View Article

  • Development and Validation of Stability-Indicating RP-HPLC Methods for Pharmaceutical Drugs: A Comprehensive Review

  • 1Department of Pharmaceutical Quality Assurance, R. G, Sapkal College of Pharmacy, Anjaneri, Nashik-422212. 
    2Department of Pharmaceutical Chemistry, R. G, Sapkal College of Pharmacy, Anjaneri, Nashik-422212. 
    3Department of Pharmacology, R. G, Sapkal College of Pharmacy, Anjaneri, Nashik-422212.

Abstract

One important feature of pharmaceutical goods that has a direct bearing on their safety, effectiveness, and shelf life is stability. The need for reliable, validated analytical techniques grows as regulatory requirements change and formulation complexity rises. Because of its excellent resolution, sensitivity, reproducibility, and adaptability, Reverse Phase Excellent-Performance Liquid Chromatography (RP-HPLC) has become the gold standard among analytical techniques for creating stability-indicating methods (SIMs). The principles, development techniques, and validation criteria for stability-indicating RP-HPLC methods in the pharmaceutical business are thoroughly examined in this review article. A thorough review of method development, including the choice of chromatographic conditions, mobile and stationary phases, and detection parameters, follows a discussion of the significance of SIMs in guaranteeing medication quality. According to ICH Q1A(R2) recommendations, forced degradation tests are crucial for assessing the method's capacity to differentiate between the active pharmaceutical ingredient (API) and its breakdown products. The article also describes the validation procedure in accordance with ICH Q2(R1), emphasizing important criteria including robustness, specificity, linearity, accuracy, precision, limit of detection (LOD), and limit of quantification (LOQ). It is also covered how to include contemporary quality paradigms like Design of Experiments (DoE) and Quality by Design (QbD) into method development, highlighting how they can improve method robustness and regulatory compliance. Overall, this review contributes to efficient quality control, regulatory approval, and safe therapeutic outcomes by highlighting the crucial role stability-indicating RP-HPLC methods play in pharmaceutical analysis and offering a scientific framework for their methodical development and validation.Method dependability and regulatory compliance are improved by incorporating contemporary methods like Design of Experiments (DoE) and Quality by Design (QbD). All things considered, RP-HPLC is still an essential tool for pharmaceutical stability testing and quality control.

Keywords

RP-HPLC, Stability-indicating method, Method validation, ICH guidelines, Forced degradation, pharmaceutical analysis, QbD, DoE

Introduction

Strict quality control and assurance procedures are required by the pharmaceutical business to guarantee the safety, effectiveness, and shelf life of drugs during their whole lifespan. Implementing stability-indicating methods (SIMs) that can identify the degradation products of active pharmaceutical ingredients (APIs) without interference from excipients, impurities, or degradation products is a fundamental requirement in the analysis of pharmaceutical formulations [1]. Since it can analyze non-volatile, thermally unstable, and polar substances and has good resolution, repeatability, and sensitivity, reverse phase high-performance liquid chromatography (RP-HPLC) is the most commonly used analytical method in the pharmaceutical industry [2]. According to the requirements of the International Conference on Harmonization (ICH), the development of stability-indicating RP-HPLC procedures is essential to guaranteeing the stability of pharmaceutical products [3].  First used to describe techniques that can differentiate the API from its breakdown products, the term "stability-indicating" was further strengthened by regulatory guidance from the ICH Q1A(R2) and Q2(R1) guidelines, which stress the significance of proving stability through validated analytical methods [4,5]. The selection of chromatographic conditions (column type, mobile phase, flow rate, and detection wavelength), forced degradation studies, method optimization, and subsequent validation for accuracy, precision, specificity, linearity, robustness, and detection limits are all crucial steps in the development of a stability-indicating RP-HPLC method [6,7]. Since they mimic the impact of environmental factors on medicinal compounds and offer insights into possible degradation routes, forced degradation studies—also referred to as stress testing—are essential to the development of SIMs [8].  The capabilities of stability-indicating techniques have been further improved by recent developments in HPLC instrumentation and column technologies, such as ultra-high-performance liquid chromatography (UHPLC), core-shell columns, and diode-array detection, which enable better separation of complex mixtures and improved analytical performance [9]. Developing strong, dependable, and regulatory-compliant RP-HPLC stability-indicating procedures is crucial given the increasing number of new chemical entities and generic formulations hitting the market. With the use of case studies and examples from the literature, this review seeks to provide a thorough overview of the tactics, difficulties, and regulatory considerations related to the creation and validation of RP-HPLC methods for stability investigations. Equally important is method validation, which is carried out in accordance with ICH Q2(R1), which lists important validation factors including robustness, specificity, accuracy, precision, linearity, range, limit of detection (LOD), limit of quantification (LOQ), robustness, and system adaptability [5,10]. Every one of these factors guarantees the method's dependability and consistency over the course of the pharmaceutical product's lifecycle. To improve method comprehension and control, there has been a growing tendency in recent years to apply Design of Experiments (DoE) and Quality by Design (QbD) approaches in method development [9]. Additionally, stability-indicating assays are highly recommended by regulatory bodies such as the US FDA and EMA to evaluate the integrity of APIs in their formulations throughout the medication approval process [11]. Therefore, the principles, development strategies, validation protocols, and regulatory viewpoints involved in the design of a reliable RP-HPLC-based stability-indicating method for pharmaceutical pharmaceuticals are all covered in detail in this review paper.

