0% found this document useful (0 votes)
95 views20 pages

1 Stability

This document provides an overview of current practices and future trends in stability testing of pharmaceuticals. It discusses key aspects of stability testing including regulatory requirements, ICH guidelines, accelerated and real-time stability studies, and photostability testing. The goals of stability testing are to ensure drug quality and safety over the shelf life and determine appropriate storage conditions. Emerging technologies may help revolutionize stability testing approaches.

Uploaded by

kanhaiyabattase
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as DOCX, PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
95 views20 pages

1 Stability

This document provides an overview of current practices and future trends in stability testing of pharmaceuticals. It discusses key aspects of stability testing including regulatory requirements, ICH guidelines, accelerated and real-time stability studies, and photostability testing. The goals of stability testing are to ensure drug quality and safety over the shelf life and determine appropriate storage conditions. Emerging technologies may help revolutionize stability testing approaches.

Uploaded by

kanhaiyabattase
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as DOCX, PDF, TXT or read online on Scribd
You are on page 1/ 20

review article on Stability testing of pharmaceuticals: An overview of

current practices and future trends


Authors name
Email id
Abstract

Stability testing is a critical component of pharmaceutical development, ensuring the quality,


safety, and efficacy of drugs throughout their shelf life. This review article provides a
comprehensive overview of the current practices and regulatory guidelines related to stability
testing in the pharmaceutical industry. Additionally, we explore emerging trends and
technologies that have the potential to revolutionize stability testing approaches in the future.
The insights provided in this article aim to enhance understanding and pave the way for more
robust and efficient stability testing protocols, thus contributing to the overall advancement of
pharmaceutical product development.

Keywords

1. Introduction

Stability is defined as a formulation's capacity to maintain its physical, chemical,


microbiological, therapeutic, and toxicological requirements during the course of its shelf life
in a specified container or closed system. The official definition of stability is "the interval of
time during which the drug product retains the same properties & characters that are
processed at the time of manufacture." When a pharmacological substance or designed
product is tested for stability, the impacts of external conditions on its quality are assessed in
order to forecast how long it will last and to determine how to store and label it. A stability
research looks at how the pharmaceutical product is affected by changes in temperature, time,
humidity, light intensity, and partial vapor pressure.[1]

The term "current" in current good manufacturing practises (cGMP) refers to the current
good manufacturing practises regulations, not the previous or following regulations. From a
regulatory standpoint, a product may be considered adulterated if the production conditions
were less stringent than those that are currently acknowledged and widely followed by the
industry. cGMP requirements refer to the circumstances in which the product is made, not to
the state in which it is delivered. If products are not made in accordance with cGMP, they

1
may be considered adulterated. Therefore, stability study is a cGMP tool that indirectly links
high-quality products to a company's reputation on the world market.[2]

Importance Of Stability Testing [3][4]

The safety of patients is ensured by drug quality stability testing. It guarantees the security of
pharmaceutical dose forms with regard to the sick patients. Developmental stability studies
offer a data base that may be helpful in choosing an appropriate formulation to estimate shelf
life, container closure mechanism, and storage conditions for new product development.
Stability tests are crucial for determining the calibre of a modified or repackaged drug
product since they assess the appearance and physical characteristics (such as colour, caking,
hardness, phase separation, and re-suspendability), potency, and purity of a drug product over
its specified shelf-life.

2. Current Practices in Stability Testing

Current trend in multifunctional pharmaceutical companies are to define condition for


stability testing for global marketing. For this companies are creating their protocols to single
set of conditions that covers extreme environmental condition. The specific changes for
global testing includes increase in duration of accelerated testing period from 6-12 months
and conduct of additional tests at 500 c/75% RH for 3 months (mischler et al 2004).It avoids
the repetition of stability testing for the region and efficient optimum use of resources as all
tests are done in one laboratory testing under combination of 3 environmental factors vise,
temperature, humidity and light has been reported to result in stronger deleterious effect on
drug substances and products than humidity condition only [5]

2.1. Regulatory Requirements

A pharmaceutical product’s shelf-life estimation by assessing through a systematic stability


study is a regulatory requirement, during this review current regulatory requirements are
elaborated. The main aim of this study is to produce an indication of how the drug substance
and drug product quality change with time by exposure to different climate conditions like
temperature, humidity, and photolytic condition. A stability study helps to establish a retest
period for the drug substance and validity of the drug product and proposed storage
conditions and proves that it meets its set specifications. [7]

