Pharmaceutical Forced Degradation Studies with Regulatory
Consideration
Namdeo G. Shinde1*, Bhaskar N. Bangar1, Sunil M. Deshmukh1,
Suyog P. Sulake1,
Dipak P. Sherekar2
1Department
of Pharmaceutics, Satara College of Pharmacy, Degaon, Satara-415004, (MS) India.
2Satara
College of Pharmacy, Degaon, Satara-415004, (MS)
India.
*Corresponding Author E-mail: pr.shindenamdeo@gmail.com
ABSTRACT:
Forced degradation
is a powerful tool used routinely in pharmaceutical development in order to
develop stability-indicating methods that lead to quality stability data and to
understand the degradation pathways of the drug substances and drug products.
These experiments generally expose the material to an external stress to assess
the stability of the constituents or formulation. External Stress mainly
includes temperature, pH, light, moisture, and even exposure to other materials
within the product formulation, and their degradation products. Conventionally,
degradation tests can take very long periods of time, because standard test
methods require the materials to be exposed to stress factors for periods of
weeks or longer, and then tested using standard analytical methods. Accelerated
testing is of clear benefit, as use of elevated temperature to increase the
rate of interactions is the most powerful factor to shorten the length of time
required for these tests. Forced degradation studies ensure appropriate
stability of final pharmaceutical products in very early stages of
pharmaceutical development.
KEYWORDS: Forced
degradation study, Active Pharmaceutical Ingredient (API), and International
Conference on Harmonization (ICH).
INTRODUCTION:
Forced degradation is the
process of subjecting drug compounds to extreme chemical and environmental
conditions to determine product breakdown levels and preliminary degradation
kinetics, and to identify degradant species. The
stress testing practices that companies use can vary significantly and can have
a serious impact on the analytical methodology used throughout the industry.
Stress testing studies can be conducted to challenge specificity of stability
indicating and impurity monitoring methods as part of validation protocol.
Forced degradation or stress testing studies are part of the development
strategy and are an integral component of validating analytical methods that
indicate stability and detect impurities. This relates to the specificity
section of the validation studies. It is important to recognize that forced
degradation studies are not designed to establish qualitative or quantitative
limits for change in drug substance or drug product.
Forced degradation studies are
used for multiple purposes, including demonstration of the specificity of
separation methods, gaining insight into degradation pathways, and discernment
of degradation products in formulations that are related to drug substances
versus those that are related to other ingredients of a formulation. Reliable
chemical stability testing data can show how a drug product changes over time
with influence of environmental factors. The stability of a drug product or a drug substance is a critical
parameter, which may affect purity, potency and safety. Changes in drug
stability can risk patient safety by formation of a toxic degradation
product(s) or deliver a lower dose than expected. Therefore, it is essential to
know the purity profile and behaviour of a drug substance under various
environmental conditions.
The main purpose of forced degradation testing
studies is to evaluate the overall photosensitivity of the material for method
development purposes and/or degradation pathway elucidation. This testing may
involve the drug substance alone and/or in simple solutions/suspensions to
validate the analytical procedures. For development and validation purposes, it
is appropriate to limit exposure and end the studies if extensive decomposition
occurs. For photostable materials, studies may be
terminated after an appropriate exposure level has been used. The
initial purpose of forced degradation studies is to investigate
stability-related properties of an API and to develop an understanding of the
degradation products and pathways. These studies should also be used to
evaluate the susceptibility of the drug substance to hydrolysis across a wide
range of pH values. Forced degradation studies are used to provide degraded
samples for the development of stability-indicating analytical methods for the
API. The information developed from a forced degradation study can be utilized
in several other areas of development, including analytical methods
development, formulation development and storage conditions, manufacturing
processing, safety toxicological, identification of possible genotoxic degradants,
identification of potential metabolites and API design/discovery.
Forced degradation is synonymous
with stress testing and purposeful degradation. Purposeful degradation can be a
useful tool to predict the stability of a drug substance or a drug product with
effects on purity, potency, and safety. It is imperative to know the impurity
profile and behaviour of a drug substance under various stress conditions.
Forced degradation also plays an important role in the development of
analytical methods, setting specifications, and design of formulations under
the quality-by-design (QbD) paradigm. The nature of
the stress testing depends on the individual drug substance and the type of
drug product (e.g., solid oral dosage, lyophilized powders, and liquid
formulations) involved. Forced
degradation studies are most beneficial if done early in the drug development
process. The reasoning for this is that these studies yield predictive
information on the nature of the degradants which are
valuable when assessing the appropriate synthesis routes, API salt selection
and formulation development. These early studies can help to provide
information needed for the following:
1.
As a predictive tool to understand
degradation pathways and stability-related issues.
2.
To predict API stability before
real-time stability data is available.
3.
Development and validation of
stability-indicating methodology.
4.
Determination of degradation pathways of
drug substances and drug products.
5.
Discernment of degradation products in
formulations that are related to drug substances versus those that are related
to non–drug substances (e.g., excipients).
6.
Structure elucidation of degradation
products.
7.
Determination of the intrinsic stability
of drug substances.
8.
Helps to identify reactions that cause degradation of
pharmaceutical products.
9.
To generate a degradation profile that mimics what would be
observed in a formal stability study under ICH conditions.
10.
To generate more stable formulation.
11.
To identify impurities related to drug substances and excipients.
12.
To understand the drug molecular chemistry.
13.
Selection of storage conditions, packaging and better
understanding of the potential liabilities of the drug molecule chemistry. [1]
Forced
degradation studies can be done on both, the solid state and aqueous solution
or suspension forms of the API. Furthermore, the use of analysis at multiple
time points allows for approximation of rates of degradation and such testing
at early time points can provide a distinction between primary and secondary
degradation products. This approach allows for better degradation pathway
determination.
Forced
degradation studies should be repeated as needed throughout the drug
development process. For example, when there are changes in API impurity
profile, API salt or polymorph form. When carried out in late development, such
studies are referred as confirmatory studies. Confirmatory studies are
quantitative in nature. Full mass accountability of the API, its impurities and
degradation products are generated from these late stage studies. Furthermore,
based upon the outcome of these studies, if necessary, new or orthogonal
methods may need to be developed to account for all observed degradation. In
addition, confirmatory studies for API are done after finalization of the
synthetic route and form of the API. Such studies are typically done in Phase
III with one of the registration batches of the API. For drug products,
confirmatory studies are done when final formulation(s) and packaging are
chosen. After the confirmatory studies are completed, a report on degradation
products and pathways is generated and included in or used to support NDA
filings.