Overview of RP-HPLC

In pharmaceutical research and development, reverse phase high-performance liquid chromatography (RP-HPLC) is a commonly used analytical method, particularly for routine quality control and stability investigations. It is the preferred technique for creating stability-indicating assays because to its exceptional resolution, repeatability, sensitivity, and suitability for a wide variety of chemicals [12,13].

1. Principle of RP-HPLC

Analytes are separated by RP-HPLC according to their hydrophobic interactions with a somewhat polar mobile phase and a non-polar stationary phase. Silica particles bound with hydrophobic alkyl chains, such C18, usually make up the stationary phase, whilst aqueous buffers combined with organic solvents, such as methanol or acetonitrile, make up the mobile phase [2]. Effective separation of structurally related substances and degradation products is made possible by hydrophobic molecules' stronger retention and later elution compared to polar molecules [1].

2.Instrumentation

An RP-HPLC system generally includes:

  • Mobile phase reservoirs
  • Degasser
  • Pump (for high-pressure solvent delivery)
  • Injector
  • Column (usually C18, 150–250 mm in length)
  • Detector (UV, PDA, or MS)

Data acquisition system

Each component contributes to method performance, and optimal configuration ensures precision, sensitivity, and method robustness [14].

3. Column and Mobile Phase Selection

Because to their hydrophobicity and wide compatibility, C18 columns are the most widely used. For the best separation, the pH and components of the mobile phase must be chosen carefully. For ionizable substances, aqueous buffers, such as phosphate buffer, are commonly used to regulate pH and enhance peak shape [18]. Either gradient or isocratic elution may be employed, depending on how complicated the sample is. When the polarity of the analytes varies greatly or when the primary drug component needs to be resolved together with contaminants and degradation products, gradient elution is the recommended method [15].

4 .Detection Techniques

Because of their sensitivity and broad range of applications, UV-visible and photodiode array (PDA) detectors are the most often used in RP-HPLC. These are perfect for substances that absorb ultraviolet light. Mass spectrometry (MS) can be used in conjunction with RP-HPLC to increase sensitivity or provide structural clarification of unknown degradation products [3].

5. Advantages of RP-HPLC in Stability Studies

The following benefits make RP-HPLC especially well-suited for the development of stability-indicating methods: • High resolution and peak capacity;

• Compatibility with a broad spectrum of medicines.

• The capacity to extract medications from excipients and breakdown products.

• Adaptability in the creation of methods (pH, solvents, gradients that can be changed).

• ICH compliance and regulatory acceptance [11].

Regulatory Framework

In order to guarantee drug safety, efficacy, and quality over the course of the product's shelf life, regulatory regulations heavily influence the development and validation of stability-indicating RP-HPLC procedures. Stability testing and thorough analytical method validation are emphasized by regulatory bodies as essential steps in the pharmaceutical development process.

1.ICH Guidelines

The International Council for Harmonisation (ICH) provides harmonized guidelines widely accepted by major regulatory agencies worldwide. Two key guidelines relevant to stability-indicating RP-HPLC method development are:

ICH Q1A(R2): Stability Testing of New Drug Substances and Products
In order to determine how the quality of a drug substance or product changes over time under environmental conditions including temperature, humidity, and light, this guideline describes how stability studies should be planned and carried out. It requires the use of stability-indicating, validated analytical methods that can identify degradation products [3].

ICH Q2(R1): Validation of Analytical Procedures: Text and Methodology
Specificity, accuracy, precision, linearity, range, detection limit, quantitation limit, robustness, and system suitability are among the parameters and requirements for validating analytical techniques that are outlined in this paper. Specificity is essential for stability-indicating techniques to differentiate the medication from contaminants and degradation products [5].

2 .US Food and Drug Administration (FDA)

The FDA emphasizes the significance of stability-indicating techniques in the drug approval process and offers guidelines on the development and validation of analytical procedures. Their Advice to Industry: Methods and Procedures for Analysis Expectations for proving method robustness, suitability for intended use, and reliability are outlined in Validation for Drugs and Biologics [11].

3. European Medicines Agency (EMA)

Stability testing standards for medications sold in the EU are standardized by the EMA. When evaluating product shelf life and degradation routes, EMA guidelines stress the importance of using proven, stability-indicating methodologies. Additionally, when developing methods, the EMA promotes the use of risk-based strategies like Quality by Design (QbD) [16].

4 .Pharmacopoeial Standards

The United States Pharmacopoeia (USP), the British Pharmacopoeia (BP), and the European Pharmacopoeia (EP) are pharmacopoeias that offer monographs that contain compendial methods or criteria for analytical procedures, including assays that indicate stability. Method approval in official quality control is ensured by adherence to these monographs [17].

5. Importance of Compliance

Adherence to these regulatory frameworks ensures that the stability-indicating RP-HPLC methods:

  • Accurately detect and quantify APIs and degradation products.
  • Provide data essential for shelf life and storage condition determination.
  • Support regulatory submissions, ensuring market authorization and patient safety.