2.2. ICH Guidelines on Stability Testing

2
To assure that optimally stable molecules and products are manufactured, distributed and
given to the patients, the regulatory authorities in several countries have made provisions in
the drug regulations for the submission of stability data by the manufacturers. Its basic
purpose was to bring in uniformity in testing from manufacturer to manufacturer. These
guidelines include basic issues related to stability, the stability data requirements for
application dossier and the steps for their execution. Such guidelines were initially issued in
1980s. These were later harmonized (made uniform) in the International Conference on
Harmonization (ICH) in order to overcome the bottleneck to market and register the products
in other countries. The ICH was a consortium formed with inputs from both regulatory and
industry from European commission, Japan and USA. The World Health Organization
(WHO), in 1996, modified the guidelines because the ICH guidelines did not address the
extreme climatic conditions found in many countries and it only covered new drug substances
and products and not the already established products that were in circulation in the WHO
umbrella countries. In June 1997, US FDA also issued a guidance document entitled
‘Expiration dating of solid oral dosage form containing Iron. WHO, in 2004, also released
guidelines for stability studies in global environment (WHO, 2004). ICH guidelines were also
extended later for veterinary products. A technical monograph on stability testing of drug
substances and products existing in India has also been released by India Drug Manufacturers
Association. Further, different test condition and requirements have been given in the
guidance documents for active pharmaceutical ingredients, drug products or formulations and
excipient. The codes and titles covered under ICH guidance have been outlined in the table
[3]

2.3. Accelerated Stability Studies

In accelerated stability testing; a product is stored at elevated stress conditions. Accelerated


stability testing should at the minimum be done at 0, 3 and 6 months. Degradation at the
recommended storage conditions can then be predicted by using known relationships between
the acceleration factor and the degradation rate. Degradation at recommended storage
conditions could be predicted based on the degradation at each stress condition and known
relationships between the acceleration factor and the degradation rate. A product may be
released based on accelerated stability data, but the real-time testing must be done in parallel
to confirm the shelf-life prediction. Sometimes the amount of error of the predicted stability
is so large that the prediction itself is not useful. Design your experiments carefully to reduce
this error. It is recommended that several production lots should be stored at various

3
acceleration levels to reduce prediction error. Increasing the number of levels is a good
strategy for reducing error. [1]

2.4. Real-Time Stability Studies

In real-time stability tests, a product is stored at recommended storage conditions and


monitored for a period of time (testimation) Real-time tests store the product at recommended
storage conditions and monitors the product until it fails the specification. Real-time stability
should typically be done at 0, 3, 6, 9, 12 months on the first year, every 6 months on the
second year and once every year afterwards. In accelerated stability studies, the product is
stored at elevated stress conditions (such as temperature and humidity). Product will degrade
below its specification, at some time, denoted t(specification), and we must also assure that it
is less than or equal to test. The estimated value of t(specification)can be obtained by
modeling the degradation pattern. Good experimental design and practices are needed to
minimize the risk of biases and reduce the amount of random error during data collection.
Testing should be performed at time intervals that encompass the target shelf life and must be
continued for a period after the product degrades below specification. It is also required that
at least three lots of material be used in stability testing to capture lot-to-lot variation, an
important source of product variability. [8]

2. 5. Photostability Testing

Photostability testing should be conducted on at least one primary batch of the drug product if
appropriate. Proof that the products do not or not significantly change within a certain period
of use must, among other things, is provided through the photo-stability test with light. For
this purpose, binder offers the complete solution on the market - the KBF with standard
equipment of ICH-conforming lighting. The special international ICH guideline Q1B was
created for proving photo-stability. Since the fulfillment of this guideline must now
mandatorily be documented by the authorities without exception, pharmaceutical companies
are faced by new challenges in this regard in their test practices. For the new photo-stability
tests, samples must be exposed to a light amount of 1.2 million Lux x hours, as well as UV
radiation of 200 Watt x hours /m2 , in climatic chambers with ICH lighting. But what is the
most objective method of proving these light values? The fundamental prerequisite for
reliable recording is the integration and display of the light values on the regulator, as in the
binder KBF series with ICH lighting. This includes the automatic shutoff of the lamps (VIS
and UV separately) when the freely selectable dosage values are reached. Reliable recording

4
of the light amounts is provided at binder with Light Quantum Control, two spherical light
sensors which, due to their direction-independent characteristics, function more precisely
than planar sensors.[1][6]

2.6. Evaluation of Impurities and Degradants

Detection and Identification

Characterization

Quantification:

Toxicological Evaluation:

Source Identification

Risk Assessment

Control Strategies

Regulatory Compliance

Stability Studies

Continued Monitoring

As per ICH

a. Organic impurity (process- and drug-related).


