studies During Early Stage Development. Part III—How to Make Science- and Risk-based Stability Testing Decisions for Drug Substance Batches Produced after Manufacturing Process Changes

  • Research Article
  • Published:
Journal of Pharmaceutical Innovation Aims and scope Submit manuscript

Abstract

During phase 1 and phase 2 drug development (early stage drug development), it is normal to continuously improve a manufacturing process, with changes made to the synthetic pathway, reagents, reaction conditions, crystallization parameters, drying conditions, or manufacturing equipment or scale or site. These manufacturing process changes (“process changes” used thereafter) may or may not affect quality attributes such as impurities, and quality attribute changes may or may not affect drug substance (DS) stability. But a common misconception is that almost all process changes and/or quality attribute changes affect DS stability, and a new (or repeat) stability study is conducted for the DS batch produced after process changes. This misconception is clearly refuted by our many years of DS stability experience. To understand how process changes might affect DS stability, we compiled and analyzed manufacturing processes, quality test results, and stability data for 48 batches from seven drug substances in recent development. Of these 48 batches, the seven first DS clinical batches were used as references against which the other respective 41 batches, which were produced after process changes, were compared for changes in manufacturing processes, quality test results, and stability data. This comparison showed that the chemical and physical stability of 36 (of the 41) batches was not affected by process changes, and the chemical or physical stability of the other 5 batches was affected by residual inorganic impurities, significant amounts of water or residual solvents, or significant changes in DS particle size distribution or surface area. These quality attributes that affect stability are called stability-related quality attributes (SRQAs). A new (or repeat) stability study is warranted only if process changes significantly affect SRQAs. We have established a procedure to systematically assess changes in manufacturing process and quality attributes (particularly impurity profiles), to identify SRQAs (risk assessment), and to make science- and risk-based stability testing decisions on whether and how stability testing for new DS batches should be conducted (risk management).

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.

Fig. 1

Similar content being viewed by others

Abbreviations

AC:

Accelerated Storage Condition (e.g., 40 °C/75 % RH)

BfArM:

Bundesinstitut für Arzneimittel und Medizinprodukte (German Health Authority)

DS (API):

Drug Substance (or Active Pharmaceutical Ingredient)

HT/HH:

High Temperature and High Humidity (e.g., 70 °C/75 % RH)

EMA:

European Medicines Agency

GMP:

Good Manufacturing Practice

ICH:

International Conference on Harmonisation

IMPD:

Investigational Medicinal Product Dossier

IND:

Investigational New Drug

LT:

Long-term storage condition, e.g., 25 °C/60 % RH

LT/AC:

Long-Term Storage Condition and Accelerated Storage Condition (e.g., 25 °C/60 % RH and 40 °C/75 % RH)

PFOS:

Powder For Oral Solution (DS in bottle, without excipients)

US FDA:

United States Food and Drug Administration

References

  1. Chan Li Q, Qiu F, Cohen K, Tougas T, Li J, McCaffrey J, et al. Best practices for drug substance stress and stability studies during early-stage development, Part I—conducting drug substance solid stress to support phase Ia clinical Trials. J Pharm Innov (JPI). 2012;7(3):214–24.

    Google Scholar 

  2. Chan Li Q, Qiu F, Cohen K, Tougas T, Li J, McCaffrey J, et al. Best practices for drug substance stress and stability studies during early-stage development Part II—conducting abbreviated long term and accelerated stability testing on the first clinical drug substance batch to confirm and adjust the drug substance retest period/powder for oral solution shelf life. J Pharm Innov (JPI). 2013;8(1):56–65.

    Article  Google Scholar 

  3. International Conference on Harmonisation Guidance (ICH) Q1B: Stability testing: photostability testing of new drug substances and products, November 1996

  4. Colgan ST, Whipple RD, Watson TJ, Nosal R, Beaman JV, DeAntonis DM. The application of quality by design's science and risk based concepts to API stability strategies. September: AAPS Stability Workshop; 2009.

    Google Scholar 

  5. Colgan ST, Watson TJ, Whipple RD, Nosal R, Beaman JV, De Antonis DM. The application of science and risk based concepts to drug substance stability strategies. J Pharm Innov (JPI). 2012;7(3–4):205–13. doi:10.1007/s12247-012-9135-9.

    Article  Google Scholar 

  6. Acken B, Alasandro M, Colgan S. Paul Curry, Frank Diana, Q. Chan Li, Z. Jane Li, Tony Mazzeo, Andy Rignall, Z. Jessica Tan, Robert Timpano. Early development GMPs for stability—an industry perspective (Part IV). Pharmaceutical Technology. 2012;29(9):64–70.

    Google Scholar 

  7. J.V. Beaman. Stability testing—doing everything or doing the right thing? Pharm. Rev. Apr July/August 2010; 73–76

  8. International Conference on Harmonisation Guidance (ICH) Q9: Quality risk management, dated 9 November 2005

  9. International Conference on Harmonisation Guidance (ICH) Q10: Pharmaceutical quality system, dated 4 June 2008

  10. International Conference on Harmonisation Guidance (ICH) Q11: Development and manufacture of drug substances (Chemical entities and biotechnological/biological entities), dated 1 May 2012

  11. International Conference on Harmonisation Guidance (ICH) Q3A(R2): Impurities in New Drug Substances, dated October 2006

  12. US FDA. Guidance for industry: INDs for Phase 2 and Phase 3 Studies Chemistry, Manufacturing, and Controls Information, May 2003. http://www.fda.gov/cder/guidance./index.htm

  13. Canada GUIDANCE DOCUMENT Quality (Chemistry and Manufacturing) Guidance: clinical trial applications (CTAs) for pharmaceuticals, Effective Date 2009/06/01. http://www.hc-sc.gc.ca/dhp-mps/alt_formats/hpfb-dgpsa/pdf/prodpharma/qual_cta_dec-eng.pdf

  14. European Medicines Agency (EMA): Guideline on the requirements to the chemical and pharmaceutical quality documentation concerning investigational medicinal products in clinical trials—CHMP/QWP/185401/2004 Final, 31 March 2006. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003484.pdf

Download references

Acknowledgments

Thanks to John Smoliga, Thomas Offerdahl, Jane Li, Soojin Kim, Bing-shou Yang, Terrence Tougas, Christian Kulinna, and many BI colleagues for constructive discussions and contributions. Special thanks go to Mr. Gordon Hansen, Dr. Chris Senanayake, Dr. Keith Horspool, and Ms. Patricia Watson for management support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Q. Chan Li.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Li, Q.C., Qiu, F., McWilliams, W. et al. Best Practices for Drug Substance Stress and Stability Studies During Early Stage Development. Part III—How to Make Science- and Risk-based Stability Testing Decisions for Drug Substance Batches Produced after Manufacturing Process Changes. J Pharm Innov 8, 229–239 (2013). https://doi.org/10.1007/s12247-013-9163-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12247-013-9163-0

Keywords

Navigation