P. R. Pote Patil College of Pharmacy, Amravati, Maharashtra 444604
Stability refers to the period during which a product maintains its integrity and quality under recommended conditions, without undergoing physical or chemical changes. It is a critical consideration in product development across industries such as pharmaceuticals, food, and cosmetics, where product safety and efficacy directly affect consumer health. Stability studies are an essential component of quality assurance, designed to evaluate how environmental factors like temperature, humidity, and light impact a product’s characteristics over time. Typically, three initial batches are tested in the first year of manufacture to establish baseline data, followed by ongoing testing of one batch per strength annually to monitor quality during the product’s market life. The primary objectives of stability studies are to determine shelf life and optimal storage conditions, ensuring the product maintains its safety, efficacy, and quality throughout its lifespan. Additionally, stability testing supports regulatory compliance and guides packaging decisions, making it a cornerstone of product quality management across diverse sectors. This article reviews on the complete evaluation of Stability study of the drug substances and drug products.
The term ‘Stability’ can be defined as the period of time in which a product remains stable under the recommended conditions (as different products require different conditions), without compromising its integrity. This means that no physical or chemical changes occur, and the product retains the same quality as when it left the manufacturer. When a company is manufacturing a new product, it needs to perform a stability study.
Stability is a crucial aspect of product development, particularly in industries such as pharmaceuticals, food and beverages, and cosmetics, where product efficacy and safety directly impact consumer health and satisfaction.
The term stability refers to the period during which a product maintains its intended physical, chemical, microbiological, therapeutic, and toxicological specifications when stored under recommended conditions [1]. In other words, a product is considered stable when no significant changes occur in its composition, performance, or appearance throughout its anticipated shelf life.
Stability studies are an integral part of the quality assurance process, designed to evaluate how environmental factors such as temperature, humidity, and light exposure affect a product’s integrity over time [2]. When a new product is manufactured, the initial phase involves testing three primary production batches during the first year of development to establish baseline data. In subsequent years, one batch per strength per year is placed on stability testing to monitor the product’s ongoing quality while on the market [3].
The primary goals of stability studies are to determine the product’s shelf life, identify optimal storage conditions, and confirm that the product remains within specified parameters throughout its lifecycle. This process ensures that consumers receive products that meet established standards of safety, quality, and efficacy up to the point of use [4].
Moreover, stability testing helps manufacturers establish regulatory compliance regarding labeling and quality control documentation. It also aids in decision-making for packaging design, as packaging materials can influence product stability by providing protection against environmental stressors. Stability studies, therefore, serve as a cornerstone of product quality management across diverse sectors.
Regulatory Framework:
Objectives of stability study
The main objective of stability testing is to ensure the quality, safety, and efficacy of a drug substance or drug product throughout its shelf life under the influence of various environmental factors such as temperature, humidity, and light. Stability testing is fundamental to understanding how a drug's characteristics evolve over time and to ensure that it maintains its intended performance until its expiry date.
1. Ensure Quality, Safety, and Efficacy
Stability testing guarantees that the drug product retains its purity, potency, safety, and overall quality during storage and use[10]. It evaluates the physical, chemical, microbiological, and biological properties of the drug to ascertain that none of these degrade beyond acceptable limits over time.
2. Determination of Shelf Life and Storage Conditions
One of the primary outcomes of stability testing is to establish a scientifically justified shelf life and to define optimal storage conditions. By studying how environmental factors such as temperature, humidity, and light affect the drug, manufacturers can recommend storage requirements like "Store below 25°C" or "Protect from light" to help maintain drug stability[10].
3. Evaluation of the Stability Profile
Stability testing involves rigorous testing of various parameters such as chemical composition (e.g., potency, degradation products), physical characteristics (e.g., appearance, dissolution rate), microbiological integrity, and biological activity. This comprehensive evaluation helps build a stability profile over time and under different conditions[10].
4. Identification of Degradation Products and Pathways
Testing allows identification of degradation products that may form during storage and helps elucidate degradation pathways. This information is critical to understanding potential risks associated with drug use after prolonged storage and to ensuring safety[10].
5. Regulatory Compliance and Documentation
Stability testing provides the essential data package for regulatory submissions such as New Drug Applications (NDA) or Abbreviated New Drug Applications (ANDA). Regulators require these data to approve marketing authorization and to verify that the product meets quality standards throughout its labeled shelf life[10].
6. Support for Labeling and Marketing Claims
Data from stability studies back up claims made on product labels, including storage instructions, expiration dates, and specific handling requirements[10]. These claims guide healthcare professionals and consumers in ensuring the drug's effectiveness and safety.
Scope of Stability Study
Stability testing in the pharmaceutical industry encompasses a broad scope, essential for ensuring the consistent quality, safety, and efficacy of drug substances and drug products throughout their shelf life. The scope of stability testing includes multiple dimensions, starting with the types of products tested, environmental conditions, packaging interactions, stability-indicating parameters, special cases, and regulatory compliance.
Stability testing applies to new drug substances (active pharmaceutical ingredients, APIs) and formulated products such as tablets, capsules, and injections. Both innovator and generic products undergo testing, including biotechnological and biological products like vaccines, peptides, and proteins[5].
Testing involves various conditions to simulate real-world storage and stress situations:
Studies assess the interaction between drug products and packaging materials (e.g., glass, plastic, blister packs), vital for maintaining product integrity and protection throughout storage and distribution [5].
Tests cover physical properties such as appearance and dissolution, chemical properties including potency and degradation products, microbiological attributes like sterility, and functional properties such as drug release behavior [2],[8].
Stability testing can include reconstituted products (post-mixing stability), in-use stability for multidose containers, photostability testing to assess light exposure effects, and freeze-thaw studies especially relevant for biological products [5].
Stability data are a regulatory requirement for Investigational New Drug Applications (INDs), New Drug Applications (NDAs), Abbreviated New Drug Applications (ANDAs), and Common Technical Documents (CTDs). Compliance with ICH guidelines (Q1A-Q1F) and regional authorities such as the US FDA, EMA, and CDSCO is mandatory to support shelf-life determination and labelling [2],[6],[7].
These comprehensive stability studies provide a scientifically based assurance that drug products remain safe and effective during their marketed life, helping manufacturers optimize storage conditions, packaging, and usage recommendations.