 Starting material or intermediate impurities
 By-products
 Degradation products
b. Inorganic impurity.
 Heavy metals
 Filter aids
c. Residual solvents.[9]
Forced degradation is a critical analytical study for the development of stability-
indicating methods to be used by pharmaceutical companies as part of regulatory
submissions to the FDA. Some of the applications of the studies are:

1. To develop and validate stability-indicating methods as per ICH guidelines.

5
2. To identify structure and toxicity and to set up specification of degradants or
impurities.
3. To propose shelf life of the product without real-time stability information.
4. To optimize formulations and to select placebos for drug product to avoid
interference.
5. To justify impurities that are process related or degradation products.
6. To support identification of root cause during out-of-specification (OOS)/lab
investigations.
7. To accompany drug master file and ANDA/NDA and IND submissions to the FDA.
[10]

2.7 Packaging Compatibility Studies


Stability testing assesses the formulation only and is carried out in glass jars whereas
compatibility testing assesses the product in the final packaging.
The data obtained from the testing is used to determine the date of minimum
durability of the product. The date of minimum durability is expressed as a ‘best used
before end of’ date and uses the ‘egg timer’ symbol. If a product has a minimum
durability greater than 30 months a ‘period after opening’ is used and the symbol for
this is the ‘open jar’. Stability tests involves incubating products at a variety of
temperatures and light conditions to mimic transportation and storage conditions
which may be encountered during warehousing, retail and with the consumer. These
tests ensure that the characteristics of the products, including the fragrance, colour,
texture, appearance and formulation do not change when subjected to these
conditions. Compatibility testing is similar to stability but would also include an
assessment of packaging functionality and stability, label stability and determine if
there are any interactions between product and packaging. [11]

2.8.Case Studies on Stability Testing

Case Studies A lean stability strategy can take many forms. It should be product specific, dependent
on the stage of development and reflect the product knowledge available at the time. In the clinical
development phase, while development is under way and changes and improvements are constant,
this may take the form of utilizing predictive tools and/or confirmatory studies to demonstrate a
change does not impact stability rather than reinitiating a long-term stability protocol, to justify re-
test period or shelf-life, or to identify which tests are stability indicating. In the registration phase, a

6
lean strategy may leverage the cumulative clinical and registration stability data to justify reducing
the tests, time points and/or storage conditions that are necessary to monitor stability on an annual
basis. In the postapproval phase, product knowledge and stability understanding are highest and
there are many opportunities to leverage lean strategies to support a change. Following are
examples of case studies collected during each phase of development.

 clinical development
Case Study 1
In this case study, the drug substance stability data were used to support drug in capsule
(DiC) product. The scope of the project spanned across 5 small molecules with drug in
capsule formulations. The FDA Guidance for Industry, cGMP for Phase 1 Investigational Drugs
(U.S. Food and Drug Administration, 2008) allows representative samples of phase 1
investigational drugs to be used to monitor stability and quality
Case Study 2
In this case study, accelerated short-term stability comparison studies were leveraged for
subsequent drug substance campaigns with minimal synthetic route changes. An internal risk
assessment was performed to determine the potential impact of the synthetic route
changes. This assessment was then used to inform the comparative stability study. The study
design consisted of a short term, typically 2 weeks to 1 month, accelerated stability study to
establish comparability of a new batch of drug substance back to the original drug substance
batch.
Case Study 3
An approach used to support many drug substance in bottle regulatory filings is provision of
3 weeks of drug substance stability data at 70 °C/75%RH in the initial clinical submission to
justify a 15-month drug substance retest period.
Case Study 4
Exclusion of assay testing has been routinely proposed in CTAs in situations where the drug
substance stability batch is the same as the reference standard batch
 registration,
Case Study 5
A registration strategy for drug product primary stability was developed to support seven
different dosage strengths (compositionally proportional) where each dosage had two
packaging configurations. An ICH Q1D bracketing design was proposed to support the NDA
submission and was agreed to during a pre-NDA type C meeting with US FDA.
Case Study 6