Method Development Process

The development of a stability-indicating RP-HPLC method involves a systematic approach to achieve reliable separation and quantification of the drug substance along with its degradation products. The process must ensure the method’s specificity, sensitivity, accuracy, and robustness to meet regulatory standards and practical analytical requirements.

1. Selection of Analyte and Sample Preparation

It is essential to correctly identify and comprehend the active pharmaceutical ingredient (API) and its formulation matrix. Techniques for preparing samples should avoid adding artifacts while maintaining the integrity of the analyte and degradation products. This could involve reproducibility-optimized dilution, filtering, or dissolution procedures [1].

2. Selection of Chromatographic Conditions

Key parameters influencing separation are carefully selected and optimized, often iteratively:

  • Column Selection: Because they can separate a variety of chemicals and have hydrophobic interactions, reversed-phase columns—which are primarily C18—are recommended. Resolution and analysis time are influenced by particle size and column diameters [13].
  • Mobile Phase Composition: To regulate analyte ionization and retention behavior, the selection of solvents (water, buffers, acetonitrile, methanol) and pH adjustment are essential. Peak form and reproducibility are improved by buffered aqueous phases at regulated pH, particularly for ionizable substances [2].
  • Elution Mode:
    • Isocratic elution offers simplicity and reproducibility for less complex mixtures.
    • Gradient elution is favored when resolving multiple degradation products with varying polarities to improve separation efficiency [3].
  • Flow Rate and Temperature: These parameters affect chromatographic resolution and peak shape and are optimized to balance sensitivity and run time [12].

3. Detection Wavelength

The analyte's and known degradants' UV absorption maxima (λmax) are used to determine the detection wavelength. Spectral analysis can be used to detect co-eluting contaminants and ensure peak purity using photodiode array (PDA) detectors [14].

4. Forced Degradation Studies

Forced degradation studies create prospective degradants by simulating several stress conditions (acid, basic, oxidative, thermal, and photolytic) in order to create a stability-indicating approach. The technique must exhibit specificity and selectivity by clearly separating the API from its degradation products [15].

5. Optimization and System Suitability

The parameters of the method are adjusted to retain sufficient sensitivity, guarantee appropriate peak symmetry, and optimize the resolution between key peaks. To verify that a method is ready for validation, system suitability tests (SST) evaluate theoretical plates, resolution, repeatability, and tailing factor [5].

Forced Degradation Studies

The development of stability-indicating RP-HPLC procedures requires the use of forced degradation experiments, commonly referred to as stress testing. By exposing the drug substance and drug product to a range of stress settings, these investigations aid in understanding their inherent stability. In order to demonstrate the analytical method's specificity and stability-indicating capability, the main goal is to produce possible degradation products and validate that it can separate and quantify the active pharmaceutical ingredient (API) clearly from its degradation products [1,15].

1. Purpose of Forced Degradation

• To determine the products and mechanisms of degradation.
• To determine the drug's inherent stability.
• To confirm the analytical method's specificity.
• To aid in the creation of formulations and the assessment of shelf life.
• To fulfill stability testing regulations (ICH Q1A(R2)) [3].

2. Common Stress Conditions

Forced degradation is conducted under controlled laboratory conditions that accelerate drug degradation beyond normal storage conditions. Typical stress conditions include:

•Acid hydrolysis is the process of promoting acid-catalyzed breakdown by treating with diluted acid (0.1 N HCl, for example).

• Basic Hydrolysis: Alkaline deterioration is simulated by treating with a diluted base (0.1 N NaOH, for example).

• Oxidative Degradation: To assess oxidative stability, expose the material to oxidizing agents such hydrogen peroxide (e.g., 3% H2O?).

• Thermal Degradation: Heat stability is assessed by subjecting the material to high temperatures (such as 60–80°C).

• Photolytic degradation: In accordance with ICH Q1B, exposure to visible and UV light is used to evaluate photostability [6,18].

• Humidity Stress: Hydrolytic breakdown is assessed by exposure to elevated relative humidity.

3. Execution and Monitoring

To track the degree of degradation and the product profile, samples are examined using the established RP-HPLC method after each stress condition is administered for predefined periods of time. The method's specificity is confirmed when all degradation products are successfully separated from the API peak. To provide adequate degradant generation without total API destruction, forced degradation should ideally lead to 10–30% degradation [19].

4. Role in Method Validation

The approach validation is supported by forced degradation data, which demonstrate specificity, a crucial ICH Q2(R1) characteristic. It proves that the technique can precisely measure the API even when its degradation products and contaminants are present, guaranteeing trustworthy stability testing and quality assurance [5].

Method Validation (ICH Q2(R1))

A crucial stage in the creation of stability-indicating RP-HPLC techniques is method validation, which guarantees that the analytical process is dependable, repeatable, and appropriate for its intended use. A thorough framework for validating analytical techniques in the pharmaceutical business is provided by International Council for Harmonization (ICH) guideline Q2(R1), which covers crucial factors to ensure method quality and regulatory compliance [5].

1. Validation Parameters

The key parameters for validating stability-indicating RP-HPLC methods as per ICH Q2(R1) include:

• Specificity: The method's capacity to clearly evaluate the analyte in the presence of contaminants, degradation products, and matrix constituents. Peak purity analysis and forced degradation tests were used to demonstrate [1].