DEGRADATION CONDITIONS [2, 3]
The
following are general conditions that should be considered when conducting
forced degradation studies:
Solid State
·
Heat
·
Heat/humidity
·
Light
Solution and/or Suspension
·
Hydrolysis at various pHs
·
Unbuffered HCl, NaOH, water
·
Buffer solutions (used to determine if
pH adjustment needed to attain
maximum stability)
·
Oxidative stress testing
·
H2O2 (to mimic
possible presence of peroxides in excipients)
·
Metal ions (to mimic possible exposure during
manufacture)
·
Radical initiators (to mimic autoxidation)
·
Light
SOURCES OF IMPURITIES AND TYPES
OF IMPURITIES:
In a typical study, relevant
stress conditions are light, heat, humidity, hydrolysis (acid / base
influence) and oxidation or even a combination of described parameters. If it
is necessary to form degradation products, the strength of stress conditions
can vary due to the chemical structure of the drug substance, the kind of drug
product, and product specific storage requirements. An individual program has
to be set up in order to reach a target degradation of 5 to 20%. A higher level
of degradation will be out of the scope of product stability requirements and
therefore unrealistic. The scope of the test is to generate degradation products
in order to facilitate a method development for determination of the relevant
products. Therefore, samples will be stressed in a solid form and/or in
solution. Typically, stress tests are carried out on one batch of material.
For drug products the placebo should be stressed in a similar way in order to
exclude those impurities that are not degradation products (e.g. impurities
arising from excipients). Table 1 shows typical
stress conditions of API and drug product. Drugs that are poorly soluble in water
can be conducted either in suspension or in solution using inert organic
co-solvents (e.g., DMSO, acetic acid or propionic
acid). It is important to avoid co-solvents that may be reactive with the drug
or complicate analysis (e.g. by LC-MS). [7, 8]
For drug substances, photostability testing should consist of two parts: forced
degradation testing and confirmatory testing. The purpose of forced degradation
testing studies is to evaluate the overall photosensitivity of the material for
method development purposes and/or degradation pathway elucidation. This
testing may involve the drug substance alone and/or in simple
solutions/suspensions to validate the analytical procedures. In these studies,
the samples should be in chemically inert and transparent containers. In these
forced degradation studies, a variety of exposure conditions may be used,
depending on the photosensitivity of the drug substance involved and the
intensity of the light sources used. For development and validation purposes it
is appropriate to limit exposure and end the studies if extensive decomposition
occurs. For photostable materials, studies may be
terminated after an appropriate exposure level has been used. The design of
these experiments is left to the applicant’s discretion although the exposure
levels used should be justified. The potential for these degradation pathways
should be assessed in both drug substance and drug product. These mechanisms
can be assessed in a systematic way by exposure to stress conditions of heat, humidity,
photo stress, oxidative conditions, and aqueous conditions across a broad range
of pH. The ICH guidelines Q3A (R) require
identification of each impurity with respect to both chemistry and safety
perspective. The chemistry perspective includes classification and
identification of impurities, report generation, listing of impurities in
specific guidance and a brief discussion of analytical procedures while safety
perspectives include specific guidance for qualifying those impurities. The
impurities usually encountered in pharmaceuticals are synthesis related,
formulation related, and degradation related. These ICH guidelines classify the
impurities into the fallowing categories.
1. Impurities associated with
active pharmaceutical ingredient (APIs).
2. Impurities that are formed
during formulation, formed with ageing and that are related to formulation
forms.
Impurities associated with
active pharmaceutical ingredients:
According to the ICH guidelines,
impurities associated with APIs are classified into the fallowing categories
A. Organic impurities
B. Inorganic impurities
Organic impurities:
·
Starting materials
·
Byproducts
·
Intermediates
·
Degradation products
·
Residual solvents
·
Reaction products with excipients
·
Leachables from container closure system
Inorganic impurities
·
Reagents, ligands, catalysts
·
Heavy metals or other residual metals
·
Inorganic salts
Other materials
·
Filter aids
·
Charcoal
Formulation related impurities:
During formulation, excipients added to API to render the product elegant. In
such cases, drug-excipients incompatibility may lead
to undesirable products, which can affect the therapeutic efficiency of the
product. In addition, other solvents, which is usually present in API or used
in the synthesis of API or excipients, may also
interact with excipients resulting in impurities.
Dosage form related impurities:
1. Precipitation of main ingredient
can occur due to various factors like pH, environment, or leaching. In general,
liquid dosage forms are very much susceptible to both degradation and microbial
contamination.
Example-
Precipitation of imipramine HCl
with sodium bisulfate and pH alteration of lidocaine HCl in presence of 5% dextrose solution.
2. Pharmaceutical solids: in the
presence of excipients and moisture topochemical and nucleation reactions occur.
Example- Presence
of sodium CMC during granulation of papaverine, aminopyrine and salicylic acid tablets causes reduced
coloration.
3. Microbial growth resulting from
the growth of bacteria, fungi, and yeast in a humid and warm environment may
result in oral liquid products that are unusable for human consumption.
Method related impurities:
A known impurity 1-(2, 6 Dichloro phenyl) indoline-2one is formed in the production
of a parental dosage form diclofenac sodium ampoules.
Formation of this impurity depends on initial pH of the preparation and the
conditions of sterilization i.e. autoclave method (1210c) that
enforces the intramolecular cyclic reaction of diclofenac sodium forming indolineone
derivative and sodium hydroxide.[5]
Environmental related
impurities:
The environmental factors that
can reduce stability are
i. Temperature:
There is many APIs that are
liable to heat or topical temperature. Ex: Vitamins as drug substances are very
heat sensitive and degradation frequently leads to loss of potency in vitamin
products, especially in liquid formulations. Degradation caused by exposure to
temperatures include bond breakage i.e. pyrolysis.
Any degradation mechanism that is enhanced at elevated temperatures are thermolytic pathways include Hydrolysis, Dehydration, Isomerization, Decorboxylation,
Rearrangement, Polymerization reactions, pyrolysis.