Mechanistic Principle of Stability Testing
The mechanistic principle of stability testing of a new drug product centre on understanding the changes in chemical, physical, microbiological, and therapeutic properties over time when subjected to environmental factors such as temperature, humidity, light, and oxygen. The principle involves exposing the drug product to controlled conditions to observe degradation mechanisms, identify degradation pathways such as hydrolysis, oxidation, and photolysis, and determine the kinetics of these degradation reactions (e.g., first-order or zero-order kinetics). This understanding helps elucidate the impact of the drug formulation and packaging on its stability. Ultimately, these studies enable the estimation of shelf life and the assignment of an expiry date, ensuring that the product maintains its intended quality and performance throughout its lifecycle [9],[10].
Stability testing employs stress and accelerated conditions to simulate and predict long-term changes in the drug substance or product. Analytical methods used are stability-indicating, capable of differentiating the drug substance from its degradation products. Kinetic modeling, often based on Arrhenius principles, allows extrapolation from accelerated data to predict behavior under normal storage conditions. This mechanistic approach is essential in establishing robust stability profiles critical for regulatory approval and label claims[9],[10].
Table 1.1.- Types of Mechanisms Considered [2]
|
Mechanism |
Description |
|
Chemical degradation
|
Breakdown of active pharmaceutical ingredient (API) through hydrolysis, oxidation, photolysis, etc. |
|
Physical degradation |
Changes in appearance, dissolution rate, hardness, moisture content, etc. Microbiological contamination Growth of microorganisms if product is not properly preserved. |
|
Toxicological changes |
Formation of harmful degradation products over time |
Order of reaction [12]
Order of a reaction is defined as the number of concentration terms on which the rate of a reaction depends when determined experimentally.
The order of reaction is established with respect to each reactant. This can be verified by plotting log concentration of a reactant on y axis and time on x axis. If a straight line is obtained, then the order will be 'one' with respect to that reactant. The overall order of a reaction is equal to the powers of the concentration terms (m + n) affecting the experimentally determined rate. In contrast to molecularity, it is possible for the order to assume fractional or zero values.
The orders that are common in pharmacy are zero, first-, pseudo first-, and second- orders. These topics will be discussed in detail in the subsequent sections.
Zero Order Reaction :
Zero order reaction is defined as a reaction in which the rate does not depend on the concentration terms of the reactants.
Example:
Mechanism : In zero order reaction, the rate must depend on some factor other than the concentration term. The rate limiting factors are solubility as in suspensions or absorption of light as in certain photochemical reactions.
First Order Reaction
First order reaction is defined as a reaction in which rate of reaction depends on the concentration of one reactant.
where c is the concentration of the reactant and kl is the specific rate constant for first order.
Examples : Decomposition of hydrogen peroxide catalyzed by 0.02 M potassium iodide.
2H2O2→2H2O+O2
The stoichiometric equation involves 2 molecules, i.e., molecularity is two. But the order is found to be first order, based on experimental observation. In the presence of stabilisers, this reaction follows zero order kinetics. In other words, same drug may exhibit different Orders of decomposition under different experimental conditions.
Examples of biological importance are as follows:
Pseudo First Order Reaction
Pseudo first order reaction is defined as a reaction which is originally a second order, but is made to behave like a first order reaction.
In second order reaction, the rate depends on the concentration terms of two reactants.
Examples:
Therefore, the rate solely depends on the concentration of ester.Second Order Reaction
Second order reaction is defined as a reaction in which the rate depends on the concentration terms of two reactants each raised to the power one.
In a second-order reaction, the rate of reaction depends on the concentration of two reactant molecules or on the square of one reactant’s concentration. The rate decreases more rapidly compared to first-order reactions, and the half-life is inversely proportional to the initial concentration. This means that as the initial concentration increases, the half-life decreases. Second-order kinetics are typically observed in bimolecular reactions and certain oxidation or substitution processes.
Examples:
Force degradation study
Forced degradation, also known as stress testing, refers to the process of deliberately subjecting a new drug substance or drug product to conditions more severe than those used in accelerated stability studies. This is done to induce degradation rapidly and extensively, which serves several critical purposes in pharmaceutical development. Forced degradation is essential for demonstrating the specificity and robustness of stability-indicating analytical methods by ensuring these methods can distinguish the drug substance from its degradation products. Moreover, it provides valuable insight into the degradation pathways and chemical behavior of the molecule, helping to elucidate the structure of degradation products and understand their formation mechanisms. This information is vital for formulation development and packaging design, helping to optimize product stability and ensure that protective measures in packaging adequately safeguard the drug during its shelf life[13],[14].
Forced degradation (also called stress testing) is an essential part of stability testing that helps:
Unlike formal stability studies conducted under prescribed regulatory conditions (such as ICH guidelines), forced degradation studies are conducted over a shorter time frame and under harsher environmental stresses, including acid/base hydrolysis, oxidation, photolysis, and thermal degradation. These conditions are carefully selected to generate about 5-20% degradation, which is sufficient to identify critical degradation products without overwhelming the analytical systems. Early implementation of forced degradation studies during drug development facilitates timely formulation improvements, appropriate storage condition recommendations, and the development of validated, stability-indicating analytical methods that are essential for regulatory submissions [13],[15].
Objective of forced degradation studies
Forced degradation studies, also known as stress testing, are purposely designed experiments where drug substances and drug products are subjected to conditions harsher than those of accelerated stability studies. The main objective of these studies is to understand the intrinsic stability and degradation behavior of pharmaceutical compounds, thereby supporting drug development, formulation, packaging, regulatory compliance, and quality control.
Forced degradation studies elucidate the specific chemical routes through which drug substances and products break down under stress. Understanding these pathways offers insight into potential degradation products and how environmental or manufacturing factors influence drug stability.
Because formulations often include excipients and other non-active components, forced degradation helps distinguish impurities that originate from the drug substance versus those arising from the formulation excipients or packaging materials, informing formulation optimizations.
Characterizing the chemical structure of degradation products is crucial to evaluate their safety and impact on drug efficacy. Forced degradation facilitates isolation and structural identification of these products by providing stressed samples for analytical methods development.
