0% found this document useful (0 votes)
19 views7 pages

24-6 RFQ 2 Tsang Part 1

Uploaded by

Maged Magdy
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
19 views7 pages

24-6 RFQ 2 Tsang Part 1

Uploaded by

Maged Magdy
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 7

Volume 4 • Number 2

Pharmaceutical stability testing, Part 1:


An overview of stability

A regulating body’s approval of a proposed drug substance retest period and pro-
posed drug product shelf life depends on the data generated from stability testing.
The stability data of pharmaceutical products are crucial because they correlate
to the quality, safety, and efficacy of the product. This article, the first of three on
pharmaceutical stability testing, provides readers with an overview of stability and
its importance in drug safety and highlights the common pitfalls of stability testing
Kerry Tsang, MSc, programs. The second article addresses stress testing, and the third, bracketing and
BA Mod matrixing designs for stability testing. The series is based on current stability guide-
lines from the International Council for Harmonisation (ICH).

Keywords – deficiencies, stability, testing

Introduction and defines the retest period for the drug


The stability of the drug substance and substance.
drug product in the pharmaceutical setting
Sherri N. Thrower,
BSc correlates to the quality, safety, and efficacy Stability testing programs are costly and
of these highly regulated pharmaceutical can be time-consuming for manufacturers.
products. In this article, we will discuss However, they generate valuable data that
the specific principles defined in the ICH can significantly benefit the manufacturer
Q1A(R2) guideline on stability testing over time and can support their continuous
protocol and explore how those principles production process improvement strategies.
contribute to the reliability of stability test- The data can benefit the company long-
ing and the safety of medications.1 term by providing years worth of data that
can support an extended expiry/shelf life
Stability, as defined in the ICH Q1A(R2) showing that the substance or product is
guideline, measures how pharmaceutical stable over a significant period of time.
products maintain their quality over time
under various environmental factors such as A full glossary of terms, including drug
temperature, humidity, and light encoun- substance, drug product, shelf life, and retest
tered during transportation and storage period, which are used the preceding text, is
before consumers can use the product.1 at the end of this article.
Stability testing is considered a routine
procedure during product development. It The importance of stability
establishes the shelf life of the drug product The purpose of stability testing is to ensure
and the recommended storage conditions that the final drug product is safe for

RegulatoryFocus.org June 2024 29


Volume 4 • Number 2 Pharmaceutical stability testing, Part 1: An overview of stability

patient use and to provide assurance for the end user extraction of chemical substances from the closure com-
that the drug product will remain at an acceptable level ponents or any adsorption of the product into the clo-
of quality while it is available on the market. Drug sure parts. At a minimum, a suitably selected container
manufacturers must take precautions to offset the en- should protect the drug product from physical damage,
vironmental factors or variables that can affect product biological contamination, and external influences, such
stability. as heat and light, that could alter the properties of the
product.
It is crucial when testing for drug substance stability to
also establish the retest date (see Glossary) for when the In addition, the container closure system would also be
material should be reexamined to ensure its suitability subjected to a range of package stability tests, including
and potency for subsequent use in the drug product checks for the presence of extractables and leachables,
has been maintained. If a batch of drug substance is container closure integrity, and conditions during ship-
destined for use in the manufacturing of a given drug ping distribution. Quality control tests of the pharma-
product, then the result of the retest should remain ceutical packaging materials will vary depending on the
within established specifications. The ICH Q1A(R2) type of packaging selected.
and ICH Q1E guidelines provide recommendations on
how to establish a drug substance retest date.1,2 Similarly, any potential incompatibility between the
drug substance and all packaging components should be
One of the main objectives of drug product stability identified. The drug substance’s reactivity to the packag-
testing is to determine the product shelf life, which is ing will determine the type of packaging that is needed
a compulsory regulatory label component that must be for storage. Once the reactivity parameters have been
displayed on the outer packaging of the product. The identified, they should be routinely tested to ensure
expiration date (see Glossary) ensures the quality and their consistency and acceptability for continued use of
potency of the product will remain consistent within the packaging during storage.
the established shelf life, provided the product is stored
according to the recommended conditions outlined in A range of adverse effects, such as the loss of potency in an
ICH Q1A(R2).1 The shelf life is established based on active drug substance, can occur if drug substance stability
the stability test data and is vital to obtaining regulatory is not achieved and maintained throughout the specified
approval from health agencies. shelf life. For example, aspirin is hydrolyzed in the human
body during normal consumption for an indicated condi-
Another important aspect of stability testing is deter- tion. However, if the tablets are stored in moist or humid
mining the suitable packaging components for the drug conditions, they could begin hydrolyzing before being used
substance or drug product. The ICH Q1A(R2) guide- by patients and become less potent and therefore less effec-
line recommends that stability testing be conducted on tive when eventually used as a treatment.
the dosage form packaged in the proposed commercial
container closure system (see Glossary) so the data gener- Types of stability
ated can reflect a real-time condition.1 Pharmaceutical stability can be divided into two
categories: premarket (developmental) and marketed
Typically, the drug product samples for these stability (commercial). The former aims to generate data to sup-
studies are kept upright rather than inverted or on their port ongoing clinical trials. The latter ensures stability
sides, which would represent the worst-case This is to assurance of postapproval batches for monitoring long-
allow the product a full interaction with the container term stability. In either case, stability-indicating tests
and would help determine whether the contact be- are performed on drug substances and drug products to
tween the drug product and the closure would result in establish the retest period and shelf life.