7
A proposal to use a bracketing design according to ICH Q1D for an NDA submission was
agreed to by US FDA at an End of Phase 2 type B meeting. This program had five different
dosage strengths with the same (compositionally proportional) formulation and packaging
configuration
Case Study 7
An NDA was submitted for a complex modified release (MR) product with multiple strengths
in capsules manufactured from common pellets. The primary stability data supported an
initial shelf life of 36 months. The NDA did not include a protocol commitment to confirm
expiry, since the primary stability batches were manufactured at the commercial site and
commercial scale.
Case Study 8
A stable, well-understood drug product (IR (Immediate Release) capsule) was filed with a
standard primary stability protocol and data to support the initial shelf-life claim. An
alternate proposal was made for the post-approval stability protocol. To limit wasted supplies
for a low-volume commercial program, the applicant proposed to utilize non-printed
capsules for 2 of the 3 lots for 2 of the 3 strengths.
 post-approval.
Case Study 9
In this case study, a lean stability strategy was applied for post approval/annual stability
batches of a small molecule oral drug substance and drug product. Only the long-term
storage condition was required, and no data was proposed to be collected for the
accelerated storage condition.
Case Study 10
This case study relates to a large product family in semipermeable containers, spanning
multiple US regulatory filings, separated into multiple categories based on formulation,
container type and filling volume.
Case Study 11
1 This case study covers the submission of a registration stability package for a biologic in
accordance with the recommended batch enrollment and timepoint frequency prescribed by
ICH Q5C [18] and ICH Q1D [2].
Case Study 12
A science- and risk-based stability strategy was developed to support a drug substance
manufacturing site change for a marketed small molecule solid oral dosage form product.

8
There were no changes to the drug substance manufacturing process associated with the site
change, and there were no proposed changes to the drug product. [12]

3. Challenges and Limitations

3.1.Complexity of Formulations

Herbal formulations are complex mixtures of different components obtained during


extraction process. Each component has variable shelf-life, activity, concentration and
consistency. It creates a problem during storage condition determination as it is not easy to
determine the stability of final herbal preparation based on the activity and stability profile of
a single component.

3.2. Sampling and Sample Handling

When the units of the population are not in homogeneous, the sampling technique will be
unscientific. In sampling, though the number of cases is small, it is not always easy to stick to
the, selected cases. The units of sample may be widely dispersed.

Some of the cases of sample may not cooperate with the researcher and some others may be
inaccessible. Because of these problems, all the cases may not be taken up. The selected cases
may have to be replaced by other cases. Changeability of units stands in the way of results of
the study

3.3. Analytical Method Suitability

The manufacture of biopharmaceuticals presents some unique challenges when ensuring


product quality and patient safety. Analytical testing can provide the data needed to produce a
safe and effective drug product. The development and validation of analytical methods is
crucial in drug development. Technologies, such as FDA’s quality-by-design (QbD) initiative,
may have a positive impact on analytical method development and validation according to
Paul Smith, EMEAI laboratory compliance productivity specialist at Agilent Technologies,
because methodologies are identified early in the development process.

3.4. Statistical Analysis of Stability Data

9
The design and execution of formal stability studies should follow the principles outlined in the parent
guideline. The purpose of a stability study is to establish, based on testing a minimum of three batches
of the drug substance or the veterinary medicinal product, a retest period or shelf life and label storage
instructions applicable to all future batches manufactured and packaged under similar circumstances.
The degree of variability of individual batches affects the confidence that a future production batch
will remain within acceptance criteria throughout its retest period or shelf life. Although normal
manufacturing and analytical variations are to be expected, it is important that the veterinary
medicinal product be formulated with the intent to provide 100 percent of the labeled amount of the
drug substance at the time of batch release. If the assay values of the batches used to support the
registration application are higher than 100 percent of label claim at the time of batch release, after
taking into account manufacturing and analytical variations, the shelf life proposed in the application
can be overestimated. On the other hand, if the assay value of a batch is lower than 100 percent of
label claim at the time of batch release, it might fall below the lower acceptance criterion before the
end of the proposed shelf life.[12]

3.5. Predictive Modeling for Shelf-Life Determination

Predictive modelling can be used alongside shelf-life testing of real-life samples, for example
using challenge testing. Challenge testing involves deliberately inoculating a product with
relevant microorganisms and assessing their growth over time. This approach considers all
factors that may influence growth. Predictive modelling, however, will usually only consider
three factors – pH, aw and temperature.