In general, rate of reaction increases with increase in temperature. Many APIs
are sensitive to heat or topical temperature such as vitamins and peptides.
Effect of temperature on thermal degradation of a substance is studied through
Arrhenius equation.
K = Ae
-Ea/RT
Where K = specific reaction rate
A = frequency factor
Ea = energy of activation
Thermal degradation study is
carried out at 400C. The mostly accepted temperature is 700C
at low and high humidity for 1-2 months. The use of high-temperatures in
predictive degradation studies assumes that the drug molecule will follow the
same pathway of decomposition at all temperatures.[9]
ii. Light (UV light): Light
is one of the means by which the formulation degrades because of photolytic
reaction. Ex: sunlight having about 8000 foot-candles can destruct nearly 34%
of vitamin B in 24 hrs.
iii. Humidity: Humidity
is one of the important key factors in case of hygroscopic compounds. Ex:
Ranitidine, Aspirin[9]
Functional group related:
1) Hydrolysis:
Drug degradation that involves
reaction with water is called hydrolysis. PH, buffer salts, ionic strength,
solvents, complexing agents, surfactants and excipients affect hydrolysis. Hydrolysis reactions are
typically acid or base catalyzed. Hydrolysis is the most common degradation
chemical reaction over wide range of pH. Water either
as solvent or as moisture in the air comes in contact with pharmaceutical
dosage form is responsible for degradation of most drugs. Hydrolytic study
under acidic and basic condition involves catalyzation
of ionisable functional groups present in the
molecule.
Example- Aspirin combined with
water and hydrolyzed to form salicylic acid and acetic acid. The hydrolytic degradation of new drug in
acidic and alkaline condition can be studied by refluxing the drug in 0.1N HCl or 0.1N NaOH. If degradation
is seen testing can be stopped at that point. In case of no degradation under
these conditions, the drug should be refluxed in acid and alkali of higher
strength and for a long duration time. Hydrolysis of most of drugs is depending
upon the relative concentration of hydronium and
hydroxyl ions.
Example- Anatrazole
– degraded in basic pH
Doxophylline
– shows degradation in acidic pH
A reaction in which water is an
reactant causing precipitation. Hydrolysis is a common phenomenon for ester
type of drugs. Esters, amides, lactones, lactams,
imides, and carbamates, are susceptible for acid base
hydrolysis.
Examples- Aspirin, Chloramphenicol, Barbiturates, Chlordiazepoxide,
Oxazepam, Examples includes Aspirin, Benzocaine, Cefolaxime, Cocaine, and Cefpodoxime.
2) Oxidation:
Many drug substances undergo autoxidation i.e. oxidation under normal conditions and
involving ground state elemental oxidation. Autoxidation
is a free radical reaction that requires free radical initiator to begin the
chain reaction. Hydrogen peroxide, metal ions, traces level of impurities in a
drug substance act as initiator for oxidation. The oxidative stress testing is
initially carried out in 3% H2O at room temperature for 6 h and it
can be increased or decreased to achieve sufficient degradation. The time can
also be increased up to 24 hr 3% or decreased up to 30 min with 1% of H2O
The mechanism of oxidative degradation of drug substance involves an electron
transfer mechanism to form reactive anions and cations.
The mechanism of oxidative degradation of drug substance involves an electron
transfer mechanism to form reactive anions and cations.
Drugs In pharmaceuticals most common form of oxidative decomposition is
oxidation through a free radical chain process.
Example- Amines, sulphide, and phenols are susceptible to electron transfer
oxidation. Drugs which prone to oxidation are hydrocortisone, methotrexate, adinazolam,
catecholamine conjugated dienes, heterocyclic
aromatic rings, nitraso and nitrate derivatives.
a) Rapamycin, an immunosuppressant drug
reacts with oxygen to form a complex mixture of monomeric
and oligomeric products. Formation of the majority of
the rapamycin degradation products could be
rationalized with free radical-mediated autoxidation
reactions involving alkenes and alcohol sites.
b) Auto-oxidation of ascorbic acid
studies reveals that cupric ion known to oxidize ascorbic acid rapidly in to dehydro ascorbic acid and potassium cyanide.
3) Photolysis:
Exposure of drug molecule to
light may produce photolytic degradation products. The rate of photo
degradation depends upon the intensity of incident light and quantity of light
absorbed by drug molecule. Photolytic degradation is carried out by exposing
the drug substance to or drug product to a combination of visible and UV light.
The photolytic degradation can occur through non-oxidative and oxidative
photolytic reaction.
a) The non-oxidative photolytic
reaction includes isomerization, diamerization,
cyclization, rearrangement, decorboxylation
and haemolytic cleavage.
b)
Oxidative photolytic reaction occurs through either single
oxidation or triple oxidation mechanism. Ex: Barnidipin.[1,10]
Photolytic cleavage on aging
includes examples of pharmaceutical drugs or products that are prone to
degradation on exposure to UV-light. During manufacturing process or packing or
on storage drugs like ergometrine, nifedipine, nitroprucide,
riboflavin, are liable to photo oxidation. Most of the compounds will degrade
as solutions when exposed to high-energy UV exposure. This photo oxidation
involves generation of free-radical intermediates, which will degrade the
product.
Example- The formulation of
ciprofloxacin eye drops 0.3% on exposure to UV-light induces photolysis, which
results in formation of ethylene di-amine analogue of
ciprofloxacin.
Selection
of Stress Conditions
Forced
degradation is normally carried out under more severe conditions than those
used for accelerated studies. The choice of stress conditions should be
consistent with the products decomposition under normal manufacturing, storage,
and use conditions, which are specific in each case. The ICH guidance recognize
that it is impossible to provide strict degradation guidelines and allows
certain freedom in selecting stress conditions for biologics. The
choice of forced degradation conditions should be based on data from
accelerated pharmaceutical studies and sound scientific understanding of the
product’s decomposition mechanism under typical use conditions. A minimal list
of stress factors suggested for forced degradation studies must include acid
and base hydrolysis, thermal degradation, photolysis, oxidation, and may
include freeze-thaw cycles and shear.
Regulatory
guidance does not specify pH, temperature ranges, specific oxidizing agents, or
conditions to use, the number of freeze-thaw cycles, or specific wavelengths
and light intensities. The design of photolysis studies is left to the
applicant's discretion although Q1B recommends that the light source should
produce combined visible and ultraviolet (UV, 320-400 nm) outputs, and that
exposure levels should be justified. Consult the appropriate regulatory
authorities on a case-by-case basis to determine guidance for light-induced
stress.