RegulatoryFocus.org June 2024 30


Volume 4 • Number 2 Pharmaceutical stability testing, Part 1: An overview of stability

In either stability category stability-indicating tests are on the climatic zone in which it will be marketed.
performed on the drug substance and drug products to
establish the retest period and shelf life. The stability of Stability requirements at the initial filing
a drug substance or product can be broadly categorized Under ICH guidance, the amount of stability data
into five types. The final drug product is deemed safe required at the time of an original submission (e.g., new
for use only if the following aspects of stability are fully drug, marketing authorization, biologics license appli-
demonstrated: cations) will depend on the type of product and type of
container closure system, intended label storage condition,
Chemical stability – analyzes how the active type of stability studies, and the storage condition. A gen-
pharmaceutical ingredient maintains its potency and eral overall requirement is presented below in Table 2.1,3
integrity;
Physical stability – refers to properties such as Stability testing on drug substances and drug
appearance; products
Microbiological stability – analyzes the resistance A robust stability protocol should be in place before any
of microbial growth; stability testing program is started. The protocol should
Therapeutic stability – ensures that therapeutic set out the executable plan of the stability program,
effect is maintained; and detailing the design of the program and the information
Toxicological stability – assesses any significant to be recorded. The scope of information outlined in the
increase in toxicity that would harm patients. protocol could include a range of aspects such as details
about the product, its packaging, manufacturing process,
Relevant analytical methodologies should be used to storage conditions, and testing schedule.
determine the complete stability-indicating profile of a
drug substance or product, such as the use of high-per- Batch selection
formance liquid chromatography, ion exchange chroma- Typically, three primary batches are used for drug
tography, and peptide mapping, among others. substance and drug product stability testing in stability
studies. For drug substances, the selected study batch-
Global stability zones es should be on a pilot scale at a minimum and the
Different global climate conditions must be taken into method used should simulate the process to be used
consideration when managing storage. In accordance during production of the drug substance batches. (Pilot
with ICH Q1A(R2) and the World Health Organiza- scale refers to a small-scale preliminary study conducted
tion’s (WHO) stability guideline, there are four stability to evaluate the feasibility, cost, and duration. It is also
zones across the world (Table 1).1,3 The same drug prod- performed to identify possible improvements to process
uct could have different storage conditions depending design/methodology before the development of a full-
Table 1. Global stability zones1,3
Temperature, °C Relative humidity, %
Zone Type of climate [range, °C] [range, %]

I Temperate 21 [±2] 45 [±5]

II Mediterranean/subtropical 25 [±2] 60 [±5]

III Hot and dry 30 [±2] 35 [±5]

IV Hot and humid/tropical 30 [±2] 65 [±5]

IVb Hot/higher humidity 30 [±2] 75 [±5]

RegulatoryFocus.org June 2024 31


Volume 4 • Number 2 Pharmaceutical stability testing, Part 1: An overview of stability

Table 2. General stability requirements for initial filing1,3

Storage condition Minimum


submission
Intended label Temperature, °C Relative humidity, % requirements,
storage condition Stability studies [range, °C] [range, %] months

