You are on page 1of 71

Stability Studies

(Role in Pharmaceutical Product Development)

(Course_405)
--------------------- Dr Md Bytul Mokaddesur Rahman
Professor of Pharmacy, RU
“More processing- less stable”;
“More contact with water – less stable”.

 Food has to be well preserved if we want it to retain its quality.


 If raw materials such as rice and wheat grains are not stored at
the proper temperature and humidity they are spoiled by
microbial contamination.
 Food is spoiled by three varieties of decomposition; physical,
chemical and microbiological.
 Rice grains left alone stay fine for years together; rice made
into flour is less stable (with time it develops a musty odor and
off color) and rice processed into food is stable only for a few
hours.
All this is true for drugs also

◉ Pure drugs, solids, liquids, or gases are usually more stable than
their formulations.
◉ When they are formulated into medicines decomposition happens
faster because of the presence of excipients, and moisture and
because of processing.
◉ So when we say stability, we actually are talking about two
varieties of stability; that of the drug and that of the formulation.
◉ For example a capsule may become soft, an emulsion may break,
a suspension may cake and an ointment may bleed.
◉ Even if solid pure powders of drugs are stored under ideal
temperature and humidity conditions – even then there is some
degradation.
For example consider Aspirin

For example consider aspirin


If you take a bottle of an old sample of pure aspirin and smell it you can
clearly feel the unmistakable odor of acetic acid.
If you granulate aspirin and make it into tablets the rate of decomposition
will be faster and if you formulate it into a suspension it will decompose
totally into acetic acid and salicylic acid in less than 25 days.
This is because the breakdown is a chemical reaction involving the
collisions of the molecules, collisions having sufficient energy and the
molecules having the proper orientation. This breakdown is affected by
various factors such as presence of oxygen, acidity, alkalinity, moisture
and light. This breakdown is what we are calling degradation.
This is the reason why the manufacturers of medicines are bound by rules
to put an expiry date on the medicine. In fact if they can not put an expiry
date then they have to explain reasons for that.
◉ So the researchers in charge of the formulation development do
extensive studies to understand the mechanism of degradation and
the rate of degradation.
◉ Degradation happens because the molecules are hitting one another
(like agitated football players in a big field running madly without
looking and hitting one another). But there is a method in this
confusion.
◉ And it is a pharmaceutical technologist’s job to find the factors that
enhance or reduce this madness (light? Heat? Air? Acidity?
Alkalinity?).
◉ And it is a pharmaceutical chemistry man’s job to find the
mechanism of this madness- how is it triggered and how is it
happening? Oxidation? Hydrolysis? Racemization? Photolysis?
Pharmaceutical Degradation

The incapacity or incapability of a particular formulation in a


specific container to remain within a particular chemical,
microbiological, therapeutical, physical & toxicological
specification in a specified period of time.
PHYSICAL
DEGRADATION

Pharmaceutical
degradation

CHEMICAL MICROBIOLOGICAL
DEGRADATION DEGRADATION
Physical Degradation

It is the degradation which results into the change of physical nature of


drug. The formulation is totally changed by way of appearance,
organoleptic properties, hardness, brittleness, particle size.

Factors effecting physical degradation are as under:


1. Loss of volatile components
2. Loss of H2O
3. Absorption of H2O
4. Crystal growth
5. Polymorphic changes
6. Color changes
Chemical Degradation

It is the separation of chemical compound into elements or simpler


compounds. Change in the chemical nature of the drug is called as
chemical degradation.