Stress conditions
Typical stress tests include
four main degradation mechanisms: heat, hydrolytic, oxidative, and photolytic
degradation. Selecting suitable reagents such as the concentration of acid,
base, or oxidizing agent and varying the conditions (e.g., temperature) and
length of exposure can achieve the preferred level of degradation.
Over-stressing a sample may lead to the formation of secondary degradants that would not be seen in formal shelf-life
stability studies and under-stressing may not serve the purpose of stress
testing. Therefore, it is necessary to control the degradation to a desired
level. A generic approach for stress testing has been proposed to achieve
purposeful degradation that is predictive of long-term and accelerated storage
conditions. The generally recommended degradation varies between 5-20%
degradation. This range covers the generally permissible 10% degradation for
small molecule pharmaceutical drug products, for which the stability limit is
90%-110% of the label claim. Although there are references in the literature
that mention a wider recommended range (e.g., 10-30%), the more extreme stress
conditions often provide data that are confounded with secondary degradation
products.
Table 1. The four climate zones
(ICH Stability guidelines)
Zone I |
Temperate |
21°C ± 2°C/ 45% RH ± 5% RH |
Zone II |
Subtropical and Mediterranean |
25°C ± 2°C/ 60% RH ± 5% RH |
Zone III |
Hot and Dry |
30°C ± 2°C/ 35% RH ± 5% RH |
Zone IV |
Hot and Humid |
30°C ± 2°C/ 65% RH ± 5% RH |
Table 2. Type of study with Storage conditions (ICH Stability guidelines)
Study |
Storage condition |
Min. time period |
Long term |
25°C ± 2°C/ 60% RH ± 5% RH or 30°C ± 2°C/ 65% RH ± 5% RH |
12 months |
Intermediate |
30°C ± 2°C/ 65% RH ± 5% RH |
6 months |
Accelerated |
40°C ± 2°C/ 75% RH ± 5% RH |
6 months |
Table 3. Drug products intended for storage in refrigerator (ICH Stability guidelines)
Study |
Storage condition |
Min. time period |
Long term |
5°C ± 3°C |
12 months |
Accelerated |
25°C ± 2°C/ 60% RH ± 5% RH |
6 months |
Table 4. Drug products intended for storage in freezer
Study |
Storage condition |
Min. time period |
Long term |
-20°C ± 5°C |
12 months |
Photostability
Photostability testing should be an integral
part of stress testing, especially for photo-labile compounds. Some recommended
conditions for photostability testing are described
in ICH Q1B Photostability Testing of New Drug Substances and Products.
Samples of drug substance, and solid/liquid drug product, should be exposed to
a minimum of 1.2 million lux hours and 200-watt hours
per square meter light. The same samples should be exposed to both white and UV
light. To minimize the effect of temperature changes during exposure,
temperature control may be necessary. The light-exposed samples should be
analyzed for any changes in physical properties such as appearance, clarity,
color of solution, and for assay and degradants. The
decision tree outlined in the ICH Q1B can be used to determine the photo
stability testing conditions for drug products. The product labelling
should reflect the appropriate storage conditions. It is also important to note
that the labelling for generic drug products should
be concordant with that of the Reference Listed Drug (RLD) and with United States Pharmacopeia (USP)
monograph recommendations, as applicable.
Heat:
Thermal stress testing (e.g.,
dry heat and wet heat) should be more strenuous than recommended ICH Q1A
accelerated testing conditions. Samples of solid-state drug substances and drug
products should be exposed to dry and wet heat, whereas liquid drug products
can be exposed to dry heat. It is recommended that the effect of temperature be
studied in 10°C increments above that for routine accelerated testing, and
humidity at 75% relative humidity or greater. Studies may be conducted at
higher temperatures for a shorter period. Testing at multiple time points could
provide information on the rate of degradation and primary and secondary
degradation products. In the event that the stress conditions produce little or
no degradation due to the stability of a drug molecule, one should ensure that
the stress applied is in excess of the energy applied by accelerated conditions
(40°C for 6 months) before terminating the stress study.
Acid and base hydrolysis:
Acid and base hydrolytic stress
testing can be carried out for drug substances and drug products in solution at
ambient temperature or at elevated temperatures. The selection of the type and
concentrations of an acid or a base depends on the stability of the drug
substance. A strategy for generating relevant stressed samples for hydrolysis
is stated as subjecting the drug substance solution to various pH (e.g., 2, 7,
10–12) at room temperature for two weeks or up to a maximum of 15%
degradation. Hydrochloric acid or sulphuric acid
(0.1M to 1M) for acid hydrolysis and sodium hydroxide or potassium hydroxide
(0.1M to 1M) for base hydrolysis are suggested as suitable reagents for
hydrolysis. For lipophilic drugs, inert co-solvents
can be used to solubilize the drug substance.
Attention should be given to the functional groups present in the drug molecule
when selecting a co-solvent. Prior knowledge of a compound can be useful in
selecting the stress conditions. For instance, if a compound contains ester
functionality and is very labile to base hydrolysis, low concentrations of a
base can be used. Analysis of samples at various intervals can provide
information on the progress of degradation and help to distinguish primary degradants from secondary degradants.
Oxidation:
Oxidative degradation can be
complex. Although hydrogen peroxide is used predominantly because it mimics
possible presence of peroxides in excipients, other
oxidizing agents such as metal ions, oxygen, and radical initiators (e.g., azobisisobutyronitrile) can also be used. Selection of an
oxidizing agent, its concentration, and conditions depends on the drug
substance. Solutions of drug substances and solid/liquid drug products can be
subjected to oxidative degradation. It is reported that subjecting the
solutions to 0.1%-3% hydrogen peroxide at neutral pH and room temperature for
seven days or up to a maximum 20% degradation could potentially generate
relevant degradation products. Samples can be analyzed at different time
intervals to determine the desired level of degradation.