Factors not accounted for in predictive modelling:

 natural antimicrobials that may be present (for example, allicin in garlic)


 differences in structure across a food sample
 change of aw and pH over time, and
 competing organisms [13][14]

4. Future Trends in Stabillty Testing

4.1. QbD (Quality by Design) Applications

There are several statements about the elements of QbD, the most widely accepted is that,
QbD consists of the following parameters

Quality Target Product Profile (QTPP): including dosage form, delivery systems, dosage
strength(s), etc. It is a prospective summary of quality characteristics of a drug product to be

10
achieved, taking into account dosage strength(s) and container closure system of the drug
product, together with the attributes affecting pharmacokinetic characteristics (e.g.,
dissolution, aerodynamic performance) and drug product quality criteria (e.g., sterility, purity,
stability and drug release) appropriate for the intended marketed product.

Critical Quality Attributes (CQAs): including physical, chemical, biological, or


microbiological properties or characteristics of an output material including finished drug
product. Potential drug product CQAs derived from the QTPP and/or prior knowledge are
used to guide the product and process development and they should be within an appropriate
limit, range, or distribution to ensure the desired product quality.

Critical Material Attributes (CMAs): including physical, chemical, biological, or


microbiological properties or characteristics of an input material. CMAs should be within an
appropriate limit, range, or distribution to ensure the desired quality of that drug substance,
excipient, or in-process material.

Critical Process Parameters (CPPs): parameters monitored before or in process that influence
the appearance, impurity, and yield of final product significantly. [15]

4.2. Application of Artificial Intelligence and Machine Learning

Automated Testing

One of the most significant applications of AI in quality assurance is in automated testing.


Traditionally, testing has been a manual process that requires a lot of time and effort.
However, with AI-powered tools, businesses can now automate the testing process, reducing
the time and effort required significantly. AI-powered testing tools can detect errors and bugs
in the software code and provide accurate reports on the system’s functionality.

Predictive Maintenance

Another application of AI in quality assurance is predictive maintenance. AI-powered


predictive maintenance systems can analyze data from machines and predict when they are
likely to fail. This allows businesses to schedule maintenance activities before the machine
breaks down, reducing downtime and improving productivity.

Quality Control

AI is also being used in quality control to identify defects in products. With AI-powered
tools, businesses can analyze data from product inspections and detect defects that would

11
otherwise go unnoticed. AI-powered quality control systems can also provide insights into the
root cause of defects, enabling businesses to make the necessary changes to prevent them
from happening again.

Image and Speech Recognition

AI-powered image and speech recognition technologies are also being used in quality
assurance. For example, image recognition can be used to identify defects in products by
analyzing images of the products. Similarly, speech recognition can be used to analyze
customer feedback and identify areas where improvements can be made.

Fraud Detection

Finally, AI is being used in quality assurance to detect fraud. AI-powered fraud detection
systems can analyze large volumes of data and identify patterns that indicate fraudulent
activity. This helps businesses to prevent fraudulent transactions and maintain the integrity of
their products and services. [16]

4.3. Advanced Analytical Techniques

High-Performance Liquid Chromatography (HPLC): HPLC is a widely used technique in


stability testing to separate, quantify, and identify drug compounds and degradation products.
It can detect and quantify impurities and degradation products at very low concentrations.

Mass Spectrometry (MS): Coupling HPLC with Mass Spectrometry allows for the
identification and structural characterization of drug compounds and degradation products.
MS is highly sensitive and provides information on molecular weight and fragmentation
patterns.

Gas Chromatography (GC): GC is used for the analysis of volatile compounds and can be
valuable in stability testing for certain drug formulations.

Nuclear Magnetic Resonance (NMR) Spectroscopy: NMR provides detailed information


about the molecular structure and dynamics of drug compounds and can be used to detect
changes in the chemical structure during stability testing.

Fourier Transform Infrared Spectroscopy (FTIR): FTIR is employed to analyze molecular


vibrations, which can reveal changes in drug compounds due to degradation.

12
Raman Spectroscopy: Raman spectroscopy is a non-destructive technique that can provide
information about molecular vibrations, offering insights into the stability of drug
formulations.