Types of chemical degradation are


1. Hydrolysis
2. Oxidation
3. Decarboxylation
4. Isomerization
5. Polymerization
Factors Affecting Drug Stability
Factors affecting drug stability:
1.Temperature: High temperature accelerate oxidation, reduction and
hydrolysis reaction which lead to drug degradation
2. pH:
 Acidic and alkaline pH influence the rate of decomposition of most
drugs.
 Many drugs are stable between pH 4 and 8.
 Weekly acidic and basic drugs show good solubility when they are
ionized and they also decompose faster when they are ionized.
 Sometimes pH can have a very serious effect on decomposition. As little
as 1 pH unit change in pH can cause a change of ten fold in rate constant.
So when we are formulating a drug into a solution we should carefully
prepare a pH - decomposition profile
3. Moisture:
a. Water catalyzes chemical reactions as oxidation, hydrolysis and
reduction reaction
b. Water promotes microbial growth
4. Light: affects drug stability through its energy or thermal effect which
lead to oxidation
5. Pharmaceutical dosage forms: Solid dosage forms are more stable
than liquid dosage forms for presence of water.
6. Concentration: Rate of drug degradation is constant for the solutions of
the same drug with different concentration. So, ratio of degraded part to
total amount of drug in diluted solution is bigger than of concentrated
solution.
7. Drug incompatibility: Reactions between components of
pharmaceutical dosage forms itself or between these components and
cover of the container .
8. Oxygen: exposure of drug formulations to oxygen affects their stability
Types of Stability

Therapeutical
Stability

Chemical Physical
Stability Stability
I
AM
STABLE

Microbiological Toxicologic
Stability Stability
Types of Stability
WHY…?

 The stability of pharmaceutical preparations


should be evaluated by exposing the product to
normal shelf conditions for a year or extended
periods. The rate of decomposition is slow at
room temperature. Such a method is time
consuming and uneconomical.
 Ideally any commercial pharmaceutical product should have a
shelf life of 5 yrs and should not fall below 90-95% potency
under recommended storage.

 In designing a solid dosage form it is necessary to know the


inherent stability of the drug substance, excipients to be used,
formulation procedure.

 For a drug substance, we need to study 3 categories of


stabilities-
1. Solid state stability of drug only (physical and chemical
stability)
2. Compatibility studies (drug + excipients)
3. Solution phase stability
SOLUTION PHASE STUDY

 This study assures that the drug substance does not


degrade intolerably when exposed to gastrointestinal
fluids.

 Stability of dissolved drug in buffers ranging from pH


1-8 is investigated.

 Example- if it degrades in acidic solutions a less


soluble form will show increased bioavailability.
TESTING SCOPE FOR SOLID DOSAGE

 Physical-chemical properties
–Appearance
– Elasticity
– Mean mass
– Moisture
– Hardness
– Disintegration
– Dissolution

 Chemical properties
– Assay
– Degradation

 Microbial properties

 Container closure system properties


– Functionality tests (e.g. extraction from blister)
TESTING SCOPE FOR LIQUID FORMS FOR
INJ. AND PARENTRAL

 Physical-chemical properties
– pH
– Loss on weight
– Color & clarity of solution
- Sterility Tests
 Chemical properties
– Assay
– Degradation products
– Degradation preservatives
– Content antioxidants,
 Microbial properties
- Pyrogen Testing
 Container closure system properties
– Functionality tests
- Leakage test
TESTING SCOPE FOR ORAL LIQUID FORM

 Physical-chemical properties
– pH
– Color & clarity of solution
– Viscosity
– Particle size distribution (for oral suspensions only)
 Chemical properties
– Assay
– Degradation products
– Degradation preservatives
– Content antioxidants
 Microbial properties

 Container closure system properties


– Functionality tests
TESTING SCOPE FOR SEMI LIQUID FORMS

 Physical-chemical properties
– Appearance, odor, homogeneity, consistency
– Loss on weight, Viscosity
– Content uniformity (within the container)
 Chemical properties
– Assay
– Degradation products & preservatives
– Content preservatives
– Degradation– Content antioxidants
 Microbial properties

 Container closure system properties


– Functionality tests
• Quality Guidelines “Q” (chemical and pharmaceutical QA)
– details see next slide

• Safety Guidelines “S” (in vitro and in vivo pre-clinical studies)


– covering Carcinogenicity Testing, Genotoxicity Testing,
Toxicokinetics and Pharmacokinetics ….. etc.