These studies reveal inherent weaknesses in the chemical stability of drug substances, allowing scientists to predict how molecules will behave in different environments and aiding in mitigating risks early in development
Forced degradation exposes mechanisms such as hydrolysis, oxidation, photolysis, thermal decomposition, and others that might be responsible for drug degradation. Understanding these enables better control through formulation or packaging measures
Validation of analytical methods that precisely measure drug and degradation products is key in regulatory submissions. Forced degradation samples ensure these methods selectively detect the drug substance without interference from degradants
Information obtained assists in understanding the physicochemical properties, reactivity, and stability profile of the drug molecules under various conditions, supporting drug design and process development
By identifying weak points in stability, forced degradation outcomes guide formulation scientists in selecting excipients, adjusting pH, or improving packaging to enhance the drug's shelf life
Forced degradation efforts strive to mimic the degradation kinetics and product profiles observed during formal ICH-guided stability studies, thus providing predictive insights earlier in development
When stability issues arise—such as unexpected impurities or loss of potency—forced degradation helps pinpoint root causes and develop suitable corrective actions[2],[16].
Fig.1.2- Forced dehydration study
Types of stability types
Stability studies of drug products (formulations) are essential to evaluate how the formulation components, excipients, and packaging affect the product’s safety, efficacy, and quality over time under various storage conditions. These studies are conducted in three main phases:
This study provides real-time data for establishing the product's shelf life or retest period under actual storage conditions recommended on the product label. Storage conditions are based on climatic zones—for example, 25 °C ± 2 °C/60% RH ± 5% RH for temperate zones (Zone I & II), and up to 30 °C ± 2 °C/75% RH ± 5% RH for hot and very humid zones (Zone IVb, including India and Southeast Asia). Data collection typically begins at 0, 3, 6, 9, and 12 months, continuing annually until the end of the proposed shelf life, which can span 24 to 60 months. A minimum of 12 months of long-term data is required at the time of regulatory submission [2],[5].
This phase aims to hasten degradation under more stressful conditions (40 °C ± 2 °C/75% RH ± 5% RH) to quickly identify potential stability issues. The duration is usually 6 months. If no significant change—defined as less than or equal to 5% degradation or no failure in physical or chemical tests—is observed, the accelerated data can be used to support shelf-life extrapolation [2],[6].
An intermediate condition study (30 °C ± 2 °C/65% RH ± 5% RH for 12 months) acts as a bridge when the accelerated conditions cause significant instability, helping to ensure stability under conditions between long-term and accelerated testing. This is particularly used to understand whether the product can withstand slightly harsher conditions than those of the long-term storage [2].
These studies collectively support the determination of appropriate storage conditions, shelf-life labelling, and packaging decisions to ensure product quality and patient safety throughout the product lifecycle.
Table 1.2- Stability Study types [12]
|
Conditions |
Temperature |
Relative Humidity |
Duration |
Purpose |
|
Long term |
25°C ± 2°C |
60% RH ± 5% |
12-36 months |
Real time stability |
|
Accelerated |
40°C ± 2°C |
75% RH ± 5% |
6 months |
Predict degradation rate |
|
Intermediate |
30°C ± 2°C |
65% RH ± 5% |
12 months |
Optional for product failing accelerated test |
Table 1.3- Key difference between Drug Substances vs Drug product
|
Aspect |
Drug substance (API) |
Drug product (Formulation) |
|
Focus |
Intrinsic stability of pure API |
Stability of final dosage form (API + excipients + packaging) |
|
Stress testing |
Identifies degradation pathways |
Evaluates effects of excipients, processes and packaging |
|
Additional studies |
Limited Photostability, stress |
In use stability, packaging compatibility |
|
Regulatory requirements |
Needed for API DMF submission |
Needed for product registration dossier (NDA/ANDA) |
Stability-Indicating Analytical Methods (SIAM).
A stability-indicating assay method (SIAM) is a validated analytical procedure that accurately measures the active pharmaceutical ingredient (API) in a drug substance or drug product without interference from degradation products, impurities, or excipients. The primary purpose of SIAM is to detect chemical degradation over time, monitor the shelf life of drug substances and products, and ensure their safety, efficacy, and quality throughout storage. APIs can degrade due to several environmental factors such as heat, humidity, light, oxidation, or pH changes. Regular assay methods may not distinguish the intact API from its degradants, making SIAM indispensable for stability studies. The International Council for Harmonisation (ICH) guidelines Q1A and Q2(R1) require stability-indicating methods for regulatory approval as they provide critical evidence of a product’s ongoing quality and integrity during its shelf life. SIAM typically involves separation techniques such as high-performance liquid chromatography (HPLC), gas chromatography, or spectroscopic methods coupled with detectors to achieve specificity and sensitivity in detecting degradation products [18],[19],[2].
These methods are vital throughout pharmaceutical development and quality control to ensure that the drug product maintains its intended potency and remains free from interfering impurities caused by degradation. Additionally, SIAM assists in investigating out-of-trend or out-of-specification results, providing a powerful tool for quality assurance processes [18].
Studies include:
Checks if container–closure system affects product stability (e.g., leachables, adsorption, permeability).
Characteristics of a Good SIAM
A strong stability-indicating analytical method (SIAM) should be built around a core set of validated performance characteristics that ensure reliability and regulatory acceptability [20].
Steps in Developing a SIAM
Step 1: Forced Degradation / Stress Testing
Forced degradation, also known as stress testing, is a critical part of stability-indicating method (SIAM) development. Its primary objective is to generate potential degradation products of the active pharmaceutical ingredient (API) to assess the specificity of an analytical method. According to Stability-Indicating Methods for Drug Analysis guideline, stress conditions should be selected based on the chemical nature of the drug substance[20],[2].Typical stress parameters include acid and base hydrolysis (using 0.1–1 N HCl or NaOH to simulate hydrolytic degradation), oxidation (commonly induced by 1–3% hydrogen peroxide), thermal degradation (temperatures ranging from 40°C to 80°C depending on API stability), photolysis (exposure to UV and visible light as per ICH Q1B), and humidity stress (usually 75% RH at 40°C). The level of degradation should ideally range between 5% and 20% to sufficiently challenge the method without completely destroying the analyte.