• Linearity: The capacity of the procedure to yield outcomes within a given range that are proportionate to the analyte concentration. To create a calibration curve, at least five concentration levels are advised [2].

• Accuracy: The degree to which measured results resemble the genuine value; this is frequently determined by recovery tests that include injecting known amounts of the drug into the matrix [14].

Precision:  To evaluate procedure consistency, repeatability (intra-day), intermediate precision (inter-day), and reproducibility are evaluated [12].

Limit of Detection (LOD) and Limit of Quantitation (LOQ): The lowest quantity of analyte that can be detected and quantified with reasonable accuracy and precision is known as the Limit of Detection (LOD) and Limit of Quantitation (LOQ). These values are often determined using response standard deviation or signal-to-noise ratios [13].
• Robustness: The ability of the procedure to withstand minor intentional changes in its parameters (such as flow rate, pH, and temperature), hence verifying its dependability under normal operating conditions [3].

System Suitability Testing (SST):Prior to sample analysis, System Suitability Testing (SST) verifies the system's performance by examining characteristics such as theoretical plates, tailing factor, resolution, and repeatability [11].

2. Importance in Stability-Indicating Methods

As a stability-indicating assay, validation guarantees that the RP-HPLC technology can accurately separate and quantify the therapeutic component from its degradation products. By offering written proof of method performance characteristics, it helps regulatory submissions and satisfies pharmacopeia and regulatory agency criteria [15].

Quality by Design (QbD) and Design of Experiments (DoE) Approaches

1. Overview of Quality by Design (QbD)

Instead, than depending just on end-product testing, Quality by Design (QbD) is a methodical, science- and risk-based approach to pharmaceutical development that stresses building quality into the product and process from the start [20]. By comprehending the connections between method variables and performance, QbD guarantees that analytical methods are resilient, dependable, and appropriate for their intended use.
Analytical Target Profile (ATP) definition, Critical Method Attributes (CMAs), and Critical Method Parameters (CMPs) are all part of QbD in RP-HPLC method development. Risk assessment and optimization are then carried out to get the intended method performance [21].

2. Role of Design of Experiments (DoE)

An essential statistical tool for QbD is the Design of Experiments (DoE), which methodically assesses how various factors affect the results of analytical methods. By assisting in the comprehension of the interplay of chromatographic parameters, including pH, mobile phase composition, flow rate, and column temperature, DoE facilitates robust performance and method optimization [22].
Factorial designs, central composite design (CCD), and Box-Behnken design (BBD) are common DoE designs used in RP-HPLC method development. These designs allow for more effective testing with fewer runs and provide greater insight into the behavior of the method [23].

3. Advantages of QbD and DoE in Stability-Indicating RP-HPLC

• Improved Method Robustness: During routine analysis, methods that explore the design space are less susceptible to minor changes in analytical settings, which lowers failure rates [24].
• Regulatory Flexibility: QbD techniques are supported by regulatory bodies such as the FDA and EMA, which permit justifiable method adjustments inside the design space without requiring extra submissions [11].
• Shorter Development Time and Cost: A methodical approach expedites method development and reduces trial-and-error.
• Better Understanding: Offers in-depth knowledge of crucial elements influencing separation, sensitivity, and specificity, which is crucial for stability-indicating techniques where accurate degradant detection is needed [25].

4 .Implementation Steps

1. Specify the RP-HPLC method's ATP and performance requirements.
2. Determine the CMPs (pH, organic solvent percentage, flow rate, etc.).
3. Prioritize parameters by conducting a risk assessment.
4. Examine factor interactions and effects using DoE.
5. Determine the Method Operable Design Region (MODR) and optimize it.
6. Verify the optimized approach in accordance with ICH recommendations [26].

Applications and Case Studies

1. Applications of Stability-Indicating RP-HPLC Methods

Stability-indicating RP-HPLC methods have become indispensable in pharmaceutical analysis due to their ability to simultaneously separate and quantify the active pharmaceutical ingredient (API) and its degradation products. These methods are widely applied in:

Quality Control: To guarantee batch-to-batch uniformity and adherence to pharmacopeial standards, routine analysis of raw materials, intermediates, and final products is necessary [1].
Stability testing: assessing a drug's stability under several storage circumstances in order to ascertain its shelf life and necessary packaging [3].
Formulation Development: Evaluating how packaging, manufacturing parameters, and excipients affect medication stability [15].
Regulatory Submissions: supplying solid stability information for dossiers requiring regulatory approval [11].
Degradation Kinetics: Examining kinetics and paths of degradation to comprehend mechanisms of drug stability [19].

2. Case Studies

Case Study 1: Stability-Indicating Method for a Non-Steroidal Anti-Inflammatory Drug (NSAID)

The NSAID diclofenac sodium was separated from its acidic, basic, oxidative, and photolytic breakdown products using a stability-indicating RP-HPLC technique. The technique used a C18 column with gradient elution using acetonitrile and phosphate buffer. Multiple degradants that were all well resolved from the API peak were found using forced degradation investigations, proving the robustness and specificity of the approach [27].