X-ray Diffraction (XRD): XRD is used to study the crystallinity of drug substances and
formulations, which can impact stability.

Differential Scanning Calorimetry (DSC): DSC measures the heat flow associated with
thermal transitions, aiding in the detection of phase changes and drug degradation.

Dynamic Light Scattering (DLS): DLS is utilized to determine the particle size distribution of
drug formulations, which can influence stability.

Stability-Indicating Assays: These assays are designed to detect and quantify specific
degradation products, ensuring that the analytical method used is selective and capable of
accurately assessing drug stability.

Accelerated Stability Studies: Advanced mathematical modeling and statistical methods are
used to extrapolate stability data obtained from accelerated conditions to predict long-term
stability.

Design of Experiments (DOE): DOE is utilized to optimize stability study designs,


determining critical factors that influence drug stability and identifying the optimal
conditions.

Multivariate Data Analysis: This technique allows for the simultaneous analysis of multiple
parameters, making it possible to identify patterns and correlations that may impact drug
stability.

4.6. Nanotechnology in Stability Assessment

Stability in terms of nanoparticle morphology can be assessed from changes in atomic crystal
lattice and surface facets. These are most often monitored using X-ray diffraction99 or high-
resolution transmission electron microscopy (HR-TEM) [17]

5. Novel Approaches for Specific Drug Types

5.1. Biologics and Biosimilars Stability

Biologics Stability Testing:


13
Biologics are large, complex molecules, such as monoclonal antibodies, vaccines, and
therapeutic proteins, which are sensitive to environmental conditions.

Stability testing for biologics involves monitoring the drug product's physical, chemical, and
biological properties over time to assess its degradation pathways and determine the
appropriate storage conditions.

It includes evaluating parameters like protein structure, potency, impurity profiles,


aggregation, and immunogenicity during the drug's shelf life.

Accelerated stability studies, where the drug is subjected to elevated temperature, humidity,
and light, are commonly used to predict long-term stability.

Advanced analytical techniques like high-performance liquid chromatography (HPLC), mass


spectrometry (MS), and bioassays are employed to analyze biologics during stability testing.

Biosimilars Stability Testing:

Biosimilars are biologic drugs that are highly similar to an already approved reference
biologic (the originator) but are not identical due to their inherent complexity.

Stability testing for biosimilars is critical to demonstrate that they maintain similar quality
and efficacy to the reference biologic over time.

The analysis includes comparisons of physicochemical attributes, biological activity, impurity


profiles, and immunogenicity between the biosimilar and the reference product.

Regulatory authorities require extensive comparative stability data between the biosimilar
and the reference product to establish similarity.

Studies may involve head-to-head comparisons, forced degradation studies, and stress testing
to evaluate the biosimilar's stability and degradation pathways. [18]

5.2. Vaccine Stability Testing

1. Importance of Vaccine Stability Testing:

Vaccine stability testing assesses the physical, chemical, and biological properties of vaccines
over time to determine their shelf life and storage conditions.It ensures that vaccines maintain
their potency and effectiveness, as the loss of efficacy can lead to inadequate protection
against diseases.

14
2. Parameters Evaluated in Vaccine Stability Testing:

Potency

Physical Properties.

Chemical Stability

Temperature Sensitivity

Packaging Compatibility

3. Types of Stability Studies

Accelerated Stability Studies

Real-Time Stability Studies

4. Regulatory Guidelines

Regulatory agencies, such as the World Health Organization (WHO), the U.S. Food and Drug
Administration (FDA), and the European Medicines Agency (EMA), provide guidelines for
vaccine stability testing.

5. Ongoing Stability Monitoring:

After a vaccine is approved and marketed, manufacturers continue to monitor its stability in
real-world storage and distribution conditions through post-marketing surveillance.

6. Cold Chain Management:

Maintaining a proper cold chain is crucial for vaccines that are sensitive to temperature. This
involves careful storage, transportation, and handling to preserve vaccine stability. [19]

6. Regulatory Perspectives and Harmonization

6.1. Challenges in Global Harmonization

The number of non active compounds

Assuring batch to batch consistency

Labelling

Impact on generics

15
Justification recommendation

Submission expectation [20]

6.2. Evolving Regulatory Landscape and ICH Updates

Harmonization Efforts: The International Council for Harmonisation (ICH) continues to work
on global harmonization of regulatory requirements to facilitate drug development and
registration.