Step 2: Selection of Analytical Technique
Choosing the appropriate analytical technique is essential for accurate identification and quantification of degradation products[21].Ultra-Performance Liquid Chromatography (UPLC) offers faster analysis with better resolution due to smaller particle sizes in the stationary phase. Coupling chromatographic techniques with mass spectrometry (LC-MS or LC-MS/MS) facilitates identification of unknown degradant structures by providing molecular weight and fragmentation data. UV-Visible spectroscopy can be used when degradation products do not interfere spectrally with the API. For rapid, preliminary assessments, Thin-Layer Chromatography (TLC) or High-Performance TLC (HPTLC) serves as a simple and cost-effective screening method.
Step 3: Method Development
Method development involves the systematic optimization of chromatographic parameters to achieve a stability-indicating profile. Key parameters include the selection of an appropriate column (e.g., C18 reverse-phase), mobile phase composition (such as buffer–organic solvent ratio), pH optimization (to stabilize ionizable drugs), flow rate, and detection wavelength based on the UV-Vis absorption spectrum of the compound[22]. The goal is complete resolution of the API peak from all degradant peaks, verified by peak purity analysis using a Diode Array Detector (DAD) or Mass Spectrometer. The method must ensure no co-elution occurs between analyte and degradation products, establishing true specificity of the method.
Step 4: Method Validation
Validation of the stability-indicating method must follow the International Council for Harmonisation guideline, which defines parameters such as specificity, linearity, accuracy, precision, detection/quantitation limits, robustness, and system suitability[23].Specificity demonstrates that the API is clearly separated from degradation products and excipients. Linearity assesses proportionality of response across a defined range (typically 50–150% of nominal concentration). Accuracy is evaluated by recovery studies, which should fall within 98–102%. Precision is verified through repeatability, ensuring relative standard deviation (%RSD) below 2%. Limits of Detection (LOD) and Quantitation (LOQ) determine the minimum detectable and quantifiable concentrations of degradants. Robustness confirms method reliability against small deliberate variations (e.g., temperature, pH, or mobile phase ratio). System suitability tests, such as evaluating resolution, tailing factor (<2), and number of theoretical plates, ensure performance consistency.
Step 5: Application in Stability Studies
A validated stability-indicating method is then employed in formal stability studies to monitor the drug’s potency and degradation profile over time under prescribed storage conditions. These include long-term (25°C ± 2°C/60% RH ± 5%), intermediate (30°C ± 2°C/65% RH ± 5%), and accelerated (40°C ± 2°C/75% RH ± 5%) conditions[1]. The method quantifies the API content and any degradation products at various time intervals, supporting determination of shelf life (t90 — time at which the API retains 90% of its initial concentration). Data generated from these studies provide a scientific basis for establishing appropriate storage recommendations, expiration dating, and regulatory filing of stability data in accordance with ICH Q1A(R2) guidelines and regulatory requirements across major health authorities[2].
Stability Study Specifications
Stability studies evaluate how a drug substance or product maintains its quality over time under specific environmental conditions (temperature, humidity, light). Specifications focus on critical quality attributes (CQAs) that ensure safety, efficacy, and quality.
Table 1.3- Drug substances specification[25],[26]
|
Specification |
Purpose/ Importance |
Typical method |
|
Appearance |
Check colour, form, crystal structure, or powder texture. Early indicator of degradation. |
Visual inspection, microscopy |
|
Identification |
Confirms correct chemical identity of API; detects chemical transformations. |
IR, UV, NMR, HPLC, TLC |
|
Assay / Potency (% API) |
Measures active content; ensures API retains efficacy. |
HPLC, UV, titration |
|
Impurities / Degradants |
Detects chemical degradation products; ensures safety and compliance with ICH limits. |
HPLC, GC, LC-MS, TLC |
|
Moisture Content |
Prevents hydrolysis, polymorphic changes, or microbial growth |
Karl Fischer titration |
|
pH / Solubility |
Ensures drug stability and suitability for formulation. |
pH meter, solubility tests |
|
Melting Point / Crystallinity |
Detects polymorphic changes affecting solubility and bioavailability. |
DSC, XRD |
Table 1.4- Dosage form specification [27],[28]
|
Specification |
Purpose/ Importance |
Typical method |
|
Physical appearance |
Checks tablet/capsule colour, shape, size, coating; indicates visual stability and uniformity |
Visual inspection, microscopy |
|
Assay / Content Uniformity |
Ensures each dosage unit contains the correct dose of API to guarantee safety and efficacy |
HPLC, UV spectroscopy, USP methods |
|
Dissolution Profile |
Predicts bioavailability and ensures consistent drug release from the dosage form |
USP dissolution apparatus |
|
Hardness / Friability |
Measures mechanical strength for durability during handling and transport |
Hardness tester, friabilator |
|
Moisture Content / Water Activity |
Controls potential degradation, microbial growth, and impacts on tablet hardness |
Karl Fischer titration, water activity meter |
|
Microbial Limits |
Ensures microbiological safety, critical in liquids, suspensions, and some solid forms |
Plate count, sterility testing |
|
Degradation Products / Impurities |
Detects and quantifies chemical changes in API or excipients over time ensuring product quality |
HPLC, LC-MS, TLC |
Stability study protocol
A stability study protocol is a documented plan that details the conditions, tests, intervals, and methods used to evaluate the stability of a drug substance or drug product over time.
Purpose of a Stability Study Protocol
1. Title Page
2. Objective [2]
3. Scope [29]
4. Reference Standards and Guidelines[30]
5. Test Items
6. Storage condition
Table 1.5- Different stability conditions [12]
|
Conditions |
Temperature |
Relative Humidity |
Duration |
Purpose |
|
Long term |
25°C ± 2°C |
60% RH ± 5% |
12-36 months |
Real time stability |
|
Accelerated |
40°C ± 2°C |
75% RH ± 5% |
6 months |
Predict degradation rate |
|
Intermediate |
30°C ± 2°C |
65% RH ± 5% |
12 months |
Optional for product failing accelerated test |
7. Analytical Methods [2]
8. Acceptance Criteria [2]
9. Data Handling and Statistical Analysis
10. Reporting
11. Appendices Copies of analytical method validation reports
Objective:
Determine the stability of Amlodipine 5 mg tablets under ICH-recommended long-term and accelerated conditions to establish shelf life and storage recommendations.