Case Study 2: Analysis of Antihypertensive Drug — Amlodipine Besylate

Following ICH criteria, an RP-HPLC technique was created and verified for amlodipine besylate in the presence of breakdown products produced by hydrolytic, oxidative, and photolytic stress. The technique provided quick analysis with outstanding accuracy and precision by using an isocratic mobile phase and UV detection at 360 nm [28].

Case Study 3: Development of Stability-Indicating Method for Anticancer Drug Imatinib Mesylate

For imatinib mesylate and its breakdown products produced by forced degradation under acid, base, oxidation, heat, and photolytic conditions, researchers created a validated RP-HPLC method. All of the degradants were successfully isolated, demonstrating the method's suitability for quality control and stability investigations [29].

Future Perspectives

As pharmaceutical sciences advance, the development and validation of stability-indicating RP-HPLC methods are expected to evolve significantly, driven by regulatory, technological, and scientific innovations. Several future trends and perspectives can be anticipated in this domain:

1. Integration of Advanced Analytical Technologies

RP-HPLC will increasingly be coupled with tandem mass spectrometry (LC-MS/MS), photodiode array detectors (PDA), and high-resolution mass spectrometry (HRMS). Particularly for complex formulations and biologics, these hybrid approaches improve the specificity and sensitivity of the method and allow for more accurate identification of breakdown products [30, 31].

2. Expanded Use of Quality by Design (QbD)

It is anticipated that the development of analytical methods in the pharmaceutical business would more widely embrace QbD concepts. The development of more reliable, adaptable, and legally acceptable RP-HPLC techniques will be aided by the integration of risk assessment tools, Design of Experiments (DoE), and multivariate data analysis (MVDA) [21].

3. Automation and Digitalization

Machine learning (ML), automation, and artificial intelligence (AI) are becoming increasingly used in method development. These tools can shorten development times, optimize method parameters, and forecast chromatographic behavior. Continuous compliance and verification will also be supported by digital systems for method lifecycle management [32].

4. Green Analytical Chemistry (GAC)

The development of analytical methods is paying more attention to environmental sustainability. Greener solvents, lower solvent consumption, and energy-efficient equipment will probably be given priority in future RP-HPLC procedures in accordance with GAC guidelines and green chemistry metrics [33, 34].

5. Regulatory Harmonization and Real-Time Release Testing (RTRT)

The development of RP-HPLC methods will be impacted by the global harmonization of guidelines and the progress of real-time release testing (RTRT). Regulatory bodies are promoting the use of analytical target profiles (ATPs) that are in line with product performance and quality as well as lifecycle-based method management [35].

6. Tailored Methods for Novel Drug Modalities

As new drug modalities like peptides, oligonucleotides, and nanomedicines hit the market, there will be a greater demand for specialized RP-HPLC techniques to evaluate their stability and degradation processes. Customizing the method will require labile structures, biological components, and complex matrices [36].

CONCLUSION

For pharmaceutical goods to be safe, effective, and of high quality, stability-indicating reversed-phase high-performance liquid chromatography (RP-HPLC) techniques must be developed and validated. As required by regulatory bodies, these analytical techniques are made to accurately measure active pharmaceutical ingredients (APIs) and concurrently identify possible degradation products produced under different stress situations. They are essential at every stage of the drug lifecycle, from early drug development to post-marketing surveillance. RP-HPLC techniques become effective tools for determining degradation pathways and evaluating drug stability under ICH-recommended stress conditions—acid, basic, oxidative, thermal, and photolytic environments—through a methodical approach that includes method development, forced degradation studies, and rigorous validation. These tests guarantee that degrading goods do not endanger patient safety in addition to assisting in the definition of suitable storage conditions and shelf life. These techniques' accuracy, precision, specificity, robustness, and reproducibility are guaranteed by their validation in accordance with ICH Q2(R1) principles. Every validation parameter verifies that, even in complicated matrices, the technique can accurately distinguish between the API and its degradation products. Moreover, these techniques offer the sensitivity needed for trace-level impurity analysis by incorporating limits of detection (LOD) and quantitation (LOQ).  Robustness and regulatory compliance are further improved when Quality by Design (QbD) and Design of Experiments (DoE) concepts are applied during method development. By identifying a Method Operable Design Region (MODR) and facilitating a thorough grasp of essential method variables, these technologies reduce the likelihood of method failures during normal use. Such scientific and risk-based methods will become more and more important as pharmaceutical development advances toward more complex formulations and delivery technologies.
The draft ICH Q14 guideline reflects the changing regulatory expectations on lifecycle management of analytical methods. In the end, stability-indicating RP-HPLC techniques created under these frameworks facilitate real-time release testing (RTRT) and provide more flexibility for post-approval modifications, which leads to quicker drug approvals and better patient outcomes. In the future, green chemistry concepts, automation, machine learning, and hybrid methods like LC-MS/MS will probably be incorporated into stability-indicating RP-HPLC. These developments hold promise for lowering the impact on the environment, increasing the effectiveness of the technique, and enabling predictive analytics for method performance.