• Efficacy Guidelines “E” (clinical studies in human subject)


– Covering clinical safety, Dose Response Studies, Good
Clinical Practices, Clinical evaluation …. etc.

• Multidisciplinary Guidelines “M”


– Covering Medical Terminology, Electronic Standards for
Transmission of Regulatory Information …… etc.
– Important for Stability !
» Guideline M4: The Common Technical Document (CTD)
Stress Testing

Stress testing may be carried out on a single batch of the API. It


should include the effect of temperature (in 10 °C increments
(e.g. 50 °C, 60 °C, etc.) above the temperature used for
accelerated testing), humidity (e.g. 75% relative humidity (RH)
or greater) and, where appropriate, oxidation and photolysis on
the API. The testing should also evaluate the susceptibility of
the API to hydrolysis across a justified range of pH values when
in solution or suspension
ICH Guidelines for Stability Testing

The intended market and the climatic conditions in the area in


which the drug product will be used should be taken into account
for the design of the stability testing programme. For the purpose
of stability testing, the whole world has been divided into four
zones (I - IV) depending upon the environmental conditions the
pharmaceutical products are likely to be subjected to during their
storage. These conditions have been derived on the basis of the
mean annual temperature and relative humidity data in these
regions.

Based upon this data, long-term or real-time stability testing


conditions and accelerated stability testing conditions have been
derived.
Test Schedule for Stability Test of
New Products
Stability and Stability Test of
Pharmaceutical Products

Stability
The ability of a pharmaceutical product to retain its chemical,
physical, microbiological and biopharmaceutical properties
within specified limits throughout its shelf-life.

Stability tests
A series of tests designed to obtain information on the stability of
a pharmaceutical product in order to define its shelf-life and
utilization period under specified packaging and storage
conditions.
Types of Stability Studies

• 25 °C ± 2 °C / • 30 °C ± 2 °C /
60% ± 5% RH 65% ± 5% RH

Long Term or
Intermediate
Real Time
Stability Study
Stability Study

Types of
Accelerated
Stability Stability Study
Study
• 40 °C ± 2 °C /
75% ± 5% RH
Long term stability studies are performed by testing the sample at specific time intervals
and conditions of external parameters are changed accordingly. Main objective of this
study is to determine shelf-life of the drug product. 30°C ± 2°C/65% RH ± 5% RH can be
a suitable alternative long-term storage condition to 25°C ± 2°C/60% RH ± 5%

Intermediate stability studies conducted at 30°C/65% RH and designed to moderately


increase the rate of chemical degradation or physical changes for a drug substance or drug
product intended to be stored long term at 25°C.

Accelerated stability studies include use of exaggerated storage conditions designed to


study increased rate of physical and chemical degradation. This is part of the formal
stability studies. Data from these studies is uses to carry out long term stability studies i.e.
to determine shelf-life of the drug product.

In-use stability studies is specifically for the drugs that are prescribed to be taken in more
than one dose or multi-dose drugs. The chemical composition and physical stability of
these drugs are such that due to repetitive opening and closing, it gets degraded due
microbial contamination. The purpose of in-use stability testing is to establish – where
applicable – a period of time during which a multi-dose product can be used until
retaining quality within an accepted specification once the container is opened.
Accelerated Stability Studies

◉ Studies designed to increase the rate of chemical degradation or


physical change of an active substance or drug product by using
exaggerated storage conditions with the purpose of monitoring
degradation reactions as part of the formal, definitive storage
programme.

◉ To evaluate the impact of short term excursions and predicting


the shelf-life under normal storage conditions.

◉ The design of accelerated studies may include elevated


temperature, high humidity and intense light.
Accelerated Stability Studies

Storage Conditions

Temperature (0C) Relative Humidity% Minimum Time

40 0C +/- 2 0C 75 +/-5% RH 3, 6 Months

◎ Storage condition of 400C and relative humidity of 75% has been


recommended for all the four zones for drug substances and drug
products.

◎ Studies carried out for 3, 6 months.