Test parameters and methods
Table 1.6- Test parameter of Amlodipine Besylate [31]
|
Parameter |
Method |
Acceptance Criteria |
|
Appearance |
Visual inspection |
No change in color, texture, or integrity |
|
Assay |
Stability-indicating HPLC |
90–110% of label claim |
|
Dissolution |
USP Apparatus II, 900 mL pH 6.8, 50 rpm |
≤80% release at 30 minutes |
|
Hardness / Friability |
Hardness tester / Friabilator |
Friability ≤1% |
|
Moisture |
Karl Fischer titration |
≤2% |
|
Impurities |
HPLC |
≤1% for any impurity |
Storage Conditions and Sampling Points:
Selection of sample
The protocols for the selection of samples are as following
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[32].
Batches
Stability studies at developmental stages are generally carried out on a single batch while studies intended for registration of new product or unstable established product are done on first three production batches, while for stable and well-established batches, even two are allowed. If the initial data is not on a full-scale production batch, first three batches of drug product manufactured post-approval should be placed on long-term studies using the same protocol as in approved drug application. Data on laboratory scale batches obtained during development of pharmaceuticals are not accepted as primary stability data but constitute supportive information. In general, the selection of batches should constitute a random sample from the population of pilot or production batches[32].
Containers and closures
The testing is done on the product in immediate containers and closures proposed for marketing. The packaging materials include aluminum strip packs, blister packs, Alu-Alu packs, HDPE bottles etc. This may also include secondary packs, but not shippers. Products in all different types of containers/closures, whether meant for distribution or for physician and promotional samples, are to be tested separately. However, for bulk containers, testing in prototype containers is allowed, if it simulates the actual packaging[32].
Orientation of storage of containers
Samples of the solutions, dispersed systems and semi solid drug products for stability studies must be kept upright and positioned either inverted or on the side to allow for full interaction of the product with the container-closure. This orientation helps to determine whether the contact between the drug product or solvent and the closure results in the extraction of chemical substances from the closure components or adsorption of product components in to the container-closure[32].
Sampling time points
Frequency of testing should be such that it is sufficient to establish the stability profile of the new drug substance. For products with a proposed shelf life of at least 12 months, the testing frequency at the long-term storage condition should be every 3 months over the first year, every 6 months over the second year and annually thereafter throughout the proposed shelf-life expiration date. In the case of accelerated storage conditions, a minimum of three time points, including the initial and end points, for example, 0, 3, and 6 months is recommended. When testing at the intermediate storage condition is necessary as a result of significant change at the accelerated storage condition, a minimum of four test points, including the initial and final time points, is recommended, for example, 0, 6, 9 and 12 months[32].
Table 1.7- Test Schedule for stability testing of new products[32].
|
Environment |
Sampling Time Points (months) |
Method & Climatic zone |
|
25°C/60% RH |
3, 6, 9, 12, 18, 24, 36 |
Long term for zones I and IV |
|
30°C/35% RH |
3, 6, 9, 12, 18, 24, 36 |
Long term for zones III |
|
30°C/65% RH |
3, 6, 9, 12, 18, 24, 36 |
Long term for zone Iva, or intermediate condition for zones I and II |
|
30°C/75% RH |
3, 6, 9, 12, 18, 24, 36 |
Long term for zone Iva, or intermediate condition for zones I and II |
|
40°C/75% RH |
3,6 |
Accelerated condition for all zones |
In case the same product of different strengths, multiple sizes, etc. is required to be tested, reduced stability testing plans can be worked out, which involves a smaller number of test points. The reduced testing plans are based on bracketing and matrixing statistical designs. Bracketing is the design of a stability schedule such that only samples on the extremes of certain design factors, e.g., strength, package size, are tested at all time points as in a full design. On the other hand, matrixing involves testing of a subset of the total number of possible samples for all combinations at a specific time point. Subsequently, another subset of samples for all factor combinations is tested. The factors that can be matrixed include batches, strengths with identical formulation, container sizes, fill sizes, and intermediate time points.
Sampling Plan
Sampling plan for stability testing involves, planning for the number of samples to be charged to the stability chambers and sampling out of the charged batch so as to cover the entire study. The first step should be the development of the sampling time points followed by the number of samples needed to be drawn at each pull point for complete evaluation of all test parameters and finally adding up to get the total number of samples. For example, there would be a requirement of about 100 tablets per pull out in a long term or accelerated stability studies including 10 each for assay, hardness and moisture determination, 6 each for dissolution and disintegration and 50 for friability. This multiplied by the total number of pull outs will give the total number of tablets required for a study. This is followed by the development of a sampling plan, which includes the selection of the containers representing the batch as a whole but in an unbiased manner. A stratification plan has been suggested whereby from a random starting point every nth container is taken from the filling or packaging line (n is chosen such that the sample is spread over the whole batch)[32].
Test storage conditions
The storage conditions to be selected are based upon the climatic zone in which the product is intended to be marketed or for which the product is proposed to be filed for regulatory approval. General recommendations on the storage conditions have been given by ICH, CPMP and WHO. The abridged/indicative ICH and WHO storage conditions for drug products have been given in Table 1.8 [32].
Table 1.8- Stability test storage conditions for drug Products[32].
|
Intended storage condition |
Stability Test Method |
ICH Test temperature and humidity (Period in months) |
WHO Test temperature and humidity (Period in months) |
|
Room temperature |
Long term
Intermediate Accelerated |
25±2°C/60±5% RH (12)
30±2°C/65±5% RH (6) 40±2°C/75±5% RH (6) |
25±2°C/60±5% RH or 30±2°C/65±5% RH 30±2°C/75±5% RH (12) 30±2°C/65±5% RH (6) 40±2°C/75±5% RH (6) |
|
Refrigerated |
Long term Accelerated |
5°C/ambient (12) 25±2°C/60±5% RH (6) |
5±3°C 25±2°C/60±5% RH or 30±2°C/65±5% RH |
|
Freezer |
Long term |
-20°C/ambient (12) |
-20°C±5°C |
Test paramètres
The stability test protocol should define the test parameters that would be used for evaluation of the stability samples. The tests that monitor the quality, purity, potency, and identity which could be expected to change upon storage are chosen as stability tests. Therefore appearance, assay, degradation products, microbiological testing, dissolution, and moisture are standard tests performed on stability test samples. Microbiological tests include sterility, preservative efficacy and microbial count as applicable e.g. for liquid injectable preparations. The batches used for stability study must meet all the testing requirements including heavy metals, residue on ignition, residual solvents etc. Some of these are required at the time of product release but not required to be repeated during stability testing[32].