Room Temperature Long term 25 [±2] 60 [±5]


or 30 [±2] or 65 [±5] 12
Intermediate 30 [±2] 65 [±5] 6
Accelerated 40 [±2] 75 [±5] 6

Refrigerator Long term 5 [±3] NA 12


Accelerated 25[±2] 60 [±5] 6

Freezer Long term -20[±2] NA 12

scale study.) In addition, the container closure compo- least 12 months, testing is required every three months for
nents used in the stability test should be the same as the first year, every six months for the second, and annually
those proposed for storage and distribution.1 for each subsequent year through the proposed retest pe-
riod. A similar testing frequency applies to a drug product
Stability testing for drug products should be carried with a shelf life of at least 12 months.
out in batches of the same formulation and packaging
intended for commercialization, meeting the same For accelerated storage conditions in a six-month study,
commercial specification intended for marketing. If the testing is required a minimum of three times over the
container closure components in stability studies are not study period: once at the start of the study and then at
the same as the intended final packaging components three and six months. For intermediate storage conditions
for either the drug substance or drug product, then in a 12-month study, a minimum of four time points is
comparability studies of the different container closure required: once at the start of the study, and then at 6, 9,
components should be conducted to ensure that the dif- and 12 months (see Glossary).
ferences will not affect the stability profile of the drug
substance and drug product. In addition, from a regulatory planning perspective, a re-
duced stability database can be submitted to support appli-
Setting specifications cation for the approval of a new dosage of a drug product
Drug substance and drug product stability testing that contains the same active ingredients as an existing
should include attributes that are susceptible to change approved drug product. In such a scenario, six months of
over time during storage and those that are likely to af- accelerated and six months of long-term data from ongo-
fect quality, safety, and/or efficacy. Typically, this testing ing studies are typically required for submission.4
should cover physical, chemical, microbiological, thera-
peutic, and toxicological attributes and should use fully Storage conditions
validated and stability-indicating analytical procedures. Both the drug substance and drug product should be
subjected to storage conditions that would test ther-
Testing frequency mal stability and sensitivity to moisture. A minimum
The requirements for testing frequency depend on the type of 12 months for long-term testing should be carried
of study and storage conditions. For long-term studies for out on three primary registration batches at the time
which the proposed retest period of a drug substance is at of submission and should be continued to include

RegulatoryFocus.org June 2024 32


Volume 4 • Number 2 Pharmaceutical stability testing, Part 1: An overview of stability

the proposed retest or shelf-life period. There may be estimate its shelf life, but there are only six months of
certain scenarios in which it is possible to submit less stability data available. By analyzing the available data,
than 12 months of long-term stability on the primary the company can extrapolate the drug’s stability over
registration batches but only with prior agreement with a longer duration by assuming the degradation rate of
the agency. Primary registration batches are defined as the drug observed in the initial six months will remain
drug product batches manufactured and used for the relatively constant.
registration and approval of the drug product.
Failure in stability testing
Evaluation of stability data and extrapolation In an ideal setting, complete stability test results are
ICH Q1E focuses on the evaluation of stability data.2 within the specified acceptance criteria. However, in
The evaluation of stability data is important because it reality, there may be times when stability batches will
helps determine the appropriate storage conditions for not meet the specification(s). During a stability study
pharmaceutical products to ensure their stability and program, the approved stability protocol must be
potency. Stability evaluation assists in establishing shelf- followed for all selected batches. If any batch shows
life specifications and providing patients and health- out-of-specification (OOS) results for any test parame-
care professionals with accurate information about the ter at any time during the study, the specific test param-
product’s expiration date. It also helps pharmaceutical eter may be retested to rule out analyst error, depending
manufacturers make informed decisions about product on company policy. If the parameter fails on retest, then
labeling, storage, and distribution. the stability study for this batch will typically continue
to the next time point. This batch will continue to be
Extrapolation is a way of extending what is known monitored for the duration of the stability protocol for
about something to make predictions about what is any stability trends to determine whether they will have
not known. It involves using existing data or patterns any impact on the quality of the drug product batch.1
to estimate how a system will behave in the future or
in situations in which data are limited. This method If, after the point at which the OOS result occurred and
assumes that the observed behavior will prevail even the remaining time-point results are still acceptable, then
outside of the known range. the batch is still considered stable, provided the OOS
results have been fully justified to ensure there is no impact
In stability studies, there may be a need to determine on the stability study. However, if all time-point results are
how a material or system will behave over an extended within specification but fail at the last time point, a justi-
period. However, running stability tests for the entire fication must be provided to support why the batch is still
expected duration may not always be feasible because acceptable. If other batches in the program show the same
of time, cost, or practical constraints. This is where issue, that is, failure at the last time point, it could result in
extrapolation can be used. While extrapolation is a a reduced expiration date or retest date.1
valuable tool, it is important to recognize its limitations.
Extrapolating too far into the future or outside the If the stability batches have any OOS or unexpected
range of available data can introduce uncertainties and trends, it is good practice to provide a justification to
inaccuracies. Factors such as environmental conditions ensure that these batches are stable and that the OOS
and/or the presence of unknown variables can affect the or unexpected trends have no impact on the quality of
accuracy of extrapolations. Therefore, it is crucial to ex- the drug substance or product.1
ercise caution and validate extrapolations with addition-
al data whenever possible. For example, consider a new Common stability deficiencies
drug product that has been developed and that needs Stability batches that fail to meet specifications are
to be evaluated for stability. The manufacturer needs to cause for concern. Equally, all requirements of the