Analysis method: The preferred method of
analysis for a stability indicating assay is reverse-phase high-performance
liquid chromatography. Reverse-phase HPLC is preferred for several reasons,
such as its compatibility with aqueous and organic solutions, high precision,
sensitivity, and ability to detect polar compounds. Separation of peaks can be
carried out by selecting appropriate column type, column temperature, and
making adjustment to mobile phase pH. Poorly
retained, highly polar impurities should be resolved from the solvent front. As
part of method development, a gradient elution method with varying mobile phase
composition (very low organic composition to high organic composition) may be
carried out to capture early eluting highly polar compounds and highly retained
nonpolar compounds. Stressed samples can also be
screened with the gradient method to assess potential elution pattern. Sample
solvent and mobile phase should be selected to afford compatibility with the
drug substance, potential impurities, and degradants.
Stress sample preparation should mimic the sample preparation outlined in the
analytical procedure as closely as possible. Neutralization or dilution of
samples may be necessary for acid and base hydrolyzed samples. Chromatographic
profiles of stressed samples should be compared to those of relevant blanks
(containing no active) and unstressed samples to determine the origin of peaks.
The blank peaks should be excluded from calculations. The amount of impurities
(known and unknown) obtained under each stress condition should be provided
along with the chromatograms (full scale and expanded scale showing all the
peaks) of blanks, unstressed, and stressed samples. Additionally, chiral drugs should be analyzed with chiral
methods to establish stereo-chemical purity and stability.
The analytical method of choice
should be sensitive enough to detect impurities at low levels (i.e., 0.05% of
the analyte of interest or lower), and the peak
responses should fall within the range of detector's linearity. The analytical
method should be capable of capturing all the impurities formed during a formal
stability study at or below ICH threshold limits. Degradation product
identification and characterization are to be performed based on formal
stability results in accordance with ICH requirements. Conventional methods
(e.g., column chromatography) or hyphenated techniques (e.g., LC–MS, LC–NMR)
can be used in the identification and characterization of the degradation
products. Use of these techniques can provide better insight into the structure
of the impurities that could add to the knowledge space of potential structural
alerts for genotoxicity and the control of such
impurities with tighter limits. It should be noted that structural
characterization of degradation products is necessary for those impurities that
are formed during formal shelf-life stability studies and are above the
qualification threshold limit. Various detection types can be used to analyze
stressed samples such as UV and mass spectroscopy. The detector should contain
3D data capabilities such as diode array detectors or mass spectrometers to be
able to detect spectral non-homogeneity. Diode array detection also offers the
possibility of checking peak profile for multiple wavelengths. The limitation
of diode array arises when the UV profiles are similar for analyte
peak and impurity or degradant peak and the noise
level of the system is high to mask the co-eluting impurities or degradants. Compounds of similar molecular weights and
functional groups such as diastereoisomers may
exhibit similar UV profiles. In such cases, attempts must be made to modify the
chromatographic parameters to achieve necessary separation. An optimal
wavelength should be selected to detect and quantitate
all the potential impurities and degradants. Use of
more than one wavelength may be necessary, if there is no overlap in the UV
profile of an analyte and impurity or degradant peaks. A valuable tool in method development is
the overlay of separation signals at different wavelengths to discover
dissimilarities in peak profiles.
Table 5. Stress conditions for drug substance
Type of study |
Conditions |
Time |
Acid hydrolysis |
0.1 N- 1.0 N HCl, RT or higher |
1-7 days |
Base hydrolysis |
0.1 N- 1.0 N NaOH, RT or higher |
1-7 days |
Thermal hydrolysis |
Aqueous solution, 70°C |
1-7 days |
Oxidative/ solution |
O2 + Initiator
(AIBN) in CAN/ H2O; 80/20,40°C |
1-7 days |
0.3-3.0 % H2O/ Ambient in the
dark |
Few hours to 7 days |
|
Thermal |
solid, 70°C |
Upto 3 weeks |
Thermal/ Humidity |
solid, 70°C/ 75% RH |
Upto 3 weeks |
Photo degradation |
Solid, fluorescent and UV
light |
>2× ICH |
Peak purity analysis:
Peak purity is used as an aid in
stability indicating method development. The spectral uniqueness of a compound
is used to establish peak purity when co-eluting compounds are present. Peak
purity or peak homogeneity of the peaks of interest of unstressed and stressed
samples should be established using spectral information from a diode array
detector. When instrument software is used for the determination of spectral
purity of a peak, relevant parameters should be set up in accordance with the
manufacturer's guidance. Attention should be given to the peak height
requirement for establishing spectral purity. UV detection becomes non linear
at higher absorbance values. thresholds should be set such that co-eluting
peaks can be detected. Optimum location of reference spectra should also be
selected. The ability of the software to automatically correct spectra for
continuously changing solvent background in gradient separations should be
ascertained.
Termination of study:
Stress testing studies are
terminated after ensuring adequate exposure to stress conditions. Typical
activation energy of drug substance molecules varies from 12–24
kcal/mol. A compound may not necessarily degrade under every single stress
condition. In circumstances, where some stable drugs do not show any
degradation under any of the stress conditions, specificity of an analytical
method can be established by spiking the drug substance or placebo with known
impurities and establishing adequate separation.
Forced degradation in QbD paradigm
A well-designed, forced
degradation study is indispensable for analytical method development in a QbD paradigm. A systematic process of manufacturing quality
drug products that meet the predefined targets for the critical quality
attributes (CQA) necessitates the use of knowledge obtained in forced degradation
studies. It helps to establish the specificity of a stability indicating method
and to predict potential degradation products that could form during formal
stability studies. Incorporating all potential impurities in the analytical
method and establishing the peak purity of the peaks of interest helps to avoid
unnecessary method re-development and revalidation. Knowledge of chemical
behaviour of drug substances under various stress conditions can also provide
useful information regarding the selection of excipients
for formulation development. Excipients compatibility
is an integral part of understanding potential formulation interactions during
product development and is a key part of product understanding. Degradation
products due to drug-excipient interaction or
drug-drug interaction in combination products can be examined by stressing
samples of drug substance, drug product, and placebo separately and comparing
the impurity profiles. Information obtained regarding drug-related peaks and
non-drug-related peaks can be used in the selection and development of more
stable formulations. For instance, if a drug substance is labile to oxidation,
addition of an antioxidant may be considered for the formulation. For drug
substances that are labile to acid or undergo stereochemical
conversion in acidic medium, delayed-release formulations may be necessary. Acid/base
hydrolysis testing can also provide useful insight in the formulation of drug
products that are liquids or suspensions. Knowledge gained in forced degradation
studies can facilitate improvements in the manufacturing process.