REFERENCES

  1. Blessy M, Patel RD, Prajapati PN, Agrawal YK. Development of forced degradation and stability indicating studies of drugs—A review. J Pharm Anal. 2014;4(3):159–165. doi:10.1016/j.jpha.2013.09.003
  2. Snyder LR, Kirkland JJ, Dolan JW. Introduction to Modern Liquid Chromatography. 3rd ed. John Wiley & Sons; 2010.
  3. ICH Harmonised Tripartite Guideline Q1A(R2): Stability Testing of New Drug Substances and Products. 2003
  4. Bakshi M, Singh S. Development of validated stability-indicating assay methods—critical review. J Pharm Biomed Anal. 2002;28(6):1011–1040
  5. ICH Guideline Q2(R1): Validation of Analytical Procedures: Text and Methodology. 2005.
  6. ICH Guideline Q1B: Photostability Testing of New Drug Substances and Products. 1996.
  7. Ravisankar P, et al. A review on analytical method development. Indian J Res Pharm Biotechnol. 2015;3(3):229–233.
  8. Singh R, Rehman Z. Current trends in forced degradation study for pharmaceutical product development. J Pharm Educ Res. 2012;3(1):54–63.
  9. Nguyen DT, et al. Development and validation of a stability-indicating method using UHPLC for pharmaceutical quality control. J Chromatogr Sci. 2011;49(10):753–759.
  10. Rao, R. N., & Nagaraju, V. (2006). An overview of the recent trends in development of HPLC methods for pharmaceutical analysis. Journal of Pharmaceutical and Biomedical Analysis, 44(2), 337–346
  11. US FDA. (2000). Guidance for Industry: Analytical Procedures and Methods Validation for Drugs and Biologics. U.S. Department of Health and Human Services.
  12. Kazakevich, Y., & LoBrutto, R. (2007). HPLC for pharmaceutical scientists. John Wiley & Sons
  13. Dong, M. W. (2006). Modern HPLC for practicing scientists. Wiley-Interscience
  14. Swartz, M. E., & Krull, I. S. (2012). Handbook of Analytical Validation. CRC Press
  15. Alsante, K. M., Ando, A., & Brown, R. (2007). The role of degradant profiling in active pharmaceutical ingredients and drug products. Advanced Drug Delivery Reviews, 59(1), 29–37.
  16. EMA. (2016). Guideline on the Chemistry of Active Substances. European Medicines Agency.
  17. United States Pharmacopeia (USP) 43-NF 38. (2020). General Chapter <1225> Validation of Compendial Procedures. USP Convention.
  18. Singh, S., & Bakshi, M. (2000). Guidance on Conduct of Stress Tests to Determine Inherent Stability of Drugs. Pharmaceutical Technology Online.
  19. Carstensen, J. T., & Rhodes, C. T. (2000). Drug Stability: Principles and Practices (3rd ed.). Marcel Dekker.
  20. Yu, L. X., Kopcha, M., & Brackett, A. (2014). Analytical Quality by Design: A Tool for Regulatory Flexibility and Robust Analytics. Pharmaceutical Technology, 38(6), 24-32.
  21. Rathore, A. S., & Winkle, H. (2009). Quality by design for biopharmaceuticals. Nature Biotechnology, 27(1), 26–34
  22. Mandal, S., Mandal, S., & Nayak, S. (2013). Analytical Quality by Design: A Tool for Robust Method Development in Pharmaceutical Analysis. International Journal of Pharmaceutical Sciences and Research, 4(7), 2587–2596.
  23. Montgomery, D. C. (2017). Design and Analysis of Experiments (9th ed.). Wiley.
  24. Shah, R. B., & Jambhekar, S. S. (2010). Implementation of QbD in analytical methods: a review. Journal of Pharmaceutical Sciences, 99(4), 1817–1834.
  25. Badawy, S. M., & El-Ahmady, S. H. (2019). Application of Design of Experiments (DoE) in Development and Validation of Stability-Indicating Methods. Journal of Chromatographic Science, 57(7), 569–579.
  26. ICH Q8(R2). Pharmaceutical Development. International Council for Harmonisation, 2009.
  27. Patel, R. M., & Patel, M. M. (2011). Development and validation of stability-indicating RP-HPLC method for determination of diclofenac sodium in pharmaceutical formulations. Journal of Pharmacy Research, 4(6), 1996–2000
  28. Sharma, P., & Kumar, S. (2015). Stability-indicating RP-HPLC method for the determination of amlodipine besylate in pharmaceutical dosage forms. International Journal of Pharmaceutical Sciences and Research, 6(8), 3413–3420.
  29. Rao, R. S., & Kumar, S. (2012). Development and validation of a stability-indicating RP-HPLC method for imatinib mesylate. Journal of Analytical Chemistry, 67(3), 245–250.
  30. Samanidou, V. F., & Zacharis, C. K. (2015). Current trends and applications of RP-HPLC in pharmaceutical analysis. Journal of Liquid Chromatography & Related Technologies, 38(13), 1286–1302.
  31. Niessen, W. M. A. (2011). Liquid Chromatography–Mass Spectrometry (LC-MS/MS): Principles and Applications. CRC Press.
  32. Skoczylas, M., & P?otka-Wasylka, J. (2021). Artificial intelligence in green analytical chemistry. TrAC Trends in Analytical Chemistry, 138, 116223
  33. Tobiszewski, M. (2018). Metrics for green analytical chemistry. Analytical Methods, 10(37), 4615–4622.
  34. Ga?uszka, A., Migaszewski, Z., & Namie?nik, J. (2013). The 12 principles of green analytical chemistry and the SIGNIFICANCE mnemonic of green analytical practices. TrAC Trends in Analytical Chemistry, 50, 78–84.
  35. ICH Q14 Draft Guideline. Analytical Procedure Development. International Council for Harmonisation, 2022.
  36. Sender, R., Bar-On, Y. M., & Milo, R. (2021). RP-HPLC and stability studies of peptides and RNA therapeutics: Challenges and future perspectives. Biotechnology Advances, 53, 107836.