ICH Q12: The development of the ICH Q12 guideline aims to promote a flexible approach to
managing post-approval changes in pharmaceutical product lifecycle.

ICH Q3D: Implementation of the ICH Q3D guideline on Elemental Impurities sets limits for
toxic elements in drug products to ensure patient safety.

ICH Q8-Q12 Implementation: Various ICH guidelines, such as Q8, Q9, Q10, and Q11, are
established to enhance pharmaceutical development and manufacturing practices.

Real-World Evidence (RWE): Regulatory agencies explore the use of real-world evidence to
support drug approvals and post-approval decision-making. [21]

6.3. Accelerated Approval and Conditional Marketing Authorization

Accelerated Approval: Used by the FDA in the U.S. and similar pathways in other regions.
Based on surrogate endpoints or reasonably likely clinical benefits. Conditional approval with
post-approval confirmatory trials.

Conditional Marketing Authorization: Used in the European Union by the EMA. Applicable
for life-threatening or seriously debilitating diseases without satisfactory treatments. Based
on preliminary data with ongoing data submission requirements for full authorization [22]

7. Case Studies and Success Stories

7.1. Industry Examples of Successful Stability Testing Practices

Pfizer's Prevnar 13® Vaccine:

Pfizer's Prevnar 13® vaccine is used to prevent pneumococcal disease in children and adults.
Stability testing played a critical role in the approval and commercial success of the vaccine.
Through extensive stability studies, Pfizer demonstrated that the vaccine remained stable and

16
maintained its efficacy throughout its shelf life, ensuring its safety and effectiveness for
patients. [23]

7.2.Case Studies of Stability Testing Failures and Lessons Learned

Pfizer's Bextra Withdrawal (2005):

Pfizer withdrew its pain reliever, Bextra, from the market due to concerns about
cardiovascular risks and adverse skin reactions. The withdrawal was followed by FDA
warnings and legal actions.

Lesson Learned: Comprehensive stability testing should include ongoing safety evaluations
and assessments of potential risks to identify and mitigate adverse effects early on in a drug's
lifecycle.[24]

8. Environmental Factors Impacting Drug Stability

8.1. Temperature and Humidity

 Temperature:

High Temperatures: Elevated temperatures can accelerate chemical reactions, leading to the
degradation of drug molecules. This degradation can result in the formation of impurities,
loss of potency, or changes in drug composition.

Low Temperatures: Extremely low temperatures, such as freezing, can cause physical
changes in drug formulations, like crystallization or phase separation, affecting the product's
appearance and stability.

Temperature Fluctuations: Repeated temperature fluctuations during storage or transportation


can lead to stress on the drug product, potentially causing physical and chemical changes.

 Humidity:

Humidity, or the moisture content in the environment, can accelerate chemical reactions,
promote microbial growth, and cause physical changes in drug formulations. Moisture-
sensitive drugs may undergo hydrolysis, where the water molecules break down the drug's
active ingredient. High humidity levels can also cause tablets or capsules to soften, dissolve,
or become moldy, compromising their stability and safety [25]

8.2. Light and Photostability

17
 Light Exposure:

Visible Light: Visible light can induce photochemical reactions in drug molecules, leading to
the formation of reactive intermediates and degradation products. This can result in a
decrease in drug potency and changes in the drug's chemical structure.

UV Light: Ultraviolet (UV) light, especially in the UV-C and UV-B ranges, can be highly
damaging to drug compounds. UV light can break chemical bonds, leading to significant
degradation and loss of therapeutic activity.

Fluorescent Light: Fluorescent light sources, commonly found in indoor environments, can
emit UV radiation that may cause drug degradation over time.

 Photostability Testing:

Photostability testing is a crucial aspect of stability testing for drugs, particularly for light-
sensitive compounds.

These studies are designed to evaluate the drug's susceptibility to photodegradation under
specific light conditions, simulating the light exposure that the drug may encounter during
storage, transportation, or use

8.3. Oxidation and Degradation Pathways

Oxidation:

Oxidation of Alcohols: Alcohols present in drug molecules can undergo oxidation to form
aldehydes or ketones.

Oxidation of Amines: Amines can be oxidized to form nitroso compounds or imines.