Table 6. Recommended labeling statements for finished
pharmaceutical products (FPP)
Test conditions of FPP |
Recommended labeling statement |
25°C/ 60 % RH (long term)40°C/
75 % RH (accelerated) |
“Do not store above 25° C” |
25°C/ 60 % RH (long term)30°C/
65 % RH (intermediate, failure of accelerated) |
“Do not store above 25° C” |
30° C/ 65 % RH
(long term)40°C/ 75 % RH (accelerated) |
“Do not store above 30° C” |
30° C/ 75 % RH ( long
term)40°C/ 75 % RH (accelerated) |
“Do not store above 30° C” |
5° C ± 3° C |
“Store in refrigerator” (2°
C to 8° C) |
-20° C ± 5° C |
“Store in freezer” |
If a photostability
study shows a drug substance to be photolabile,
caution should be taken during the manufacturing process of the drug product.
Useful information regarding process development (e.g., wet versus dry
processing, temperature selection) can be obtained from thermal stress testing
of drug substance and drug product. Additionally, increased scientific
understanding of degradation products and mechanisms may help to determine the
factors that could contribute to stability failures such as ambient
temperature, humidity, and light. Appropriate selection of packaging materials
can be made to protect against such factors.
GUIDELINES FOR FORCED DEGRADATON STUDY
Impurities in new drug substances
(CPMP/ICH/2737/99)
This document are intended to
provide guidance for registration applications on the content and qualification
of impurities in new drug substances produced by chemical syntheses and not
previously registered in a region or member state. It is not intended to apply
to new drug substances used during the clinical research stage of development.
The following types of drug substances are not covered in this guideline:
·
Biological/biotechnological products
·
Peptides
·
Oligonucleotide
·
Radiopharmaceuticals
·
Fermentation products
·
Semi-synthetic products
·
Herbal products, crude products of animal or plant origin.
Impurities in new drug products (CPMP/ICH/2738/99)
This guideline addresses only
those impurities in new drug products classified as degradation products of the
drug substance or reaction products of the drug substance with an excipients and/or immediate container closure system
(referred to as "degradation products"). Generally, impurities
present in the new drug substance need not be monitored or specified in the new
drug product, unless they are also degradation products. Impurities arising
from excipients present in the new drug product,
extracted, or leached from the container closure system are not covered by this
guideline. [5]
Stability Testing of New Drug Substances and Products (ICH Q1A
(R2))
Drug substance:
Stress testing a drug substance
can help identify the likely degradation products, which can in turn help
establish the degradation pathways and the intrinsic stability of the molecule
and validate the stability-indicating power of the analytical procedures used.
The nature of the stress testing will depend on the individual drug substance
and the type of drug product. Stress testing is likely to be carried out on a
single batch of the drug substance. It should include the effect of
temperatures (in 100C increments [e.g., 500C, 600C]
above that for accelerated testing), humidity (e.g., 75% RH or greater) where
appropriate, oxidation, and photolysis on the drug substance. Testing should
evaluate the susceptibility of the drug substance to hydrolysis across a wide
range of pH values when in solution or suspension. Photostability
testing should be an integral part of stress testing. Examining degradation
products under stress conditions is useful for establishing degradation
pathways, developing and validating suitable analytical procedures. It may not
be necessary, however, to examine for specific degradation products if previous
studies have demonstrated that these products are not formed under accelerated
or long-term storage conditions.
Drug product:
The design of formal stability
studies for a drug product should be based on the behavior and properties of
the drug substance, the results from stability studies on the drug substance,
and the experience gained from clinical formulation studies. The likely changes
to storage conditions and the rationale for the selection of attributes to be
tested in the formal stability studies should be stated. Photostability
testing should be conducted on at least one primary batch of the drug product
if appropriate.
Stability Testing: Photostability
Testing of New Drug Substances and Products (ICH Q1B)
Drug substance:
Photostability testing should consist of two
parts: forced-degradation testing and confirmatory testing. The purpose of
forced-degradation testing is to evaluate the overall photosensitivity of the
material for method-development purposes and/or degradation pathway
elucidation. This testing may involve the drug substance alone and/or the
substance in simple solutions and suspensions to validate the analytical
procedures. In these studies, the samples should be in chemically inert and transparent
containers. For forced-degradation studies, various exposure conditions may be
used, depending on the photosensitivity of the drug substance and the intensity
of the light sources. For development and validation purposes, it is
appropriate to limit exposure and end the studies if extensive decomposition
occurs. For photostable materials, studies may be
terminated after an appropriate exposure level has been used. The design of
these experiments is left to the applicant’s discretion although the exposure
levels used should be justified. Under forcing conditions, decomposition
products may be observed that are unlikely to be formed under the conditions
used for confirmatory studies. This information may be useful in developing and
validating suitable analytical methods. If in practice it has been demonstrated
that they are not formed in the confirmatory studies, these degradation
products need not be further examined.
Validation of Analytical Procedures: Methodology (ICH Q2B)
Drug substance/drug product:
If impurity or degradation
product standards are unavailable, specificity can be demonstrated by comparing
the test results of samples containing impurities or degradation products with
a second well-characterized procedure; e.g., pharmacopeial
method or other validated analytical procedure (independent procedure). As
appropriate, this should include samples stored under relevant stress
conditions (light, heat, humidity, acid–base hydrolysis, and oxidation).
Impurities in New Drug Substances (ICH Q3A(R))
Drug substance:
The applicant should summarize
the actual and potential impurities most likely to arise during the synthesis,
purification, and storage of the new drug substance. This summary should be
based on sound scientific appraisal of the chemical reactions involved in the
synthesis, impurities associated with raw materials that could contribute to
the impurity profile of the new drug substance, and possible degradation
products. This discussion can be limited to those impurities that might reasonably
be expected based on knowledge of the chemical reactions and conditions
involved. In addition, the applicant should summarize the laboratory studies
conducted to detect impurities in the new drug substance. This summary should
include test results of batches manufactured during the development process and
batches from the proposed commercial processes well as the results of stress
testing (see ICH Guideline Q1A on Stability) used to identify potential
impurities arising during storage. The impurity profile of the drug substance
batches intended for marketing should be compared with those used in
development and any differences discussed.