Test methodology
It is always recommended to follow the procedures given in the official compendia, as the results obtained using the official tests, in general find better acceptance. If alternate methods are used, they are required to be duly validated. However, the assay of the drug should be carried out using a stability-indicating method, established by carrying out stress tests on the drug under forced decomposition conditions. This method should be validated for specificity, accuracy, precision and linearity, in the range to which the drug is expected to fall during stability studies. For the assay of degradation products, the validated method should also include the limits of detection/quantification. The methods reported in literature should be used after confirming reproducibility and carrying out minimal validation, say of linearity, range, etc. It is always recommended to prepare a standard test protocol (STP) for each test[32].
Acceptance criteria
All analytical methods are required to be validated before initiating the stability studies. Similarly, the acceptance criteria for the analytical results as well as that for the presence of degradation products should also be fixed beforehand. The acceptance criteria for each test in the stability study are fixed in the form of numerical limits for the results expressed in quantitative terms e.g., moisture pick-up, viscosity, particle size, assay, degradation products, etc. and pass or fail for qualitative tests e.g., odor, color, appearance, cracking, microbial growth, etc. These acceptance criteria should also include individual and total upper limits for degradation products. ICH guideline Q3B(R2) related to impurities in new drug products addresses degradation products in new drug formulations. The degradation products of the active or interaction products from the active ingredients and excipients and/or active and container component should be reported, identified, and/or qualified when the proposed thresholds are exceeded. The reporting threshold of impurities is based upon the intended dose. If the maximum daily dose is less than or equal to 1gm, the limit is 0.1% and if greater than 1, the limit is 0.05%. The identification threshold of impurities is between 1.0-0.1% for the maximum daily dose ranging between 1mg and 2gm [32].
Table 1.9- Selection of samples [2]
|
Parameter |
Description |
|
Sample Type |
Finished drug product or drug substance in final packaging intended for marketing |
|
Number of Batches |
Minimum 3 batches (2 pilot-scale + 1 production-scale OR 3 production-scale) |
|
Batch Size |
Pilot-scale (≥10% of production batch or ≥100,000 units for solid orals) |
|
Manufacturing |
Manufactured using GMP, same process, equipment, and formulation as marketed |
|
Packaging System |
Same container-closure system intended for commercial distribution |
|
Storage Conditions |
- Long-term: 25°C ± 2°C / 60% RH ± 5% RH - Accelerated: 40°C ± 2°C / 75% RH ± 5% RH - Intermediate (if needed): 30°C ± 2°C / 65% RH ± 5% RH |
|
Time Points |
- Long-term: 0, 3, 6, 9, 12, 18, 24, 36 months - Accelerated: 0, 3, 6 months |
|
Sample Quantity |
Sufficient for: - All time-point tests - Retesting - Investigation if needed |
|
Labeling |
Must include Batch No., Mfg. Date, Expiry Date, storage condition, etc. |
|
Retain Samples |
Stored for 1 year after expiry under recommended conditions for future reference/testing |
Storage condition
The storage conditions to be applied to different stability studies must be strictly controlled within the ranges defined by regulatory guidelines to ensure reliable stability data. The equipment used for stability storage should have the capability to maintain temperature and humidity accurately according to these specifications. During the study, actual temperature and humidity should be continuously monitored, especially when humidity control is required [2],[4].
Short-term deviations or spikes, such as those caused by opening the doors of the storage facility, are generally considered unavoidable and acceptable. However, any excursions due to equipment failure or other reasons should be thoroughly documented and addressed in the study report if they are judged to impact the stability results.
Specifically, excursions that exceed the defined tolerances for more than 24 hours must be described in detail in the stability study report, and an assessment of their effect on product quality should be conducted. This ensures transparency and allows regulators to evaluate the risk of instability under real-world storage conditions.
These requirements are consistent with ICH Q1A(R2) guidelines which state that:
This comprehensive approach helps ensure that the integrity and quality of pharmaceutical products are maintained throughout their marketed shelf-life periods, safeguarding patient safety.
Zones of stability
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. The break-up of the environmental conditions in each zone and also the derived long-term stability test storage conditions, as given by WHO have also been presented. The stability conditions have also been harmonized and adjusted to make them more practical for industry application and rugged for generalized application [32].
The International Council for Harmonisation (ICH) has established five climatic zones recognized globally for stability testing:[2]
These zones are critical to simulate the specific climatic conditions drugs may be exposed to in different regions so that stability testing ensures the drug’s efficacy and safety under those environmental stresses. Testing products under the appropriate climatic zone conditions helps establish reliable shelf life and storage recommendations globally.
Table 1.10- Climatic zone storage conditions [2]
Case study 1
Testing frequency determination in stability studies is a critical aspect of drug development and registration, as detailed by the International Council for Harmonization (ICH) Q1A and Q1D guidelines [33]. The frequency of testing depends on the type of study (long-term vs. accelerated) and the use of reduced designs like bracketing and matrixing, which optimize resources based on sound scientific justification.
Full design testing frequency
The standard testing frequency provides the baseline for stability testing and is applied to all samples in a full study design.
Determining testing frequency with bracketing
Bracketing simplifies the stability protocol by reducing the number of different samples that must be tested at each time point, not the time points themselves. The testing schedule remains the standard full design, but it is applied only to the extreme samples.
Case study: Bracketing testing frequency
A company manufactures an oral solution in three concentrations: 10 mg/mL, 20 mg/mL, and 50 mg/ml. The formulations are identical, differing only in the amount of active pharmaceutical ingredient (API). The proposed shelf life is 24 months.
Result: The testing frequency for the tested batches remains the same as the standard schedule. The resource savings come from eliminating the intermediate sample from all testing time points.
Determining testing frequency with matrixing
Matrixing involves a more complex reduction in testing frequency by testing different subsets of samples at different time points. This is particularly useful when dealing with multiple batches, strengths, or container sizes.
Case study: Matrixing testing frequency
A company produces tablets in three strengths (S1, S2, S3) from three different batches (B1, B2, B3) for a total of nine factor combinations. The company wants to implement a one-third matrixing design for a proposed 36-month shelf life.
Drug Product Example
Objective: Establish stability profile while reducing unnecessary testing.
Full Factorial Design (Baseline)
If all combinations were tested:
Total pulls = 3 × 3 × (7+5+3) = 135 stability pulls.