Reference

  1. Blessy M, Patel RD, Prajapati PN, Agrawal YK. Development of forced degradation and stability indicating studies of drugs—A review. J Pharm Anal. 2014;4(3):159–165. doi:10.1016/j.jpha.2013.09.003
  2. Snyder LR, Kirkland JJ, Dolan JW. Introduction to Modern Liquid Chromatography. 3rd ed. John Wiley & Sons; 2010.
  3. ICH Harmonised Tripartite Guideline Q1A(R2): Stability Testing of New Drug Substances and Products. 2003
  4. Bakshi M, Singh S. Development of validated stability-indicating assay methods—critical review. J Pharm Biomed Anal. 2002;28(6):1011–1040
  5. ICH Guideline Q2(R1): Validation of Analytical Procedures: Text and Methodology. 2005.
  6. ICH Guideline Q1B: Photostability Testing of New Drug Substances and Products. 1996.
  7. Ravisankar P, et al. A review on analytical method development. Indian J Res Pharm Biotechnol. 2015;3(3):229–233.
  8. Singh R, Rehman Z. Current trends in forced degradation study for pharmaceutical product development. J Pharm Educ Res. 2012;3(1):54–63.
  9. Nguyen DT, et al. Development and validation of a stability-indicating method using UHPLC for pharmaceutical quality control. J Chromatogr Sci. 2011;49(10):753–759.
  10. Rao, R. N., & Nagaraju, V. (2006). An overview of the recent trends in development of HPLC methods for pharmaceutical analysis. Journal of Pharmaceutical and Biomedical Analysis, 44(2), 337–346
  11. US FDA. (2000). Guidance for Industry: Analytical Procedures and Methods Validation for Drugs and Biologics. U.S. Department of Health and Human Services.
  12. Kazakevich, Y., & LoBrutto, R. (2007). HPLC for pharmaceutical scientists. John Wiley & Sons
  13. Dong, M. W. (2006). Modern HPLC for practicing scientists. Wiley-Interscience
  14. Swartz, M. E., & Krull, I. S. (2012). Handbook of Analytical Validation. CRC Press
  15. Alsante, K. M., Ando, A., & Brown, R. (2007). The role of degradant profiling in active pharmaceutical ingredients and drug products. Advanced Drug Delivery Reviews, 59(1), 29–37.
  16. EMA. (2016). Guideline on the Chemistry of Active Substances. European Medicines Agency.
  17. United States Pharmacopeia (USP) 43-NF 38. (2020). General Chapter <1225> Validation of Compendial Procedures. USP Convention.
  18. Singh, S., & Bakshi, M. (2000). Guidance on Conduct of Stress Tests to Determine Inherent Stability of Drugs. Pharmaceutical Technology Online.
  19. Carstensen, J. T., & Rhodes, C. T. (2000). Drug Stability: Principles and Practices (3rd ed.). Marcel Dekker.
  20. Yu, L. X., Kopcha, M., & Brackett, A. (2014). Analytical Quality by Design: A Tool for Regulatory Flexibility and Robust Analytics. Pharmaceutical Technology, 38(6), 24-32.
  21. Rathore, A. S., & Winkle, H. (2009). Quality by design for biopharmaceuticals. Nature Biotechnology, 27(1), 26–34
  22. Mandal, S., Mandal, S., & Nayak, S. (2013). Analytical Quality by Design: A Tool for Robust Method Development in Pharmaceutical Analysis. International Journal of Pharmaceutical Sciences and Research, 4(7), 2587–2596.
  23. Montgomery, D. C. (2017). Design and Analysis of Experiments (9th ed.). Wiley.
  24. Shah, R. B., & Jambhekar, S. S. (2010). Implementation of QbD in analytical methods: a review. Journal of Pharmaceutical Sciences, 99(4), 1817–1834.
  25. Badawy, S. M., & El-Ahmady, S. H. (2019). Application of Design of Experiments (DoE) in Development and Validation of Stability-Indicating Methods. Journal of Chromatographic Science, 57(7), 569–579.
  26. ICH Q8(R2). Pharmaceutical Development. International Council for Harmonisation, 2009.
  27. Patel, R. M., & Patel, M. M. (2011). Development and validation of stability-indicating RP-HPLC method for determination of diclofenac sodium in pharmaceutical formulations. Journal of Pharmacy Research, 4(6), 1996–2000
  28. Sharma, P., & Kumar, S. (2015). Stability-indicating RP-HPLC method for the determination of amlodipine besylate in pharmaceutical dosage forms. International Journal of Pharmaceutical Sciences and Research, 6(8), 3413–3420.
  29. Rao, R. S., & Kumar, S. (2012). Development and validation of a stability-indicating RP-HPLC method for imatinib mesylate. Journal of Analytical Chemistry, 67(3), 245–250.
  30. Samanidou, V. F., & Zacharis, C. K. (2015). Current trends and applications of RP-HPLC in pharmaceutical analysis. Journal of Liquid Chromatography & Related Technologies, 38(13), 1286–1302.
  31. Niessen, W. M. A. (2011). Liquid Chromatography–Mass Spectrometry (LC-MS/MS): Principles and Applications. CRC Press.
  32. Skoczylas, M., & P?otka-Wasylka, J. (2021). Artificial intelligence in green analytical chemistry. TrAC Trends in Analytical Chemistry, 138, 116223
  33. Tobiszewski, M. (2018). Metrics for green analytical chemistry. Analytical Methods, 10(37), 4615–4622.
  34. Ga?uszka, A., Migaszewski, Z., & Namie?nik, J. (2013). The 12 principles of green analytical chemistry and the SIGNIFICANCE mnemonic of green analytical practices. TrAC Trends in Analytical Chemistry, 50, 78–84.
  35. ICH Q14 Draft Guideline. Analytical Procedure Development. International Council for Harmonisation, 2022.
  36. Sender, R., Bar-On, Y. M., & Milo, R. (2021). RP-HPLC and stability studies of peptides and RNA therapeutics: Challenges and future perspectives. Biotechnology Advances, 53, 107836.