◎ In general, the accelerated stability conditions must be at least 15’C


above the actual storage temperature and appropriate relative
humidity.
Limitations of Accelerated Stability
Studies

1) ACCSS are valid only when the break down depends on


temperature.
2) It is not useful when degradation is due to:
a) Microbial contamination
b) Photochemical reactions
c) Diffusion
d) Excessive agitation
3) Stability studies are meaningless when the product looses its
physical integrity at higher temperatures. For example, coagulation
of suspending agents, cracking of emulsion, loss of consistency of
ointment etc.
4) It is not useful when the order changes at elevated temperatures.
A `Significant Change` in Stability based on ICH and
FDA Guidelines

In general, “significant change” for a drug product is defined as:

1. 5% change in assay from its initial value; or failure to meet the acceptance
criteria for potency when using biological or immunological procedures;
2. Any degradation product’s exceeding its acceptance criterion;
3. Failure to meet the acceptance criteria for appearance, physical attributes,
and functionality test (e.g., color, phase separation, re-suspendibility, cakin
hardness, dose delivery per actuation); however, some changes in physical
attributes (e.g., softening of suppositories, melting of creams) may be
expected under accelerated conditions; and, as appropriate for the dosage
form:
4. Failure to meet the acceptance criterion for pH; or
5. Failure to meet the acceptance criteria for dissolution for 12 dosage units.
Shelf life and Expire date of
Pharmaceutical Products
Shelf-life
The period of time during which a drug product, if stored correctly, is expected to
comply with the specification as determined by stability studies on a number of
batches of the product. The shelf-life is used to establish the expiry date of each batch.

Shelf life is the time during which the product, if stored appropriately as per the
manufacturer’s instructions, will retain fitness for use (>90% of label claim of
potency).

Shelf life is the time taken by the drug to get reduced up to 90% or degradation of 10
% of active drug.

Shelf life is defined as the time necessary for the drug to decay to 90% of its original
concentration. Product will remain stable when stored under recommended storage
conditions. Thus, an expiration date is the date beyond which it is predicted that the
product may no longer retain fitness for use.
Shelf life and Expire date of
Pharmaceutical Products
Expiration Date
Expiration Date is the date beyond which it is predicted that the product
may no longer retain fitness for use because of the potency of the drug is
reduced and it may have less therapeutic action and duration of action.

An expiration date is defined as the time up to which the product will


remain stable when stored under recommended storage conditions.

The expiration date is also defined as the date placed on the


container/labels of a drug product designating the time during which a
batch of the product is expected to remain within the approved shelf life
specifications, if stored under defined conditions and after which it should
not be used
Shelf life of Pharmaceutical Products

For new drugs, it is a general practice to grant only two-year expiry


initially, which is based on satisfactory one year long-term and 6
months accelerated stability data. The expiry date for third and later
years is allowed only on production of real-time data for the
subsequent years.

Most pharmaceutical products are characterized by only one shelf life.


However, in some cases a product may have two e.g. a freeze-dried
(lyophilized) protein product may have only 1 shelf life, say 2 years,
for the product stored in the dry condition and a 2nd shelf life, say 2
days, for the product when it has been reconstituted with the
appropriate vehicle and is ready for injection.
Shelf life Estimation of Pharmaceutical
Products

How the shelf life of the pharmaceutical products and substances is determined according
the ICH guidelines for stability testing.

All pharmaceutical drugs degrade with the time forming the byproducts. These byproducts
may harmful to the health of the patients consuming the drug. The shelf life of the
pharmaceutical products is the time period for which the product maintains its identity and
quality when stored at the conditions defined on the label of the product.

It is important to set the time frame to consume the pharmaceutical products. ICH Q1E
guideline provides guidance for the estimation of the shelf life of the pharmaceutical
products and substances. Shelf life is determined by the evaluation of whole stability
data of the product.