Step 1 – Bracketing Application
Revised pulls = 2 strengths × 2 packs × (7+5+3) = 60 pulls.
Bracketing reduced testing by ~55%.
Step 2 – Matrixing Application
Even after bracketing, 60 pulls remain. To optimize further:
Matrixing Plan:
Revised pulls ≈ 45 (instead of 135).
Table 1.11- Testing frequency table [35]
|
Condition |
Timepoints |
Configurations Tested (Example) |
|
25°C/60%RH (Long-term) |
0, 6, 12, 24 3, 9, 18 |
All 4 configs Only 2 configs (rotating) |
|
30°C/65%RH (Intermediate) |
0, 6, 12 3, 9 |
All 4 configs Only 2 configs |
|
40°C/75%RH (Accelerated) |
0, 3, 6 |
All 4 configs (no reduction, short study) |
Case study 2
Drug :- Amlodipine besylate
Background
Amlodipine besylate is a dihydropyridine calcium channel blocker used in the management of hypertension and angina pectoris [2]. Like many drug substances, its stability is influenced by temperature and humidity. Being slightly hygroscopic, Amlodipine may undergo physical softening and increased impurity formation under high humidity conditions.
Objective
To determine the appropriate storage condition statement for labeling of Amlodipine Besylate Tablets (5 mg, blister packed) using accelerated and long-term stability data.
Study Design
Dosage Form & Packaging
Storage Condition Decision
Proposed Label Statement
“Store below 30 °C. Protect from moisture. Keep in the original package.”
Case study 3
How to decide suitable packing material [2]
Step 1 — API characterization
Step 2 — Forced degradation / stress studies (targeted experiments)
Purpose: Reveal the most damaging stressors quickly so you know which packaging properties matter most.
Standard forced tests (use a small representative batch):
Output: Identify primary degradants, mechanisms, and analytical markers to follow during packaging tests. ICH Q1A requires stress testing to support stability-indicating methods.
Step 3 — Photostability (ICH Q1B) — required if photolabile risk exists
Use the confirmatory photostability exposures specified by ICH Q1B: ≥1.2 million lux hours visible light and ≥200 Wh/m² near-UV (or equivalent validated actinometric exposure). If the API shows unacceptable changes, packaging must provide light protection (amber glass, opaque container, alu–alu blister, secondary carton).
Decision rule: any unacceptable change → include “protect from light” in label and select light-protective packaging.
Step 4 — Shortlist candidate container materials:
Stability Results
|
Packaging |
6M Accelerated |
12M Long-term |
Observations |
|
HDPE + Silica Gel |
98% assay, no degradation products |
97% assay |
Good moisture protection, minor peroxide interaction risk |
|
Amber Glass |
99% assay, stable |
98% assay |
Excellent light protection, good moisture barrier |
|
Alu-Alu Blister |
99% assay, stable |
98% assay |
Best protection, no degradation |
|
PVC Blister |
92% assay, >2% degradation products |
88% assay |
Poor protection against humidity, failed |
Decision making:-
Step 5 — Extractables & Leachables (E&L) screening (USP framework)
Two-tiered approach (recommended best practice/ regulatory expectation):
Use USP chapters and FDA/industry E&L frameworks for study design, AET calculations, and toxicological assessment. If extractables include pro-oxidants or plasticizers at relevant levels, the material may be unsuitable.
Step 6 — Container-Closure Integrity (CCI)
When to run: initial, during stability timepoints (select points), and final shelf-life check.
Step 7 — Formal ICH stability studies in candidate materials
Run parallel stability programs (same batches, same aliquots) in each shortlisted material to generate comparative data.
ICH practical points to follow:
Tests at each Timepoints:
Deliverable: comparative stability reports (assay + impurities vs time) for each packaging candidate.
Step 8 — Evaluate data and apply decision rules
Decision hierarchy (practical):
If more than one candidate passes, prefer the simplest/commercially robust option (lowest risk for supplier variability).
Regulatory note: justify material choice in regulatory filings with the stress data, E&L reports, CCI data, and stability results.
Step 9 — Toxicology & safety assessment for leachables
For any identified leachable, quantify and compare to a toxicological threshold (AET/PDE) and perform identification + safety assessment. Follow USP/FDA E&L recommendations. If an unknown leachable is present at concerning levels, consider changing material.
Step 10 — Final selection, labelling & submission package
Final packaging selection should be supported by: stress maps, photostability report (if relevant), extractables & leachables study, CCI validation report, full stability in final packaging (assay, impurities, water), supplier specs, and a justification narrative. Include required labelling such as “Protect from light” or “Keep tightly closed; store below X °C” where data support it.
Case study 4
Determination of Shelf Life Drug :- Amlodipine Besylate [36]
1. Objective
The purpose of this study is to determine the shelf life of Amlodipine Besylate bulk drug substance by evaluating:
Definition: Shelf life is the time during which a drug substance remains within its specification limits for potency and purity under specified storage conditions.
2. Drug Substance and Sample Selection
|
Parameter |
Details |
|
Drug |
Amlodipine Besylate |
|
Form |
Bulk powder |
|
Batch size |
10 kg (representative batch) |
|
Packaging |
Amber glass bottles, tightly closed to prevent moisture/light exposure |
Rationale for selection: Bulk drug is used in formulation; stability data is required for regulatory submissions.
Sampling plan:
3. Storage Conditions
Based on ICH guidelines:
|
Condition Type |
Temperature |
Relative Humidity |
Duration |
Purpose |
|
Long-term |
25°C ± 2°C |
60% ± 5% RH |
Up to 24 months |
Determine real shelf life |
|
Accelerated |
40°C ± 2°C |
75% ± 5% RH |
6 months |
Predict long-term stability |
|
Intermediate |
30°C ± 2°C |
65% ± 5% RH |
6–12 months |
Optional, if accelerated degradation is rapid |
4. Analytical Testing
Objective: Detect chemical and physical changes over time.
|
Parameter |
Method |
Purpose |
|
Assay (potency) |
HPLC |
Quantify active drug content |
|
Degradation products |
HPLC / LC-MS |
Identify and quantify impurities |
|
Moisture content |
Karl Fischer titration |
Monitor hygroscopic degradation |
|
Appearance |
Visual inspection |
Detect discoloration or physical changes |
|
Polymorphic changes |
DSC / XRD |
Detect solid-state transformations |
Acceptance criteria:
5. Data Collection
|
Time (Months) |
Assay (%) |
|
0 |
100 |
|
1 |
99.5 |
|
2 |
99.0 |
|
3 |
98.5 |
|
6 |
97.0 |
Observation: Gradual decrease in potency; degradation is linear in ln(% assay) → first-order kinetics.