Photo
Poonam Kasar
Corresponding author

Department of Pharmaceutical Quality Assurance, R. G, Sapkal College of Pharmacy, Anjaneri, Nashik-422212.

Photo
Smita Aher
Co-author

Department of Pharmaceutical Chemistry, R. G, Sapkal College of Pharmacy, Anjaneri, Nashik-422212.

Photo
Rishikesh Bachhav
Co-author

Department of Pharmacology, R. G, Sapkal College of Pharmacy, Anjaneri, Nashik-422212.

Poonam Kasar*, Smita Aher, Rishikesh Bachhav, Development and Validation of Stability-Indicating RP-HPLC Methods for Pharmaceutical Drugs: A Comprehensive Review, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 8, 1361-1372. https://doi.org/10.5281/zenodo.16849573

More related articles
Effect Of Different Flavouring Agents In Drug Pala...
Sujal S. Shete, Tanvi T. Bhosale , Tejashri V. kamble , Nilesh B....
Formulation And Evaluation of Floating Drug Delive...
Nagendra R., Divyashree P., Nanditha V. V., Venkatesh, K. Hanuman...
Formulation And Evaluation of Anti-Dandruff Herbal...
Dileep Singh, Kavita Lovanshi, Rita Mourya, ...
Ocular and Systemic Manifestations of Behçet’s Disease: Exploring Pathogenesi...
Khadeejathul Mauseera, Diya Anzita, M. Mallikarjuna Gouda , ...
Moringa Oleifera: A Herbal Remedy for the Management of Diabetes...
Shivam Dange, Dewanshu Sathone, Ganesh Derkar, ...
Insilico Studies On Herbal Phytoconstituents For The Treatment Of Covid-19: A Re...
Umarani Savita Bhimashankar, H. M. Mallikarjuna, Meghana H. E. , Muni Naga Gayathri S. , Naveenkumar...
Related Articles
Anthelmintic Activity of Hymenocallis Littoralis Leaves Extract on Indian Earthw...
Hemant Suryawanshi, Sami Khan, Bhavana Sonar, Vaishnavi Mali, Himanshu Malve, R. A. Ahirrao, ...
Six Sigma in Pharmaceutical Industry – A Comprehensive Review...
Ashwini Chakote, Omkar Chougale, Sairaj Patil, Akash Malakane, ...
Effect Of Different Flavouring Agents In Drug Palatability Rate Or Sale...
Sujal S. Shete, Tanvi T. Bhosale , Tejashri V. kamble , Nilesh B. Chougule, ...
More related articles
Effect Of Different Flavouring Agents In Drug Palatability Rate Or Sale...
Sujal S. Shete, Tanvi T. Bhosale , Tejashri V. kamble , Nilesh B. Chougule, ...
Formulation And Evaluation of Floating Drug Delivery System of Benazepril...
Nagendra R., Divyashree P., Nanditha V. V., Venkatesh, K. Hanumanthachar Joshi, ...
Formulation And Evaluation of Anti-Dandruff Herbal Shampoo...
Dileep Singh, Kavita Lovanshi, Rita Mourya, ...
Effect Of Different Flavouring Agents In Drug Palatability Rate Or Sale...
Sujal S. Shete, Tanvi T. Bhosale , Tejashri V. kamble , Nilesh B. Chougule, ...
Formulation And Evaluation of Floating Drug Delivery System of Benazepril...
Nagendra R., Divyashree P., Nanditha V. V., Venkatesh, K. Hanumanthachar Joshi, ...
Formulation And Evaluation of Anti-Dandruff Herbal Shampoo...
Dileep Singh, Kavita Lovanshi, Rita Mourya, ...