Minimum data of three batches are used to estimate the shelf life of pharmaceutical
products. For the products stored at room temperature (Long or Real Time Stability Study),
the assessment should be started from the occurrence of the significant change in product
stored in accelerated conditions. If no significant change is found in the accelerated
conditions then shelf life would depend upon the long-term storage conditions data.
Shelf life Estimation of Pharmaceutical
Products

If long-term studies are conducted at 25 °C ± 2 °C / 60% RH ± 5% RH and


“significant change” occurs at any time during six months’ testing at the
accelerated storage condition (40 °C ± 2 °C / 75% ± 5% RH), additional
testing at the intermediate storage condition (30 °C ± 2 °C / 65% ± 5% RH)
should be conducted and evaluated against significant change criteria.
Testing at the intermediate storage condition should be a minimum of four
time points, including the initial and final time points (e.g. 0, 6, 9 and 12
months), from a 12-month study is recommended.
Shelf life Estimation of Pharmaceutical
Products
If long-term and accelerated conditions data show very little change or no change at all
then it might be assumed that the product would be stable during the proposed shelf
life period.

In this case, there is no requirement of the statistical analysis and the justification for
the same should be given. In this case, the shelf life of the product can be proposed
twice but should not be more than 12 months of the period covered by the long-
term evaluation data.

If long-term condition or accelerated condition data shows change then intermediate


stability study is used to estimate the shelf-life of the drug product. If there is no
change in intermediate study data, the shelf life period can be defined 1.5 times of the
period covered by the long-term data but should not be more than 6 months of the
period covered by the long-term evaluation data.

When the data is sufficient for the statistical calculations shelf-life would be defined
double but not more than 12 months from the period covered by the long-term data.
Layout for the Process of Determination of the Shelf Life of the
Pharmaceutical Products.

Tabulate stability data on Y Shelf Life


all parameters at all
storage conditions and
Months of the period covered by
evaluate separately X the long-term Study

Any Significant
change at Any Significant No extension; Shelf
accelerated
Yes change at Yes life can be assigned
condition within 6 intermediate based on long term
months? conditions? stability data only

No
No

Y= up to 2X but Y= up to 1.5X but


not exceeding not exceeding X+6
X+12 months months
Determination of the Shelf Life If no significant change at
Accelerated stability data

36 Accelerated Data Long Term Data Proposed Self Life


30
30
24
24
Months

18
18
12 12
12
6 6 6 6
6
0

CLAUSE I

Y= up to 2X but Y= Shelf Life;


not exceeding X= Actual Stability Period
X+12 months Covered in Long Term
Determination of the Shelf Life If a significant change at Accelerated
stability data and No significant change at Intermediate data

CLAUSE II
30

MONTHS
18

6
1 2 3
Intermediate Study 12 12 12
Long Term Study 12 18 24
Proposed Self Life 18 24 30

Y= up to 1.5X but
Y= Shelf Life;
not exceeding X+6
X= Actual Stability Period
months
Covered in Long Term
ICHQ1D (Bracketing & Matrixing Design
for Stability Study)

Bracketing
The design of a stability schedule such that only samples on the
extremes of certain design factors (e.g., strength and package size)
are tested at all time points as in a full design. The design assumes
that the stability of any intermediate level is represented by the
stability of the extremes tested.

For example, this indicates that for a product with three different
strengths, say tablets at 2, 4 and 6 mg, it may be possible to omit
testing of the 4 mg tablets.
The use of a bracketing design would not be considered appropriate
if it cannot be demonstrated that the strengths or container sizes
and/or fills selected for testing are indeed the extremes.
1. Design Factors
Design factors are variables (e.g., strength, container size and/or fill) to be evaluated in
a study design for their effect on product stability.
◉ Strength:- Bracketing can be applied to studies with multiple strengths of identical
or closely related formulations. Examples include but are not limited to
1. capsules of different strengths made with different fill plug sizes from the
same powder blend,
2. tablets of different strengths manufactured by compressing varying amounts
of the same granulation, and
3. oral solutions of different strengths with formulations that differ only in minor
excipients (e.g., colourants, flavourings).
With justification, bracketing can be applied to studies with multiple strengths where
the relative amounts of drug substance and excipients change in a formulation. Such
justification can include a demonstration of comparable stability profiles among the
different strengths of clinical or development batches. In cases where different
excipients are used among strengths, bracketing generally should not be applied.
◉ Container Closure Sizes and/or Fills

Bracketing can be applied to studies of the same container closure system where
either container size or fill varies while the other remains constant.