6. Kinetic Analysis
Step 1: Determine reaction order
Step 2: Calculate degradation rate constant (k)
|
Time (Months) |
Assay (%) |
ln(Assay) |
|
0 |
100 |
4.605 |
|
1 |
99.5 |
4.600 |
|
2 |
99.0 |
4.595 |
|
3 |
98.5 |
4.590 |
|
6 |
97.0 |
4.574 |
Step 3: Calculate shelf life (t90)
Step 4: Extrapolation (if needed)
7. Statistical Analysis
8. Shelf Life Assignment & Storage
Case study 5
Evaluation of Stability Study [37]
Objective
The purpose of evaluating a stability study is to:
Evaluation Process
A. For Drug Substance
Step 1: Study Design
Step 2: Testing Parameters
|
Test |
Purpose |
|
Appearance |
Detect physical changes |
|
Assay (Potency) |
Measure degradation |
|
Impurity profile |
Identify degradation products |
|
Moisture content |
Check hygroscopicity |
|
pH (if in solution) |
Detect hydrolysis |
|
Specific optical rotation |
For chiral substances |
B. For Drug Product
Step 1: Study Design
Step 2: Testing Parameters
|
Test |
Purpose |
|
Appearance |
Detect discoloration, caking, etc. |
|
Assay |
Check active content |
|
Degradation products |
Check purity/stability |
|
Dissolution |
Evaluate drug release profile |
|
Water content |
Detect moisture sensitivity |
|
pH |
Monitor solution stability |
|
Microbial limits |
For liquid/semi-solid products |
Study Design
1. Drug Substance – Amlodipine Besylate
|
Study Type |
Storage Conditions |
Duration |
Frequency of Testing |
|
Long-term |
25°C ± 2°C / 60% RH ± 5% RH |
12 months |
0, 3, 6, 9, 12 months |
|
Intermediate |
30°C ± 2°C / 65% RH ± 5% RH |
6 months |
0, 3, 6 months |
|
Accelerated |
40°C ± 2°C / 75% RH ± 5% RH |
6 months |
0, 3, 6 months |
Packaging: Double polyethylene bags inside HDPE container with screw cap.
Parameters Tested:
2. Drug Product – Amlodipine Tablets 5 mg
|
Study Type |
Storage Conditions |
Duration |
Frequency of Testing |
|
Long-term |
25°C ± 2°C / 60% RH ± 5% RH |
12 months |
0, 3, 6, 9, 12 months |
|
Intermediate |
30°C ± 2°C / 65% RH ± 5% RH |
6 months |
0, 3, 6 months |
|
Accelerated |
40°C ± 2°C / 75% RH ± 5% RH |
6 months |
0, 3, 6 months |
Packaging: PVC/Aluminum foil blister packs.
Parameters Tested:
5. Analytical Methodology
All analytical methods were validated according to ICH Q2(R1) guidelines for:
Chromatographic Conditions for Assay:
6. Observed Results
6.1 Drug Substance
|
Storage Condition |
Time (months) |
Assay (%) |
Total Impurities (%) |
Moisture (%) |
|
25°C/60% RH |
0 |
100.0 |
0.10 |
0.12 |
|
25°C/60% RH |
12 |
99.4 |
0.28 |
0.18 |
|
30°C/65% RH |
6 |
99.2 |
0.30 |
0.20 |
|
40°C/75% RH |
6 |
98.3 |
0.44 |
0.24 |
Interpretation:
6.2 Drug Product (Amlodipine Tablets 5 mg)
|
Storage Condition |
Time (months) |
Assay (%) |
Related Substances (%) |
Dissolution (%) |
Remarks |
|
25°C/60% RH |
0 |
100.0 |
0.12 |
99.2 |
No change |
|
25°C/60% RH |
12 |
99.0 |
0.28 |
98.7 |
Stable |
|
30°C/65% RH |
6 |
98.8 |
0.33 |
98.1 |
Stable |
|
40°C/75% RH |
6 |
98.4 |
0.46 |
97.9 |
Slight degradation |
Physical Appearance: No discoloration or caking observed.
7. Kinetic Analysis and Shelf-Life Calculation
The degradation follows first-order kinetics (since log concentration decreases linearly with time).
Equation:
From regression analysis of assay values at 40°C/75% RH:
Slope = –0.0009 month?¹
Thus,
Shelf Life (t??) Calculation
Practical shelf life = 24 months (2 years) (as per ICH recommendation to round down based on statistical confidence).
8. Statistical Evaluation (ICH Q1E)
Conclusion: Shelf life of 24 months supported statistically.
Storage Condition and Labeling
|
Condition |
Observation |
Recommended Label |
|
Long-term |
Stable |
Store below 30°C |
|
Accelerated |
Slight degradation |
Protect from light and moisture |
|
Photostability |
No significant change |
Avoid direct sunlight |
CONCLUSION
Stability is a fundamental attribute that ensures a product maintains its intended quality, safety, and efficacy throughout its shelf life under recommended storage conditions. Stability testing plays a critical role in product development across pharmaceuticals, food, and cosmetics by systematically evaluating how environmental factors like temperature, humidity, and light affect product integrity over time. Through stability studies, manufacturers can establish scientifically justified shelf lives, define optimal storage and handling conditions, identify degradation pathways, and meet regulatory requirements for product approval and labeling. The mechanistic approach to stability testing, involving controlled environmental exposures and stability-indicating analytical methods, supports robust shelf-life estimation and ensures consumers receive safe, effective, and high-quality products. Thus, stability testing constitutes an indispensable pillar of quality assurance, regulatory compliance, and consumer protection in product lifecycle management.
REFERENCES
Abhijeet Welankiwar, Abhishek Jambhe, Evaluation of Stability Study of Drug Substance and Drug Product as per Regulatory Guidelines, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 11, 607-638. https://doi.org/10.5281/zenodo.17531392
10.5281/zenodo.17531392