Refrence
1. Parkar bhagyashree, et al., 2015 Recent trends in stability testing of pharmaceutical
products a review RJPBS 6 (1) 1557-1569
2. The GCC Guidelines for Stability Testing of Drug Substance and pharmaceutical
products edition 2 1428H 2007 G
3. Ajay Malik ,Vipin Kumar,Renu,Sunil,Tarun Kumar. J Chem Pharm Res
2011;3(2):892-898.
4. Agraval Vipul, Sharma Devesh. J Pharm Sci Innov 2012;1(2); 18-23
5. Sanjay Bajaj. J App Pharm Sci 2012;2(3):129-138
6. Asean Guideline on stability study of drug,Version 6.0,Update Version May 2013

18
7. Dr. Venkat Shinde, https://veeprho.com/author/venkatshinde/
8. Maninderjit Kaur, Gurpreet Kaur, Harpreet Kaur,Saurav Sharma, Overview on
stability studies Institute of Pharmaceutical Sciences, District . Jalandhar, Punjab,
India
9. Singh, Anita & Afreen, Sadaf & Dhirendra, Pratap & Singh, & Kumar, Rajeev.
(2017). A REVIEW ON PHARMACEUTICAL IMPURITIES AND THEIR
IMPORTANCE *Corresponding Author. WORLD JOURNAL OF PHARMACY
AND PHARMACEUTICAL SCIENCES. 6647. 1337-1354. 10.20959/wjpps201710-
10280.
10. An Introduction To Forced Degradation Studies For Drug Substance & Drug Product
By Vinubhai N. Patolia January 9, 2020
11. https://www.melbecmicrobiology.co.uk/services/stability-compatibility-testing/
12. Carstensen, J.T., “Stability and Dating of Solid Dosage Forms” Pharmaceutics of
Solids and Solid Dosage Forms, Wiley-Interscience, 182-185, 1977
13. E book of https://www.campdenbri.co.uk/blogs/predictive-modelling.php
14. Rasane, P., Jha, A., & Sharma, N. (2015). Predictive modelling for shelf life
determination of nutricereal based fermented baby food. Journal of food science and
technology, 52(8), 5003–5011. https://doi.org/10.1007/s13197-014-1545-x
15. Lan zhang, shirui mao (2017) Application of quality by design in the current drug
development AJPS 12, 1 ; 1-8
16. https://www.aeologic.com/blog/the-use-of-artificial-intelligence-in-quality-assurance/
17. Phan, H. T., & Haes, A. J. (2019). What Does Nanoparticle Stability Mean?. The
journal of physical chemistry. C, Nanomaterials and interfaces, 123(27), 16495–
16507. https://doi.org/10.1021/acs.jpcc.9b00913
18. QUALITY OF BIOTECHNOLOGICAL PRODUCTS: STABILITY TESTING OF
BIOTECHNOLOGICAL/BIOLOGICAL PRODUCTS Annex to the ICH Harmonised
Tripartite Guideline for the Stability Testing of New Drug Substances and Products.
1996 , 1-9
19. Stability testing of biotechnological /biological veterinary medicinal products the
eropean agency for the evalvation of medicinal products veterinary medicines and
information technology unit 2001, 1-10
20. Mahesh Ramanadham, Pharm.D./M.B.A., USPHS Deputy Director, Global Regulatory
Harmonization Challenges and Opportunities: FDA Perspective (ramanadham-slides)
21. Danielle J. Beetler, Damian N. Di Florio, Ethan W. Law, Chris M. Groen, Anthony J.
Windebank, Quinn P. Peterson, DeLisa Fairweather, The evolving regulatory
landscape in regenerative medicine, Molecular Aspects of Medicine, Volume 91,2023.
22. Vokinger, K. N., Kesselheim, A. S., Glaus, C. E. G., & Hwang, T. J. (2022).
Therapeutic Value of Drugs Granted Accelerated Approval or Conditional Marketing
Authorization in the US and Europe From 2007 to 2021. JAMA health forum, 3(8),
e222685. https://doi.org/10.1001/jamahealthforum.2022.2685
23. https://www.theguardian.com/business/2005/apr/08/health.society
24. https://www.pfizer.com/news/press-release/press-release-detail/
pfizer_receives_fda_approval_for_prevnar_13_in_adults_age_18_through_49
25. Bobbili naga pavani 2023 Factors Influencing Drug Stability and Degradation - A
Comprehensive Overview

19
https://www.icliniq.com/articles/drug-and-supplements/factors-influencing-drug-
stability-and-degradation

20

You might also like