RegulatoryFocus.org June 2024 33


Volume 4 • Number 2 Pharmaceutical stability testing, Part 1: An overview of stability

specification; WHO, World Health Organization.


stability study must be fulfilled to ensure a successful
stability study program. Some of the common stability
About the authors
pitfalls are lack of:5
Kerry Tsang, MSc, BA Mod, is a regulatory affairs
consultant at Parexel with a background in biochemistry and
A valid sample size and test intervals based on
biotechnology. She has more than seven years’ experience
statistical criteria for each attribute examined;
in regulatory affairs labeling and chemistry, manufacturing,
Appropriate storage conditions for samples retained
and control (CMC), covering areas of small molecules and
for testing;
biologics, supporting clients with marketing authorization
Reliable, meaningful, and specific test methods; applications, and regulatory lifecycle maintenance for
Testing of the drug product in the same emerging markets. In addition to project management, she
container closure system that is intended for has experience in technical CMC writing for regulatory
commercialization; and applications. Tsang has most recently worked in CMC
Testing of drug products for reconstitution at the lifecycle management for vaccine products. She can be
time of dispensing as well as after reconstitution. reached at kerry.tsang@parexel.com

It is important to note that an adequate number of drug Sherri N. Thrower, BSc, is a regulatory affairs senior
product batches must be tested to determine an appro- consultant at Parexel with a background in biology. She has
priate expiration date. 23 years’ experience in regulatory affairs, with a focus on
CMC. That experience includes defining strategy and the
Conclusion managing of the quality sections for small molecule and
biologic products. She has substantial experience in writing
As increasingly complex pharmaceutical products are
initial marketing and development applications and extensive
being developed and manufactured, regulations will also
writing experience with postapproval supplement applications.
be adapted to ensure that the quality, safety, and efficacy
She can be reached at sherri.thrower@parexel.com
of these regulated products remain adequate. From the
regulatory agencies’ perspective, the stability aspect will Acknowledgment This article was previously published online
always be taken into detailed consideration during the on 16 April 2024.
review process. Therefore, manufacturers should aim to
fulfill all the stability requirements to ensure a successful Citation Tsang K, Thrower SN. Pharmaceutical stability
and fully justified stability program. testing, Part 1: An overview of stability. RF Quarterly.
2024;4(2):29-35. Published online 7 June 2024. https://
Abbreviations www.raps.org/News-and-Articles/News-Articles/2024/6/
ICH, International Council for Harmonisation; OOS, out of Pharmaceutical-stability-testing,-Part-1- An-overv

References
All references were last accessed and/or verified on 11 May 2024.