However, if a bracketing design is considered where both container size and fill vary,
it should not be assumed that the largest and smallest containers represent the
extremes of all packaging configurations. Care should be taken to select the extremes
by comparing the various characteristics of the container closure system that may
affect product stability. These characteristics include container wall thickness, closure
geometry, surface area to volume ratio, headspace to volume ratio, water vapour
permeation rate or oxygen permeation rate per dosage unit or unit fill volume, as
appropriate. With justification, bracketing can be applied to studies for the same
container when the closure varies. Justification could include a discussion of the
relative permeation rates of the bracketed container closure systems.
2. Design Considerations and Potential Risks

If, after starting the studies, one of the extremes is no longer expected to be
marketed, the study design can be maintained to support the bracketed
intermediates. A commitment should be provided to carry out stability studies
on the marketed extremes post-approval. Before a bracketing design is applied,
its effect on the retest period or shelf life estimation should be assessed. If the
stability of the extremes is shown to be different, the intermediates should be
considered no more stable than the least stable extreme (i.e., the shelf life for
the intermediates should not exceed that for the least stable extreme).
Example of Bracketing Design for
Stability Study

Strength 10 mg 20 mg 30 mg
Batch 1 2 3 1 2 3 1 2 3
25 ml T T T T T T
Container Size 50 ml
Or Fill Volume
75 ml T T T T T T

Key: T _ Tested Sample

This example is based on a product available in three strengths and three container
sizes. In this example, it should be demonstrated that the 25 ml and 75 ml high-density
polyethylene container sizes truly represent the extremes. The batches for each
selected combination should be tested at each time point as in a full design.
Example of Bracketing Design for
Stability Study

Batch

X= Batch Tested; 0= Batch not tested


Matrixing Design for Stability Study

Matrixing
The design of a stability schedule such that a selected subset of the
total number of possible samples for all factor combinations is
tested at a specified time point. At a subsequent time point, another
subset of samples for all factor combinations is tested. The design
assumes that the stability of each subset of samples tested
represents the stability of all samples at a given time point.

Therefore, at a given time point (other than the initial or final ones)
not every batch on stability needs to be tested.
Example of Matrixing Design for Stability Study
Protocol for stability testing

The protocol for stability testing is a pre-requisite for starting stability


testing and is necessarily a written document that describes the key
components of a regulated and well-controlled stability study. Because
the testing condition is based on inherent stability of the compound, the
type of dosage form and the proposed container-closure system, the
protocol depends on the type of drug substance or the product. In
addition, the protocol can depend on whether the drug is new or is
already in the market. The protocol should also reflect the regions where
the product is proposed to be marketed e.g. if the product is planned to be
used in climatic zones I-III, IVa and IVb, the stability program must
include all these zones. A well designed stability protocol should contain
the following information.
Protocol for Stability Testing

⦿ Containers and closures


⦿ Orientation of storage container
⦿ Sampling interval
⦿ Type, size and number of batches
⦿ Plan of sampling
⦿ Storage conditions
⦿ Test methodology
⦿ Acceptance criteria
Addition of Overages

1. Excess amount of the drug can be added to the preparation to maintain 100%
of the labeled amount during the shelf life of the product.
2. Overages are calculated from the accelerated stability studies and added to
the preparation at the time of manufacture.
3. They should be within the limits compatible with the therapeutic
requirement.
4. Addition of overages doubles the shelf life of the product.
5. Overages are added in multi vitamin preparations
Importance of Addition of
Overages

110%

1 Year

2 years 100%

90%

You might also like