Impurities in New Drug Products (ICH Q3B(R))
Drug product:
Analytical procedures should be
validated to demonstrate specificity for the specified and unspecified
degradation products. As appropriate, this validation should include samples
stored under relevant stress conditions: light, heat, humidity, acid/base
hydrolysis, and oxidation.
FDA Guidance for Industry: Stability Testing of Drug Substances
and Drug Products
Drug substance/drug product:
Stress testing helps determine
the intrinsic stability characteristics of a molecule by establishing
degradation pathways to identify the likely degradation products, and to
validate the stability-indicating power of the analytical procedures used.
Stress testing provides data about the forced-decomposition products and
decomposition mechanisms of the drug substance. The severe conditions that may
be encountered during distribution can be covered by stress testing definitive
batches of the drug substance. These studies should establish the inherent
stability characteristics of the molecule such as the degradation pathways,
lead to identification of degradation products, and hence support the
suitability of the proposed analytical procedures. The detailed nature of the
studies will depend on the particular drug substance and type of drug product.
This testing is likely to be carried out on a single batch of a drug substance.
Testing should include the effects of temperatures in 100C
increments above the accelerated temperature test condition (e.g., 500C,
600C) and humidity, where appropriate (e.g., 75% or greater). In
addition, oxidation and photolysis on the drug substance plus its
susceptibility to hydrolysis across a wide range of pH values when in solution
or suspension should be evaluated. Results from these studies will form an
integral part of the information provided to regulatory authorities. Light
testing should be an integral part of stress testing. Some degradation pathways
can be complex and under forced conditions, decomposition products may be
observed that are unlikely to be formed under accelerated or long-term testing.
This information may be useful in developing and validating suitable analytical
methods, but it may no always be necessary to examine
specifically for all degradation products if, in practice, it has been
demonstrated that these are not formed.
FDA Guidance for Industry: Analytical Procedures and Methods
Validation
Drug substance/drug product:
Degradation information obtained
from stress studies (e.g., products of acid and base hydrolysis, thermal
degradation, photolysis, and oxidation) for the drug substance and for the
active ingredient in the drug product should be provided to demonstrate the
specificity of the assay and analytical procedures for impurities. The stress
studies should demonstrate that impurities and degradants
from the active ingredient and drug product excipients
do not interfere with the quantization of the active ingredient.
FDA Guidance for Industry: Submitting Documentation for the
Stability of Human Drugs and Biologics
Drug substance:
A program for the stability
assessment might include storage at ambient temperature and under stress
conditions. Stress testing conditions ordinarily include temperature (e.g., 5,
50, and 750C), humidity, where appropriate (e.g., 75% or greater)
and exposure to various wavelengths of electromagnetic radiation (e.g., 190–780
nm UV and visible ranges), preferably in open containers, where applicable. It
is also suggested that the following conditions be evaluated in stability
studies on solutions or suspensions of the bulk-drug substance: acidic and
alkaline pH, high oxygen atmosphere, and the presence of added substances under
consideration for product formulation.
Drug product:
Stress testing the drug product
helps identify potential problems during storage and transportation and
provides an estimate of the expiration-dating period.
FDA Guidance for Industry: INDs for Phase II and Phase III
Studies, Chemistry Manufacturing, and Controls Information
Drug substance:
Performance of stability-stress
studies with the drug substance early in drug development is encouraged because
these studies provide information crucial to selecting stability indicating
analytical procedures for real-time studies. If not performed earlier, stress
studies should be conducted during Phase 3 to demonstrate the inherent
stability of the drug substance, potential degradation pathways, and the
capability and suitability of proposed analytical procedures. Stress studies
should assess the stability of the drug substance in different pH solutions, in
the presence of oxygen and light, and at elevated levels of temperatures and
humidity. These one-time stress studies on a single batch are not considered
part of the formal stability program.
Drug product:
Drug products, one-time stress
testing can be warranted to assess the potential for changes in the physical
(e.g., phase separation, precipitation, aggregation, changes in particular-size
distribution) and/or chemical (e.g., degradation and/or interaction of
components) characteristics of the drug product. The studies could include
testing to assess the effect of high temperature, humidity, oxidation,
photolysis and/or thermal cycling. [6]
CONCLUSION:
Forced degradation studies, plays important role in formulation
development process. A well-designed stress study can provide insight in
choosing the appropriate formulation for a proposed product prior to intensive
formulation development studies. A thorough knowledge of degradation, including
mechanistic understanding of potential degradation pathways, stress conditions
etc. are essential for success of these studies. Stress testing can provide
useful insight into the selection of physical form, stereo-chemical stability
of a drug substance, packaging, and storage conditions. It is important to
perform stress testing for generic drugs due to allowable qualitative and
quantitative differences in formulation with respect to the RLD, selection of
manufacturing process, processing parameters, and packaging materials.
ACKNOWLEDGEMENT:
The authors express their sense of gratitude towards
management of Satara College of Pharmacy, Degaon, Satara for providing all
obligatory facilities necessary to carry out present work.
REFERENCES:
1.
Kallam.Vinusha, Srinath
Nissankararao, Ramadevi Bhimavarapu, K.Renuka, J. Naga Silpa, S. Lakshmi Sravanthi. Retrospect
of impurity profiling – an industry perspective. int.j.inv.pharm.sci, 1 (2) 2013; 89-98.
2.
J Menoutis, A Paris. Technical Guide Series Forced
Degradation Studies. Quantex Laboratory, 2012.
3.
Kishor K Hotha,
sattho phani kumar reddy, V Kishor Raju. Forced degradation
studies: practical approach: overview of regulatory guidance and literature for
the drug products and drug substances. Int. Res. J. Pharm. 4 (5): 2013.
4.
Gabriel
K Kaddu. Stability studies. National Drug Authority,
WHO, Uganda, 2009: 1-38.
5.
Stefanie Seitz. Purity testing and analytical
method requirements. Pharma solutions, 2012: 1-46.
6.
Kathy Waddle, Wai
Pan. Stability studies in pharmaceutical development. Catalant
Pharma Solutions, 2009: 1-56.
7.
Hildegard Brummer.
How to approach a forced degradation study. Technical bulletin, (31) 2011: 1-4.
8.
Michael Kats. Forced Degradation Studies:
Regulatory Considerations and Implementation. BioPharm
International,
2005: 1-08.