1. International Council for Harmonisation. Stability testing active pharmaceutical ingredients and finished pharmaceu-
of new drug substances and products Q1A(R2). Dated 6 tical products. 2018. Accessed 25 April 2023. https://
February 2003. Accessed 21 April 2023. https://database. database.ich.org/sites/default/files/Q1F_Stability_Guide-
ich.org/sites/default/files/Q1A%28R2%29%20Guideline. line_WHO_2018.pdf
pdf 4. International Council for Harmonisation. Stability testing
2. International Council for Harmonisation. Evaluation for for new dosage forms – Annex to the ICH harmonised
stability data Q1E. Dated 6 February 2003. Accessed 21 tripartite guideline on stability testing for new drugs and
April 2023. https://database.ich.org/sites/default/files/ products Q1C. Dated 6 November 1996. Accessed 21
Q1E%20Guideline.pdf April 2023 https://database.ich.org/sites/default/files/
3. World Health Organization. Annex 10. Stability testing of Q1C%20Guideline.pdf

RegulatoryFocus.org June 2024 34


Volume 4 • Number 2 Pharmaceutical stability testing, Part 1: An overview of stability

5. Therapeutic Goods Administration [Australia]. Common before which the product is known to remain stable, that is,
deficiencies in stability data and trial design. Updated 6 it retains its strength, quality, and purity when it is stored
March 2017. Accessed 21 April 2023. https://www.tga.gov. according to its labeled storage conditions.
au/resources/resource/guidance/stability-testing-prescrip-
tion-medicines/145-common-deficiencies-stability-da- Intermediate storage condition. Intermediate conditions are
ta-and-trial-design generally 30°C/65% relative humidity to moderately increase
rate of chemical degradation or physical changes in the
Glossary1 drug substance or drug product (suitable long-term storage,
Accelerated storage condition. The condition under which 25°C/69% relative humidity). Testing is recommended in the
a drug substance or drug product is stored at elevated intermediate storage condition if there is significant change
stress conditions – such as temperature, humidity, and in the accelerated storage condition. The testing frequency
pH – to provide data for predicting the degradation rate should be at a minimum of four time points, including the
or physical changes. (Temperature and relative humidity in initial and final time points (e.g., 0, 6, 9, and 12 months), from
the accelerated storage condition would be 40°C and 75%, a 12-month study.
respectively, versus 25°C and 69% for suitable long-term
storage conditions.) In the accelerated storage condition, Pilot scale. A pilot scale study is a small-scale preliminary
testing is recommended at a minimum of three time points, study conducted to evaluate the feasibility, cost, and duration.
including the initial and final time points (e.g., at zero, three, It is also performed to identify possible improvements to
and six months in a six-month study). process design/methodology before the development of a
full-scale study.
Container closure system. A container closure system
comprises the primary and secondary packaging components Retest period. The period during which a drug substance
that contain and protect the dosage form. The primary or product can be considered suitable for use, provided it
packaging container closure system is made up of the meets specific quality criteria. It is determined based on
packaging components that contain and protect the dosage stability data obtained from formal stability studies. A batch
form. The secondary packaging refers to the exterior of drug substance must be retested after this period to ensure
packaging that groups the primary packages, provides it still meets the criteria, and, if it does, it should be used
additional information about the dosage form, and carries the immediately.
identifying branding for the dosage form.
Retest date. The date after which a drug substance should
Drug substance. An active, unformulated ingredient with be re-examined to ensure it remains suitable for use. It is
a specific pharmacological activity. The drug stance can be determined based on stability data generated during formal
formulated with inactive ingredients to produce the dosage stability studies. The retest date for a drug product is related
form. to its quality control testing. It indicates when the product
should be retested to ensure it still meets the required
Dosage form. The physical form in which a drug is specifications.
administered (e.g., tablet, capsule, lozenge, powder, ointment,
solution, eye drops, lotion, inhaler-administered medications). Shelf life. The shelf life of a pharmaceutical product is the
period for which the product maintains its identity and
Drug product. A finished dosage form (e.g., tablet or quality when stored at the conditions defined on the label of
solution) that contains an active drug ingredient and usually, the product.
but not always, inactive ingredients.
Time point. The designated test date for a drug substance or
Expiration date. The drug expiration date marks the date product in a stability study.

RegulatoryFocus.org June 2024 35

You might also like

pFad - Phonifier reborn

Pfad - The Proxy pFad of © 2024 Garber Painting. All rights reserved.

Note: This service is not intended for secure transactions such as banking, social media, email, or purchasing. Use at your own risk. We assume no liability whatsoever for broken pages.


Alternative Proxies:

Alternative Proxy

pFad Proxy

pFad v3 Proxy

pFad v4 Proxy