9.
Yande
Huang, Wei Ding. Forced degradation study of BMS582664 a prodrug
of Brivanib for the treatment of cancer. Bristol Mayers Squib, USA, 2013: 1-23.
10.
Doredla Narasimha rao, M. Prasada rao, J. Naga Hussain, S. Lakshmi Sumanoja and V. Rajeswara rao. Method development
and validation of forced degradation studies of metformin
hydrochloride by using UV spectroscopy. IJPCBS, 3(3) 2013: 546-553.
11.
Michael Kats. Forced Degradation Studies: Regulatory
Considerations and Implementation Stress testing studies are conducted to
challenge specificity of stability- indicating and impurity- monitoring methods
as part of validation protocol. BioPharm Int, (1) 2005: 1-7.
12.
FDA. Guidance for Industry. Analytical Procedures and Methods
Validation: Chemistry, Manufacturing, and Controls Documentation, Draft
Guidance, 2000.
13.
ICH Guidance for Industry, Quality of Biotechnological Products:
Stability Testing of Biotechnological/Biological Products. ICH-Q5C. 1996 July
10.
14.
Kovaleski J, Kraut B, Matiuz
A, Giangiulio M, Brosbst G,
Cango W. Impurities in generic pharmaceutical
development. Adverse Drug Rev (59) 2007: 56-63.
15.
Rao RN, Nagaraju V.
A overview of the recent trends in development of HPLC methods for
determination of impurities in drugs. J Phramaceut
Biomed, 33, 2003: 335-337.
16.
Zhou L, Mao B, Novak T, Ge Z. Impurity
profile tracking for act pharmaceutical ingredients: Case reports. J Pharmaceut Biomed, 44, 2007: 421-429.
17.
ICH Q3A (R2) (October 2006). Impurities in New Drug Substances and
Products (Step 4), International Conference on Harmonization.
18.
ICH Q3B (R2) (June 2006). Impurities in New Drug.
19.
Substances and Products (Step 4), International Conference on
Harmonization.
20. Ragine Maheswaran
Ragine. Scientific Considerations of Forced
Degradation Studies in ANDA. Pharmaceutical Technology 36 (5), 2012: 73-80.
21.
ICH Q3C (R3) (September 2002). Impurities: Guidelines for Residual
solvents (Step 5), International Conference on Harmonization.
22.
Ahuja S. Impurities Evaluation of
Pharmaceuticals. Marcel Dekker, New York, 1998.
23.
Shirazi D, Ware E, Harper T, Pflaumer
A. The Benefits of Using AAI Pharma Services for
Elemental Impurity Analysis Using ICP-MS Technology. Am Pharmaceut
Rev 2012: 1-10.
24.
Qiu F Norwood DL, Identification of
pharmaceutical impurities. J Liq Chromatogr
R T 2007; 30: 877-935.
25.
Matthews BR. Regulatory aspects of stability testing in Europe.
Drug Dev Ind Pharm 1999;
25: 831-856.
26.
Kunzle IS, Swartz ME. Validated viewpoint:
Laboratory Notebook Documuentation. Lc Gc N. Am 1997; 15: 1122-1129.
27.
Reynolds DW, Fachine KL, Mullaney JF, Alsante KM, Hatajik TD, Motto MG. Availiable
guidance and best practices for conducting forced degradation studies. Pharm Tech, 26, 2002: 48-56.
28.
ICH Q1A (R2) (2003). Stability Testing of New Drug Substances and
Products. International Conference on
Harmonization , IFPMA, Geneva.
29.
Kovarikova P, Jiri K, Jiri D, Lucie T. HPLC study of glimepiride
under hydrolytic stress conditions. J. Pharmaceut
Biomed 36, 2004: 205-209.
30.
Singh S, Bakshi M. Guidance on conduct
of stress tests to determine inherent stability of drugs. Phrama
Tech 24, 2000: 1-14.
31.
Skoog DA, West DM, Holler FJ. Analytical
Chemistry, An; Introduction, Saunders College Publishing, Philadelphia. 2002:
3-11
32.
Cheekatla S, Ravichandrababu
R, Reddy BN. Determination and characterization of degradation products of Anastrozole by LC–MS/MS and NMR spectroscopy. J. Pharmaceut Biomed, 56, 2011: 962-968.
33.
Gupta A, Yadav JS, Rawat
S, Gandhi M. Method Development and Hydrolytic Degradation Study of Doxofylline by RPHPLC and LC-MS/MS. Asian J Pharm Ana , 11, 2011:14-18.
34.
Skoog, DA, West DM. Principles of Instrumental
Analysis. Saunderg Golden, Japan. 1980: 2-3.
35.
Boccardi G. Oxidative susceptibility testing. In;
pharmaceutical Stress Testing-Predicting Drug Degradation; Baertschi
SW, editors, Taylor and Francis, New York. 2005: 220.
36.
Alsante KM, Hatajik TD, Lohr LL, Santafianos D Sharp TR.
Solving impurity/degradation problems: case studies, In; handbook of Isolation
and Characterization of impurities in Pharmaceutical, Ahuja
S, Alsante K, editors, Academics Press, New York.
2003: 380.
37.
Alan RO, Brigitte ES, Yanqiu SA, Polshyna NM, Dunphy R, Barbara
LM, et al. Forced degradation studies of rapamycin:
Identification of autoxidation products. J Pharmaceut Biomed 2012; 59: 194-200
38.
Bojana P, Markus D, Kappe
CO. Microwave-assisted forced degradation using high-throughput microtiter platforms. J Pharmaceut
Biomed 56, 2011: 867-873
39.
Susanne CM, Lisa AM, Amina SW, Victor
VL, Vladimir MD, Robert JC. Atmospheric pressure matrix-assisted laser
desorption/ionization (AP MALDI) on a quadrupole ion
trap mass spectrometer. J Mass Spectrom, 226, 2003:
133-150.
40.
Takats Z, Wiseman JM, Gologan
B, Cooks RG. Mass spectroscopy sampling under ambient conditions with
desorption electrospray ionization. Science, 306,
2004: 471- 473.
Received on 30.10.2013 Accepted on 25.11.2013
© Asian Pharma
Press All Right Reserved
Asian J. Res.
Pharm. Sci. 2013; Vol. 3: Issue 4, Pg 178-188